Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cancer Cell ; 42(6): 1067-1085.e11, 2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38759655

RESUMEN

In acral melanoma (AM), progression from in situ (AMis) to invasive AM (iAM) leads to significantly reduced survival. However, evolutionary dynamics during this process remain elusive. Here, we report integrative molecular and spatial characterization of 147 AMs using genomics, bulk and single-cell transcriptomics, and spatial transcriptomics and proteomics. Vertical invasion from AMis to iAM displays an early and monoclonal seeding pattern. The subsequent regional expansion of iAM exhibits two distinct patterns, clonal expansion and subclonal diversification. Notably, molecular subtyping reveals an aggressive iAM subset featured with subclonal diversification, increased epithelial-mesenchymal transition (EMT), and spatial enrichment of APOE+/CD163+ macrophages. In vitro and ex vivo experiments further demonstrate that APOE+CD163+ macrophages promote tumor EMT via IGF1-IGF1R interaction. Adnexal involvement can predict AMis with higher invasive potential whereas APOE and CD163 serve as prognostic biomarkers for iAM. Altogether, our results provide implications for the early detection and treatment of AM.


Asunto(s)
Antígenos CD , Antígenos de Diferenciación Mielomonocítica , Transición Epitelial-Mesenquimal , Melanoma , Invasividad Neoplásica , Neoplasias Cutáneas , Humanos , Melanoma/genética , Melanoma/inmunología , Melanoma/patología , Transición Epitelial-Mesenquimal/genética , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Antígenos de Diferenciación Mielomonocítica/metabolismo , Antígenos de Diferenciación Mielomonocítica/genética , Antígenos CD/metabolismo , Antígenos CD/genética , Apolipoproteínas E/genética , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Femenino , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Microambiente Tumoral/inmunología , Microambiente Tumoral/genética , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Regulación Neoplásica de la Expresión Génica , Análisis Espacial , Persona de Mediana Edad , Pronóstico , Progresión de la Enfermedad , Anciano , Receptores de Superficie Celular
2.
Nature ; 612(7938): 141-147, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36352227

RESUMEN

The heterogeneity of the tumour immune microenvironment (TIME), organized by various immune and stromal cells, is a major contributing factor of tumour metastasis, relapse and drug resistance1-3, but how different TIME subtypes are connected to the clinical relevance in liver cancer remains unclear. Here we performed single-cell RNA-sequencing (scRNA-seq) analysis of 189 samples collected from 124 patients and 8 mice with liver cancer. With more than 1 million cells analysed, we stratified patients into five TIME subtypes, including immune activation, immune suppression mediated by myeloid or stromal cells, immune exclusion and immune residence phenotypes. Different TIME subtypes were spatially organized and associated with chemokine networks and genomic features. Notably, tumour-associated neutrophil (TAN) populations enriched in the myeloid-cell-enriched subtype were associated with an unfavourable prognosis. Through in vitro induction of TANs and ex vivo analyses of patient TANs, we showed that CCL4+ TANs can recruit macrophages and that PD-L1+ TANs can suppress T cell cytotoxicity. Furthermore, scRNA-seq analysis of mouse neutrophil subsets revealed that they are largely conserved with those of humans. In vivo neutrophil depletion in mouse models attenuated tumour progression, confirming the pro-tumour phenotypes of TANs. With this detailed cellular heterogeneity landscape of liver cancer, our study illustrates diverse TIME subtypes, highlights immunosuppressive functions of TANs and sheds light on potential immunotherapies targeting TANs.


Asunto(s)
Neoplasias Hepáticas , Neutrófilos , Microambiente Tumoral , Animales , Humanos , Ratones , Inmunoterapia , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/inmunología , Recurrencia Local de Neoplasia , Neutrófilos/citología , Neutrófilos/inmunología , Microambiente Tumoral/inmunología , Linfocitos T/inmunología , Macrófagos/inmunología , Pronóstico , Progresión de la Enfermedad
3.
Gastroenterology ; 162(1): 238-252, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34481846

RESUMEN

BACKGROUND & AIMS: Copy number alterations (CNAs), elicited by genome instability, are a major source of intratumor heterogeneity. How CNAs evolve in hepatocellular carcinoma (HCC) remains unknown. METHODS: We performed single-cell DNA sequencing (scDNA-seq) on 1275 cells isolated from 10 patients with HCC, ploidy-resolved scDNA-seq on 356 cells from 1 additional patient, and single-cell RNA sequencing on 27,344 cells from 3 additional patients. Three statistical fitting models were compared to investigate the CNA accumulation pattern. RESULTS: Cells in the tumor were categorized into the following 3 subpopulations: euploid, pseudoeuploid, and aneuploid. Our scDNA-seq analysis revealed that CNA accumulation followed a dual-phase copy number evolution model, that is, a punctuated phase followed by a gradual phase. Patients who exhibited prolonged gradual phase showed higher intratumor heterogeneity and worse disease-free survival. Integrating bulk RNA sequencing of 17 patients with HCC, published datasets of 1196 liver tumors, and immunohistochemical staining of 202 HCC tumors, we found that high expression of CAD, a gene involved in pyrimidine synthesis, was correlated with rapid tumorigenesis and reduced survival. The dual-phase copy number evolution model was validated by our single-cell RNA sequencing data and published scDNA-seq datasets of other cancer types. Furthermore, ploidy-resolved scDNA-seq revealed the common clonal origin of diploid- and polyploid-aneuploid cells, suggesting that polyploid tumor cells were generated by whole genome doubling of diploid tumor cells. CONCLUSIONS: Our work revealed a novel dual-phase copy number evolution model, showed HCC with longer gradual phase was more severe, identified CAD as a promising biomarker for early recurrence of HCC, and supported the diploid origin of polyploid HCC.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma Hepatocelular/genética , Evolución Clonal , Heterogeneidad Genética , Neoplasias Hepáticas/genética , Análisis de Secuencia de ADN , Análisis de la Célula Individual , Adulto , Anciano , Carcinoma Hepatocelular/metabolismo , Variaciones en el Número de Copia de ADN , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Femenino , Dosificación de Gen , Regulación Neoplásica de la Expresión Génica , Inestabilidad Genómica , Humanos , Neoplasias Hepáticas/mortalidad , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/terapia , Masculino , Persona de Mediana Edad , Modelos Genéticos , Recurrencia Local de Neoplasia , Ploidias , Factores de Tiempo
4.
Adv Sci (Weinh) ; 8(17): e2101230, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34250753

RESUMEN

Intrahepatic cholangiocarcinoma (ICC) is highly heterogeneous. Here, the authors perform exome sequencing and bulk RNA sequencing on 73 tumor regions from 14 ICC patients to portray the multi-faceted intratumor heterogeneity (ITH) landscape of ICC. The authors show that ITH is highly concordant across genomic, transcriptomic, and immune levels. Comparison of these data to 8 published datasets reveals significantly higher degrees of ITH in ICC than hepatocellular carcinoma. Remarkably, the authors find that high-ITH tumors highly overlap with the IDH (isocitrate dehydrogenase)-mutant subgroup (IDH-SG), comprising of IDH-mutated tumors and IDH-like tumors, that is, those IDH-wildtype tumors that exhibit similar molecular profiles to the IDH-mutated ones. Furthermore, IDH-SG exhibits less T cell infiltration and lower T cell cytotoxicity, indicating a colder tumor microenvironment (TME). The higher ITH and colder TME of IDH-SG are successfully validated by single-cell RNA sequencing on 17 503 cells from 4 patients. Collectively, the study shows that IDH mutant subgroup status, rather than IDH mutation alone, is associated with ITH and the TME of ICC tumors. The results highlight that IDH-like patients may also benefit from IDH targeted therapies and provide important implications for the diagnosis and treatment of ICC.


Asunto(s)
Neoplasias de los Conductos Biliares/genética , Colangiocarcinoma/genética , Isocitrato Deshidrogenasa/genética , Mutación/genética , Microambiente Tumoral/genética , Humanos
5.
Int J Biol Sci ; 16(15): 2835-2852, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33061800

RESUMEN

As one of the most ominous malignancies, hepatocellular carcinoma (HCC) is frequently diagnosed at an advanced stage, owing to its aggressive invasion and metastatic spread. Emerging evidence has demonstrated that Rictor, as a unique component of the mTORC2, plays a role in cell migration, as it is dysregulated in various cancers, including HCC. However, the underlying molecular mechanism has not been well-characterized. Here, evaluation on a tissue-array panel and bioinformatics analysis revealed that Rictor is highly expressed in HCC tissues. Moreover, increased Rictor expression predicts poor survival of HCC patients. Rictor knockdown significantly suppressed cell migration and actin polymerization, thereby leading to decreased nuclear accumulation of MKL1 and subsequent inactivation of SRF/MKL1-dependent gene transcription, i.e. Arp3 and c-Fos. Mechanistically, we identified ABLIM1 as a previously unknown phosphorylation target of Rictor. Rictor interacts with ABLIM1 and regulates its serine phosphorylation in HCC cells. We generated ABLIM1 knockout cell lines of HCC, in which dominant negative mutations of Ser 214 and Ser 431 residues inhibited the ABLIM1-mediated actin polymerization and the MKL1 signaling pathway. Overall, ABLIM1 phosphorylation induced by Rictor plays an important role in controlling actin polymerization in HCC cells.


Asunto(s)
Carcinoma Hepatocelular , Proteínas con Dominio LIM , Neoplasias Hepáticas , Proteínas de Microfilamentos , Proteína Asociada al mTOR Insensible a la Rapamicina , Actinas/genética , Actinas/metabolismo , Carcinoma Hepatocelular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Humanos , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/metabolismo , Neoplasias Hepáticas/genética , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Fosforilación , Polimerizacion , Proteína Asociada al mTOR Insensible a la Rapamicina/genética , Proteína Asociada al mTOR Insensible a la Rapamicina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA