Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
1.
EBioMedicine ; 106: 105260, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39067134

RESUMO

BACKGROUND: Deeper insights into ERBB2-driven cancers are essential to develop new treatment approaches for ERBB2+ breast cancers (BCs). We employed the Collaborative Cross (CC) mouse model to unearth genetic factors underpinning Erbb2-driven mammary tumour development and metastasis. METHODS: 732 F1 hybrid female mice between FVB/N MMTV-Erbb2 and 30 CC strains were monitored for mammary tumour phenotypes. GWAS pinpointed SNPs that influence various tumour phenotypes. Multivariate analyses and models were used to construct the polygenic score and to develop a mouse tumour susceptibility gene signature (mTSGS), where the corresponding human ortholog was identified and designated as hTSGS. The importance and clinical value of hTSGS in human BC was evaluated using public datasets, encompassing TCGA, METABRIC, GSE96058, and I-SPY2 cohorts. The predictive power of mTSGS for response to chemotherapy was validated in vivo using genetically diverse MMTV-Erbb2 mice. FINDINGS: Distinct variances in tumour onset, multiplicity, and metastatic patterns were observed in F1-hybrid female mice between FVB/N MMTV-Erbb2 and 30 CC strains. Besides lung metastasis, liver and kidney metastases emerged in specific CC strains. GWAS identified specific SNPs significantly associated with tumour onset, multiplicity, lung metastasis, and liver metastasis. Multivariate analyses flagged SNPs in 20 genes (Stx6, Ramp1, Traf3ip1, Nckap5, Pfkfb2, Trmt1l, Rprd1b, Rer1, Sepsecs, Rhobtb1, Tsen15, Abcc3, Arid5b, Tnr, Dock2, Tti1, Fam81a, Oxr1, Plxna2, and Tbc1d31) independently tied to various tumour characteristics, designated as a mTSGS. hTSGS scores (hTSGSS) based on their transcriptional level showed prognostic values, superseding clinical factors and PAM50 subtype across multiple human BC cohorts, and predicted pathological complete response independent of and superior to MammaPrint score in I-SPY2 study. The power of mTSGS score for predicting chemotherapy response was further validated in an in vivo mouse MMTV-Erbb2 model, showing that, like findings in human patients, mouse tumours with low mTSGS scores were most likely to respond to treatment. INTERPRETATION: Our investigation has unveiled many new genes predisposing individuals to ERBB2-driven cancer. Translational findings indicate that hTSGS holds promise as a biomarker for refining treatment strategies for patients with BC. FUNDING: The U.S. Department of Defense (DoD) Breast Cancer Research Program (BCRP) (BC190820), United States; MCIN/AEI/10.13039/501100011039 (PID2020-118527RB-I00, PDC2021-121735-I00), the "European Union Next Generation EU/PRTR," the Regional Government of Castile and León (CSI144P20), European Union.


Assuntos
Camundongos de Cruzamento Colaborativo , Predisposição Genética para Doença , Polimorfismo de Nucleotídeo Único , Receptor ErbB-2 , Animais , Feminino , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Camundongos , Humanos , Camundongos de Cruzamento Colaborativo/genética , Estudo de Associação Genômica Ampla , Metástase Neoplásica , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Transcriptoma , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Neoplasias da Mama/metabolismo , Regulação Neoplásica da Expressão Gênica , Biomarcadores Tumorais/genética
2.
Science ; 384(6699): eadi7453, 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38815020

RESUMO

Stem cells play a critical role in cancer development by contributing to cell heterogeneity, lineage plasticity, and drug resistance. We created gene expression networks from hundreds of mouse tissue samples (both normal and tumor) and integrated these with lineage tracing and single-cell RNA-seq, to identify convergence of cell states in premalignant tumor cells expressing markers of lineage plasticity and drug resistance. Two of these cell states representing multilineage plasticity or proliferation were inversely correlated, suggesting a mutually exclusive relationship. Treatment of carcinomas in vivo with chemotherapy repressed the proliferative state and activated multilineage plasticity whereas inhibition of differentiation repressed plasticity and potentiated responses to cell cycle inhibitors. Manipulation of this cell state transition point may provide a source of potential combinatorial targets for cancer therapy.


Assuntos
Carcinoma de Células Escamosas , Linhagem da Célula , Células-Tronco Neoplásicas , Neoplasias Cutâneas , Animais , Camundongos , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Células-Tronco Neoplásicas/patologia , Análise de Célula Única , Diferenciação Celular , Resistencia a Medicamentos Antineoplásicos/genética , Plasticidade Celular , Proliferação de Células , Redes Reguladoras de Genes , RNA-Seq , Regulação Neoplásica da Expressão Gênica
3.
Cell ; 187(7): 1589-1616, 2024 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-38552609

RESUMO

The last 50 years have witnessed extraordinary developments in understanding mechanisms of carcinogenesis, synthesized as the hallmarks of cancer. Despite this logical framework, our understanding of the molecular basis of systemic manifestations and the underlying causes of cancer-related death remains incomplete. Looking forward, elucidating how tumors interact with distant organs and how multifaceted environmental and physiological parameters impinge on tumors and their hosts will be crucial for advances in preventing and more effectively treating human cancers. In this perspective, we discuss complexities of cancer as a systemic disease, including tumor initiation and promotion, tumor micro- and immune macro-environments, aging, metabolism and obesity, cancer cachexia, circadian rhythms, nervous system interactions, tumor-related thrombosis, and the microbiome. Model systems incorporating human genetic variation will be essential to decipher the mechanistic basis of these phenomena and unravel gene-environment interactions, providing a modern synthesis of molecular oncology that is primed to prevent cancers and improve patient quality of life and cancer outcomes.


Assuntos
Neoplasias , Humanos , Carcinogênese , Microbiota , Neoplasias/genética , Neoplasias/patologia , Neoplasias/terapia , Obesidade/complicações , Qualidade de Vida
4.
Artigo em Inglês | MEDLINE | ID: mdl-38052482

RESUMO

The transition from a single, initiated cell to a full-blown malignant tumor involves significant genomic evolution. Exposure to carcinogens-whether directly mutagenic or not-can drive progression toward malignancy, as can stochastic acquisition of cancer-promoting genetic events. Mouse models using both carcinogens and germline genetic manipulations have enabled precise inquiry into the evolutionary dynamics that take place as a tumor progresses from benign to malignant to metastatic stages. Tumor progression is characterized by changes in somatic point mutations and copy-number alterations, even though any single tumor can itself have a high or low burden of genomic alterations. Further, lineage-tracing, single-cell analyses and CRISPR barcoding have revealed the distinct clonal dynamics within benign and malignant tumors. Application of these tools in a range of mouse models can shed unique light on the patterns of clonal evolution that take place in both mouse and human tumors.


Assuntos
Carcinógenos Ambientais , Humanos , Animais , Camundongos , Processos Neoplásicos , Genômica , Mutação , Evolução Clonal , Modelos Animais de Doenças
5.
bioRxiv ; 2023 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-37215032

RESUMO

Adult mammalian stem cells play critical roles in normal tissue homeostasis, as well as in tumor development, by contributing to cell heterogeneity, plasticity, and development of drug resistance. The relationship between different types of normal and cancer stem cells is highly controversial and poorly understood. Here, we carried out gene expression network analysis of normal and tumor samples from genetically heterogeneous mice to create network metagenes for visualization of stem-cell networks, rather than individual stem-cell markers, at the single-cell level during multistage carcinogenesis. We combined this approach with lineage tracing and single-cell RNASeq of stem cells and their progeny, identifying a previously unrecognized hierarchy in which Lgr6+ stem cells from tumors generate progeny that express a range of other stem-cell markers including Sox2, Pitx1, Foxa1, Klf5, and Cd44. Our data identify a convergence of multiple stem-cell and tumor-suppressor pathways in benign tumor cells expressing markers of lineage plasticity and oxidative stress. This same single-cell population expresses network metagenes corresponding to markers of cancer drug resistance in human tumors of the skin, lung and prostate. Treatment of mouse squamous carcinomas in vivo with the chemotherapeutic cis-platin resulted in elevated expression of the genes that mark this cell population. Our data have allowed us to create a simplified model of multistage carcinogenesis that identifies distinct stem-cell states at different stages of tumor progression, thereby identifying networks involved in lineage plasticity, drug resistance, and immune surveillance, providing a rich source of potential targets for cancer therapy.

8.
NPJ Breast Cancer ; 7(1): 118, 2021 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-34508103

RESUMO

Polymorphisms in the PER3 gene have been associated with several human disease phenotypes, including sleep disorders and cancer. In particular, the long allele of a variable number of tandem repeat (VNTR) polymorphism has been previously linked to an increased risk of breast cancer. Here we carried out a combined germline and somatic genetic analysis of the role of the PER3VNRT polymorphism in breast cancer. The combined data from 8284 individuals showed a non-significant trend towards increased breast cancer risk in the 5-repeat allele homozygous carriers (OR = 1.17, 95% CI: 0.97-1.42). We observed allelic imbalance at the PER3 locus in matched blood and tumor DNA samples, showing a significant retention of the long variant (risk) allele in tumor samples, and a preferential loss of the short repetition allele (p = 0.0005). Gene co-expression analysis in healthy and tumoral breast tissue samples uncovered significant associations between PER3 expression levels with those from genes which belong to several cancer-associated pathways. Finally, relapse-free survival (RFS) analysis showed that low expression levels of PER3 were linked to a significant lower RSF in luminal A (p = 3 × 10-12) but not in the rest of breast cancer subtypes.

9.
Nat Commun ; 12(1): 4288, 2021 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-34257283

RESUMO

The commonly mutated human KRAS oncogene encodes two distinct KRAS4A and KRAS4B proteins generated by differential splicing. We demonstrate here that coordinated regulation of both isoforms through control of splicing is essential for development of Kras mutant tumors. The minor KRAS4A isoform is enriched in cancer stem-like cells, where it responds to hypoxia, while the major KRAS4B is induced by ER stress. KRAS4A splicing is controlled by the DCAF15/RBM39 pathway, and deletion of KRAS4A or pharmacological inhibition of RBM39 using Indisulam leads to inhibition of cancer stem cells. Our data identify existing clinical drugs that target KRAS4A splicing, and suggest that levels of the minor KRAS4A isoform in human tumors can be a biomarker of sensitivity to some existing cancer therapeutics.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Células-Tronco Neoplásicas/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Proteínas de Ligação a RNA/metabolismo , Células A549 , Animais , Western Blotting , Proliferação de Células , Citometria de Fluxo , Xenoenxertos , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas de Ligação a RNA/genética
10.
Nat Genet ; 52(11): 1139-1143, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33106632

RESUMO

Cancer is driven by genomic mutations in 'cancer driver' genes, which have essential roles in tumor development. These mutations may be caused by exposure to mutagens in the environment or by endogenous DNA-replication errors in tissue stem cells. Recent observations of abundant mutations, including cancer driver mutations, in histologically normal human tissues suggest that mutations alone are not sufficient for tumor development, thus prompting the question of how single mutant cells give rise to neoplasia. In a concept supported by decades-old data from mouse tumor models, non-mutagenic tumor-promoting agents have been posited to activate the proliferation of dormant mutated cells, thus generating actively growing lesions, with the promotion stage as the rate-limiting step in tumor formation. Non-mutagenic promoting agents, either endogenous or environmental, may therefore have a more important role in human cancer etiology than previously thought.


Assuntos
Poluentes Ambientais/toxicidade , Mutagênicos/toxicidade , Neoplasias/etiologia , Animais , Transformação Celular Neoplásica , Humanos , Camundongos , Mutação , Neoplasias/induzido quimicamente , Neoplasias/genética , Neoplasias Experimentais , Fatores de Risco , Neoplasias Cutâneas/etiologia , Neoplasias Cutâneas/genética
11.
Nat Genet ; 52(11): 1189-1197, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32989322

RESUMO

Epidemiological studies have identified many environmental agents that appear to significantly increase cancer risk in human populations. By analyzing tumor genomes from mice chronically exposed to 1 of 20 known or suspected human carcinogens, we reveal that most agents do not generate distinct mutational signatures or increase mutation burden, with most mutations, including driver mutations, resulting from tissue-specific endogenous processes. We identify signatures resulting from exposure to cobalt and vinylidene chloride and link distinct human signatures (SBS19 and SBS42) with 1,2,3-trichloropropane, a haloalkane and pollutant of drinking water, and find these and other signatures in human tumor genomes. We define the cross-species genomic landscape of tumors induced by an important compendium of agents with relevance to human health.


Assuntos
Carcinógenos/toxicidade , Mutação , Animais , Carcinogênese/genética , Análise Mutacional de DNA , Poluentes Ambientais/toxicidade , Feminino , Genoma , Humanos , Masculino , Camundongos , Taxa de Mutação , Propano/análogos & derivados , Propano/toxicidade , Especificidade da Espécie
12.
Nat Commun ; 11(1): 2375, 2020 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-32398776

RESUMO

Synthetic lethal screens have the potential to identify new vulnerabilities incurred by specific cancer mutations but have been hindered by lack of agreement between studies. In the case of KRAS, we identify that published synthetic lethal screen hits significantly overlap at the pathway rather than gene level. Analysis of pathways encoded as protein networks could identify synthetic lethal candidates that are more reproducible than those previously reported. Lack of overlap likely stems from biological rather than technical limitations as most synthetic lethal phenotypes are strongly modulated by changes in cellular conditions or genetic context, the latter determined using a pairwise genetic interaction map that identifies numerous interactions that suppress synthetic lethal effects. Accounting for pathway, cellular and genetic context nominates a DNA repair dependency in KRAS-mutant cells, mediated by a network containing BRCA1. We provide evidence for why most reported synthetic lethals are not reproducible which is addressable using a multi-faceted testing framework.


Assuntos
Biologia Computacional/métodos , Análise de Dados , Redes Reguladoras de Genes , Neoplasias/genética , Mutações Sintéticas Letais , Animais , Proteína BRCA1/genética , Linhagem Celular Tumoral , Biologia Computacional/normas , Modelos Animais de Doenças , Humanos , Camundongos , Mapas de Interação de Proteínas/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Reprodutibilidade dos Testes
13.
Nat Commun ; 11(1): 394, 2020 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-31959748

RESUMO

Ionising radiation (IR) is a recognised carcinogen responsible for cancer development in patients previously treated using radiotherapy, and in individuals exposed as a result of accidents at nuclear energy plants. However, the mutational signatures induced by distinct types and doses of radiation are unknown. Here, we analyse the genetic architecture of mammary tumours, lymphomas and sarcomas induced by high (56Fe-ions) or low (gamma) energy radiation in mice carrying Trp53 loss of function alleles. In mammary tumours, high-energy radiation is associated with induction of focal structural variants, leading to genomic instability and Met amplification. Gamma-radiation is linked to large-scale structural variants and a point mutation signature associated with oxidative stress. The genomic architecture of carcinomas, sarcomas and lymphomas arising in the same animals are significantly different. Our study illustrates the complex interactions between radiation quality, germline Trp53 deficiency and tissue/cell of origin in shaping the genomic landscape of IR-induced tumours.


Assuntos
Carcinogênese/efeitos da radiação , Instabilidade Genômica/efeitos da radiação , Neoplasias Induzidas por Radiação/genética , Lesões Experimentais por Radiação/genética , Proteína Supressora de Tumor p53/genética , Animais , Carcinogênese/genética , Dano ao DNA/efeitos da radiação , Análise Mutacional de DNA , Relação Dose-Resposta à Radiação , Feminino , Amplificação de Genes/efeitos da radiação , Mutação em Linhagem Germinativa , Humanos , Masculino , Camundongos , Camundongos Knockout , Neoplasias Induzidas por Radiação/patologia , Mutação Puntual/efeitos da radiação , Proteínas Proto-Oncogênicas c-met/genética , Lesões Experimentais por Radiação/patologia , Sequenciamento Completo do Genoma
14.
Nature ; 576(7787): 482-486, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31827279

RESUMO

The most frequently mutated oncogene in cancer is KRAS, which uses alternative fourth exons to generate two gene products (KRAS4A and KRAS4B) that differ only in their C-terminal membrane-targeting region1. Because oncogenic mutations occur in exons 2 or 3, two constitutively active KRAS proteins-each capable of transforming cells-are encoded when KRAS is activated by mutation2. No functional distinctions among the splice variants have so far been established. Oncogenic KRAS alters the metabolism of tumour cells3 in several ways, including increased glucose uptake and glycolysis even in the presence of abundant oxygen4 (the Warburg effect). Whereas these metabolic effects of oncogenic KRAS have been explained by transcriptional upregulation of glucose transporters and glycolytic enzymes3-5, it is not known whether there is direct regulation of metabolic enzymes. Here we report a direct, GTP-dependent interaction between KRAS4A and hexokinase 1 (HK1) that alters the activity of the kinase, and thereby establish that HK1 is an effector of KRAS4A. This interaction is unique to KRAS4A because the palmitoylation-depalmitoylation cycle of this RAS isoform enables colocalization with HK1 on the outer mitochondrial membrane. The expression of KRAS4A in cancer may drive unique metabolic vulnerabilities that can be exploited therapeutically.


Assuntos
Hexoquinase/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Regulação Alostérica , Animais , Linhagem Celular Tumoral , Ativação Enzimática , Glicólise , Guanosina Trifosfato/metabolismo , Hexoquinase/química , Humanos , Técnicas In Vitro , Isoenzimas/metabolismo , Lipoilação , Masculino , Camundongos , Mitocôndrias/enzimologia , Mitocôndrias/metabolismo , Membranas Mitocondriais/enzimologia , Membranas Mitocondriais/metabolismo , Neoplasias/enzimologia , Neoplasias/metabolismo , Ligação Proteica , Transporte Proteico
15.
Dev Cell ; 51(3): 326-340.e4, 2019 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-31564613

RESUMO

Oil-secreting sebaceous glands (SGs) are critical for proper skin function; however, it remains unclear how different factors act together to modulate SG stem cells. Here, we provide functional evidence that each SG lobe is serviced by its own dedicated stem cell population. Upon ablating Notch signaling in different skin subcompartments, we find that this pathway exerts dual counteracting effects on SGs. Suppressing Notch in SG progenitors traps them in a hybrid state where stem and differentiation features become intermingled. In contrast, ablating Notch outside of the SG stem cell compartment indirectly drives SG expansion. Finally, we report that a K14:K5→K14:K79 keratin shift occurs during SG differentiation. Deleting K79 destabilizes K14 in sebocytes, and attenuates SGs and eyelid meibomian glands, leading to corneal ulceration. Altogether, our findings demonstrate that SGs integrate diverse signals from different niches and suggest that mutations incurred within one stem cell compartment can indirectly influence another.


Assuntos
Glândulas Sebáceas/citologia , Pele/citologia , Nicho de Células-Tronco , Células-Tronco/citologia , Animais , Diferenciação Celular , Feminino , Proteínas Hedgehog/metabolismo , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Queratinas/metabolismo , Masculino , Glândulas Tarsais/metabolismo , Camundongos Knockout , Mutação/genética , Receptores Notch/genética
16.
Nat Rev Cancer ; 18(12): 767-777, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30420765

RESUMO

The three RAS genes - HRAS, NRAS and KRAS - are collectively mutated in one-third of human cancers, where they act as prototypic oncogenes. Interestingly, there are rather distinct patterns to RAS mutations; the isoform mutated as well as the position and type of substitution vary between different cancers. As RAS genes are among the earliest, if not the first, genes mutated in a variety of cancers, understanding how these mutation patterns arise could inform on not only how cancer begins but also the factors influencing this event, which has implications for cancer prevention. To this end, we suggest that there is a narrow window or 'sweet spot' by which oncogenic RAS signalling can promote tumour initiation in normal cells. As a consequence, RAS mutation patterns in each normal cell are a product of the specific RAS isoform mutated, as well as the position of the mutation and type of substitution to achieve an ideal level of signalling.


Assuntos
Genes ras , Mutação , Neoplasias/genética , Animais , Transformação Celular Neoplásica/genética , GTP Fosfo-Hidrolases/genética , Humanos , Proteínas de Membrana/genética , Camundongos , Modelos Genéticos , Mutagênese , Neoplasias/metabolismo , Isoformas de Proteínas/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Transdução de Sinais/genética
17.
Nat Cell Biol ; 20(6): 699-709, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29802408

RESUMO

Tumour cells are subjected to evolutionary selection pressures during progression from initiation to metastasis. We analysed the clonal evolution of squamous skin carcinomas induced by DMBA/TPA treatment using the K5CreER-Confetti mouse and stage-specific lineage tracing. We show that benign tumours are polyclonal, but only one population contains the Hras driver mutation. Thus, benign papillomas are monoclonal in origin but recruit neighbouring epithelial cells during growth. Papillomas that never progress to malignancy retain several distinct clones, whereas progression to carcinoma is associated with a clonal sweep. Newly generated clones within carcinomas demonstrate intratumoural invasion and clonal intermixing, often giving rise to metastases containing two or more distinct clones derived from the matched primary tumour. These data demonstrate that late-stage tumour progression and dissemination are governed by evolutionary selection pressures that operate at a multicellular level and, therefore, differ from the clonal events that drive initiation and the benign-malignant transition.


Assuntos
Carcinoma de Células Escamosas/genética , Linhagem da Célula , Movimento Celular/genética , Transformação Celular Neoplásica/genética , Evolução Clonal , Células Epiteliais/patologia , Neoplasias Experimentais/genética , Neoplasias Cutâneas/genética , 9,10-Dimetil-1,2-benzantraceno , Animais , Carcinoma de Células Escamosas/induzido quimicamente , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/secundário , Proliferação de Células/genética , Transformação Celular Neoplásica/induzido quimicamente , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Células Epiteliais/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Genes ras , Predisposição Genética para Doença , Masculino , Camundongos Transgênicos , Mutação , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Fenótipo , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Acetato de Tetradecanoilforbol , Fatores de Tempo , Carga Tumoral/genética
18.
Oncoimmunology ; 6(11): e1356142, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29147603

RESUMO

Chronic inflammation promotes breast tumor growth and invasion by accelerating angiogenesis and tissue remodeling in the tumor microenvironment. There is a complex relationship between inflammation and estrogen, which drives the growth of 70 percent of breast tumors. While low levels of estrogen exposure stimulate macrophages and other inflammatory cell populations, very high levels are immune suppressive. Breast tumor incidence is increased by obesity and age, which interact to influence inflammatory cell populations in normal breast tissue. To characterize the impact of these factors on tumors and the tumor microenvironment, we measured gene expression in 195 breast adenocarcinomas and matched adjacent normal breast tissue samples collected at Akershus University Hospital (AHUS). Age and Body Mass Index (BMI) were independently associated with inflammation in adjacent normal tissue but not tumors. Estrogen Receptor (ER)-negative tumors had elevated macrophage expression compared with matched normal tissue, but ER-positive tumors showed an unexpected decrease in macrophage expression. We found an inverse relationship between the increase in tumor estrogen pathway expression compared with adjacent normal tissue and tumor macrophage score. We validated this finding in 126 breast tumor-normal pairs from the previously published METABRIC cohort. We developed a novel statistic, the Rewiring Coefficient, to quantify the rewiring of gene co-expression networks at the level of individual genes. Differential correlation analysis demonstrated distinct pathways were rewired during tumorigenesis. Our data support an immune suppressive effect of high doses of estrogen signaling in breast tumor microenvironment, suggesting that this effect contributes to the greater presence of prognostic and therapeutically relevant immune cells in ER-negative tumors.

19.
Nat Genet ; 49(11): 1624-1632, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28945253

RESUMO

The G-protein-coupled receptors LGR4, LGR5 and LGR6 are Wnt signaling mediators, but their functions in squamous cell carcinoma (SCC) are unclear. Using lineage tracing in Lgr5-EGFP-CreERT2/Rosa26-Tomato and Lgr6-EGFP-CreERT2/Rosa26-Tomato reporter mice, we demonstrate that Lgr6, but not Lgr5, acts as an epithelial stem cell marker in SCCs in vivo. We identify, by single-molecule in situ hybridization and cell sorting, rare cells positive for Lgr6 expression in immortalized keratinocytes and show that their frequency increases in advanced SCCs. Lgr6 expression is enriched in cells with stem cell characteristics, and Lgr6 downregulation in vivo causes increased epidermal proliferation with expanded lineage tracing from epidermal stem cells positive for Lgr6 expression. Surprisingly, mice with germline knockout of Lgr6 are predisposed to SCC development, through a mechanism that includes compensatory upregulation of Lgr5. These data provide a model for human patients with germline loss-of-function mutations in Wnt pathway genes, including RSPO1 or LGR4, who show increased susceptibility to squamous tumor development.


Assuntos
Carcinoma de Células Escamosas/genética , Regulação Neoplásica da Expressão Gênica , Queratinócitos/metabolismo , Células-Tronco Neoplásicas/metabolismo , Receptores Acoplados a Proteínas G/genética , Neoplasias Cutâneas/genética , Animais , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular Transformada , Epiderme/metabolismo , Epiderme/patologia , Humanos , Queratinócitos/patologia , Camundongos , Camundongos Transgênicos , Células-Tronco Neoplásicas/patologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Trombospondinas/genética , Trombospondinas/metabolismo
20.
Genome Med ; 8(1): 83, 2016 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-27506198

RESUMO

BACKGROUND: Body mass index (BMI) has been implicated as a primary factor influencing cancer development. However, understanding the relationship between these two complex traits has been confounded by both environmental and genetic heterogeneity. METHODS: In order to gain insight into the genetic factors linking BMI and cancer, we performed chemical carcinogenesis on a genetically heterogeneous cohort of interspecific backcross mice ((Mus Spretus × FVB/N) F1 × FVB/N). Using this cohort, we performed quantitative trait loci (QTL) analysis to identify regions linked to BMI. We then performed an integrated analysis incorporating gene expression, sequence comparison between strains, and gene expression network analysis to identify candidate genes influencing both tumor development and BMI. RESULTS: Analysis of QTL linked to tumorigenesis and BMI identified several loci associated with both phenotypes. Exploring these loci in greater detail revealed a novel relationship between the Pannexin 3 gene (Panx3) and both BMI and tumorigenesis. Panx3 is positively associated with BMI and is strongly tied to a lipid metabolism gene expression network. Pre-treatment Panx3 gene expression levels in normal skin are associated with tumor susceptibility and inhibition of Panx function strongly influences inflammation. CONCLUSIONS: These studies have identified several genetic loci that influence both BMI and carcinogenesis and implicate Panx3 as a candidate gene that links these phenotypes through its effects on inflammation and lipid metabolism.


Assuntos
Carcinogênese/genética , Conexinas/genética , Regulação Neoplásica da Expressão Gênica , Metabolismo dos Lipídeos/genética , Locos de Características Quantitativas , Neoplasias Cutâneas/genética , 9,10-Dimetil-1,2-benzantraceno , Animais , Índice de Massa Corporal , Carcinogênese/metabolismo , Carcinogênese/patologia , Carcinógenos , Cruzamentos Genéticos , Feminino , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Predisposição Genética para Doença , Humanos , Inflamação , Masculino , Camundongos , Camundongos Endogâmicos , Fatores Sexuais , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Acetato de Tetradecanoilforbol/análogos & derivados
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA