Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
J Hypertens ; 42(1): 118-128, 2024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-37711097

RESUMEN

BACKGROUND: The stroke-prone spontaneously hypertensive rat (SHRSP) is a genetic model for cerebral stroke. Although a recent study on a congenic SHRSP suggested that a nonsense mutation in stromal interaction molecule 1 ( Stim1 ) encoding a major component of store-operated Ca 2+ entry was a causal variant for stroke in SHRSP, this was not conclusive because the congenic region including Stim1 in that rat was too wide. On the other hand, we demonstrated that the Wistar-Kyoto (WKY)-derived congenic fragment adjacent to Stim1 exacerbated stroke susceptibility in a congenic SHRSP called SPwch1.71. In the present study, we directly examined the effects of the Stim1 genotype on stroke susceptibility using SHRSP in which wild-type Stim1 was knocked in (called Stim1 -KI SHRSP). The combined effects of Stim1 and the congenic fragment of SPwch1.71 were also investigated. METHODS: Stroke susceptibility was assessed by the stroke symptom-free and survival periods based on observations of behavioral symptoms and reductions in body weight. RESULTS: Stim1 -KI SHRSP was more resistant to, while SPwch1.71 was more susceptible to stroke than the original SHRSP. Introgression of the wild-type Stim1 of Stim1 -KI SHRSP into SPwch1.71 by the generation of F1 rats ameliorated stroke susceptibility in SPwch1.71. Gene expression, whole-genome sequencing, and biochemical analyses identified Art2b , Folr1 , and Pde2a as possible candidate genes accelerating stroke in SPwch1.71. CONCLUSION: The substitution of SHRSP-type Stim1 to wild-type Stim1 ameliorated stroke susceptibility in both SHRSP and SPwch1.71, indicating that the nonsense mutation in Stim1 is causally related to stroke susceptibility in SHRSP.


Asunto(s)
Hipertensión , Accidente Cerebrovascular , Humanos , Ratas , Animales , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Hipertensión/complicaciones , Hipertensión/genética , Hipertensión/metabolismo , Cromosomas Humanos Par 1/metabolismo , Molécula de Interacción Estromal 1/genética , Codón sin Sentido , Genotipo , Accidente Cerebrovascular/etiología
2.
Theor Appl Genet ; 137(1): 2, 2023 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-38072878

RESUMEN

KEY MESSAGE: Two wheat-Ae. longissima translocation chromosomes (1BS·1SlL and 1SlS·1BL) were transferred into three commercial wheat varieties, and the new advanced lines showed improved bread-making quality compared to their recurrent parents. Aegilops longissima chromosome 1Sl encodes specific types of gluten subunits that may positively affect wheat bread-making quality. The most effective method of introducing 1Sl chromosomal fragments containing the target genes into wheat is chromosome translocation. Here, a wheat-Ae. longissima 1BS·1SlL translocation line was developed using molecular marker-assisted chromosome engineering. Two types of translocation chromosomes developed in a previous study, 1BS·1SlL and 1SlS·1BL, were introduced into three commercial wheat varieties (Ningchun4, Ningchun50, and Westonia) via backcrossing with marker-assisted selection. Advanced translocation lines were confirmed through chromosome in situ hybridization and genotyping by target sequencing using the wheat 40 K system. Bread-making quality was found to be improved in the two types of advanced translocation lines compared to the corresponding recurrent parents. Furthermore, 1SlS·1BL translocation lines displayed better bread-making quality than 1BS·1SlL translocation lines in each genetic background. Further analysis revealed that high molecular weight glutenin subunit (HMW-GS) contents and expression levels of genes encoding low molecular weight glutenin subunits (LMW-GSs) were increased in 1SlS·1BL translocation lines. Gliadin and gluten-related transcription factors were also upregulated in the grains of the two types of advanced translocation lines compared to the recurrent parents. This study clarifies the impacts of specific glutenin subunits on bread-making quality and provides novel germplasm resources for further improvement of wheat quality through molecular breeding.


Asunto(s)
Aegilops , Triticum , Humanos , Triticum/genética , Triticum/metabolismo , Aegilops/genética , Aegilops/metabolismo , Translocación Genética , Pan/análisis , Cromosomas Humanos Par 1/metabolismo , Glútenes/genética , Glútenes/metabolismo
3.
Virchows Arch ; 483(6): 821-833, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37938323

RESUMEN

In the spectrum of oncocytic renal neoplasms, a subset of tumors with high-grade-appearing histologic features harboring pathogenic mutations in mammalian target of rapamycin (mTOR) and hitherto clinical indolent behavior has been described. Three cases (2F,1 M) with histologically documented metastases (lymph node, skull, and liver) were retrieved and extensively investigated by immunohistochemistry, FISH, and next-generation sequencing. Tumors were composed of eosinophilic cells with prominent nucleoli (G3 by ISUP/WHO) arranged in solid to nested architecture. Additionally, there were larger cells with perinuclear cytoplasmic shrinkage and sparse basophilic Nissl-like granules, superficially resembling the so-called spider cells of cardiac rhabdomyomas. The renal tumors, including the skull and liver metastases, showed immunoexpression PAX8, CK8-18, and cathepsin-K, and negativity for vimentin. NGS identified mTOR genetic alterations in the three cases, including the skull and liver metastases. One patient was then treated with Everolimus (mTOR inhibitors) with clinical response (metastatic tumor shrinkage). We present a distinct renal tumor characterized by high-grade eosinophilic cells, cathepsin-K immunohistochemical expression, and harboring mTOR gene mutations demonstrating a malignant potential and showing responsiveness to mTOR inhibitors.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Neoplasias Hepáticas , Humanos , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Cromosomas Humanos Par 1/metabolismo , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Inhibidores mTOR , Mutación , Serina-Treonina Quinasas TOR/genética
4.
Bioinformatics ; 39(1)2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36637205

RESUMEN

MOTIVATION: Many studies have shown that IDH mutation and 1p/19q co-deletion can serve as prognostic signatures of glioma. Although these genetic variations affect the expression of one or more genes, the prognostic value of gene expression related to IDH and 1p/19q status is still unclear. RESULTS: We constructed an ensemble gene pair signature for the risk evaluation and survival prediction of glioma based on the prior knowledge of the IDH and 1p/19q status. First, we separately built two gene pair signatures IDH-GPS and 1p/19q-GPS and elucidated that they were useful transcriptome markers projecting from corresponding genome variations. Then, the gene pairs in these two models were assembled to develop an integrated model named Glioma Prognostic Gene Pair Signature (GPGPS), which demonstrated high area under the curves (AUCs) to predict 1-, 3- and 5-year overall survival (0.92, 0.88 and 0.80) of glioma. GPGPS was superior to the single GPSs and other existing prognostic signatures (avg AUC = 0.70, concordance index = 0.74). In conclusion, the ensemble prognostic signature with 10 gene pairs could serve as an independent predictor for risk stratification and survival prediction in glioma. This study shed light on transferring knowledge from genetic alterations to expression changes to facilitate prognostic studies. AVAILABILITY AND IMPLEMENTATION: Codes are available at https://github.com/Kimxbzheng/GPGPS.git. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Neoplasias Encefálicas , Glioma , Humanos , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Pronóstico , Glioma/genética , Aberraciones Cromosómicas , Mutación , Cromosomas Humanos Par 1/genética , Cromosomas Humanos Par 1/metabolismo
5.
EBioMedicine ; 87: 104410, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36525723

RESUMEN

BACKGROUND: Human oligodendroglioma presents as a heterogeneous disease, primarily characterized by the isocitrate dehydrogenase (IDH) mutation and 1p/19q co-deletion. Therapy development for this tumor is hindered by incomplete knowledge of somatic driving alterations and suboptimal disease classification. We herein aim to identify intrinsic molecular subtypes through integrated analysis of transcriptome, genome and methylome. METHODS: 137 oligodendroglioma patients from the Cancer Genome Atlas (TCGA) dataset were collected for unsupervised clustering analysis of immune gene expression profiles and comparative analysis of genome and methylome. Two independent datasets containing 218 patients were used for validation. FINDINGS: We identified and independently validated two reproducible subtypes associated with distinct molecular characteristics and clinical outcomes. The proliferative subtype, named Oligo1, was characterized by more tumors of CNS WHO grade 3, as well as worse prognosis compared to the Oligo2 subtype. Besides the clinicopathologic features, Oligo1 exhibited enrichment of cell proliferation, regulation of cell cycle and Wnt signaling pathways, and significantly altered genes, such as EGFR, NOTCH1 and MET. In contrast, Oligo2, with favorable outcome, presented increased activation of immune response and metabolic process. Higher T cell/APC co-inhibition and inhibitory checkpoint levels were observed in Oligo2 tumors. Finally, multivariable analysis revealed our classification was an independent prognostic factor in oligodendrogliomas, and the robustness of these molecular subgroups was verified in the validation cohorts. INTERPRETATION: This study provides further insights into patient stratification as well as presents opportunities for therapeutic development in human oligodendrogliomas. FUNDING: The funders are listed in the Acknowledgement.


Asunto(s)
Neoplasias Encefálicas , Oligodendroglioma , Humanos , Oligodendroglioma/genética , Oligodendroglioma/metabolismo , Oligodendroglioma/patología , Neoplasias Encefálicas/patología , Mutación , Aberraciones Cromosómicas , Transcriptoma , Pronóstico , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/metabolismo , Cromosomas Humanos Par 1/metabolismo
6.
Blood ; 139(13): 1939-1953, 2022 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-35015835

RESUMEN

Understanding the biological and clinical impact of copy number aberrations (CNAs) on the development of precision therapies in cancer remains an unmet challenge. Genetic amplification of chromosome 1q (chr1q-amp) is a major CNA conferring an adverse prognosis in several types of cancer, including in the blood cancer multiple myeloma (MM). Although several genes across chromosome 1 (chr1q) portend high-risk MM disease, the underpinning molecular etiology remains elusive. Here, with reference to the 3-dimensional (3D) chromatin structure, we integrate multi-omics data sets from patients with MM with genetic variables to obtain an associated clinical risk map across chr1q and to identify 103 adverse prognosis genes in chr1q-amp MM. Prominent among these genes, the transcription factor PBX1 is ectopically expressed by genetic amplification and epigenetic activation of its own preserved 3D regulatory domain. By binding to reprogrammed superenhancers, PBX1 directly regulates critical oncogenic pathways and a FOXM1-dependent transcriptional program. Together, PBX1 and FOXM1 activate a proliferative gene signature that predicts adverse prognosis across multiple types of cancer. Notably, pharmacological disruption of the PBX1-FOXM1 axis with existing agents (thiostrepton) and a novel PBX1 small molecule inhibitor (T417) is selectively toxic against chr1q-amp myeloma and solid tumor cells. Overall, our systems medicine approach successfully identifies CNA-driven oncogenic circuitries, links them to clinical phenotypes, and proposes novel CNA-targeted therapy strategies in MM and other types of cancer.


Asunto(s)
Mieloma Múltiple , Cromosomas Humanos Par 1/metabolismo , Proteína Forkhead Box M1/genética , Humanos , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Factor de Transcripción 1 de la Leucemia de Células Pre-B/genética , Pronóstico , Análisis de Sistemas , Factores de Transcripción/genética
7.
Cells ; 10(6)2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34205916

RESUMEN

Multiple myeloma (MM) is characterized by an accumulation of malignant plasma cells (PCs) in the bone marrow (BM). The amplification of 1q21 is one of the most common cytogenetic abnormalities occurring in around 40% of de novo patients and 70% of relapsed/refractory MM. Patients with this unfavorable cytogenetic abnormality are considered to be high risk with a poor response to standard therapies. The gene(s) driving amplification of the 1q21 amplicon has not been fully studied. A number of clear candidates are under investigation, and some of them (IL6R, ILF2, MCL-1, CKS1B and BCL9) have been recently proposed to be potential drivers of this region. However, much remains to be learned about the biology of the genes driving the disease progression in MM patients with 1q21 amp. Understanding the mechanisms of these genes is important for the development of effective targeted therapeutic approaches to treat these patients for whom effective therapies are currently lacking. In this paper, we review the current knowledge about the pathological features, the mechanism of 1q21 amplification, and the signal pathway of the most relevant candidate genes that have been suggested as possible therapeutic targets for the 1q21 amplicon.


Asunto(s)
Cromosomas Humanos Par 1 , Amplificación de Genes , Mieloma Múltiple , Proteínas de Neoplasias , Transducción de Señal , Cromosomas Humanos Par 1/genética , Cromosomas Humanos Par 1/metabolismo , Humanos , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Mieloma Múltiple/terapia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo
8.
Adv Anat Pathol ; 28(4): 251-257, 2021 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-34009776

RESUMEN

Recent advances in molecular genetics have expanded our knowledge of renal tumors and enabled a better classification. These studies have revealed that renal tumors with predominantly "eosinophilic/oncocytic" cytoplasm include several novel biological subtypes beyond the traditionally well-recognized renal oncocytoma and an eosinophilic variant of chromophobe renal cell carcinoma. Herein, we present a comprehensive review of the eosinophilic vacuolated tumor (EVT) building upon a case report including radiology, histopathology, electron microscopy, and next-generation sequencing. EVTs are characterized by mTORC1 activation. We speculate that loss of chromosome 1 in EVT with MTOR mutation may be driven in part by an advantage conferred by loss of the remaining MTOR wild-type allele. mTORC1 is best known for its role in promoting protein translation and it is interesting that dilated cisterns of rough endoplasmic reticulum (ER) likely account for the cytoplasmic vacuoles seen by light microscopy. We present an integrated view of EVT as well as cues that can assist in the differential diagnosis.


Asunto(s)
Carcinoma de Células Renales/patología , Aberraciones Cromosómicas , Cromosomas Humanos Par 1/genética , Neoplasias Renales/patología , Mutación , Serina-Treonina Quinasas TOR/genética , Biomarcadores de Tumor , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Cromosomas Humanos Par 1/metabolismo , Humanos , Riñón/metabolismo , Riñón/patología , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Masculino , Persona de Mediana Edad , Serina-Treonina Quinasas TOR/metabolismo
9.
Hum Mol Genet ; 30(7): 595-602, 2021 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-33772547

RESUMEN

Deletions of chromosome 1p36 are the most common telomeric deletions in humans and are associated with an increased risk of orofacial clefting. Deletion/phenotype mapping, combined with data from human and mouse studies, suggests the existence of multiple 1p36 genes associated with orofacial clefting including SKI, PRDM16, PAX7 and GRHL3. The arginine-glutamic acid dipeptide (RE) repeats gene (RERE) is located in the proximal critical region for 1p36 deletion syndrome and encodes a nuclear receptor co-regulator. Pathogenic RERE variants have been shown to cause neurodevelopmental disorder with or without anomalies of the brain, eye or heart (NEDBEH). Cleft lip has previously been described in one individual with NEDBEH. Here we report the first individual with NEDBEH to have a cleft palate. We confirm that RERE is broadly expressed in the palate during mouse embryonic development, and we demonstrate that the majority of RERE-deficient mouse embryos on C57BL/6 background have cleft palate. We go on to show that ablation of Rere in cranial neural crest (CNC) cells, mediated by a Wnt1-Cre, leads to delayed elevation of the palatal shelves and cleft palate and that proliferation of mesenchymal cells in the palatal shelves is significantly reduced in Rereflox/flox; Wnt1-Cre embryos. We conclude that loss of RERE function contributes to the development of orofacial clefts in individuals with proximal 1p36 deletions and NEDBEH and that RERE expression in CNC cells and their derivatives is required for normal palatal development.


Asunto(s)
Trastornos de los Cromosomas/genética , Labio Leporino/genética , Fisura del Paladar/genética , Modelos Animales de Enfermedad , Desarrollo Embrionario/genética , Proteínas del Tejido Nervioso/genética , Proteínas Represoras/genética , Animales , Proliferación Celular/genética , Deleción Cromosómica , Trastornos de los Cromosomas/metabolismo , Cromosomas Humanos Par 1/genética , Cromosomas Humanos Par 1/metabolismo , Labio Leporino/embriología , Labio Leporino/metabolismo , Fisura del Paladar/embriología , Fisura del Paladar/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Mesodermo/citología , Mesodermo/embriología , Mesodermo/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas del Tejido Nervioso/deficiencia , Cresta Neural/embriología , Cresta Neural/metabolismo , Fenotipo , Proteínas Represoras/deficiencia , Proteína Wnt1/genética , Proteína Wnt1/metabolismo
10.
J Mol Biol ; 433(6): 166700, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33160979

RESUMEN

Significant efforts have been recently made to obtain the three-dimensional (3D) structure of the genome with the goal of understanding how structures may affect gene regulation and expression. Chromosome conformational capture techniques such as Hi-C, have been key in uncovering the quantitative information needed to determine chromatin organization. Complementing these experimental tools, co-polymers theoretical methods are necessary to determine the ensemble of three-dimensional structures associated to the experimental data provided by Hi-C maps. Going beyond just structural information, these theoretical advances also start to provide an understanding of the underlying mechanisms governing genome assembly and function. Recent theoretical work, however, has been focused on single chromosome structures, missing the fact that, in the full nucleus, interactions between chromosomes play a central role in their organization. To overcome this limitation, MiChroM (Minimal Chromatin Model) has been modified to become capable of performing these multi-chromosome simulations. It has been upgraded into a fast and scalable software version, which is able to perform chromosome simulations using GPUs via OpenMM Python API, called Open-MiChroM. To validate the efficiency of this new version, analyses for GM12878 individual autosomes were performed and compared to earlier studies. This validation was followed by multi-chain simulations including the four largest human chromosomes (C1-C4). These simulations demonstrated the full power of this new approach. Comparison to Hi-C data shows that these multiple chromosome interactions are essential for a more accurate agreement with experimental results. Without any changes to the original MiChroM potential, it is now possible to predict experimentally observed inter-chromosome contacts. This scalability of Open-MiChroM allow for more audacious investigations, looking at interactions of multiple chains as well as moving towards higher resolution chromosomes models.


Asunto(s)
Cromatina/química , Cromosomas Humanos Par 1/química , Cromosomas Humanos Par 2/química , Cromosomas Humanos Par 3/química , Cromosomas Humanos Par 4/química , Simulación de Dinámica Molecular , Programas Informáticos , Animales , Línea Celular Tumoral , Cromatina/metabolismo , Cromatina/ultraestructura , Cromosomas Humanos Par 1/metabolismo , Cromosomas Humanos Par 1/ultraestructura , Cromosomas Humanos Par 2/metabolismo , Cromosomas Humanos Par 2/ultraestructura , Cromosomas Humanos Par 3/metabolismo , Cromosomas Humanos Par 3/ultraestructura , Cromosomas Humanos Par 4/metabolismo , Cromosomas Humanos Par 4/ultraestructura , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Humanos , Linfocitos/citología , Linfocitos/metabolismo , Saccharum/genética , Saccharum/metabolismo , Termodinámica , Triticum/genética , Triticum/metabolismo
11.
Diabetes ; 69(10): 2206-2216, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32651240

RESUMEN

A chromosome 1q25 variant (rs10911021) has been associated with coronary heart disease (CHD) in type 2 diabetes. In human umbilical vein endothelial cells (HUVECs), the risk allele "C" is associated with lower expression of the adjacent gene GLUL encoding glutamine synthase, converting glutamic acid to glutamine. To further investigate the mechanisms through which this locus affects CHD risk, we measured 35 intracellular metabolites involved in glutamic acid metabolism and the γ-glutamyl cycle in 62 HUVEC strains carrying different rs10911021 genotypes. Eight metabolites were positively associated with the risk allele (17-58% increase/allele copy, P = 0.046-0.002), including five γ-glutamyl amino acids, ß-citryl-glutamate, N-acetyl-aspartyl-glutamate, and ophthalmate-a marker of γ-glutamyl cycle malfunction. Consistent with these findings, the risk allele was also associated with decreased glutathione-to-glutamate ratio (-9%, P = 0.012), decreased S-lactoylglutathione (-41%, P = 0.019), and reduced detoxification of the atherogenic compound methylglyoxal (+54%, P = 0.008). GLUL downregulation by shRNA caused a 40% increase in the methylglyoxal level, which was completely prevented by glutamine supplementation. In summary, we have identified intracellular metabolic traits associated with the 1q25 risk allele in HUVECs, including impairments of the γ-glutamyl cycle and methylglyoxal detoxification. Glutamine supplementation abolishes the latter abnormality, suggesting that such treatment may prevent CHD in 1q25 risk allele carriers.


Asunto(s)
Enfermedad Coronaria/metabolismo , Células Endoteliales/metabolismo , Cromosomas Humanos Par 1/metabolismo , Enfermedad Coronaria/genética , Dipéptidos , Endoftalmitis/metabolismo , Glutamato-Amoníaco Ligasa/metabolismo , Glutamatos/metabolismo , Glutamina/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Piruvaldehído/metabolismo , ARN Interferente Pequeño/metabolismo
12.
Pediatr Blood Cancer ; 67(9): e28426, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32614133

RESUMEN

BACKGROUND: Relapse occurs in 50% of pediatric ependymoma cases and has poor prognosis. Few studies have investigated the clinical progress of relapsed disease, and treatment lacks a standardized approach. METHODS AND MATERIALS: We analyzed 302 pediatric ependymoma cases. Tumor, demographic, and treatment variables were investigated for association with relapse risk, time to recurrence, and survival after relapse. DNA methylation profiling was performed for 135/302 cases, and predominant subgroups were EPN_PFA (n = 95) and EPN_RELA (n = 24). Chromosome 1q status was ascertained for 185/302 cases by fluorescent in-situ hybridization (FISH), multiplex ligation-dependent probe amplification (MLPA), and DNA methylation profiles. RESULTS: Sixty-two percent of cases relapsed, with a median of two recurrences with no difference between posterior fossa and supratentorial locations (66% vs 55% relapse rate). One hundred seventeen (38%) cases relapsed within two years and five (2%) beyond 10 years. The late relapses were clinically heterogeneous. Tumor grade and treatment affected risk and time to relapse variably across subgroups. After relapse, surgery and irradiation delayed disease progression with a minimal impact on survival across the entire cohort. In the EPN_PFA and EPN_RELA groups, 1q gain was independently associated with relapse risk (subhazard ratio [SHR] 4.307, P = 0.027 and SHR 1.982, P = 0.010, respectively) while EPN_PFA had increased relapse risk compared with EPN_RELA (SHR = 0.394, P = 0.018). CONCLUSIONS: Recurrent pediatric ependymoma is an aggressive disease with poor outcomes, for which current treatments are inadequate. We report that chromosome 1q gain increases relapse risk in common molecular subgroups in children but a deeper understanding of the underlying biology at relapse and novel therapeutic approaches are urgently needed.


Asunto(s)
Neoplasias Encefálicas , Cromosomas Humanos Par 1 , Metilación de ADN , ADN de Neoplasias , Ependimoma , Recurrencia Local de Neoplasia , Adolescente , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/terapia , Niño , Preescolar , Cromosomas Humanos Par 1/genética , Cromosomas Humanos Par 1/metabolismo , ADN de Neoplasias/genética , ADN de Neoplasias/metabolismo , Ependimoma/genética , Ependimoma/metabolismo , Ependimoma/mortalidad , Ependimoma/terapia , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/mortalidad , Recurrencia Local de Neoplasia/terapia , Estudios Retrospectivos , Factores de Riesgo
13.
Br J Haematol ; 190(6): e329-e332, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32572949
14.
Sci Transl Med ; 12(527)2020 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-31969487

RESUMEN

Heightened secretion of protumorigenic effector proteins is a feature of malignant cells. Yet, the molecular underpinnings and therapeutic implications of this feature remain unclear. Here, we identify a chromosome 1q region that is frequently amplified in diverse cancer types and encodes multiple regulators of secretory vesicle biogenesis and trafficking, including the Golgi-dedicated enzyme phosphatidylinositol (PI)-4-kinase IIIß (PI4KIIIß). Molecular, biochemical, and cell biological studies show that PI4KIIIß-derived PI-4-phosphate (PI4P) synthesis enhances secretion and accelerates lung adenocarcinoma progression by activating Golgi phosphoprotein 3 (GOLPH3)-dependent vesicular release from the Golgi. PI4KIIIß-dependent secreted factors maintain 1q-amplified cancer cell survival and influence prometastatic processes in the tumor microenvironment. Disruption of this functional circuitry in 1q-amplified cancer cells with selective PI4KIIIß antagonists induces apoptosis and suppresses tumor growth and metastasis. These results support a model in which chromosome 1q amplifications create a dependency on PI4KIIIß-dependent secretion for cancer cell survival and tumor progression.


Asunto(s)
Adenocarcinoma del Pulmón/metabolismo , Cromosomas Humanos Par 1/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Adenocarcinoma del Pulmón/genética , Animales , Cromosomas Humanos Par 1/genética , Ensayo de Inmunoadsorción Enzimática , Aparato de Golgi/metabolismo , Humanos , Técnicas In Vitro , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Microtomografía por Rayos X
15.
Taiwan J Obstet Gynecol ; 58(6): 859-863, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31759543

RESUMEN

OBJECTIVE: We present detection of a familial 1q21.1 microdeletion and concomitant CHD1L mutation in a fetus with oligohydramnios and bilateral renal dysplasia on prenatal ultrasound. CASE REPORT: A 37-year-old, primigravid woman was referred for level II ultrasound examination at 16 weeks of gestation because of oligohydramnios. The parents were phenotypically normal, and there were no congenital malformations in the family. Prenatal ultrasound at 17 weeks of gestation revealed a fetus with fetal growth biometry equivalent to 16 weeks, oligohydramnios with an amniotic fluid index (AFI) of 1.4 cm and bilateral renal dysplasia without sonographic demonstration of bilateral renal arteries. The pregnancy was subsequently terminated, and a 137-g fetus was delivered without characteristic facial dysmorphism. Postnatal cytogenetic analysis of the umbilical cord and parental bloods revealed normal karyotypes. However, array comparative genomic hybridization (aCGH) analysis on the DNA extracted from the umbilical cord revealed a 2.038-Mb microdeletion of 1q21.1-q21.2 encompassing 11 [Online Mendelian Inheritance in Man (OMIM)] genes of PRKAB2, FMO5, CHD1L, BCL9, ACP6, GJA5, GJA8, GPR89B, NBPF14, TRN-GTT2-1 and NBPF20. The mother was found to carry the same microdeletion. A missense mutation of c.2353T > G, p.Ser785Ala in CHD1L was detected in the umbilical cord. The father was found to carry a heterozygous mutation of c.2353T > G, p.Ser785Ala in CHD1L. CONCLUSION: Fetuses with a 1q21.1 microdeletion and concomitant CHD1L mutation may present oligohydramnios and bilateral renal dysplasia on prenatal ultrasound.


Asunto(s)
Anomalías Múltiples/diagnóstico , ADN Helicasas/genética , Proteínas de Unión al ADN/genética , Túbulos Renales Proximales/anomalías , Megalencefalia/diagnóstico , Mutación Missense , Oligohidramnios/diagnóstico , Ultrasonografía Prenatal/métodos , Anomalías Urogenitales/diagnóstico , Anomalías Múltiples/genética , Anomalías Múltiples/metabolismo , Adulto , Deleción Cromosómica , Cromosomas Humanos Par 1/genética , Cromosomas Humanos Par 1/metabolismo , ADN/genética , ADN Helicasas/metabolismo , Análisis Mutacional de ADN , Proteínas de Unión al ADN/metabolismo , Femenino , Humanos , Megalencefalia/genética , Megalencefalia/metabolismo , Oligohidramnios/genética , Embarazo , Anomalías Urogenitales/genética
16.
Biosci Rep ; 39(10)2019 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-31652459

RESUMEN

Cancer incidence rate has increased so much that it is the second leading cause of deaths worldwide after cardiovascular diseases. Sensitive and specific biomarkers are needed for an early diagnosis of cancer and in-time treatment. Recent studies have found that long non-coding RNAs (lncRNAs) participate in cancer tumorigenesis. LncRNA P73 antisense RNA 1T (TP73-AS1), also known as KIAA0495 and p53-dependent apoptosis modulator (PDAM), is located in human chromosomal band 1p36.32 and plays a crucial role in many different carcinomas. This review summarizes current findings on the role of TP73-AS1 and its signaling pathways in various cancers, including glioma, esophageal squamous cell carcinoma (ESCC), hepatocellular carcinoma (HCC), colorectal cancer (CRC), osteosarcoma, gastric cancer (GC), clear cell renal cell carcinoma (ccRCC), breast cancer (BC), bladder cancer, ovarian cancer, cholangiocarcinoma (CCA), lung cancer, and pancreatic cancer. Its aberrant expression generally correlates with clinicopathological characterization of patients. Moreover, TP73-AS1 regulates proliferation, migration, invasion, apoptosis, and chemoresistance cancer mechanisms, both in vivo and in vitro, through different signaling pathways. Therefore, TP73-AS1 may be considered as a marker for diagnosis and prognosis, also as a target for cancer treatment.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias/metabolismo , ARN Largo no Codificante/metabolismo , ARN Neoplásico/metabolismo , Apoptosis/genética , Biomarcadores de Tumor/genética , Movimiento Celular/genética , Proliferación Celular/genética , Cromosomas Humanos Par 1/genética , Cromosomas Humanos Par 1/metabolismo , Resistencia a Antineoplásicos/genética , Humanos , Invasividad Neoplásica , Neoplasias/diagnóstico , Neoplasias/genética , Neoplasias/patología , ARN Largo no Codificante/genética , ARN Neoplásico/genética , Transducción de Señal/genética
17.
Nucleic Acids Res ; 47(13): 6668-6684, 2019 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-31114908

RESUMEN

Rearrangement of the 1q12 pericentromeric heterochromatin and subsequent amplification of the 1q arm is commonly associated with cancer development and progression and may result from epigenetic deregulation. In many premalignant and malignant cells, loss of 1q12 satellite DNA methylation causes the deposition of polycomb factors and formation of large polycomb aggregates referred to as polycomb bodies. Here, we show that SSX proteins can destabilize 1q12 pericentromeric heterochromatin in melanoma cells when it is present in the context of polycomb bodies. We found that SSX proteins deplete polycomb bodies and promote the unfolding and derepression of 1q12 heterochromatin during replication. This further leads to segregation abnormalities during anaphase and generation of micronuclei. The structural rearrangement of 1q12 pericentromeric heterochromatin triggered by SSX2 is associated with loss of polycomb factors, but is not mediated by diminished polycomb repression. Instead, our studies suggest a direct effect of SSX proteins facilitated though a DNA/chromatin binding, zinc finger-like domain and a KRAB-like domain that may recruit chromatin modifiers or activate satellite transcription. Our results demonstrate a novel mechanism for generation of 1q12-associated genomic instability in cancer cells.


Asunto(s)
Ensamble y Desensamble de Cromatina , Cromosomas Humanos Par 1/metabolismo , Heterocromatina/metabolismo , Proteínas de Neoplasias/fisiología , Proteínas Represoras/fisiología , Empalme Alternativo , Línea Celular Tumoral , ADN de Neoplasias/genética , ADN Satélite/genética , Represión Epigenética , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Inestabilidad Genómica , Humanos , Melanoma/patología , Proteínas de Neoplasias/genética , Mutación Puntual , Complejo Represivo Polycomb 1/antagonistas & inhibidores , Complejo Represivo Polycomb 1/genética , Dominios Proteicos , Pliegue de Proteína , ARN Interferente Pequeño/genética , Proteínas Recombinantes/metabolismo , Proteínas Represoras/genética , Eliminación de Secuencia , Transcripción Genética , Dedos de Zinc/fisiología
18.
Comput Biol Med ; 109: 218-225, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31078126

RESUMEN

Recent analysis identified distinct genomic subtypes of lower-grade glioma tumors which are associated with shape features. In this study, we propose a fully automatic way to quantify tumor imaging characteristics using deep learning-based segmentation and test whether these characteristics are predictive of tumor genomic subtypes. We used preoperative imaging and genomic data of 110 patients from 5 institutions with lower-grade gliomas from The Cancer Genome Atlas. Based on automatic deep learning segmentations, we extracted three features which quantify two-dimensional and three-dimensional characteristics of the tumors. Genomic data for the analyzed cohort of patients consisted of previously identified genomic clusters based on IDH mutation and 1p/19q co-deletion, DNA methylation, gene expression, DNA copy number, and microRNA expression. To analyze the relationship between the imaging features and genomic clusters, we conducted the Fisher exact test for 10 hypotheses for each pair of imaging feature and genomic subtype. To account for multiple hypothesis testing, we applied a Bonferroni correction. P-values lower than 0.005 were considered statistically significant. We found the strongest association between RNASeq clusters and the bounding ellipsoid volume ratio (p < 0.0002) and between RNASeq clusters and margin fluctuation (p < 0.005). In addition, we identified associations between bounding ellipsoid volume ratio and all tested molecular subtypes (p < 0.02) as well as between angular standard deviation and RNASeq cluster (p < 0.02). In terms of automatic tumor segmentation that was used to generate the quantitative image characteristics, our deep learning algorithm achieved a mean Dice coefficient of 82% which is comparable to human performance.


Asunto(s)
Aprendizaje Profundo , Genoma Humano , Glioma , Procesamiento de Imagen Asistido por Computador , Modelos Biológicos , Secuencia de Bases , Deleción Cromosómica , Cromosomas Humanos Par 1/genética , Cromosomas Humanos Par 1/metabolismo , Cromosomas Humanos Par 19/genética , Cromosomas Humanos Par 19/metabolismo , Metilación de ADN , ADN de Neoplasias/genética , ADN de Neoplasias/metabolismo , Regulación Neoplásica de la Expresión Génica , Glioma/diagnóstico por imagen , Glioma/genética , Glioma/metabolismo , Humanos , MicroARNs/biosíntesis , MicroARNs/genética , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética
19.
Gene ; 701: 161-168, 2019 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-30849537

RESUMEN

Glial tumors are malignant brain tumors that arise from glial cells of brain or spine and have genetic aberrations in their genome. 1p/19q co-deletion is associated with increased Overall Survival (OS) time with enhanced response to chemo- and radio-therapy in oligodendrogliomas. However, prognostic significance of 1p/19q co-polysomy is still unclear. We evaluated 1p/19q status of 221 patients with glial tumor by Fluorescent in situ Hybridization (FISH). Records of the patients were collected retrospectively. Our results demonstrated that 1p/19q co-polysomy was associated with decreased OS time, high P53 expression and frequently located in temporal lobe, whereas 1p/19q co-deletion was associated with increased overall survival time, low P53 expression and frontal lobe location. Furthermore, classification of patients based on both 1p/19q status and P53 expression revealed that patients with 1p/19q co-polysomy and high P53 expression had the worst prognosis. Lastly, our bioinformatic survival analysis revealed that high expression of SRM, ICMT, and FTL located in 1p36.13-p36.31 and 19q13.2-q13.33 region were related with decreased OS time in patients with Low Grade Glioma (LGG). The study demonstrated that 1p/19q co-polysomy is a poor prognostic marker for glial tumor.


Asunto(s)
Biomarcadores de Tumor , Neoplasias Encefálicas , Aberraciones Cromosómicas , Cromosomas Humanos Par 1 , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias , Oligodendroglioma , Adulto , Biomarcadores de Tumor/biosíntesis , Biomarcadores de Tumor/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Cromosomas Humanos Par 1/genética , Cromosomas Humanos Par 1/metabolismo , Supervivencia sin Enfermedad , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Oligodendroglioma/genética , Oligodendroglioma/metabolismo , Oligodendroglioma/mortalidad , Oligodendroglioma/patología , Estudios Retrospectivos , Tasa de Supervivencia
20.
Mol Hum Reprod ; 25(4): 194-205, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30770928

RESUMEN

Endometriotic lesions are composed in part of endometrial-like stromal cells, however, there is a shortage of immortalized human endometrial stromal cultures available for research. As genetic factors play a role in endometriosis risk, it is important that genotype is also incorporated into analysis of pathological mechanisms. Human telomerase reverse transcriptase (hTERT) immortalization (using Lenti-hTERT-green fluorescent protein virus) took place following genotype selection; 13 patients homozygous for either the risk or non-risk 'other' allele for one or more important endometriosis risk single nucleotide polymorphism on chromosome 1p36.12 (rs3820282, rs56318008, rs55938609, rs12037376, rs7521902 or rs12061255). Short tandem repeat DNA profiling validated that donor tissue matched that of the immortalized cell lines and confirmed that cultures were genetically novel. Expression of morphological markers (vimentin and cytokeratin) and key genes of interest (telomerase, estrogen and progesterone receptors and LINC00339) were examined and functional assays for cell proliferation, steroid hormone and inflammatory responses were performed for 7/13 cultures. All endometrial stromal cell lines maintained their fibroblast-like morphology (vimentin-positive) and homozygous endometriosis-risk genotype following introduction of hTERT. Furthermore, the new stromal cultures demonstrated positive and diverse responses to hormones (proliferation and decidualisation changes) and inflammation (dose-dependent response), while maintaining hormone receptor expression. In conclusion, we successfully developed a range of human endometrial stromal cell lines that carry important endometriosis-risk alleles. The wider implications of this approach go beyond advancing endometriosis research; these cell lines will be valuable tools for multiple endometrial pathologies offering a level of genetic and phenotypic diversity not previously available.


Asunto(s)
Endometriosis/genética , Efecto Fundador , Genotipo , Células del Estroma/metabolismo , Telomerasa/genética , Adulto , Biomarcadores/metabolismo , Línea Celular Transformada , Proliferación Celular , Cromosomas Humanos Par 1/química , Cromosomas Humanos Par 1/metabolismo , Endometriosis/metabolismo , Endometriosis/patología , Endometrio/metabolismo , Endometrio/patología , Femenino , Expresión Génica , Homocigoto , Humanos , Queratinas/genética , Queratinas/metabolismo , Repeticiones de Microsatélite , Polimorfismo de Nucleótido Simple , ARN Largo no Codificante , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Riesgo , Células del Estroma/patología , Telomerasa/metabolismo , Vimentina/genética , Vimentina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...