Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 19.956
Filtrar
Más filtros

Intervalo de año de publicación
1.
Mol Cell ; 73(2): 212-223.e7, 2019 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-30554942

RESUMEN

Cohesin subunits are frequently mutated in cancer, but how they function as tumor suppressors is unknown. Cohesin mediates sister chromatid cohesion, but this is not always perturbed in cancer cells. Here, we identify a previously unknown role for cohesin. We find that cohesin is required to repress transcription at DNA double-strand breaks (DSBs). Notably, cohesin represses transcription at DSBs throughout interphase, indicating that this is distinct from its known role in mediating DNA repair through sister chromatid cohesion. We identified a cancer-associated SA2 mutation that supports sister chromatid cohesion but is unable to repress transcription at DSBs. We further show that failure to repress transcription at DSBs leads to large-scale genome rearrangements. Cancer samples lacking SA2 display mutational patterns consistent with loss of this pathway. These findings uncover a new function for cohesin that provides insights into its frequent loss in cancer.


Asunto(s)
Neoplasias Óseas/genética , Proteínas de Ciclo Celular/genética , Proteínas Cromosómicas no Histona/genética , Roturas del ADN de Doble Cadena , Inestabilidad Genómica , Interfase , Osteosarcoma/genética , Transcripción Genética , Antígenos Nucleares/genética , Antígenos Nucleares/metabolismo , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proteínas Cromosómicas no Histona/metabolismo , Segregación Cromosómica , Reparación del ADN , Regulación hacia Abajo , Fase G1 , Fase G2 , Regulación Neoplásica de la Expresión Génica , Humanos , Osteosarcoma/metabolismo , Osteosarcoma/patología , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Cohesinas
2.
Mol Cell ; 76(1): 27-43.e11, 2019 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-31447390

RESUMEN

Cancer cells acquire unlimited proliferative capacity by either re-expressing telomerase or inducing alternative lengthening of telomeres (ALT), which relies on telomere recombination. Here, we show that ALT recombination requires coordinate regulation of the SMX and BTR complexes to ensure the appropriate balance of resolution and dissolution activities at recombining telomeres. Critical to this control is SLX4IP, which accumulates at ALT telomeres and interacts with SLX4, XPF, and BLM. Loss of SLX4IP increases ALT-related phenotypes, which is incompatible with cell growth following concomitant loss of SLX4. Inactivation of BLM is sufficient to rescue telomere aggregation and the synthetic growth defect in this context, suggesting that SLX4IP favors SMX-dependent resolution by antagonizing promiscuous BLM activity during ALT recombination. Finally, we show that SLX4IP is inactivated in a subset of ALT-positive osteosarcomas. Collectively, our findings uncover an SLX4IP-dependent regulatory mechanism critical for telomere maintenance in ALT cancer cells.


Asunto(s)
Neoplasias Óseas/enzimología , Proteínas Portadoras/metabolismo , Osteosarcoma/enzimología , RecQ Helicasas/metabolismo , Homeostasis del Telómero , Telómero/metabolismo , Animales , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Proteínas Portadoras/genética , Proliferación Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Células HeLa , Humanos , Ratones Noqueados , Ratones SCID , Osteosarcoma/genética , Osteosarcoma/patología , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , RecQ Helicasas/genética , Recombinasas/genética , Recombinasas/metabolismo , Transducción de Señal , Telómero/genética , Telómero/patología
3.
Am J Hum Genet ; 110(3): 427-441, 2023 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-36787739

RESUMEN

Ewing sarcoma (EwS) is a rare bone and soft tissue malignancy driven by chromosomal translocations encoding chimeric transcription factors, such as EWSR1-FLI1, that bind GGAA motifs forming novel enhancers that alter nearby expression. We propose that germline microsatellite variation at the 6p25.1 EwS susceptibility locus could impact downstream gene expression and EwS biology. We performed targeted long-read sequencing of EwS blood DNA to characterize variation and genomic features important for EWSR1-FLI1 binding. We identified 50 microsatellite alleles at 6p25.1 and observed that EwS-affected individuals had longer alleles (>135 bp) with more GGAA repeats. The 6p25.1 GGAA microsatellite showed chromatin features of an EWSR1-FLI1 enhancer and regulated expression of RREB1, a transcription factor associated with RAS/MAPK signaling. RREB1 knockdown reduced proliferation and clonogenic potential and reduced expression of cell cycle and DNA replication genes. Our integrative analysis at 6p25.1 details increased binding of longer GGAA microsatellite alleles with acquired EWSR-FLI1 to promote Ewing sarcomagenesis by RREB1-mediated proliferation.


Asunto(s)
Neoplasias Óseas , Sarcoma de Ewing , Humanos , Alelos , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Proteína Proto-Oncogénica c-fli-1/genética , Proteína Proto-Oncogénica c-fli-1/metabolismo , Proteína EWS de Unión a ARN/genética , Proteína EWS de Unión a ARN/metabolismo , Sarcoma de Ewing/genética , Sarcoma de Ewing/metabolismo , Sarcoma de Ewing/patología
4.
Mol Cell ; 69(4): 581-593.e7, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29452638

RESUMEN

The bioenergetics and molecular determinants of the metabolic response to mitochondrial dysfunction are incompletely understood, in part due to a lack of appropriate isogenic cellular models of primary mitochondrial defects. Here, we capitalize on a recently developed cell model with defined levels of m.8993T>G mutation heteroplasmy, mTUNE, to investigate the metabolic underpinnings of mitochondrial dysfunction. We found that impaired utilization of reduced nicotinamide adenine dinucleotide (NADH) by the mitochondrial respiratory chain leads to cytosolic reductive carboxylation of glutamine as a new mechanism for cytosol-confined NADH recycling supported by malate dehydrogenase 1 (MDH1). We also observed that increased glycolysis in cells with mitochondrial dysfunction is associated with increased cell migration in an MDH1-dependent fashion. Our results describe a novel link between glycolysis and mitochondrial dysfunction mediated by reductive carboxylation of glutamine.


Asunto(s)
Citosol/metabolismo , Glutamina/metabolismo , Malato Deshidrogenasa/metabolismo , Mitocondrias/patología , NAD/metabolismo , Osteosarcoma/patología , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Movimiento Celular , Ciclo del Ácido Cítrico , ADN Mitocondrial/genética , Metabolismo Energético , Femenino , Glucosa/metabolismo , Glucólisis , Humanos , Mitocondrias/metabolismo , Osteosarcoma/genética , Osteosarcoma/metabolismo , Oxidación-Reducción , Células Tumorales Cultivadas
5.
Genes Dev ; 32(15-16): 1008-1019, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-30042132

RESUMEN

Various types of repetitive sequences are dysregulated in cancer. In Ewing sarcoma, the oncogenic fusion protein EWS-FLI1 induces chromatin features typical of active enhancers at GGAA microsatellite repeats, but the function of these sites has not been directly demonstrated. Here, by combining nascent transcription profiling with epigenome editing, we found that a subset of GGAA microsatellite repeats is transcriptionally active in Ewing sarcoma and that silencing individual repeats abolishes local nascent transcription and leads to markedly reduced expression of putative target genes. Epigenome silencing of these repeat sites does not affect gene expression in unrelated cells, can prevent the induction of gene expression by EWS-FLI1, and, in the case of a GGAA repeat that controls SOX2 expression from a distance of 470 kb, is sufficient to impair the growth of Ewing sarcoma xenografts. Using an experimental approach that is broadly applicable to testing different types of repetitive genomic elements, our study directly demonstrates that specific repeat microsatellites can have critical gene regulation functions in cancer and thus represent tumor-specific vulnerabilities that may be exploited to develop new therapies.


Asunto(s)
Neoplasias Óseas/genética , Elementos de Facilitación Genéticos , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Repeticiones de Microsatélite , Proteínas de Fusión Oncogénica/metabolismo , Proteína Proto-Oncogénica c-fli-1/metabolismo , Proteína EWS de Unión a ARN/metabolismo , Sarcoma de Ewing/genética , Animales , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Células Cultivadas , Cromatina/metabolismo , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodominio/biosíntesis , Proteínas de Homeodominio/genética , Humanos , Células Madre Mesenquimatosas/metabolismo , Ratones , ARN no Traducido/biosíntesis , Sarcoma de Ewing/metabolismo , Sarcoma de Ewing/patología , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Transcripción Genética , Células Tumorales Cultivadas , Proteínas de Pez Cebra
6.
J Pathol ; 262(2): 147-160, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38010733

RESUMEN

TP53 is the most frequently mutated gene in human cancer. This gene shows not only loss-of-function mutations but also recurrent missense mutations with gain-of-function activity. We have studied the primary bone malignancy osteosarcoma, which harbours one of the most rearranged genomes of all cancers. This is odd since it primarily affects children and adolescents who have not lived the long life thought necessary to accumulate massive numbers of mutations. In osteosarcoma, TP53 is often disrupted by structural variants. Here, we show through combined whole-genome and transcriptome analyses of 148 osteosarcomas that TP53 structural variants commonly result in loss of coding parts of the gene while simultaneously preserving and relocating the promoter region. The transferred TP53 promoter region is fused to genes previously implicated in cancer development. Paradoxically, these erroneously upregulated genes are significantly associated with the TP53 signalling pathway itself. This suggests that while the classical tumour suppressor activities of TP53 are lost, certain parts of the TP53 signalling pathway that are necessary for cancer cell survival and proliferation are retained. In line with this, our data suggest that transposition of the TP53 promoter is an early event that allows for a new normal state of genome-wide rearrangements in osteosarcoma. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Asunto(s)
Neoplasias Óseas , Osteosarcoma , Niño , Adolescente , Humanos , Genes p53 , Osteosarcoma/genética , Osteosarcoma/patología , Mutación , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Regiones Promotoras Genéticas/genética , Fusión Génica , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
7.
Exp Cell Res ; 436(2): 113978, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38382805

RESUMEN

Osteosarcoma (OS) is one of the most prevalent primary bone tumors with a high degree of metastasis and poor prognosis. Epithelial-to-mesenchymal transition (EMT) is a cellular mechanism that contributes to the invasion and metastasis of cancer cells, and OS cells have been reported to exhibit EMT-like characteristics. Our previous studies have shown that the interaction between tumor necrosis factor superfamily member 11 (TNFRSF11A; also known as RANK) and its ligand TNFSF11 (also known as RANKL) promotes the EMT process in breast cancer cells. However, whether the interaction between RANK and RANKL enhances aggressive behavior by inducing EMT in OS cells has not yet been elucidated. In this study, we showed that the interaction between RANK and RANKL increased the migration, invasion, and metastasis of OS cells by promoting EMT. Importantly, we clarified that the RANK/RANKL axis induces EMT by activating the nuclear factor-kappa B (NF-κB) pathway. Furthermore, the NF-κB inhibitor dimethyl fumarate (DMF) suppressed migration, invasion, and EMT in OS cells. Our results suggest that the RANK/RANKL axis may serve as a potential tumor marker and promising therapeutic target for OS metastasis. Furthermore, DMF may have clinical applications in the treatment of lung metastasis in patients with OS.


Asunto(s)
Neoplasias Óseas , Osteosarcoma , Humanos , FN-kappa B/genética , FN-kappa B/metabolismo , Transducción de Señal , Receptor Activador del Factor Nuclear kappa-B/genética , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Línea Celular Tumoral , Invasividad Neoplásica , Osteosarcoma/patología , Neoplasias Óseas/patología , Transición Epitelial-Mesenquimal/genética , Movimiento Celular/genética
8.
Mol Cell ; 68(1): 224-232.e4, 2017 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-28985506

RESUMEN

Although ARF can suppress tumor growth by activating p53 function, the mechanisms by which it suppresses tumor growth independently of p53 are not well understood. Here, we identified ARF as a key regulator of nuclear factor E2-related factor 2 (NRF2) through complex purification. ARF inhibits the ability of NRF2 to transcriptionally activate its target genes, including SLC7A11, a component of the cystine/glutamate antiporter that regulates reactive oxygen species (ROS)-induced ferroptosis. As a consequence, ARF expression sensitizes cells to ferroptosis in a p53-independent manner while ARF depletion induces NRF2 activation and promotes cancer cell survival in response to oxidative stress. Moreover, the ability of ARF to induce p53-independent tumor growth suppression in mouse xenograft models is significantly abrogated upon NRF2 overexpression. These results demonstrate that NRF2 is a major target of p53-independent tumor suppression by ARF and also suggest that the ARF-NRF2 interaction acts as a new checkpoint for oxidative stress responses.


Asunto(s)
Sistema de Transporte de Aminoácidos y+/genética , Neoplasias Óseas/genética , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Regulación Neoplásica de la Expresión Génica , Factor 2 Relacionado con NF-E2/genética , Sistema de Transporte de Aminoácidos y+/metabolismo , Animales , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Línea Celular Tumoral , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Fibroblastos/citología , Fibroblastos/metabolismo , Células HEK293 , Xenoinjertos , Humanos , Ratones , Ratones Desnudos , Factor 2 Relacionado con NF-E2/metabolismo , Osteoblastos/metabolismo , Osteoblastos/patología , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
9.
Cell Mol Life Sci ; 81(1): 236, 2024 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-38795203

RESUMEN

Chemoresistance is the main obstacle in the clinical treatment of osteosarcoma (OS). In this study, we investigated the role of EF-hand domain-containing protein 1 (EFHD1) in OS chemotherapy resistance. We found that the expression of EFHD1 was highly correlated with the clinical outcome after chemotherapy. We overexpressed EFHD1 in 143B cells and found that it increased their resistance to cell death after drug treatment. Conversely, knockdown of EFHD1 in 143BR cells (a cisplatin-less-sensitive OS cell line derived from 143B cells) increased their sensitivity to treatment. Mechanistically, EFHD1 bound to adenine nucleotide translocase-3 (ANT3) and inhibited its conformational change, thereby inhibiting the opening of the mitochondrial membrane permeability transition pore (mPTP). This effect could maintain mitochondrial function, thereby favoring OS cell survival. The ANT3 conformational inhibitor carboxyatractyloside (CATR), which can promote mPTP opening, enhanced the chemosensitivity of EFHD1-overexpressing cells when combined with cisplatin. The ANT3 conformational inhibitor bongkrekic acid (BKA), which can inhibit mPTP opening, restored the resistance of EFHD1 knockdown cells. In conclusion, our results suggest that EFHD1-ANT3-mPTP might be a promising target for OS therapy in the future.


Asunto(s)
Proliferación Celular , Cisplatino , Resistencia a Antineoplásicos , Proteínas de Transporte de Membrana Mitocondrial , Poro de Transición de la Permeabilidad Mitocondrial , Osteosarcoma , Humanos , Osteosarcoma/metabolismo , Osteosarcoma/patología , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/genética , Poro de Transición de la Permeabilidad Mitocondrial/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/genética , Línea Celular Tumoral , Cisplatino/farmacología , Neoplasias Óseas/patología , Neoplasias Óseas/metabolismo , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/genética , Translocador 3 del Nucleótido Adenina/metabolismo , Translocador 3 del Nucleótido Adenina/genética , Antineoplásicos/farmacología , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Animales , Ratones , Unión Proteica
10.
Proc Natl Acad Sci U S A ; 119(16): e2117857119, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-35412907

RESUMEN

The RB1 gene is frequently mutated in human cancers but its role in tumorigenesis remains incompletely defined. Using an induced pluripotent stem cell (iPSC) model of hereditary retinoblastoma (RB), we report that the spliceosome is an up-regulated target responding to oncogenic stress in RB1-mutant cells. By investigating transcriptomes and genome occupancies in RB iPSC­derived osteoblasts (OBs), we discover that both E2F3a, which mediates spliceosomal gene expression, and pRB, which antagonizes E2F3a, coregulate more than one-third of spliceosomal genes by cobinding to their promoters or enhancers. Pharmacological inhibition of the spliceosome in RB1-mutant cells leads to global intron retention, decreased cell proliferation, and impaired tumorigenesis. Tumor specimen studies and genome-wide TCGA (The Cancer Genome Atlas) expression profile analyses support the clinical relevance of pRB and E2F3a in modulating spliceosomal gene expression in multiple cancer types including osteosarcoma (OS). High levels of pRB/E2F3a­regulated spliceosomal genes are associated with poor OS patient survival. Collectively, these findings reveal an undiscovered connection between pRB, E2F3a, the spliceosome, and tumorigenesis, pointing to the spliceosomal machinery as a potentially widespread therapeutic vulnerability of pRB-deficient cancers.


Asunto(s)
Neoplasias Óseas , Carcinogénesis , Factor de Transcripción E2F3 , Regulación Neoplásica de la Expresión Génica , Células Madre Pluripotentes Inducidas , Osteosarcoma , Proteínas de Unión a Retinoblastoma , Empalmosomas , Ubiquitina-Proteína Ligasas , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Carcinogénesis/genética , Factor de Transcripción E2F3/genética , Factor de Transcripción E2F3/metabolismo , Genes de Retinoblastoma , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Mutación , Osteosarcoma/genética , Osteosarcoma/patología , Neoplasias de la Retina/genética , Retinoblastoma/genética , Proteínas de Unión a Retinoblastoma/genética , Proteínas de Unión a Retinoblastoma/metabolismo , Empalmosomas/genética , Empalmosomas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
11.
Genes Dev ; 31(18): 1847-1857, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-29021240

RESUMEN

TP53 is the most frequently mutated gene in human cancer. Many mutant p53 proteins exert oncogenic gain-of-function (GOF) properties that contribute to metastasis, but the mechanisms mediating these functions remain poorly defined in vivo. To elucidate how mutant p53 GOF drives metastasis, we developed a traceable somatic osteosarcoma mouse model that is initiated with either a single p53 mutation (p53R172H) or p53 loss in osteoblasts. Our study confirmed that p53 mutant mice developed osteosarcomas with increased metastasis as compared with p53-null mice. Comprehensive transcriptome RNA sequencing (RNA-seq) analysis of 16 tumors identified a cluster of small nucleolar RNAs (snoRNAs) that are highly up-regulated in p53 mutant tumors. Regulatory element analysis of these deregulated snoRNA genes identified strong enrichment of a common Ets2 transcription factor-binding site. Homozygous deletion of Ets2 in p53 mutant mice resulted in strong down-regulation of snoRNAs and reversed the prometastatic phenotype of mutant p53 but had no effect on osteosarcoma development, which remained 100% penetrant. In summary, our studies identify Ets2 inhibition as a potential therapeutic vulnerability in p53 mutant osteosarcomas.


Asunto(s)
Neoplasias Óseas/genética , Neoplasias Óseas/patología , Regulación Neoplásica de la Expresión Génica , Osteosarcoma/genética , Osteosarcoma/secundario , Proteína Proto-Oncogénica c-ets-2/genética , ARN Nucleolar Pequeño/genética , Proteína p53 Supresora de Tumor/genética , Animales , Regulación hacia Abajo , Perfilación de la Expresión Génica , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundario , Ratones , Ratones Noqueados , Mutación , Metástasis de la Neoplasia , Análisis de Secuencia por Matrices de Oligonucleótidos , Osteoblastos/metabolismo , Osteoblastos/patología , Regulación hacia Arriba
12.
Genes Chromosomes Cancer ; 63(5): e23241, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38738966

RESUMEN

Osteoblastomas (OBs) are benign neoplasms constituting approximately 1% of primary bone tumors with a predilection for the spine and sacrum. We describe an OB of the proximal phalanx of the left thumb in a 38-year-old female. MRI of left hand demonstrated a 29-mm mildly expansile enhancing lesion involving the entire proximal phalanx of the first digit. Histology displayed a bone-forming tumor consisting of trabeculae of remodeled woven bone framed by plump osteoblasts in a vascularized background. Next-generation sequencing analysis identified a PRSS44::ALK fusion gene.


Asunto(s)
Neoplasias Óseas , Osteoblastoma , Pulgar , Humanos , Femenino , Adulto , Pulgar/patología , Pulgar/anomalías , Osteoblastoma/genética , Osteoblastoma/patología , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Proteínas de Fusión Oncogénica/genética
13.
Genes Chromosomes Cancer ; 63(1): e23206, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37819540

RESUMEN

We present two cases of malignant ossifying fibromyxoid tumor (OFMT) which eluded diagnosis due to compelling clinicopathologic mimicry, compounded by similarly elusive underlying molecular drivers. The first is of a clavicle mass in a 69 year-old female, which histologically showed an infiltrative nested and trabeculated proliferation of monomorphic cells giving rise to scattered spicules of immature woven bone. Excepting SATB2 positivity, the lesion showed an inconclusive immunoprofile which along with negative PHF1 FISH led to an initial diagnosis of high-grade osteosarcoma. Next generation sequencing (NGS) revealed a particularly rare CREBBP::BCORL1 fusion. The second illustrates the peculiar presentation of a dural-based mass in a 52 year-old female who presented with neurologic dyscrasias. Sections showed a sheeted monotonous proliferation of ovoid to spindle cells, but in contrast to Case #1, the tumor contained an exuberance of reticular osteoid and woven bone deposition mimicking malignant osteogenic differentiation. NGS showed a novel CREBZF::PHF1 fusion. Both tumors recurred locally less than 1 year post-operatively. As such we reiterate that careful morphologic examination is axiomatic to any diagnosis in our discipline, but this paradigm must shift to recognize that molecular diagnostics can provide closure where traditional tools have notable limitations.


Asunto(s)
Neoplasias Óseas , Fibroma Osificante , Fibroma , Osteosarcoma , Sarcoma , Neoplasias de los Tejidos Blandos , Femenino , Humanos , Anciano , Persona de Mediana Edad , Proteínas de Unión al ADN , Fibroma Osificante/diagnóstico , Fibroma Osificante/genética , Fibroma Osificante/patología , Osteogénesis , Proteínas del Grupo Polycomb , Recurrencia Local de Neoplasia , Fibroma/patología , Osteosarcoma/diagnóstico , Osteosarcoma/genética , Neoplasias Óseas/diagnóstico , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Neoplasias de los Tejidos Blandos/patología , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico
14.
Genes Chromosomes Cancer ; 63(7): e23254, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38979775

RESUMEN

An aneurysmal bone cyst (ABC) is a benign bone neoplasm that typically occurs during the first and second decades of life. ABC usually presents as a rapidly growing intramedullary expansile mass with multiple blood-filled cysts in the metaphysis of the long tubular bones. Here, we report a case of a periosteal solid ABC that was initially diagnosed as a high-grade surface osteosarcoma. A 10-year-old male was referred to our hospital for swelling and tenderness of the left upper arm. Radiography revealed periosteal mass without fluid-fluid levels. On performing open biopsy, the tumor showed hypercellular proliferation of uniform spindle to epithelioid cells with brisk mitotic activity (up to 12/2 mm2) and lace-like osteoid formation, which was diagnosed as a high-grade surface osteosarcoma. After one course of chemotherapy using adriamycin and cisplatin, peripheral sclerosis was conspicuous, which led to pathological review and revision of diagnosis as "possibly osteoblastoma." The patient was disease-free for 4 years after marginal resection and curettage. Retrospective nanopore DNA sequencing unexpectedly detected a PAFAH1B1::USP6 rearrangement. The fusion gene was further validated using reverse transcription-polymerase chain reaction and the diagnosis was revised to ABC. Chromothripsis involving chromosome 17 has also been identified. Methylation analysis classified the present tumor as an ABC or non-ossifying fibroma using t-distributed stochastic neighbor embedding and unsupervised hierarchical clustering. This case report highlights the utility of nanopore DNA sequencing for soft tissue and bone tumor diagnosis.


Asunto(s)
Quistes Óseos Aneurismáticos , Cromotripsis , Secuenciación de Nanoporos , Osteosarcoma , Ubiquitina Tiolesterasa , Humanos , Masculino , Quistes Óseos Aneurismáticos/genética , Quistes Óseos Aneurismáticos/patología , Quistes Óseos Aneurismáticos/diagnóstico , Osteosarcoma/genética , Osteosarcoma/patología , Osteosarcoma/diagnóstico , Ubiquitina Tiolesterasa/genética , Niño , Secuenciación de Nanoporos/métodos , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Neoplasias Óseas/diagnóstico , Reordenamiento Génico
15.
J Cell Mol Med ; 28(11): e18462, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38847478

RESUMEN

Osteosarcoma (OS) is the most common primary malignant bone tumour in children and young adults. Account for 80% of all OS cases, conventional OS are characterized by the presence of osteoblastic, chondroblastic and fibroblastic cell types. Despite this heterogeneity, therapeutic treatment and prognosis of OS are essentially the same for all OS subtypes. Here, we report that DEC2, a transcriptional repressor, is expressed at higher levels in chondroblastic OS compared with osteoblastic OS. This difference suggests that DEC2 is disproportionately involved in the progression of chondroblastic OS, and thus, DEC2 may represent a possible molecular target for treating this type of OS. In the human chondroblastic-like OS cell line MNNG/HOS, we found that overexpression of DEC2 affects the proliferation of the cells by activating the VEGFC/VEGFR2 signalling pathway. Enhanced expression of DEC2 increased VEGFR2 expression, as well as increased the phosphorylation levels at sites Y951 and Y1175 of VEGFR2. On the one hand, activation of VEGFR2Y1175 enhanced cell proliferation through VEGFR2Y1175-PLCγ1-PKC-SPHK-MEK-ERK signalling. On the other hand, activation of VEGFR2Y951 decreased mitochondria-dependent apoptosis rate through VEGFR2Y951-VARP-PI3K-AKT signalling. Activation of these two signalling pathways resulted in enhanced progression of chondroblastic OS. In conclusion, DEC2 plays a pivotal role in cell proliferation and apoptosis-resistance in chondroblastic OS via the VEGFC/VEGFR2 signalling pathway. These findings lay the groundwork for developing focused treatments that target specific types of OS.


Asunto(s)
Neoplasias Óseas , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Osteosarcoma , Transducción de Señal , Factor C de Crecimiento Endotelial Vascular , Receptor 2 de Factores de Crecimiento Endotelial Vascular , Humanos , Osteosarcoma/metabolismo , Osteosarcoma/patología , Osteosarcoma/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Línea Celular Tumoral , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Neoplasias Óseas/genética , Factor C de Crecimiento Endotelial Vascular/metabolismo , Factor C de Crecimiento Endotelial Vascular/genética , Animales , Apoptosis/genética , Fosforilación
16.
J Cell Mol Med ; 28(9): e18286, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38742843

RESUMEN

Osteosarcoma, the primary bone cancer in adolescents and young adults, is notorious for its aggressive growth and metastatic potential. Our study delved into the prognostic impact of inflammasome-related gene signatures in osteosarcoma patients, employing comprehensive genetic profiling to uncover signatures linked with patient outcomes. We identified three patient subgroups through consensus clustering, with one showing worse survival rates correlated with high FGFR3 and RARB expressions. Immune profiling revealed significant immune cell infiltration differences among these subgroups, affecting survival. Utilising advanced machine learning, including StepCox and gradient boosting machine algorithms, we developed a prognostic model with a notable c-index of 0.706, highlighting CD36 and MYD88 as key genes. Higher inflammasome risk scores from our model were associated with poorer survival, corroborated across datasets. In vitro experiments validated CD36 and MYD88's roles in promoting osteosarcoma cell proliferation, invasion and migration, emphasising their therapeutic potential. This research offers new insights into inflammasomes' role in osteosarcoma, introducing novel biomarkers for risk assessment and potential therapeutic targets. Our findings suggest a pathway towards personalised treatment strategies, potentially improving patient outcomes in osteosarcoma.


Asunto(s)
Biomarcadores de Tumor , Neoplasias Óseas , Regulación Neoplásica de la Expresión Génica , Inflamasomas , Osteosarcoma , Humanos , Osteosarcoma/genética , Osteosarcoma/patología , Osteosarcoma/inmunología , Osteosarcoma/mortalidad , Inflamasomas/metabolismo , Inflamasomas/genética , Biomarcadores de Tumor/genética , Pronóstico , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Neoplasias Óseas/mortalidad , Neoplasias Óseas/inmunología , Neoplasias Óseas/diagnóstico , Perfilación de la Expresión Génica , Femenino , Masculino , Transcriptoma/genética , Línea Celular Tumoral , Proliferación Celular/genética , Adolescente , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo
17.
J Cell Mol Med ; 28(10): e18400, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38780513

RESUMEN

Osteosarcoma is the most common primary bone malignancy in children and adolescents. Overexpression of polo-like kinase 1 (PLK1) is frequent in osteosarcoma and drives disease progression and metastasis, making it a promising therapeutic target. In this study, we explored PLK1 knockdown in osteosarcoma cells using RNA interference mediated by high-fidelity Cas13d (hfCas13d). PLK1 was found to be significantly upregulated in osteosarcoma tumour tissues compared to normal bone. sgRNA-mediated PLK1 suppression via hfCas13d transfection inhibited osteosarcoma cell proliferation, induced G2/M cell cycle arrest, promoted apoptosis, reduced cell invasion and increased expression of the epithelial marker E-cadherin. Proximity labelling by TurboID coupled with co-immunoprecipitation identified novel PLK1 interactions with Smad3, a key intracellular transducer of TGF-ß signalling. PLK1 knockdown impaired Smad2/3 phosphorylation and modulated TGF-ß/Smad3 pathway inactivation. Finally, in vivo delivery of hfCas13d vectors targeting PLK1 substantially attenuated osteosarcoma xenograft growth in nude mice. Taken together, this study highlights PLK1 as a potential therapeutic target and driver of disease progression in osteosarcoma. It also demonstrates the utility of hfCas13d-mediated gene knockdown as a strategy for targeted therapy. Further optimization of PLK1 suppression approaches may ultimately improve clinical outcomes for osteosarcoma patients.


Asunto(s)
Apoptosis , Proteínas de Ciclo Celular , Proliferación Celular , Ratones Desnudos , Osteosarcoma , Quinasa Tipo Polo 1 , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas , Interferencia de ARN , Transducción de Señal , Proteína smad3 , Factor de Crecimiento Transformador beta , Osteosarcoma/patología , Osteosarcoma/genética , Osteosarcoma/metabolismo , Humanos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Animales , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteína smad3/metabolismo , Proteína smad3/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Factor de Crecimiento Transformador beta/metabolismo , Ratones , Apoptosis/genética , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Neoplasias Óseas/metabolismo , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Ensayos Antitumor por Modelo de Xenoinjerto , Femenino
18.
J Cell Mol Med ; 28(10): e18395, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38774995

RESUMEN

Tumour-associated macrophages (TAMs), encompassing M1 and M2 subtypes, exert significant effects on osteosarcoma (OS) progression and immunosuppression. However, the impacts of TAM-derived biomarkers on the progression of OS remains limited. The GSE162454 profile was subjected to single-cell RNA (scRNA) sequencing analysis to identify crucial mediators between TAMs and OS cells. The clinical features, effects and mechanisms of these mediators on OS cells and tumour microenvironment were evaluated via biological function experiments and molecular biology experiments. Phosphodiesterase 4C (PDE4C) was identified as a pivotal mediator in the communication between M2 macrophages and OS cells. Elevated levels of PDE4C were detected in OS tissues, concomitant with M2 macrophage level, unfavourable prognosis and metastasis. The expression of PDE4C was observed to increase during the conversion process of THP-1 cells to M2 macrophages, which transferred the PDE4C mRNA to OS cells through exosome approach. PDE4C increased OS cell proliferation and mobility via upregulating the expression of collagens. Furthermore, a positive correlation was observed between elevated levels of PDE4C and increased TIDE score, decreased response rate following immune checkpoint therapy, reduced TMB and diminished PDL1 expression. Collectively, PDE4C derived from M2 macrophages has the potential to enhance the proliferation and mobility of OS cells by augmenting collagen expression. PDE4C may serve as a valuable biomarker for prognosticating patient outcomes and response rates following immunotherapy.


Asunto(s)
Neoplasias Óseas , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4 , Inmunoterapia , Macrófagos , Osteosarcoma , Microambiente Tumoral , Humanos , Biomarcadores de Tumor/metabolismo , Biomarcadores de Tumor/genética , Neoplasias Óseas/inmunología , Neoplasias Óseas/patología , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/genética , Regulación Neoplásica de la Expresión Génica , Inmunoterapia/métodos , Macrófagos/metabolismo , Macrófagos/inmunología , Metástasis de la Neoplasia , Osteosarcoma/patología , Osteosarcoma/inmunología , Osteosarcoma/genética , Osteosarcoma/metabolismo , Osteosarcoma/terapia , Pronóstico , Microambiente Tumoral/inmunología , Macrófagos Asociados a Tumores/inmunología , Macrófagos Asociados a Tumores/metabolismo
19.
J Cell Physiol ; 239(3): e31068, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37357526

RESUMEN

N6-methyladenosine (m6 A) is one of the main epitranscriptomic modifications that accelerates the progression of malignant tumors by modifying RNA. Methyltransferase-like 16 (METTL16) is a newly identified methyltransferase that has been found to play an important oncogenic role in a few malignancies; however, its function in osteosarcoma (OS) remains unclear. In this study, METTL16 was found to be upregulated in OS tissues, and associated with poor prognosis in OS patients. Functionally, METTL16 substantially promoted OS cell proliferation, migration, and invasion in vitro and OS growth in vivo. Mechanistically, vacuolar protein sorting protein 33b (VPS33B) was identified as the downstream target of METTL16, which induced m6 A modification of VPS33B and impaired the stability of the VPS33B transcript, thereby degrading VPS33B. In addition, VPS33B was found to be downregulated in OS tissues, VPS33B knockdown markedly attenuated shMETTL16-mediated inhibition on OS progression. Finally, METTL16/VPS33B might facilitate OS progression through PI3K/AKT pathway. In summary, this study revealed an important role for the METTL16-mediated m6 A modification in OS progression, implying it as a promising target for OS treatment.


Asunto(s)
Adenosina , Neoplasias Óseas , Metiltransferasas , Osteosarcoma , Fosfatidilinositol 3-Quinasas , Proteínas de Transporte Vesicular , Humanos , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Metiltransferasas/genética , Metiltransferasas/metabolismo , Osteosarcoma/genética , Osteosarcoma/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Transporte de Proteínas , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo , Adenosina/análogos & derivados , Adenosina/metabolismo , Línea Celular Tumoral
20.
J Cell Physiol ; 239(5): e31256, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38591855

RESUMEN

Osteosarcoma (OS) cancer treatments include systemic chemotherapy and surgical resection. In the last years, novel treatment approaches have been proposed, which employ a drug-delivery system to prevent offside effects and improves treatment efficacy. Locally delivering anticancer compounds improves on high local concentrations with more efficient tumour-killing effect, reduced drugs resistance and confined systemic effects. Here, the synthesis of injectable strontium-doped calcium phosphate (SrCPC) scaffold was proposed as drug delivery system to combine bone tissue regeneration and anticancer treatment by controlled release of methotrexate (MTX) and doxorubicin (DOX), coded as SrCPC-MTX and SrCPC-DOX, respectively. The drug-loaded cements were tested in an in vitro model of human OS cell line SAOS-2, engineered OS cell line (SAOS-2-eGFP) and U2-OS. The ability of doped scaffolds to induce OS cell death and apoptosis was assessed analysing cell proliferation and Caspase-3/7 activities, respectively. To determine if OS cells grown on doped-scaffolds change their migratory ability and invasiveness, a wound-healing assay was performed. In addition, the osteogenic potential of SrCPC material was evaluated using human adipose derived-mesenchymal stem cells. Osteogenic markers such as (i) the mineral matrix deposition was analysed by alizarin red staining; (ii) the osteocalcin (OCN) protein expression was investigated by enzyme-linked immunosorbent assay test, and (iii) the osteogenic process was studied by real-time polymerase chain reaction array. The delivery system induced cell-killing cytotoxic effects and apoptosis in OS cell lines up to Day 7. SrCPC demonstrates a good cytocompatibility and it induced upregulation of osteogenic genes involved in the skeletal development pathway, together with OCN protein expression and mineral matrix deposition. The proposed approach, based on the local, sustained release of anticancer drugs from nanostructured biomimetic drug-loaded cements is promising for future therapies aiming to combine bone regeneration and anticancer local therapy.


Asunto(s)
Antineoplásicos , Apoptosis , Neoplasias Óseas , Fosfatos de Calcio , Doxorrubicina , Metotrexato , Osteogénesis , Osteosarcoma , Andamios del Tejido , Humanos , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/patología , Fosfatos de Calcio/administración & dosificación , Fosfatos de Calcio/química , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Doxorrubicina/administración & dosificación , Doxorrubicina/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/patología , Osteosarcoma/metabolismo , Estroncio/farmacología , Estroncio/química , Andamios del Tejido/química , Sistemas de Liberación de Medicamentos , Metotrexato/administración & dosificación , Metotrexato/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA