Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 185(14): 2591-2608.e30, 2022 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-35803246

RESUMO

Melanoma brain metastasis (MBM) frequently occurs in patients with advanced melanoma; yet, our understanding of the underlying salient biology is rudimentary. Here, we performed single-cell/nucleus RNA-seq in 22 treatment-naive MBMs and 10 extracranial melanoma metastases (ECMs) and matched spatial single-cell transcriptomics and T cell receptor (TCR)-seq. Cancer cells from MBM were more chromosomally unstable, adopted a neuronal-like cell state, and enriched for spatially variably expressed metabolic pathways. Key observations were validated in independent patient cohorts, patient-derived MBM/ECM xenograft models, RNA/ATAC-seq, proteomics, and multiplexed imaging. Integrated spatial analyses revealed distinct geography of putative cancer immune evasion and evidence for more abundant intra-tumoral B to plasma cell differentiation in lymphoid aggregates in MBM. MBM harbored larger fractions of monocyte-derived macrophages and dysfunctional TOX+CD8+ T cells with distinct expression of immune checkpoints. This work provides comprehensive insights into MBM biology and serves as a foundational resource for further discovery and therapeutic exploration.


Assuntos
Neoplasias Encefálicas , Melanoma , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/secundário , Linfócitos T CD8-Positivos/patologia , Ecossistema , Humanos , RNA-Seq
2.
Cell ; 174(6): 1347-1360, 2018 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-30193109

RESUMO

Chromosomal instability (CIN) is a hallmark of human cancer, and it is associated with poor prognosis, metastasis, and therapeutic resistance. CIN results from errors in chromosome segregation during mitosis, leading to structural and numerical chromosomal abnormalities. In addition to generating genomic heterogeneity that acts as a substrate for natural selection, CIN promotes inflammatory signaling by introducing double-stranded DNA into the cytosol, engaging the cGAS-STING anti-viral pathway. These multipronged effects distinguish CIN as a central driver of tumor evolution and as a genomic source for the crosstalk between the tumor and its microenvironment, in the course of immune editing and evasion.


Assuntos
Instabilidade Cromossômica , Aneuploidia , Humanos , Imunidade Inata , Interferon Tipo I/metabolismo , Proteínas de Membrana/metabolismo , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/patologia , Nucleotidiltransferases/metabolismo , Transdução de Sinais , Microambiente Tumoral
3.
Cell ; 173(1): 276-276.e1, 2018 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-29570996

RESUMO

CGAS responds to cytosolic DNA by initiating a STING-dependent response that ultimately engages innate immune effectors to ensure the preservation of organismal homeostasis.


Assuntos
Proteínas de Membrana/metabolismo , Nucleotidiltransferases/metabolismo , Microambiente Celular , Humanos , Imunidade Inata , NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Quinase Induzida por NF-kappaB
4.
Cell ; 171(5): 987-989, 2017 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-29149612

RESUMO

In this issue of Cell, Martincorena et al. and Campbell et al. interrogated the selection dynamics during tumor evolution using large-scale genomics datasets. They found that somatic mutations in cancer are largely neutral, highlighting a near-complete absence of negative selection. Neutral evolution enables tolerance of hypermutation, which defines a surprisingly large fraction of adult cancer.


Assuntos
Neoplasias , Adulto , Genômica , Humanos
5.
Nature ; 618(7967): 1041-1048, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37165191

RESUMO

Complex genome rearrangements can be generated by the catastrophic pulverization of missegregated chromosomes trapped within micronuclei through a process known as chromothripsis1-5. As each chromosome contains a single centromere, it remains unclear how acentric fragments derived from shattered chromosomes are inherited between daughter cells during mitosis6. Here we tracked micronucleated chromosomes with live-cell imaging and show that acentric fragments cluster in close spatial proximity throughout mitosis for asymmetric inheritance by a single daughter cell. Mechanistically, the CIP2A-TOPBP1 complex prematurely associates with DNA lesions within ruptured micronuclei during interphase, which poises pulverized chromosomes for clustering upon mitotic entry. Inactivation of CIP2A-TOPBP1 caused acentric fragments to disperse throughout the mitotic cytoplasm, stochastically partition into the nucleus of both daughter cells and aberrantly misaccumulate as cytoplasmic DNA. Mitotic clustering facilitates the reassembly of acentric fragments into rearranged chromosomes lacking the extensive DNA copy-number losses that are characteristic of canonical chromothripsis. Comprehensive analysis of pan-cancer genomes revealed clusters of DNA copy-number-neutral rearrangements-termed balanced chromothripsis-across diverse tumour types resulting in the acquisition of known cancer driver events. Thus, distinct patterns of chromothripsis can be explained by the spatial clustering of pulverized chromosomes from micronuclei.


Assuntos
Cromossomos Humanos , Cromotripsia , Micronúcleos com Defeito Cromossômico , Mitose , Humanos , Centrômero , Cromossomos Humanos/genética , DNA/genética , DNA/metabolismo , Variações do Número de Cópias de DNA , Interfase , Mitose/genética , Neoplasias/genética
6.
Nature ; 620(7976): 1080-1088, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37612508

RESUMO

Chromosomal instability (CIN) is a driver of cancer metastasis1-4, yet the extent to which this effect depends on the immune system remains unknown. Using ContactTracing-a newly developed, validated and benchmarked tool to infer the nature and conditional dependence of cell-cell interactions from single-cell transcriptomic data-we show that CIN-induced chronic activation of the cGAS-STING pathway promotes downstream signal re-wiring in cancer cells, leading to a pro-metastatic tumour microenvironment. This re-wiring is manifested by type I interferon tachyphylaxis selectively downstream of STING and a corresponding increase in cancer cell-derived endoplasmic reticulum (ER) stress response. Reversal of CIN, depletion of cancer cell STING or inhibition of ER stress response signalling abrogates CIN-dependent effects on the tumour microenvironment and suppresses metastasis in immune competent, but not severely immune compromised, settings. Treatment with STING inhibitors reduces CIN-driven metastasis in melanoma, breast and colorectal cancers in a manner dependent on tumour cell-intrinsic STING. Finally, we show that CIN and pervasive cGAS activation in micronuclei are associated with ER stress signalling, immune suppression and metastasis in human triple-negative breast cancer, highlighting a viable strategy to identify and therapeutically intervene in tumours spurred by CIN-induced inflammation.


Assuntos
Instabilidade Cromossômica , Progressão da Doença , Neoplasias , Humanos , Benchmarking , Comunicação Celular , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Melanoma/tratamento farmacológico , Melanoma/genética , Melanoma/imunologia , Melanoma/patologia , Microambiente Tumoral , Interferon Tipo I/imunologia , Metástase Neoplásica , Estresse do Retículo Endoplasmático , Transdução de Sinais , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/imunologia , Neoplasias de Mama Triplo Negativas/patologia , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/patologia
7.
Nature ; 619(7968): 176-183, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37286593

RESUMO

Chromosomal instability (CIN) and epigenetic alterations are characteristics of advanced and metastatic cancers1-4, but whether they are mechanistically linked is unknown. Here we show that missegregation of mitotic chromosomes, their sequestration in micronuclei5,6 and subsequent rupture of the micronuclear envelope7 profoundly disrupt normal histone post-translational modifications (PTMs), a phenomenon conserved across humans and mice, as well as in cancer and non-transformed cells. Some of the changes in histone PTMs occur because of the rupture of the micronuclear envelope, whereas others are inherited from mitotic abnormalities before the micronucleus is formed. Using orthogonal approaches, we demonstrate that micronuclei exhibit extensive differences in chromatin accessibility, with a strong positional bias between promoters and distal or intergenic regions, in line with observed redistributions of histone PTMs. Inducing CIN causes widespread epigenetic dysregulation, and chromosomes that transit in micronuclei experience heritable abnormalities in their accessibility long after they have been reincorporated into the primary nucleus. Thus, as well as altering genomic copy number, CIN promotes epigenetic reprogramming and heterogeneity in cancer.


Assuntos
Instabilidade Cromossômica , Segregação de Cromossomos , Cromossomos , Epigênese Genética , Micronúcleos com Defeito Cromossômico , Neoplasias , Animais , Humanos , Camundongos , Cromatina/genética , Instabilidade Cromossômica/genética , Cromossomos/genética , Cromossomos/metabolismo , Histonas/química , Histonas/metabolismo , Neoplasias/genética , Neoplasias/patologia , Mitose , Variações do Número de Cópias de DNA , Processamento de Proteína Pós-Traducional
8.
Nature ; 595(7865): 114-119, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33915568

RESUMO

Respiratory failure is the leading cause of death in patients with severe SARS-CoV-2 infection1,2, but the host response at the lung tissue level is poorly understood. Here we performed single-nucleus RNA sequencing of about 116,000 nuclei from the lungs of nineteen individuals who died of COVID-19 and underwent rapid autopsy and seven control individuals. Integrated analyses identified substantial alterations in cellular composition, transcriptional cell states, and cell-to-cell interactions, thereby providing insight into the biology of lethal COVID-19. The lungs from individuals with COVID-19 were highly inflamed, with dense infiltration of aberrantly activated monocyte-derived macrophages and alveolar macrophages, but had impaired T cell responses. Monocyte/macrophage-derived interleukin-1ß and epithelial cell-derived interleukin-6 were unique features of SARS-CoV-2 infection compared to other viral and bacterial causes of pneumonia. Alveolar type 2 cells adopted an inflammation-associated transient progenitor cell state and failed to undergo full transition into alveolar type 1 cells, resulting in impaired lung regeneration. Furthermore, we identified expansion of recently described CTHRC1+ pathological fibroblasts3 contributing to rapidly ensuing pulmonary fibrosis in COVID-19. Inference of protein activity and ligand-receptor interactions identified putative drug targets to disrupt deleterious circuits. This atlas enables the dissection of lethal COVID-19, may inform our understanding of long-term complications of COVID-19 survivors, and provides an important resource for therapeutic development.


Assuntos
COVID-19/patologia , COVID-19/virologia , Pulmão/patologia , SARS-CoV-2/patogenicidade , Análise de Célula Única , Idoso , Idoso de 80 Anos ou mais , Células Epiteliais Alveolares/patologia , Células Epiteliais Alveolares/virologia , Atlas como Assunto , Autopsia , COVID-19/imunologia , Estudos de Casos e Controles , Feminino , Fibroblastos/patologia , Fibrose/patologia , Fibrose/virologia , Humanos , Inflamação/patologia , Inflamação/virologia , Macrófagos/patologia , Macrófagos/virologia , Macrófagos Alveolares/patologia , Macrófagos Alveolares/virologia , Masculino , Pessoa de Meia-Idade , Plasmócitos/imunologia , Linfócitos T/imunologia
9.
Trends Biochem Sci ; 46(6): 446-460, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33461879

RESUMO

The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway has been primarily characterized as an inflammatory mechanism in higher eukaryotes in response to cytosolic double-stranded DNA (dsDNA). Since its initial discovery, detailed mechanisms delineating the dynamic subcellular localization of its different components and downstream signaling have been uncovered, leading to attempts to harness its proinflammatory properties for therapeutic benefit in cancer. Emerging evidence, however, indicates that a crucial primordial function of STING is to promote autophagy, and that downstream interferon (IFN) signaling emerged recently in its evolutionary history. Furthermore, studies suggest that this pathway is a crucial regulator of cellular metabolism that potentially couples inflammation to nutrient availability. We focus on the evolutionarily conserved functions of STING, and we discuss how a broader understanding of this pathway can help us to better appreciate its potential role in cancer and harness it for therapeutic benefit.


Assuntos
Proteínas de Membrana , Neoplasias , Transdução de Sinais , Autofagia , Humanos , Proteínas de Membrana/genética , Nucleotidiltransferases/metabolismo
10.
Nature ; 559(7714): 350-355, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29995854

RESUMO

The selective pressures that shape clonal evolution in healthy individuals are largely unknown. Here we investigate 8,342 mosaic chromosomal alterations, from 50 kb to 249 Mb long, that we uncovered in blood-derived DNA from 151,202 UK Biobank participants using phase-based computational techniques (estimated false discovery rate, 6-9%). We found six loci at which inherited variants associated strongly with the acquisition of deletions or loss of heterozygosity in cis. At three such loci (MPL, TM2D3-TARSL2, and FRA10B), we identified a likely causal variant that acted with high penetrance (5-50%). Inherited alleles at one locus appeared to affect the probability of somatic mutation, and at three other loci to be objects of positive or negative clonal selection. Several specific mosaic chromosomal alterations were strongly associated with future haematological malignancies. Our results reveal a multitude of paths towards clonal expansions with a wide range of effects on human health.


Assuntos
Aberrações Cromossômicas , Células Clonais/citologia , Células Clonais/metabolismo , Hematopoese/genética , Mosaicismo , Adulto , Idoso , Alelos , Bancos de Espécimes Biológicos , Quebra Cromossômica , Sítios Frágeis do Cromossomo/genética , Cromossomos Humanos Par 10/genética , Feminino , Saúde , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/mortalidade , Humanos , Masculino , Pessoa de Meia-Idade , Penetrância , Reino Unido
11.
Nature ; 553(7689): 467-472, 2018 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-29342134

RESUMO

Chromosomal instability is a hallmark of cancer that results from ongoing errors in chromosome segregation during mitosis. Although chromosomal instability is a major driver of tumour evolution, its role in metastasis has not been established. Here we show that chromosomal instability promotes metastasis by sustaining a tumour cell-autonomous response to cytosolic DNA. Errors in chromosome segregation create a preponderance of micronuclei whose rupture spills genomic DNA into the cytosol. This leads to the activation of the cGAS-STING (cyclic GMP-AMP synthase-stimulator of interferon genes) cytosolic DNA-sensing pathway and downstream noncanonical NF-κB signalling. Genetic suppression of chromosomal instability markedly delays metastasis even in highly aneuploid tumour models, whereas continuous chromosome segregation errors promote cellular invasion and metastasis in a STING-dependent manner. By subverting lethal epithelial responses to cytosolic DNA, chromosomally unstable tumour cells co-opt chronic activation of innate immune pathways to spread to distant organs.


Assuntos
Instabilidade Cromossômica , Citosol/metabolismo , DNA de Neoplasias/metabolismo , Metástase Neoplásica/genética , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/secundário , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Neoplasias da Mama/secundário , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Linhagem Celular , Instabilidade Cromossômica/genética , Segregação de Cromossomos , Citosol/enzimologia , Feminino , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Inflamação/genética , Inflamação/metabolismo , Proteínas de Membrana/metabolismo , Mesoderma/metabolismo , Camundongos , Micronúcleos com Defeito Cromossômico , NF-kappa B/metabolismo , Nucleotidiltransferases/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Nat Mater ; 22(5): 532-533, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37138010
14.
PLoS Comput Biol ; 14(9): e1006447, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30204765

RESUMO

Cancer cells frequently undergo chromosome missegregation events during mitosis, whereby the copies of a given chromosome are not distributed evenly among the two daughter cells, thus creating cells with heterogeneous karyotypes. A stochastic model tracing cellular karyotypes derived from clonal populations over hundreds of generations was recently developed and experimentally validated, and it was capable of predicting favorable karyotypes frequently observed in cancer. Here, we construct and study a Markov chain that precisely describes karyotypic evolution during clonally expanding cancer cell populations. The Markov chain allows us to directly predict the distribution of karyotypes and the expected size of the tumor after many cell divisions without resorting to computationally expensive simulations. We determine the limiting karyotype distribution of an evolving tumor population, and quantify its dependency on several key parameters including the initial karyotype of the founder cell, the rate of whole chromosome missegregation, and chromosome-specific cell viability. Using this model, we confirm the existence of an optimal rate of chromosome missegregation probabilities that maximizes karyotypic heterogeneity, while minimizing the occurrence of nullisomy. Interestingly, karyotypic heterogeneity is significantly more dependent on chromosome missegregation probabilities rather than the number of cell divisions, so that maximal heterogeneity can be reached rapidly (within a few hundred generations of cell division) at chromosome missegregation rates commonly observed in cancer cell lines. Conversely, at low missegregation rates, heterogeneity is constrained even after thousands of cell division events. This leads us to conclude that chromosome copy number heterogeneity is primarily constrained by chromosome missegregation rates and the risk for nullisomy and less so by the age of the tumor. This model enables direct integration of karyotype information into existing models of tumor evolution based on somatic mutations.


Assuntos
Instabilidade Cromossômica , Cromossomos , Cariotipagem , Mitose , Neoplasias/genética , Algoritmos , Sobrevivência Celular , Células Cultivadas , Análise Mutacional de DNA , Humanos , Cadeias de Markov , Modelos Estatísticos , Mutação , Neoplasias/patologia , Probabilidade , Processos Estocásticos
15.
N Engl J Med ; 371(26): 2477-87, 2014 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-25426838

RESUMO

BACKGROUND: Cancers arise from multiple acquired mutations, which presumably occur over many years. Early stages in cancer development might be present years before cancers become clinically apparent. METHODS: We analyzed data from whole-exome sequencing of DNA in peripheral-blood cells from 12,380 persons, unselected for cancer or hematologic phenotypes. We identified somatic mutations on the basis of unusual allelic fractions. We used data from Swedish national patient registers to follow health outcomes for 2 to 7 years after DNA sampling. RESULTS: Clonal hematopoiesis with somatic mutations was observed in 10% of persons older than 65 years of age but in only 1% of those younger than 50 years of age. Detectable clonal expansions most frequently involved somatic mutations in three genes (DNMT3A, ASXL1, and TET2) that have previously been implicated in hematologic cancers. Clonal hematopoiesis was a strong risk factor for subsequent hematologic cancer (hazard ratio, 12.9; 95% confidence interval, 5.8 to 28.7). Approximately 42% of hematologic cancers in this cohort arose in persons who had clonality at the time of DNA sampling, more than 6 months before a first diagnosis of cancer. Analysis of bone marrow-biopsy specimens obtained from two patients at the time of diagnosis of acute myeloid leukemia revealed that their cancers arose from the earlier clones. CONCLUSIONS: Clonal hematopoiesis with somatic mutations is readily detected by means of DNA sequencing, is increasingly common as people age, and is associated with increased risks of hematologic cancer and death. A subset of the genes that are mutated in patients with myeloid cancers is frequently mutated in apparently healthy persons; these mutations may represent characteristic early events in the development of hematologic cancers. (Funded by the National Human Genome Research Institute and others.).


Assuntos
Sangue , Transformação Celular Neoplásica/genética , Neoplasias Hematológicas/genética , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Mutação , Adulto , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Células Clonais , Análise Mutacional de DNA , Exoma , Neoplasias Hematológicas/fisiopatologia , Humanos , Pessoa de Meia-Idade , Fatores de Risco , Adulto Jovem
16.
Cancer ; 120(11): 1733-42, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24604319

RESUMO

BACKGROUND: Persistent chromosome segregation errors represent a conspicuous feature of human neoplasms. It is widely accepted that this chromosomal instability is associated with poor prognosis; however, its effect on therapeutic response is a matter of conjecture. METHODS: Here, the role of chromosome segregation errors in the response of patients with rectal adenocarcinoma to chemoradiation therapy (CRT) was examined. Pretreatment samples from 62 patients were surveyed for evidence of chromosome mis-segregation and mis-segregation frequency was correlated to the pathological response to CRT as determined by the tumor regression grade after surgical resection of irradiated tumors. RESULTS: Surprisingly, it was found that errors in chromosome segregation predicted enhanced pathological response of rectal adenocarcinoma to CRT (odds ratio, 3.9; P = .02). Furthermore, tumor response inversely correlated with the frequency of cells that exhibited segregation errors during anaphase (correlation coefficient, 0.94; P < .05). Strikingly, elevated chromosome mis-segregation combined with decreased levels of the DNA damage repair protein Mre11 portended a markedly enhanced response (odds ratio, 54.0; P = .008). CONCLUSIONS: The results of the current study demonstrate that chromosomal instability is a favorable predictor of response to CRT in patients with locally invasive rectal adenocarcinoma. Therefore, the authors propose that downstream structural damage to chromosomes resulting from segregation errors potentiates the effect of DNA-damaging therapies and synergizes with deficiencies in the DNA repair machinery. This work identifies a novel mechanistic marker that foretells treatment response to CRT and suggests that concomitant targeting of whole-chromosome segregation and DNA repair may constitute an effective therapeutic strategy.


Assuntos
Adenocarcinoma/terapia , Quimiorradioterapia , Instabilidade Cromossômica , Neoplasias Retais/terapia , Adenocarcinoma/genética , Idoso , Reparo do DNA , Proteínas de Ligação a DNA/análise , Feminino , Humanos , Proteína Homóloga a MRE11 , Masculino , Pessoa de Meia-Idade , Neoplasias Retais/genética
17.
EMBO J ; 29(20): 3531-43, 2010 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-20852589

RESUMO

Accurate chromosome segregation during mitosis requires precise coordination of various processes, such as chromosome alignment, maturation of proper kinetochore-microtubule (kMT) attachments, correction of erroneous attachments, and silencing of the spindle assembly checkpoint (SAC). How these fundamental aspects of mitosis are coordinately and temporally regulated is poorly understood. In this study, we show that the temporal regulation of kMT attachments by CLASP1, astrin and Kif2b is central to mitotic progression and chromosome segregation fidelity. In early mitosis, a Kif2b-CLASP1 complex is recruited to kinetochores to promote chromosome movement, kMT turnover, correction of attachment errors, and maintenance of SAC signalling. However, during metaphase, this complex is replaced by an astrin-CLASP1 complex, which promotes kMT stability, chromosome alignment, and silencing of the SAC. We show that these two complexes are differentially recruited to kinetochores and are mutually exclusive. We also show that other kinetochore proteins, such as Kif18a, affect kMT attachments and chromosome movement through these proteins. Thus, CLASP1-astrin-Kif2b complex act as a central switch at kinetochores that defines mitotic progression and promotes fidelity by temporally regulating kMT attachments.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Cinesinas/metabolismo , Cinetocoros/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Mitose/fisiologia , Fuso Acromático/metabolismo , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Proteína B de Centrômero/genética , Proteína B de Centrômero/metabolismo , Cromossomos/metabolismo , Humanos , Cinesinas/genética , Proteínas Associadas aos Microtúbulos/genética , Interferência de RNA , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
18.
Cancer Discov ; 14(2): 214-226, 2024 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-38197599

RESUMO

Chromosome-containing micronuclei are a feature of human cancer. Micronuclei arise from chromosome mis-segregation and characterize tumors with elevated rates of chromosomal instability. Although their association with cancer has been long recognized, only recently have we broadened our understanding of the mechanisms that govern micronuclei formation and their role in tumor progression. In this review, we provide a brief historical account of micronuclei, depict the mechanisms underpinning their creation, and illuminate their capacity to propel tumor evolution through genetic, epigenetic, and transcriptional transformations. We also posit the prospect of leveraging micronuclei as biomarkers and therapeutic targets in chromosomally unstable cancers. SIGNIFICANCE: Micronuclei in chromosomally unstable cancer cells serve as pivotal catalysts for cancer progression, instigating transformative genomic, epigenetic, and transcriptional alterations. This comprehensive review not only synthesizes our present comprehension but also outlines a framework for translating this knowledge into pioneering biomarkers and therapeutics, thereby illuminating novel paths for personalized cancer management.


Assuntos
Neoplasias , Humanos , Neoplasias/genética , Instabilidade Cromossômica , Segregação de Cromossomos , Biomarcadores
19.
Cancer Immunol Res ; 12(6): 673-686, 2024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38408184

RESUMO

Chromosomal instability is a hallmark of human cancer that is associated with aggressive disease characteristics. Chromosome mis-segregations help fuel natural selection, but they risk provoking a cGAS-STING immune response through the accumulation of cytosolic DNA. The mechanisms of how tumors benefit from chromosomal instability while mitigating associated risks, such as enhanced immune surveillance, are poorly understood. Here, we identify cGAS-STING-dependent upregulation of the nuclease TREX1 as an adaptive, negative feedback mechanism that promotes immune evasion through digestion of cytosolic DNA. TREX1 loss diminishes tumor growth, prolongs survival of host animals, increases tumor immune infiltration, and potentiates response to immune checkpoint blockade selectively in tumors capable of mounting a type I IFN response downstream of STING. Together, these data demonstrate that TREX1 induction shields chromosomally unstable tumors from immune surveillance by dampening type I IFN production and suggest that TREX1 inhibitors might be used to selectively target tumors that have retained the inherent ability to mount an IFN response downstream of STING. See related article by Lim et al., p. 663.


Assuntos
Exodesoxirribonucleases , Interferon Tipo I , Fosfoproteínas , Exodesoxirribonucleases/genética , Exodesoxirribonucleases/metabolismo , Interferon Tipo I/metabolismo , Fosfoproteínas/metabolismo , Fosfoproteínas/genética , Animais , Humanos , Camundongos , Neoplasias/imunologia , Neoplasias/genética , Evasão da Resposta Imune , Linhagem Celular Tumoral , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Evasão Tumoral
20.
Clin Cancer Res ; 30(12): 2672-2683, 2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38502113

RESUMO

PURPOSE: Targeted therapies have improved outcomes for patients with metastatic colorectal cancer, but their impact is limited by rapid emergence of resistance. We hypothesized that an understanding of the underlying genetic mechanisms and intrinsic tumor features that mediate resistance to therapy will guide new therapeutic strategies and ultimately allow the prevention of resistance. EXPERIMENTAL DESIGN: We assembled a series of 52 patients with paired pretreatment and progression samples who received therapy targeting EGFR (n = 17), BRAF V600E (n = 17), KRAS G12C (n = 15), or amplified HER2 (n = 3) to identify molecular and clinical factors associated with time on treatment (TOT). RESULTS: All patients stopped treatment for progression and TOT did not vary by oncogenic driver (P = 0.5). Baseline disease burden (≥3 vs. <3 sites, P = 0.02), the presence of hepatic metastases (P = 0.02), and gene amplification on baseline tissue (P = 0.03) were each associated with shorter TOT. We found evidence of chromosomal instability (CIN) at progression in patients with baseline MAPK pathway amplifications and those with acquired gene amplifications. At resistance, copy-number changes (P = 0.008) and high number (≥5) of acquired alterations (P = 0.04) were associated with shorter TOT. Patients with hepatic metastases demonstrated both higher number of emergent alterations at resistance and enrichment of mutations involving receptor tyrosine kinases. CONCLUSIONS: Our genomic analysis suggests that high baseline CIN or effective induction of enhanced mutagenesis on targeted therapy underlies rapid progression. Longer response appears to result from a progressive acquisition of genomic or chromosomal instability in the underlying cancer or from the chance event of a new resistance alteration.


Assuntos
Neoplasias Colorretais , Resistencia a Medicamentos Antineoplásicos , Terapia de Alvo Molecular , Proteínas Proto-Oncogênicas B-raf , Humanos , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Neoplasias Colorretais/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Masculino , Proteínas Proto-Oncogênicas B-raf/genética , Pessoa de Meia-Idade , Idoso , Proteínas Proto-Oncogênicas p21(ras)/genética , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Mutação , Progressão da Doença , Receptores ErbB/genética , Receptores ErbB/antagonistas & inibidores , Adulto , Instabilidade Cromossômica , Idoso de 80 Anos ou mais , Amplificação de Genes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA