Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 18.179
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 187(7): 1574-1577, 2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38552606

RESUMEN

Discoveries of the roles of RAS oncogenes in cancer development four decades ago opened the door to proving that tumor development is driven by somatic mutations' altering the genomes of cancer cells. These discoveries led to illusions about the simplicity of cancer pathogenesis and how cancer could be cured.


Asunto(s)
Genes ras , Neoplasias , Humanos , Neoplasias/genética , Neoplasias/terapia , Oncogenes , Mutación
2.
Cell ; 186(18): 3921-3944.e25, 2023 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-37582357

RESUMEN

Cancer driver events refer to key genetic aberrations that drive oncogenesis; however, their exact molecular mechanisms remain insufficiently understood. Here, our multi-omics pan-cancer analysis uncovers insights into the impacts of cancer drivers by identifying their significant cis-effects and distal trans-effects quantified at the RNA, protein, and phosphoprotein levels. Salient observations include the association of point mutations and copy-number alterations with the rewiring of protein interaction networks, and notably, most cancer genes converge toward similar molecular states denoted by sequence-based kinase activity profiles. A correlation between predicted neoantigen burden and measured T cell infiltration suggests potential vulnerabilities for immunotherapies. Patterns of cancer hallmarks vary by polygenic protein abundance ranging from uniform to heterogeneous. Overall, our work demonstrates the value of comprehensive proteogenomics in understanding the functional states of oncogenic drivers and their links to cancer development, surpassing the limitations of studying individual cancer types.


Asunto(s)
Neoplasias , Proteogenómica , Humanos , Neoplasias/genética , Oncogenes , Transformación Celular Neoplásica/genética , Variaciones en el Número de Copia de ADN
3.
Cell ; 186(2): 363-381.e19, 2023 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-36669472

RESUMEN

Advanced solid cancers are complex assemblies of tumor, immune, and stromal cells characterized by high intratumoral variation. We use highly multiplexed tissue imaging, 3D reconstruction, spatial statistics, and machine learning to identify cell types and states underlying morphological features of known diagnostic and prognostic significance in colorectal cancer. Quantitation of these features in high-plex marker space reveals recurrent transitions from one tumor morphology to the next, some of which are coincident with long-range gradients in the expression of oncogenes and epigenetic regulators. At the tumor invasive margin, where tumor, normal, and immune cells compete, T cell suppression involves multiple cell types and 3D imaging shows that seemingly localized 2D features such as tertiary lymphoid structures are commonly interconnected and have graded molecular properties. Thus, while cancer genetics emphasizes the importance of discrete changes in tumor state, whole-specimen imaging reveals large-scale morphological and molecular gradients analogous to those in developing tissues.


Asunto(s)
Adenocarcinoma , Neoplasias Colorrectales , Humanos , Adenocarcinoma/patología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Procesamiento de Imagen Asistido por Computador , Oncogenes , Microambiente Tumoral
4.
Cell ; 186(25): 5554-5568.e18, 2023 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-38065080

RESUMEN

Cancer cells are regulated by oncogenic mutations and microenvironmental signals, yet these processes are often studied separately. To functionally map how cell-intrinsic and cell-extrinsic cues co-regulate cell fate, we performed a systematic single-cell analysis of 1,107 colonic organoid cultures regulated by (1) colorectal cancer (CRC) oncogenic mutations, (2) microenvironmental fibroblasts and macrophages, (3) stromal ligands, and (4) signaling inhibitors. Multiplexed single-cell analysis revealed a stepwise epithelial differentiation phenoscape dictated by combinations of oncogenes and stromal ligands, spanning from fibroblast-induced Clusterin (CLU)+ revival colonic stem cells (revCSCs) to oncogene-driven LRIG1+ hyper-proliferative CSCs (proCSCs). The transition from revCSCs to proCSCs is regulated by decreasing WNT3A and TGF-ß-driven YAP signaling and increasing KRASG12D or stromal EGF/Epiregulin-activated MAPK/PI3K flux. We find that APC loss and KRASG12D collaboratively limit access to revCSCs and disrupt stromal-epithelial communication-trapping epithelia in the proCSC fate. These results reveal that oncogenic mutations dominate homeostatic differentiation by obstructing cell-extrinsic regulation of cell-fate plasticity.


Asunto(s)
Proteínas Proto-Oncogénicas p21(ras) , Transducción de Señal , Diferenciación Celular , Oncogenes , Proteínas Proto-Oncogénicas p21(ras)/genética , Células Madre , Humanos , Animales , Ratones , Linaje de la Célula
5.
Cell ; 186(17): 3674-3685.e14, 2023 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-37494934

RESUMEN

Epigenetic lesions that disrupt regulatory elements represent potential cancer drivers. However, we lack experimental models for validating their tumorigenic impact. Here, we model aberrations arising in isocitrate dehydrogenase-mutant gliomas, which exhibit DNA hypermethylation. We focus on a CTCF insulator near the PDGFRA oncogene that is recurrently disrupted by methylation in these tumors. We demonstrate that disruption of the syntenic insulator in mouse oligodendrocyte progenitor cells (OPCs) allows an OPC-specific enhancer to contact and induce Pdgfra, thereby increasing proliferation. We show that a second lesion, methylation-dependent silencing of the Cdkn2a tumor suppressor, cooperates with insulator loss in OPCs. Coordinate inactivation of the Pdgfra insulator and Cdkn2a drives gliomagenesis in vivo. Despite locus synteny, the insulator is CpG-rich only in humans, a feature that may confer human glioma risk but complicates mouse modeling. Our study demonstrates the capacity of recurrent epigenetic lesions to drive OPC proliferation in vitro and gliomagenesis in vivo.


Asunto(s)
Neoplasias Encefálicas , Epigénesis Genética , Glioma , Animales , Humanos , Ratones , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Metilación de ADN , Glioma/genética , Glioma/patología , Isocitrato Deshidrogenasa/genética , Mutación , Oncogenes , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética
6.
Cell ; 184(14): 3812-3828.e30, 2021 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-34214472

RESUMEN

We study a patient with the human papilloma virus (HPV)-2-driven "tree-man" phenotype and two relatives with unusually severe HPV4-driven warts. The giant horns form an HPV-2-driven multifocal benign epithelial tumor overexpressing viral oncogenes in the epidermis basal layer. The patients are unexpectedly homozygous for a private CD28 variant. They have no detectable CD28 on their T cells, with the exception of a small contingent of revertant memory CD4+ T cells. T cell development is barely affected, and T cells respond to CD3 and CD2, but not CD28, costimulation. Although the patients do not display HPV-2- and HPV-4-reactive CD4+ T cells in vitro, they make antibodies specific for both viruses in vivo. CD28-deficient mice are susceptible to cutaneous infections with the mouse papillomavirus MmuPV1. The control of HPV-2 and HPV-4 in keratinocytes is dependent on the T cell CD28 co-activation pathway. Surprisingly, human CD28-dependent T cell responses are largely redundant for protective immunity.


Asunto(s)
Antígenos CD28/deficiencia , Patrón de Herencia/genética , Papillomaviridae/fisiología , Piel/virología , Linfocitos T/inmunología , Adulto , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Antígenos CD28/genética , Antígenos CD28/metabolismo , Linfocitos T CD4-Positivos/inmunología , Niño , Endopeptidasas/metabolismo , Femenino , Genes Recesivos , Células HEK293 , Homocigoto , Humanos , Inmunidad Humoral , Memoria Inmunológica , Células Jurkat , Queratinocitos/patología , Masculino , Ratones Endogámicos C57BL , Oncogenes , Papiloma/patología , Papiloma/virología , Linaje , Señales de Clasificación de Proteína , ARN Mensajero/genética , ARN Mensajero/metabolismo
7.
Cell ; 184(20): 5215-5229.e17, 2021 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-34559986

RESUMEN

Estrogen receptor α (ERα) is a hormone receptor and key driver for over 70% of breast cancers that has been studied for decades as a transcription factor. Unexpectedly, we discover that ERα is a potent non-canonical RNA-binding protein. We show that ERα RNA binding function is uncoupled from its activity to bind DNA and critical for breast cancer progression. Employing genome-wide cross-linking immunoprecipitation (CLIP) sequencing and a functional CRISPRi screen, we find that ERα-associated mRNAs sustain cancer cell fitness and elicit cellular responses to stress. Mechanistically, ERα controls different steps of RNA metabolism. In particular, we demonstrate that ERα RNA binding mediates alternative splicing of XBP1 and translation of the eIF4G2 and MCL1 mRNAs, which facilitates survival upon stress conditions and sustains tamoxifen resistance of cancer cells. ERα is therefore a multifaceted RNA-binding protein, and this activity transforms our knowledge of post-transcriptional regulation underlying cancer development and drug response.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Resistencia a Antineoplásicos , Receptor alfa de Estrógeno/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Secuencia de Bases , Neoplasias de la Mama/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Progresión de la Enfermedad , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Receptor alfa de Estrógeno/química , Factor 4G Eucariótico de Iniciación/genética , Factor 4G Eucariótico de Iniciación/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genómica , Humanos , Ratones Endogámicos NOD , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Oncogenes , Unión Proteica/efectos de los fármacos , Dominios Proteicos , Empalme del ARN/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Estrés Fisiológico/efectos de los fármacos , Estrés Fisiológico/genética , Tamoxifeno/farmacología , Proteína 1 de Unión a la X-Box/metabolismo
8.
Cell ; 184(18): 4753-4771.e27, 2021 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-34388391

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is characterized by notorious resistance to current therapies attributed to inherent tumor heterogeneity and highly desmoplastic and immunosuppressive tumor microenvironment (TME). Unique proline isomerase Pin1 regulates multiple cancer pathways, but its role in the TME and cancer immunotherapy is unknown. Here, we find that Pin1 is overexpressed both in cancer cells and cancer-associated fibroblasts (CAFs) and correlates with poor survival in PDAC patients. Targeting Pin1 using clinically available drugs induces complete elimination or sustained remissions of aggressive PDAC by synergizing with anti-PD-1 and gemcitabine in diverse model systems. Mechanistically, Pin1 drives the desmoplastic and immunosuppressive TME by acting on CAFs and induces lysosomal degradation of the PD-1 ligand PD-L1 and the gemcitabine transporter ENT1 in cancer cells, besides activating multiple cancer pathways. Thus, Pin1 inhibition simultaneously blocks multiple cancer pathways, disrupts the desmoplastic and immunosuppressive TME, and upregulates PD-L1 and ENT1, rendering PDAC eradicable by immunochemotherapy.


Asunto(s)
Inmunoterapia , Terapia Molecular Dirigida , Peptidilprolil Isomerasa de Interacción con NIMA/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/inmunología , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Aloinjertos/inmunología , Secuencias de Aminoácidos , Animales , Apoptosis/efectos de los fármacos , Antígeno B7-H1/metabolismo , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Desoxicitidina/uso terapéutico , Sinergismo Farmacológico , Endocitosis/efectos de los fármacos , Tranportador Equilibrativo 1 de Nucleósido/metabolismo , Humanos , Terapia de Inmunosupresión , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Ratones , Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/metabolismo , Oncogenes , Organoides/efectos de los fármacos , Organoides/patología , Transducción de Señal/efectos de los fármacos , Análisis de Supervivencia , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
9.
Cell ; 182(6): 1531-1544.e15, 2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32846158

RESUMEN

The fidelity of intracellular signaling hinges on the organization of dynamic activity architectures. Spatial compartmentation was first proposed over 30 years ago to explain how diverse G protein-coupled receptors achieve specificity despite converging on a ubiquitous messenger, cyclic adenosine monophosphate (cAMP). However, the mechanisms responsible for spatially constraining this diffusible messenger remain elusive. Here, we reveal that the type I regulatory subunit of cAMP-dependent protein kinase (PKA), RIα, undergoes liquid-liquid phase separation (LLPS) as a function of cAMP signaling to form biomolecular condensates enriched in cAMP and PKA activity, critical for effective cAMP compartmentation. We further show that a PKA fusion oncoprotein associated with an atypical liver cancer potently blocks RIα LLPS and induces aberrant cAMP signaling. Loss of RIα LLPS in normal cells increases cell proliferation and induces cell transformation. Our work reveals LLPS as a principal organizer of signaling compartments and highlights the pathological consequences of dysregulating this activity architecture.


Asunto(s)
Carcinogénesis/metabolismo , Carcinoma Hepatocelular/genética , Compartimento Celular/genética , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Proteínas del Choque Térmico HSP40/genética , Neoplasias Hepáticas/genética , Transducción de Señal , Animales , Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Carcinoma Hepatocelular/metabolismo , Compartimento Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , AMP Cíclico/farmacología , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Citoplasma/metabolismo , Humanos , Neoplasias Hepáticas/metabolismo , Ratones , Oncogenes/genética , Dominios Proteicos , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes de Fusión , Espectroscopía Infrarroja por Transformada de Fourier , Imagen de Lapso de Tiempo/métodos
10.
Nat Rev Mol Cell Biol ; 23(1): 74-88, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34508254

RESUMEN

Cancer is a group of diseases in which cells divide continuously and excessively. Cell division is tightly regulated by multiple evolutionarily conserved cell cycle control mechanisms, to ensure the production of two genetically identical cells. Cell cycle checkpoints operate as DNA surveillance mechanisms that prevent the accumulation and propagation of genetic errors during cell division. Checkpoints can delay cell cycle progression or, in response to irreparable DNA damage, induce cell cycle exit or cell death. Cancer-associated mutations that perturb cell cycle control allow continuous cell division chiefly by compromising the ability of cells to exit the cell cycle. Continuous rounds of division, however, create increased reliance on other cell cycle control mechanisms to prevent catastrophic levels of damage and maintain cell viability. New detailed insights into cell cycle control mechanisms and their role in cancer reveal how these dependencies can be best exploited in cancer treatment.


Asunto(s)
Puntos de Control del Ciclo Celular , Neoplasias/patología , Animales , Daño del ADN/genética , Replicación del ADN/genética , Humanos , Neoplasias/genética , Neoplasias/terapia , Oncogenes , Huso Acromático/metabolismo
11.
Cell ; 177(6): 1375-1383, 2019 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-31150618

RESUMEN

Recent studies of the tumor genome seek to identify cancer pathways as groups of genes in which mutations are epistatic with one another or, specifically, "mutually exclusive." Here, we show that most mutations are mutually exclusive not due to pathway structure but to interactions with disease subtype and tumor mutation load. In particular, many cancer driver genes are mutated preferentially in tumors with few mutations overall, causing mutations in these cancer genes to appear mutually exclusive with numerous others. Researchers should view current epistasis maps with caution until we better understand the multiple cause-and-effect relationships among factors such as tumor subtype, positive selection for mutations, and gross tumor characteristics including mutational signatures and load.


Asunto(s)
Epistasis Genética/genética , Genes Relacionados con las Neoplasias/genética , Neoplasias/genética , Algoritmos , Biología Computacional/métodos , Epistasis Genética/fisiología , Genes Relacionados con las Neoplasias/fisiología , Humanos , Modelos Genéticos , Mutación/genética , Oncogenes/genética
12.
Cell ; 179(5): 1033-1055, 2019 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-31730848

RESUMEN

For decades, research into cancer biology focused on the involvement of protein-coding genes. Only recently was it discovered that an entire class of molecules, termed non-coding RNA (ncRNA), plays key regulatory roles in shaping cellular activity. An explosion of studies into ncRNA biology has since shown that they represent a diverse and prevalent group of RNAs, including both oncogenic molecules and those that work in a tumor suppressive manner. As a result, hundreds of cancer-focused clinical trials involving ncRNAs as novel biomarkers or therapies have begun and these are likely just the beginning.


Asunto(s)
Oncología Médica , ARN no Traducido/metabolismo , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Ensayos Clínicos como Asunto , Humanos , Neoplasias/genética , Oncogenes , ARN no Traducido/genética
13.
Cell ; 179(6): 1330-1341.e13, 2019 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-31761532

RESUMEN

Non-coding regions amplified beyond oncogene borders have largely been ignored. Using a computational approach, we find signatures of significant co-amplification of non-coding DNA beyond the boundaries of amplified oncogenes across five cancer types. In glioblastoma, EGFR is preferentially co-amplified with its two endogenous enhancer elements active in the cell type of origin. These regulatory elements, their contacts, and their contribution to cell fitness are preserved on high-level circular extrachromosomal DNA amplifications. Interrogating the locus with a CRISPR interference screening approach reveals a diversity of additional elements that impact cell fitness. The pattern of fitness dependencies mirrors the rearrangement of regulatory elements and accompanying rewiring of the chromatin topology on the extrachromosomal amplicon. Our studies indicate that oncogene amplifications are shaped by regulatory dependencies in the non-coding genome.


Asunto(s)
Cromosomas Humanos/genética , Elementos de Facilitación Genéticos , Amplificación de Genes , Oncogenes , Acetilación , Sistemas CRISPR-Cas/genética , Línea Celular Tumoral , Supervivencia Celular/genética , Cromatina/metabolismo , ADN de Neoplasias/genética , Receptores ErbB/genética , Receptores ErbB/metabolismo , Genes Relacionados con las Neoplasias , Sitios Genéticos , Glioblastoma/genética , Glioblastoma/patología , Histonas/metabolismo , Humanos , Neuroglía/metabolismo
14.
Cell ; 172(4): 645-647, 2018 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-29425486

RESUMEN

K-Ras is the undisputed champion of oncogenes, yet our ability to interfere with its oncogenic function is hampered by insufficient mechanistic understanding. In this issue of Cell, Ambrogio and colleagues connect the ability of K-Ras to dimerize to the ability of wild-type K-Ras to limit the oncogenic properties of the mutant.


Asunto(s)
Proteínas Proto-Oncogénicas p21(ras) , Hermanos , Carcinogénesis , Dimerización , Humanos , Masculino , Oncogenes
15.
Cell ; 173(2): 499-514.e23, 2018 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-29576454

RESUMEN

Genomics has provided a detailed structural description of the cancer genome. Identifying oncogenic drivers that work primarily through dosage changes is a current challenge. Unrestrained proliferation is a critical hallmark of cancer. We constructed modular, barcoded libraries of human open reading frames (ORFs) and performed screens for proliferation regulators in multiple cell types. Approximately 10% of genes regulate proliferation, with most performing in an unexpectedly highly tissue-specific manner. Proliferation drivers in a given cell type showed specific enrichment in somatic copy number changes (SCNAs) from cognate tumors and helped predict aneuploidy patterns in those tumors, implying that tissue-type-specific genetic network architectures underlie SCNA and driver selection in different cancers. In vivo screening confirmed these results. We report a substantial contribution to the catalog of SCNA-associated cancer drivers, identifying 147 amplified and 107 deleted genes as potential drivers, and derive insights about the genetic network architecture of aneuploidy in tumors.


Asunto(s)
Aneuploidia , Neoplasias/patología , Animales , Línea Celular Tumoral , Proliferación Celular , Mapeo Cromosómico , Cromosomas/genética , Factor de Transcripción E2F1/antagonistas & inhibidores , Factor de Transcripción E2F1/genética , Factor de Transcripción E2F1/metabolismo , Femenino , Biblioteca de Genes , Genómica , Humanos , Queratinas/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Oncogenes , Sistemas de Lectura Abierta/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo
16.
Cell ; 170(3): 577-592.e10, 2017 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-28753431

RESUMEN

Elucidation of the mutational landscape of human cancer has progressed rapidly and been accompanied by the development of therapeutics targeting mutant oncogenes. However, a comprehensive mapping of cancer dependencies has lagged behind and the discovery of therapeutic targets for counteracting tumor suppressor gene loss is needed. To identify vulnerabilities relevant to specific cancer subtypes, we conducted a large-scale RNAi screen in which viability effects of mRNA knockdown were assessed for 7,837 genes using an average of 20 shRNAs per gene in 398 cancer cell lines. We describe findings of this screen, outlining the classes of cancer dependency genes and their relationships to genetic, expression, and lineage features. In addition, we describe robust gene-interaction networks recapitulating both protein complexes and functional cooperation among complexes and pathways. This dataset along with a web portal is provided to the community to assist in the discovery and translation of new therapeutic approaches for cancer.


Asunto(s)
Neoplasias/genética , Neoplasias/patología , Interferencia de ARN , Línea Celular Tumoral , Biblioteca de Genes , Redes Reguladoras de Genes , Humanos , Complejos Multiproteicos/metabolismo , Neoplasias/metabolismo , Oncogenes , ARN Interferente Pequeño , Transducción de Señal , Factores de Transcripción/metabolismo
17.
Mol Cell ; 84(7): 1365-1376.e7, 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38452764

RESUMEN

Enhancer-gene communication is dependent on topologically associating domains (TADs) and boundaries enforced by the CCCTC-binding factor (CTCF) insulator, but the underlying structures and mechanisms remain controversial. Here, we investigate a boundary that typically insulates fibroblast growth factor (FGF) oncogenes but is disrupted by DNA hypermethylation in gastrointestinal stromal tumors (GISTs). The boundary contains an array of CTCF sites that enforce adjacent TADs, one containing FGF genes and the other containing ANO1 and its putative enhancers, which are specifically active in GIST and its likely cell of origin. We show that coordinate disruption of four CTCF motifs in the boundary fuses the adjacent TADs, allows the ANO1 enhancer to contact FGF3, and causes its robust induction. High-resolution micro-C maps reveal specific contact between transcription initiation sites in the ANO1 enhancer and FGF3 promoter that quantitatively scales with FGF3 induction such that modest changes in contact frequency result in strong changes in expression, consistent with a causal relationship.


Asunto(s)
Cromatina , Elementos de Facilitación Genéticos , Factor de Unión a CCCTC/genética , Factor de Unión a CCCTC/metabolismo , Cromatina/genética , Oncogenes , ADN/química
18.
Nat Immunol ; 20(7): 835-851, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31160797

RESUMEN

How tumor cells genetically lose antigenicity and evade immune checkpoints remains largely elusive. We report that tissue-specific expression of the human long noncoding RNA LINK-A in mouse mammary glands initiates metastatic mammary gland tumors, which phenotypically resemble human triple-negative breast cancer (TNBC). LINK-A expression facilitated crosstalk between phosphatidylinositol-(3,4,5)-trisphosphate and inhibitory G-protein-coupled receptor (GPCR) pathways, attenuating protein kinase A-mediated phosphorylation of the E3 ubiquitin ligase TRIM71. Consequently, LINK-A expression enhanced K48-polyubiquitination-mediated degradation of the antigen peptide-loading complex (PLC) and intrinsic tumor suppressors Rb and p53. Treatment with LINK-A locked nucleic acids or GPCR antagonists stabilized the PLC components, Rb and p53, and sensitized mammary gland tumors to immune checkpoint blockers. Patients with programmed ccll death protein-1(PD-1) blockade-resistant TNBC exhibited elevated LINK-A levels and downregulated PLC components. Hence we demonstrate lncRNA-dependent downregulation of antigenicity and intrinsic tumor suppression, which provides the basis for developing combinational immunotherapy treatment regimens and early TNBC prevention.


Asunto(s)
Presentación de Antígeno/inmunología , Regulación Neoplásica de la Expresión Génica , Neoplasias/genética , Neoplasias/inmunología , Oncogenes , ARN Largo no Codificante/genética , Escape del Tumor/genética , Escape del Tumor/inmunología , Adenoma/genética , Adenoma/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Ratones , Neoplasias/metabolismo , Neoplasias/patología , Fosforilación , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitinación , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Cell ; 166(3): 740-754, 2016 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-27397505

RESUMEN

Systematic studies of cancer genomes have provided unprecedented insights into the molecular nature of cancer. Using this information to guide the development and application of therapies in the clinic is challenging. Here, we report how cancer-driven alterations identified in 11,289 tumors from 29 tissues (integrating somatic mutations, copy number alterations, DNA methylation, and gene expression) can be mapped onto 1,001 molecularly annotated human cancer cell lines and correlated with sensitivity to 265 drugs. We find that cell lines faithfully recapitulate oncogenic alterations identified in tumors, find that many of these associate with drug sensitivity/resistance, and highlight the importance of tissue lineage in mediating drug response. Logic-based modeling uncovers combinations of alterations that sensitize to drugs, while machine learning demonstrates the relative importance of different data types in predicting drug response. Our analysis and datasets are rich resources to link genotypes with cellular phenotypes and to identify therapeutic options for selected cancer sub-populations.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Análisis de Varianza , Línea Celular Tumoral , Metilación de ADN , Resistencia a Antineoplásicos/genética , Dosificación de Gen , Humanos , Modelos Genéticos , Mutación , Neoplasias/genética , Oncogenes , Medicina de Precisión
20.
Cell ; 166(5): 1132-1146.e7, 2016 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-27565343

RESUMEN

Cancers are distributed unevenly across the body, but the importance of cell intrinsic factors such as stem cell function in determining organ cancer risk is unknown. Therefore, we used Cre-recombination of conditional lineage tracing, oncogene, and tumor suppressor alleles to define populations of stem and non-stem cells in mouse organs and test their life-long susceptibility to tumorigenesis. We show that tumor incidence is determined by the life-long generative capacity of mutated cells. This relationship held true in the presence of multiple genotypes and regardless of developmental stage, strongly supporting the notion that stem cells dictate organ cancer risk. Using the liver as a model system, we further show that damage-induced activation of stem cell function markedly increases cancer risk. Therefore, we propose that a combination of stem cell mutagenesis and extrinsic factors that enhance the proliferation of these cell populations, creates a "perfect storm" that ultimately determines organ cancer risk. VIDEO ABSTRACT.


Asunto(s)
Carcinogénesis/genética , Carcinogénesis/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Oncogenes , Células Madre , Alelos , Animales , Genes Supresores de Tumor , Humanos , Integrasas , Ratones , Modelos Biológicos , Mutagénesis , Recombinación Genética , Riesgo , Células Madre/metabolismo , Células Madre/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA