Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Rep Med ; 5(3): 101471, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38508142

RESUMO

Drug-tolerant persisters (DTPs) are a rare subpopulation of cells within a tumor that can survive therapy through nongenetic adaptive mechanisms to develop relapse and repopulate the tumor following drug withdrawal. Using a cancer cell line with an engineered suicide switch to kill proliferating cells, we perform both genetic screens and compound screens to identify the inhibition of bromodomain and extraterminal domain (BET) proteins as a selective vulnerability of DTPs. BET inhibitors are especially detrimental to DTPs that have reentered the cell cycle (DTEPs) in a broad spectrum of cancer types. Mechanistically, BET inhibition induces lethal levels of ROS through the suppression of redox-regulating genes highly expressed in DTPs, including GPX2, ALDH3A1, and MGST1. In vivo BET inhibitor treatment delays tumor relapse in both melanoma and lung cancer. Our study suggests that combining standard of care therapy with BET inhibitors to eliminate residual persister cells is a promising therapeutic strategy.


Assuntos
Neoplasias Pulmonares , Recidiva Local de Neoplasia , Humanos , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/genética , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética
2.
Nat Cancer ; 3(11): 1284-1299, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36414711

RESUMO

Senolytics, drugs that kill senescent cells, have been proposed to improve the response to pro-senescence cancer therapies; however, this remains challenging due to a lack of broadly acting senolytic drugs. Using CRISPR/Cas9-based genetic screens in different senescent cancer cell models, we identify loss of the death receptor inhibitor cFLIP as a common vulnerability of senescent cancer cells. Senescent cells are primed for apoptotic death by NF-κB-mediated upregulation of death receptor 5 (DR5) and its ligand TRAIL, but are protected from death by increased cFLIP expression. Activation of DR5 signaling by agonistic antibody, which can be enhanced further by suppression of cFLIP by BRD2 inhibition, leads to efficient killing of a variety of senescent cancer cells. Moreover, senescent cells sensitize adjacent non-senescent cells to killing by DR5 agonist through a bystander effect mediated by secretion of cytokines. We validate this 'one-two punch' cancer therapy by combining pro-senescence therapy with DR5 activation in different animal models.


Assuntos
Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD , Neoplasias , Animais , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/genética , Apoptose , NF-kappa B/metabolismo , Transdução de Sinais , Neoplasias/tratamento farmacológico
3.
PLoS One ; 17(9): e0273182, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36067171

RESUMO

Inducing senescence in cancer cells is emerging as a new therapeutic strategy. In order to find ways to enhance senescence induction by palbociclib, a CDK4/6 inhibitor approved for treatment of metastatic breast cancer, we performed functional genetic screens in palbociclib-resistant cells. Using this approach, we found that loss of CDK2 results in strong senescence induction in palbociclib-treated cells. Treatment with the CDK2 inhibitor indisulam, which phenocopies genetic CDK2 inactivation, led to sustained senescence induction when combined with palbociclib in various cell lines and lung cancer xenografts. Treating cells with indisulam led to downregulation of cyclin H, which prevented CDK2 activation. Combined treatment with palbociclib and indisulam induced a senescence program and sensitized cells to senolytic therapy. Our data indicate that inhibition of CDK2 through indisulam treatment can enhance senescence induction by CDK4/6 inhibition.


Assuntos
Quinase 6 Dependente de Ciclina , Inibidores de Proteínas Quinases , Linhagem Celular Tumoral , Quinase 2 Dependente de Ciclina , Quinase 4 Dependente de Ciclina/metabolismo , Quinase 6 Dependente de Ciclina/metabolismo , Humanos , Piperazinas , Inibidores de Proteínas Quinases/farmacologia , Piridinas , Sulfonamidas
4.
ACS Chem Biol ; 16(9): 1770-1778, 2021 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-34427427

RESUMO

The utility of in vitro human disease models is mainly dependent on the availability and functional maturity of tissue-specific cell types. We have previously screened for and identified small molecules that can enhance hepatocyte function in vitro. Here, we characterize the functional effects of one of the hits, FH1, on primary human hepatocytes in vitro, and also in vivo on primary hepatocytes in a zebrafish model. Furthermore, we conducted an analogue screen to establish the structure-activity relationship of FH1. We performed affinity-purification proteomics that identified NQO2 to be a potential binding target for this small molecule, revealing a possible link between inflammatory signaling and hepatocellular function in zebrafish and human hepatocyte model systems.


Assuntos
Biomarcadores/metabolismo , Inibidores Enzimáticos/química , Hepatócitos/metabolismo , Quinona Redutases/antagonistas & inibidores , Animais , Inibidores Enzimáticos/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Ensaios de Triagem em Larga Escala , Humanos , Interleucina-6/genética , Fígado , Simulação de Acoplamento Molecular , Ligação Proteica , Fator de Transcrição STAT3/genética , Transdução de Sinais , Relação Estrutura-Atividade , Fatores de Necrose Tumoral/genética , Peixe-Zebra
5.
Cell Rep ; 36(4): 109441, 2021 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-34320349

RESUMO

Cellular senescence is characterized as a stable proliferation arrest that can be triggered by multiple stresses. Most knowledge about senescent cells is obtained from studies in primary cells. However, senescence features may be different in cancer cells, since the pathways that are involved in senescence induction are often deregulated in cancer. We report here a comprehensive analysis of the transcriptome and senolytic responses in a panel of 13 cancer cell lines rendered senescent by two distinct compounds. We show that in cancer cells, the response to senolytic agents and the composition of the senescence-associated secretory phenotype are more influenced by the cell of origin than by the senescence trigger. Using machine learning, we establish the SENCAN gene expression classifier for the detection of senescence in cancer cell samples. The expression profiles and senescence classifier are available as an interactive online Cancer SENESCopedia.


Assuntos
Senescência Celular , Neoplasias/patologia , Compostos de Anilina/farmacologia , Azepinas/farmacologia , Linhagem Celular Tumoral , Senescência Celular/efeitos dos fármacos , Etoposídeo/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias/genética , Pirimidinas/farmacologia , Reprodutibilidade dos Testes , Fenótipo Secretor Associado à Senescência/efeitos dos fármacos , Fenótipo Secretor Associado à Senescência/genética , Senoterapia/farmacologia , Sulfonamidas/farmacologia
6.
Mol Cancer Res ; 19(10): 1613-1621, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34158393

RESUMO

Pro-senescence therapies are increasingly being considered for the treatment of cancer. Identifying additional targets to induce senescence in cancer cells could further enable such therapies. However, screening for targets whose suppression induces senescence on a genome-wide scale is challenging, as senescent cells become growth arrested, and senescence-associated features can take 1 to 2 weeks to develop. For a screen with a whole-genome CRISPR library, this would result in billions of undesirable proliferating cells by the time the senescent features emerge in the growth arrested cells. Here, we present a suicide switch system that allows genome-wide CRISPR screening in growth-arrested subpopulations by eliminating the proliferating cells during the screen through activation of a suicide switch in proliferating cells. Using this system, we identify in a genome-scale CRISPR screen several autophagy-related proteins as targets for senescence induction. We show that inhibiting macroautophagy with a small molecule ULK1 inhibitor can induce senescence in cancer cell lines of different origin. Finally, we show that combining ULK1 inhibition with the senolytic drug ABT-263 leads to apoptosis in a panel of cancer cell lines. IMPLICATIONS: Our suicide switch approach allows for genome-scale identification of pro-senescence targets, and can be adapted to simplify other screens depending on the nature of the promoter used to drive the switch.


Assuntos
Proteínas Relacionadas à Autofagia/genética , Autofagia/genética , Sistemas CRISPR-Cas/genética , Senescência Celular/genética , Células A549 , Apoptose/efeitos dos fármacos , Apoptose/genética , Autofagia/efeitos dos fármacos , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/genética , Sistemas CRISPR-Cas/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Senescência Celular/efeitos dos fármacos , Células HEK293 , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Bibliotecas de Moléculas Pequenas/farmacologia
7.
Nature ; 574(7777): 268-272, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31578521

RESUMO

Liver cancer remains difficult to treat, owing to a paucity of drugs that target critical dependencies1,2; broad-spectrum kinase inhibitors such as sorafenib provide only a modest benefit to patients with hepatocellular carcinoma3. The induction of senescence may represent a strategy for the treatment of cancer, especially when combined with a second drug that selectively eliminates senescent cancer cells (senolysis)4,5. Here, using a kinome-focused genetic screen, we show that pharmacological inhibition of the DNA-replication kinase CDC7 induces senescence selectively in liver cancer cells with mutations in TP53. A follow-up chemical screen identified the antidepressant sertraline as an agent that kills hepatocellular carcinoma cells that have been rendered senescent by inhibition of CDC7. Sertraline suppressed mTOR signalling, and selective drugs that target this pathway were highly effective in causing the apoptotic cell death of hepatocellular carcinoma cells treated with a CDC7 inhibitor. The feedback reactivation of mTOR signalling after its inhibition6 is blocked in cells that have been treated with a CDC7 inhibitor, which leads to the sustained inhibition of mTOR and cell death. Using multiple in vivo mouse models of liver cancer, we show that treatment with combined inhibition of of CDC7 and mTOR results in a marked reduction of tumour growth. Our data indicate that exploiting an induced vulnerability could be an effective treatment for liver cancer.


Assuntos
Apoptose/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/patologia , Terapia de Alvo Molecular , Sertralina/farmacologia , Animais , Proteínas de Ciclo Celular/antagonistas & inibidores , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Sertralina/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/antagonistas & inibidores , Proteína Supressora de Tumor p53/genética
8.
Gastroenterology ; 156(6): 1788-1804.e13, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30641053

RESUMO

BACKGROUND & AIMS: Patients with cirrhosis are at high risk for hepatocellular carcinoma (HCC) and often have increased serum levels of estrogen. It is not clear how estrogen promotes hepatic growth. We investigated the effects of estrogen on hepatocyte proliferation during zebrafish development, liver regeneration, and carcinogenesis. We also studied human hepatocytes and liver tissues. METHODS: Zebrafish were exposed to selective modifiers of estrogen signaling at larval and adult stages. Liver growth was assessed by gene expression, fluorescent imaging, and histologic analyses. We monitored liver regeneration after hepatocyte ablation and HCC development after administration of chemical carcinogens (dimethylbenzanthrazene). Proliferation of human hepatocytes was measured in a coculture system. We measured levels of G-protein-coupled estrogen receptor (GPER1) in HCC and nontumor liver tissues from 68 patients by immunohistochemistry. RESULTS: Exposure to 17ß-estradiol (E2) increased proliferation of hepatocytes and liver volume and mass in larval and adult zebrafish. Chemical genetic and epistasis experiments showed that GPER1 mediates the effects of E2 via the phosphoinositide 3-kinase-protein kinase B-mechanistic target of rapamycin pathway: gper1-knockout and mtor-knockout zebrafish did not increase liver growth in response to E2. HCC samples from patients had increased levels of GPER1 compared with nontumor tissue samples; estrogen promoted proliferation of human primary hepatocytes. Estrogen accelerated hepatocarcinogenesis specifically in male zebrafish. Chemical inhibition or genetic loss of GPER1 significantly reduced tumor development in the zebrafish. CONCLUSIONS: In an analysis of zebrafish and human liver cells and tissues, we found GPER1 to be a hepatic estrogen sensor that regulates liver growth during development, regeneration, and tumorigenesis. Inhibitors of GPER1 might be developed for liver cancer prevention or treatment. TRANSCRIPT PROFILING: The accession number in the Gene Expression Omnibus is GSE92544.


Assuntos
Carcinoma Hepatocelular/metabolismo , Estradiol/farmacologia , Estrogênios/farmacologia , Neoplasias Hepáticas/metabolismo , Fígado/crescimento & desenvolvimento , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteínas de Peixe-Zebra/metabolismo , 9,10-Dimetil-1,2-benzantraceno , Animais , Carcinogênese/efeitos dos fármacos , Carcinoma Hepatocelular/patologia , Proliferação de Células/efeitos dos fármacos , Feminino , Expressão Gênica/efeitos dos fármacos , Hepatócitos , Humanos , Fígado/metabolismo , Cirrose Hepática/metabolismo , Neoplasias Hepáticas/patologia , Regeneração Hepática , Masculino , Tamanho do Órgão/efeitos dos fármacos , Fosfatidilinositol 3-Quinase/metabolismo , Receptores Acoplados a Proteínas G/genética , Fatores Sexuais , Transdução de Sinais , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Carga Tumoral/efeitos dos fármacos , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
9.
Carcinogenesis ; 35(1): 237-46, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23996931

RESUMO

Although Apc mutation is widely considered an initiating event in colorectal cancer, little is known about the earliest stages of tumorigenesis following sporadic Apc loss. Therefore, we have utilized a novel mouse model that facilitates the sporadic inactivation of Apc via frameshift reversion of Cre in single, isolated cells and subsequently tracks the fates of Apc-deficient intestinal cells. Our results suggest that consistent with Apc being a 'gatekeeper', loss of Apc early in life during intestinal growth leads to adenomas or increased crypt fission, manifested by fields of mutant but otherwise normal-appearing crypts. In contrast, Apc loss occurring later in life has minimal consequences, with mutant crypts being less prone to either increased crypt fission or adenoma formation. Using the stem cell-specific Lgr5-CreER mouse, we generated different sized fields of Apc-deficient crypts via independent recombination events and found that field size correlates with progression to adenoma. To evaluate this early stage prior to adenoma formation as a therapeutic target, we examined the chemopreventive effects of sulindac on Apc-deficient occult crypt fission. We found that sulindac treatment started early in life inhibits the morphologically occult spread of Apc-deficient crypts and thus reduces adenoma numbers. Taken together these results suggest that: (i) earlier Apc loss promotes increased crypt fission, (ii) a field of Apc-deficient crypts, which can form via occult crypt fission or independent neighboring events, is an important intermediate between loss of Apc and adenoma formation and (iii) normal-appearing Apc-deficient crypts are potential unappreciated targets for cancer screening and chemoprevention.


Assuntos
Focos de Criptas Aberrantes/prevenção & controle , Adenoma/genética , Genes APC , Neoplasias Intestinais/genética , Sulindaco/farmacologia , Focos de Criptas Aberrantes/tratamento farmacológico , Adenoma/patologia , Fatores Etários , Animais , Quimioprevenção , Reparo do DNA/genética , Neoplasias Intestinais/patologia , Intestinos/patologia , Camundongos , Camundongos Transgênicos , Mutação , Células-Tronco/patologia , beta-Galactosidase/genética , beta-Galactosidase/metabolismo
10.
EMBO Rep ; 15(1): 62-9, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24355609

RESUMO

The concept of 'field cancerization' describes the clonal expansion of genetically altered, but morphologically normal cells that predisposes a tissue to cancer development. Here, we demonstrate that biased stem cell competition in the mouse small intestine can initiate the expansion of such clones. We quantitatively analyze how the activation of oncogenic K-ras in individual Lgr5(+) stem cells accelerates their cell division rate and creates a biased drift towards crypt clonality. K-ras mutant crypts then clonally expand within the epithelium through enhanced crypt fission, which distributes the existing Paneth cell niche over the two new crypts. Thus, an unequal competition between wild-type and mutant intestinal stem cells initiates a biased drift that leads to the clonal expansion of crypts carrying oncogenic mutations.


Assuntos
Células-Tronco Adultas/fisiologia , Neoplasias Colorretais/patologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Receptores Acoplados a Proteínas G/metabolismo , Animais , Ciclo Celular , Proliferação de Células , Transformação Celular Neoplásica , Neoplasias Colorretais/genética , Mucosa Intestinal/patologia , Intestino Delgado/patologia , Camundongos , Camundongos Transgênicos , Mutação de Sentido Incorreto , Oncogenes , Receptores Acoplados a Proteínas G/genética , Nicho de Células-Tronco
11.
Science ; 337(6095): 730-5, 2012 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-22855427

RESUMO

The concept that tumors are maintained by dedicated stem cells, the so-called cancer stem cell hypothesis, has attracted great interest but remains controversial. Studying mouse models, we provide direct, functional evidence for the presence of stem cell activity within primary intestinal adenomas, a precursor to intestinal cancer. By "lineage retracing" using the multicolor Cre-reporter R26R-Confetti, we demonstrate that the crypt stem cell marker Lgr5 (leucine-rich repeat-containing heterotrimeric guanine nucleotide-binding protein-coupled receptor 5) also marks a subpopulation of adenoma cells that fuel the growth of established intestinal adenomas. These Lgr5(+) cells, which represent about 5 to 10% of the cells in the adenomas, generate additional Lgr5(+) cells as well as all other adenoma cell types. The Lgr5(+) cells are intermingled with Paneth cells near the adenoma base, a pattern reminiscent of the architecture of the normal crypt niche.


Assuntos
Adenoma/patologia , Neoplasias Intestinais/patologia , Células-Tronco Neoplásicas/patologia , Células-Tronco Neoplásicas/fisiologia , Receptores Acoplados a Proteínas G/análise , Adenoma/genética , Adenoma/metabolismo , Animais , Biomarcadores/análise , Linhagem da Célula , Transformação Celular Neoplásica , Perfilação da Expressão Gênica , Técnicas de Introdução de Genes , Genes Reporter , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Neoplasias Intestinais/genética , Camundongos , Células-Tronco Multipotentes/patologia , Células-Tronco Multipotentes/fisiologia , Celulas de Paneth/patologia , Nicho de Células-Tronco , Tamoxifeno/farmacologia , Ensaio Tumoral de Célula-Tronco
12.
EMBO J ; 31(14): 3079-91, 2012 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-22692129

RESUMO

Two types of stem cells are currently defined in small intestinal crypts: cycling crypt base columnar (CBC) cells and quiescent '+4' cells. Here, we combine transcriptomics with proteomics to define a definitive molecular signature for Lgr5(+) CBC cells. Transcriptional profiling of FACS-sorted Lgr5(+) stem cells and their daughters using two microarray platforms revealed an mRNA stem cell signature of 384 unique genes. Quantitative mass spectrometry on the same cell populations identified 278 proteins enriched in intestinal stem cells. The mRNA and protein data sets showed a high level of correlation and a combined signature of 510 stem cell-enriched genes was defined. Spatial expression patterns were further characterized by mRNA in-situ hybridization, revealing that approximately half of the genes were expressed in a gradient with highest levels at the crypt bottom, while the other half was expressed uniquely in Lgr5(+)stem cells. Lineage tracing using a newly established knock-in mouse for one of the signature genes, Smoc2, confirmed its stem cell specificity. Using this resource, we find-and confirm by independent approaches-that the proposed quiescent/'+4' stem cell markers Bmi1, Tert, Hopx and Lrig1 are robustly expressed in CBC cells.


Assuntos
Antígenos de Diferenciação/metabolismo , Regulação da Expressão Gênica , Mucosa Intestinal/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Células-Tronco/metabolismo , Animais , Antígenos de Diferenciação/genética , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Perfilação da Expressão Gênica , Intestinos/citologia , Camundongos , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores Acoplados a Proteínas G/genética , Células-Tronco/citologia
13.
Cold Spring Harb Perspect Biol ; 4(4): a007989, 2012 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-22474007

RESUMO

The Wnt signaling pathway was originally uncovered as one of the prototype developmental signaling cascades in invertebrates as well as in vertebrates. The first indication that Wnt signaling also plays a role in the adult animal came from the study of the intestine of Tcf-4 (Tcf7L2) knockout mice. The gastrointestinal epithelium continuously self-renews over the lifetime of an organism and is, in fact, the most rapidly self-renewing tissue of the mammalian body. Recent studies indicate that Wnt signaling plays a central role in the biology of gastrointestinal stem cells. Furthermore, mutational activation of the Wnt cascade is the principle cause of colon cancer.


Assuntos
Neoplasias Gastrointestinais/metabolismo , Transdução de Sinais , Células-Tronco/metabolismo , Proteínas Wnt/metabolismo , Animais , Neoplasias Gastrointestinais/patologia , Humanos
14.
EMBO J ; 30(6): 1104-9, 2011 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-21297579

RESUMO

Somatic cells have been proposed to be limited in the number of cell divisions they can undergo. This is thought to be a mechanism by which stem cells retain their integrity preventing disease. However, we have recently discovered intestinal crypt stem cells that persist for the lifetime of a mouse, yet divide every day. We now demonstrate biochemically that primary isolated Lgr5+ve stem cells contain significant telomerase activity. Telomerase activity rapidly decreases in the undifferentiated progeny of these stem cells and is entirely lost in differentiated villus cells. Conversely, asymmetric segregation of chromosomes has been proposed as a mechanism for stem cells to protect their genomes against damage. We determined the average cell cycle length of Lgr5+ve stem cells at 21.5 h and find that Lgr5+ve intestinal stem cells randomly segregate newly synthesized DNA strands, opposing the 'immortal strand' hypothesis.


Assuntos
Segregação de Cromossomos , Mucosa Intestinal/citologia , Mucosa Intestinal/enzimologia , Nicho de Células-Tronco , Células-Tronco/enzimologia , Células-Tronco/fisiologia , Telomerase/metabolismo , Animais , Ciclo Celular , Diferenciação Celular , Células Epiteliais/enzimologia , Células Epiteliais/fisiologia , Camundongos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA