Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Evol Appl ; 13(7): 1569-1580, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32821273

RESUMO

Tumors result from genetic and epigenetic alterations that change cellular survival and differentiation probabilities, promoting clonal dominance. Subsequent genetic and selection processes in tumors allow cells to lose their tissue fidelity and migrate to other parts of the body, turning tumors into cancer. However, the relationship between genetic damage and cancer is not linear, showing remarkable and sometimes seemingly counterintuitive patterns for different tissues and across animal taxa. In the present paper, we attempt to integrate our understanding of somatic evolution and cancer as a product of three major orthogonal processes: occurrence of somatic mutations, evolution of species-specific life-history traits, and physiological aging. Patterns of cancer risk have been shaped by selective pressures experienced by animal populations over millions of years, influencing and influenced by selection acting on traits ranging from mutation rate to reproductive strategies to longevity. We discuss how evolution of species shapes their cancer profiles alongside and in connection with other evolving life-history traits and how this process explains the patterns of cancer incidence we observe in humans and other animals.

2.
Sci Rep ; 10(1): 12198, 2020 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-32699207

RESUMO

Aneuploidy is a feature of many cancers. Recent studies demonstrate that in the hematopoietic stem and progenitor cell (HSPC) compartment aneuploid cells have reduced fitness and are efficiently purged from the bone marrow. However, early phases of hematopoietic reconstitution following bone marrow transplantation provide a window of opportunity whereby aneuploid cells rise in frequency, only to decline to basal levels thereafter. Here we demonstrate by Monte Carlo modeling that two mechanisms could underlie this aneuploidy peak: rapid expansion of the engrafted HSPC population and bone marrow microenvironment degradation caused by pre-transplantation radiation treatment. Both mechanisms reduce the strength of purifying selection acting in early post-transplantation bone marrow. We explore the contribution of other factors such as alterations in cell division rates that affect the strength of purifying selection, the balance of drift and selection imposed by the HSPC population size, and the mutation-selection balance dependent on the rate of aneuploidy generation per cell division. We propose a somatic evolutionary model for the dynamics of cells with aneuploidy or other fitness-reducing mutations during hematopoietic reconstitution following bone marrow transplantation. Similar alterations in the strength of purifying selection during cancer development could help explain the paradox of aneuploidy abundance in tumors despite somatic fitness costs.


Assuntos
Evolução Clonal , Células-Tronco Hematopoéticas/citologia , Modelos Biológicos , Aneuploidia , Animais , Células da Medula Óssea/citologia , Transplante de Medula Óssea , Divisão Celular , Microambiente Celular , Feminino , Raios gama , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/efeitos da radiação , Camundongos , Irradiação Corporal Total
3.
Sci Adv ; 5(2): eaav2437, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30801016

RESUMO

While a fraction of cancer patients treated with anti-PD-1 show durable therapeutic responses, most remain unresponsive, highlighting the need to better understand and improve these therapies. Using an in vivo screening approach with a customized shRNA pooled library, we identified DDR2 as a leading target for the enhancement of response to anti-PD-1 immunotherapy. Using isogenic in vivo murine models across five different tumor histologies-bladder, breast, colon, sarcoma, and melanoma-we show that DDR2 depletion increases sensitivity to anti-PD-1 treatment compared to monotherapy. Combination treatment of tumor-bearing mice with anti-PD-1 and dasatinib, a tyrosine kinase inhibitor of DDR2, led to tumor load reduction. RNA-seq and CyTOF analysis revealed higher CD8+ T cell populations in tumors with DDR2 depletion and those treated with dasatinib when either was combined with anti-PD-1 treatment. Our work provides strong scientific rationale for targeting DDR2 in combination with PD-1 inhibitors.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Dasatinibe/farmacologia , Receptor com Domínio Discoidina 2/antagonistas & inibidores , Sistemas de Liberação de Medicamentos , Imunidade Celular , Imunoterapia , Neoplasias Experimentais/terapia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Animais , Linfócitos T CD8-Positivos/patologia , Linhagem Celular Tumoral , Receptor com Domínio Discoidina 2/imunologia , Feminino , Camundongos , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Receptor de Morte Celular Programada 1/imunologia
4.
Mol Cancer Res ; 17(6): 1351-1364, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30808730

RESUMO

Mutations in oncogenes and tumor suppressor genes engender unique metabolic phenotypes crucial to the survival of tumor cells. EGFR signaling has been linked to the rewiring of tumor metabolism in non-small cell lung cancer (NSCLC). We have integrated the use of a functional genomics screen and metabolomics to identify metabolic vulnerabilities induced by EGFR inhibition. These studies reveal that following EGFR inhibition, EGFR-driven NSCLC cells become dependent on the urea cycle and, in particular, the urea cycle enzyme CPS1. Combining knockdown of CPS1 with EGFR inhibition further reduces cell proliferation and impedes cell-cycle progression. Profiling of the metabolome demonstrates that suppression of CPS1 potentiates the effects of EGFR inhibition on central carbon metabolism, pyrimidine biosynthesis, and arginine metabolism, coinciding with reduced glycolysis and mitochondrial respiration. We show that EGFR inhibition and CPS1 knockdown lead to a decrease in arginine levels and pyrimidine derivatives, and the addition of exogenous pyrimidines partially rescues the impairment in cell growth. Finally, we show that high expression of CPS1 in lung adenocarcinomas correlated with worse patient prognosis in publicly available databases. These data collectively reveal that NSCLC cells have a greater dependency on the urea cycle to sustain central carbon metabolism, pyrimidine biosynthesis, and arginine metabolism to meet cellular energetics upon inhibition of EGFR. IMPLICATIONS: Our results reveal that the urea cycle may be a novel metabolic vulnerability in the context of EGFR inhibition, providing an opportunity to develop rational combination therapies with EGFR inhibitors for the treatment of EGFR-driven NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Mutação/genética , Ureia/metabolismo , Carbamoil-Fosfato Sintase (Amônia)/genética , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/genética , Respiração Celular/genética , Receptores ErbB/genética , Glicólise/genética , Células HEK293 , Humanos , Metabolômica/métodos , Mitocôndrias/genética , Prognóstico , Pirimidinas/metabolismo , Transdução de Sinais/genética
5.
Sci Signal ; 9(450): rs12, 2016 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-27811184

RESUMO

Patients with lung cancers harboring anaplastic lymphoma kinase (ALK) gene fusions benefit from treatment with ALK inhibitors, but acquired resistance inevitably arises. A better understanding of proximal ALK signaling mechanisms may identify sensitizers to ALK inhibitors that disrupt the balance between prosurvival and proapoptotic effector signals. Using affinity purification coupled with mass spectrometry in an ALK fusion lung cancer cell line (H3122), we generated an ALK signaling network and investigated signaling activity using tyrosine phosphoproteomics. We identified a network of 464 proteins composed of subnetworks with differential response to ALK inhibitors. A small hairpin RNA screen targeting 407 proteins in this network revealed 64 and 9 proteins that when knocked down sensitized cells to crizotinib and alectinib, respectively. Among these, knocking down fibroblast growth factor receptor substrate 2 (FRS2) or coiled-coil and C2 domain-containing protein 1A (CC2D1A), both scaffolding proteins, sensitized multiple ALK fusion cell lines to the ALK inhibitors crizotinib and alectinib. Collectively, our data set provides a resource that enhances our understanding of signaling and drug resistance networks consequent to ALK fusions and identifies potential targets to improve the efficacy of ALK inhibitors in patients.


Assuntos
Carbazóis/farmacologia , Proteínas de Ciclo Celular , Neoplasias Pulmonares , Proteínas Associadas aos Microtúbulos , Piperidinas/farmacologia , Pirazóis/farmacologia , Piridinas/farmacologia , Interferência de RNA , Receptores Proteína Tirosina Quinases , Serina Endopeptidases , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Quinase do Linfoma Anaplásico , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Crizotinibe , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo
6.
Proc Natl Acad Sci U S A ; 113(43): E6669-E6678, 2016 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-27791036

RESUMO

Activating mutations in FMS-like tyrosine kinase 3 (FLT3) are common in acute myeloid leukemia (AML) and drive leukemic cell growth and survival. Although FLT3 inhibitors have shown considerable promise for the treatment of AML, they ultimately fail to achieve long-term remissions as monotherapy. To identify genetic targets that can sensitize AML cells to killing by FLT3 inhibitors, we performed a genome-wide RNA interference (RNAi)-based screen that identified ATM (ataxia telangiectasia mutated) as being synthetic lethal with FLT3 inhibitor therapy. We found that inactivating ATM or its downstream effector glucose 6-phosphate dehydrogenase (G6PD) sensitizes AML cells to FLT3 inhibitor induced apoptosis. Examination of the cellular metabolome showed that FLT3 inhibition by itself causes profound alterations in central carbon metabolism, resulting in impaired production of the antioxidant factor glutathione, which was further impaired by ATM or G6PD inactivation. Moreover, FLT3 inhibition elicited severe mitochondrial oxidative stress that is causative in apoptosis and is exacerbated by ATM/G6PD inhibition. The use of an agent that intensifies mitochondrial oxidative stress in combination with a FLT3 inhibitor augmented elimination of AML cells in vitro and in vivo, revealing a therapeutic strategy for the improved treatment of FLT3 mutated AML.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/genética , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Leucêmica da Expressão Gênica , Glucosefosfato Desidrogenase/genética , Leucemia Mieloide Aguda/genética , Tirosina Quinase 3 Semelhante a fms/genética , Animais , Antineoplásicos/farmacologia , Proteínas Mutadas de Ataxia Telangiectasia/antagonistas & inibidores , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Benzotiazóis/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Quimioterapia Combinada , Feminino , Glucosefosfato Desidrogenase/antagonistas & inibidores , Glucosefosfato Desidrogenase/metabolismo , Humanos , Hidrazinas/farmacologia , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/mortalidade , Leucemia Mieloide Aguda/patologia , Camundongos Endogâmicos NOD , Pessoa de Meia-Idade , Oxirredução , Compostos de Fenilureia/farmacologia , Inibidores de Proteínas Quinases/farmacologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Análise de Sobrevida , Ensaios Antitumorais Modelo de Xenoenxerto , Tirosina Quinase 3 Semelhante a fms/antagonistas & inibidores , Tirosina Quinase 3 Semelhante a fms/metabolismo
7.
Cancer Res ; 76(22): 6495-6506, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27671678

RESUMO

Using a three-dimensional coculture model, we identified significant subtype-specific changes in gene expression, metabolic, and therapeutic sensitivity profiles of breast cancer cells in contact with cancer-associated fibroblasts (CAF). CAF-induced gene expression signatures predicted clinical outcome and immune-related differences in the microenvironment. We found that fibroblasts strongly protect carcinoma cells from lapatinib, attributable to its reduced accumulation in carcinoma cells and an elevated apoptotic threshold. Fibroblasts from normal breast tissues and stromal cultures of brain metastases of breast cancer had similar effects as CAFs. Using synthetic lethality approaches, we identified molecular pathways whose inhibition sensitizes HER2+ breast cancer cells to lapatinib both in vitro and in vivo, including JAK2/STAT3 and hyaluronic acid. Neoadjuvant lapatinib therapy in HER2+ breast tumors lead to a significant increase of phospho-STAT3+ cancer cells and a decrease in the spatial proximity of proliferating (Ki67+) cells to CAFs impacting therapeutic responses. Our studies identify CAF-induced physiologically and clinically relevant changes in cancer cells and offer novel approaches for overcoming microenvironment-mediated therapeutic resistance. Cancer Res; 76(22); 6495-506. ©2016 AACR.


Assuntos
Neoplasias da Mama/metabolismo , Fibroblastos/metabolismo , Perfilação da Expressão Gênica/métodos , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Humanos , Resultado do Tratamento
8.
Proc Natl Acad Sci U S A ; 113(4): 1050-5, 2016 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-26755588

RESUMO

Young children have higher rates of leukemia than young adults. This fact represents a fundamental conundrum, because hematopoietic cells in young children should have fewer mutations (including oncogenic ones) than such cells in adults. Here, we present the results of stochastic modeling of hematopoietic stem cell (HSC) clonal dynamics, which demonstrated that early HSC pools were permissive to clonal evolution driven by drift. We show that drift-driven clonal expansions cooperate with faster HSC cycling in young children to produce conditions that are permissive for accumulation of multiple driver mutations in a single cell. Later in life, clonal evolution was suppressed by stabilizing selection in the larger young adult pools, and it was driven by positive selection at advanced ages in the presence of microenvironmental decline. Overall, our results indicate that leukemogenesis is driven by distinct evolutionary forces in children and adults.


Assuntos
Evolução Clonal , Leucemia/etiologia , Fatores Etários , Criança , Células-Tronco Hematopoéticas/fisiologia , Humanos , Mutação , Processos Estocásticos
9.
Trends Cancer ; 2(10): 552-560, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-28439564

RESUMO

The Armitage-Doll multi-stage model of carcinogenesis tremendously refocused cancer science by postulating that carcinogenesis is driven by a sequence of genetic changes in cells. Age-dependent cancer incidence thus has been explained in terms of the time necessary for oncogenic mutations to occur. While the multi-step nature of cancer evolution is well-supported by evidence, the mutation-centric theory is unable to explain a number of phenomena, such as the disproportion between cancer frequency and animal body size or the scaling of cancer incidence to animal lifespan. In this paper, we present a theoretical review of the current paradigm and discuss some fundamental evolutionary theory postulates that explain why cancer incidence is a function of lifespan and physiological, not chronological, aging.


Assuntos
Envelhecimento , Carcinogênese , Modelos Biológicos , Animais , Evolução Biológica , Humanos , Neoplasias , Risco
10.
Cancer Prev Res (Phila) ; 8(9): 762-4, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26122457

RESUMO

Tomasetti and Vogelstein (1) argue that lifetime cancer risk for particular tissues is mostly determined by the total number of stem cell (SC) divisions within the tissue, whereby most cancers arise due to "bad luck"­mutations occurring during DNA replication. We argue that the poorly substantiated estimations of SC division parameters and assumptions that oversimplify somatic evolution prevent such a conclusion from being drawn.


Assuntos
Divisão Celular/genética , Neoplasias/epidemiologia , Neoplasias/genética , Células-Tronco/fisiologia , Humanos
11.
Proc Natl Acad Sci U S A ; 112(29): 8914-21, 2015 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-26195756

RESUMO

Our understanding of cancer has greatly advanced since Nordling [Nordling CO (1953) Br J Cancer 7(1):68-72] and Armitage and Doll [Armitage P, Doll R (1954) Br J Cancer 8(1):1-12] put forth the multistage model of carcinogenesis. However, a number of observations remain poorly understood from the standpoint of this paradigm in its contemporary state. These observations include the similar age-dependent exponential rise in incidence of cancers originating from stem/progenitor pools differing drastically in size, age-dependent cell division profiles, and compartmentalization. This common incidence pattern is characteristic of cancers requiring different numbers of oncogenic mutations, and it scales to very divergent life spans of mammalian species. Also, bigger mammals with larger underlying stem cell pools are not proportionally more prone to cancer, an observation known as Peto's paradox. Here, we present a number of factors beyond the occurrence of oncogenic mutations that are unaccounted for in the current model of cancer development but should have significant impacts on cancer incidence. Furthermore, we propose a revision of the current understanding for how oncogenic and other functional somatic mutations affect cellular fitness. We present evidence, substantiated by evolutionary theory, demonstrating that fitness is a dynamic environment-dependent property of a phenotype and that oncogenic mutations should have vastly different fitness effects on somatic cells dependent on the tissue microenvironment in an age-dependent manner. Combined, this evidence provides a firm basis for understanding the age-dependent incidence of cancers as driven by age-altered systemic processes regulated above the cell level.


Assuntos
Evolução Biológica , Modelos Biológicos , Mutação/genética , Neoplasias/genética , Animais , Deriva Genética , Aptidão Genética , Humanos , Dinâmica não Linear , Seleção Genética , Células-Tronco/metabolismo , Fatores de Tempo
12.
Aging (Albany NY) ; 6(12): 1033-48, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25564763

RESUMO

Age-dependent tissue decline and increased cancer incidence are widely accepted to be rate-limited by the accumulation of somatic mutations over time. Current models of carcinogenesis are dominated by the assumption that oncogenic mutations have defined advantageous fitness effects on recipient stem and progenitor cells, promoting and rate-limiting somatic evolution. However, this assumption is markedly discrepant with evolutionary theory, whereby fitness is a dynamic property of a phenotype imposed upon and widely modulated by environment. We computationally modeled dynamic microenvironment-dependent fitness alterations in hematopoietic stem cells (HSC) within the Sprengel-Liebig system known to govern evolution at the population level. Our model for the first time integrates real data on age-dependent dynamics of HSC division rates, pool size, and accumulation of genetic changes and demonstrates that somatic evolution is not rate-limited by the occurrence of mutations, but instead results from aged microenvironment-driven alterations in the selective/fitness value of previously accumulated genetic changes. Our results are also consistent with evolutionary models of aging and thus oppose both somatic mutation-centric paradigms of carcinogenesis and tissue functional decline. In total, we demonstrate that aging directly promotes HSC fitness decline and somatic evolution via non-cell-autonomous mechanisms.


Assuntos
Envelhecimento/patologia , Evolução Biológica , Transformação Celular Neoplásica/patologia , Senescência Celular , Células-Tronco Hematopoéticas/patologia , Modelos Biológicos , Neoplasias/patologia , Células-Tronco Neoplásicas/patologia , Envelhecimento/genética , Envelhecimento/metabolismo , Animais , Proliferação de Células , Sobrevivência Celular , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Simulação por Computador , Genótipo , Células-Tronco Hematopoéticas/metabolismo , Humanos , Mutação , Neoplasias/genética , Neoplasias/metabolismo , Células-Tronco Neoplásicas/metabolismo , Fenótipo , Transdução de Sinais , Nicho de Células-Tronco , Processos Estocásticos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA