Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Virchows Arch ; 483(6): 821-833, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37938323

RESUMO

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.


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , Neoplasias Hepáticas , Humanos , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/metabolismo , Cromossomos Humanos Par 1/metabolismo , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/genética , Neoplasias Renais/metabolismo , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/genética , Inibidores de MTOR , Mutação , Serina-Treonina Quinases TOR/genética
2.
Bioinformatics ; 39(1)2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36637205

RESUMO

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.


Assuntos
Neoplasias Encefálicas , Glioma , Humanos , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Prognóstico , Glioma/genética , Aberrações Cromossômicas , Mutação , Cromossomos Humanos Par 1/genética , Cromossomos Humanos Par 1/metabolismo
3.
EBioMedicine ; 87: 104410, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36525723

RESUMO

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.


Assuntos
Neoplasias Encefálicas , Oligodendroglioma , Humanos , Oligodendroglioma/genética , Oligodendroglioma/metabolismo , Oligodendroglioma/patologia , Neoplasias Encefálicas/patologia , Mutação , Aberrações Cromossômicas , Transcriptoma , Prognóstico , Isocitrato Desidrogenase/genética , Isocitrato Desidrogenase/metabolismo , Cromossomos Humanos Par 1/metabolismo
4.
Blood ; 139(13): 1939-1953, 2022 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-35015835

RESUMO

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.


Assuntos
Mieloma Múltiplo , Cromossomos Humanos Par 1/metabolismo , Proteína Forkhead Box M1/genética , Humanos , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Fator de Transcrição 1 de Leucemia de Células Pré-B/genética , Prognóstico , Análise de Sistemas , Fatores de Transcrição/genética
5.
Cells ; 10(6)2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-34205916

RESUMO

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.


Assuntos
Cromossomos Humanos Par 1 , Amplificação de Genes , Mieloma Múltiplo , Proteínas de Neoplasias , Transdução de Sinais , Cromossomos Humanos Par 1/genética , Cromossomos Humanos Par 1/metabolismo , Humanos , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/terapia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo
6.
Adv Anat Pathol ; 28(4): 251-257, 2021 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-34009776

RESUMO

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.


Assuntos
Carcinoma de Células Renais/patologia , Aberrações Cromossômicas , Cromossomos Humanos Par 1/genética , Neoplasias Renais/patologia , Mutação , Serina-Treonina Quinases TOR/genética , Biomarcadores Tumorais , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/metabolismo , Cromossomos Humanos Par 1/metabolismo , Humanos , Rim/metabolismo , Rim/patologia , Neoplasias Renais/genética , Neoplasias Renais/metabolismo , Masculino , Pessoa de Meia-Idade , Serina-Treonina Quinases TOR/metabolismo
7.
Hum Mol Genet ; 30(7): 595-602, 2021 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-33772547

RESUMO

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.


Assuntos
Transtornos Cromossômicos/genética , Fenda Labial/genética , Fissura Palatina/genética , Modelos Animais de Doenças , Desenvolvimento Embrionário/genética , Proteínas do Tecido Nervoso/genética , Proteínas Repressoras/genética , Animais , Proliferação de Células/genética , Deleção Cromossômica , Transtornos Cromossômicos/metabolismo , Cromossomos Humanos Par 1/genética , Cromossomos Humanos Par 1/metabolismo , Fenda Labial/embriologia , Fenda Labial/metabolismo , Fissura Palatina/embriologia , Fissura Palatina/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Mesoderma/citologia , Mesoderma/embriologia , Mesoderma/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas do Tecido Nervoso/deficiência , Crista Neural/embriologia , Crista Neural/metabolismo , Fenótipo , Proteínas Repressoras/deficiência , Proteína Wnt1/genética , Proteína Wnt1/metabolismo
8.
J Mol Biol ; 433(6): 166700, 2021 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-33160979

RESUMO

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.


Assuntos
Cromatina/química , Cromossomos Humanos Par 1/química , Cromossomos Humanos Par 2/química , Cromossomos Humanos Par 3/química , Cromossomos Humanos Par 4/química , Simulação de Dinâmica Molecular , Software , Animais , Linhagem Celular Tumoral , Cromatina/metabolismo , Cromatina/ultraestrutura , Cromossomos Humanos Par 1/metabolismo , Cromossomos Humanos Par 1/ultraestrutura , Cromossomos Humanos Par 2/metabolismo , Cromossomos Humanos Par 2/ultraestrutura , Cromossomos Humanos Par 3/metabolismo , Cromossomos Humanos Par 3/ultraestrutura , Cromossomos Humanos Par 4/metabolismo , Cromossomos Humanos Par 4/ultraestrutura , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Humanos , Linfócitos/citologia , Linfócitos/metabolismo , Saccharum/genética , Saccharum/metabolismo , Termodinâmica , Triticum/genética , Triticum/metabolismo
9.
Pediatr Blood Cancer ; 67(9): e28426, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32614133

RESUMO

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.


Assuntos
Neoplasias Encefálicas , Cromossomos Humanos Par 1 , Metilação de DNA , DNA de Neoplasias , Ependimoma , Recidiva Local de Neoplasia , Adolescente , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/terapia , Criança , Pré-Escolar , Cromossomos Humanos Par 1/genética , Cromossomos Humanos Par 1/metabolismo , DNA de Neoplasias/genética , DNA de Neoplasias/metabolismo , Ependimoma/genética , Ependimoma/metabolismo , Ependimoma/mortalidade , Ependimoma/terapia , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/metabolismo , Recidiva Local de Neoplasia/mortalidade , Recidiva Local de Neoplasia/terapia , Estudos Retrospectivos , Fatores de Risco
10.
Br J Haematol ; 190(6): e329-e332, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32572949
11.
Sci Transl Med ; 12(527)2020 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-31969487

RESUMO

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.


Assuntos
Adenocarcinoma de Pulmão/metabolismo , Cromossomos Humanos Par 1/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Adenocarcinoma de Pulmão/genética , Animais , Cromossomos Humanos Par 1/genética , Ensaio de Imunoadsorção Enzimática , Complexo de Golgi/metabolismo , Humanos , Técnicas In Vitro , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Microtomografia por Raio-X
12.
Taiwan J Obstet Gynecol ; 58(6): 859-863, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31759543

RESUMO

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.


Assuntos
Anormalidades Múltiplas/diagnóstico , DNA Helicases/genética , Proteínas de Ligação a DNA/genética , Túbulos Renais Proximais/anormalidades , Megalencefalia/diagnóstico , Mutação de Sentido Incorreto , Oligo-Hidrâmnio/diagnóstico , Ultrassonografia Pré-Natal/métodos , Anormalidades Urogenitais/diagnóstico , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/metabolismo , Adulto , Deleção Cromossômica , Cromossomos Humanos Par 1/genética , Cromossomos Humanos Par 1/metabolismo , DNA/genética , DNA Helicases/metabolismo , Análise Mutacional de DNA , Proteínas de Ligação a DNA/metabolismo , Feminino , Humanos , Megalencefalia/genética , Megalencefalia/metabolismo , Oligo-Hidrâmnio/genética , Gravidez , Anormalidades Urogenitais/genética
13.
Biosci Rep ; 39(10)2019 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-31652459

RESUMO

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.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias/metabolismo , RNA Longo não Codificante/metabolismo , RNA Neoplásico/metabolismo , Apoptose/genética , Biomarcadores Tumorais/genética , Movimento Celular/genética , Proliferação de Células/genética , Cromossomos Humanos Par 1/genética , Cromossomos Humanos Par 1/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética , Humanos , Invasividade Neoplásica , Neoplasias/diagnóstico , Neoplasias/genética , Neoplasias/patologia , RNA Longo não Codificante/genética , RNA Neoplásico/genética , Transdução de Sinais/genética
14.
Nucleic Acids Res ; 47(13): 6668-6684, 2019 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-31114908

RESUMO

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.


Assuntos
Montagem e Desmontagem da Cromatina , Cromossomos Humanos Par 1/metabolismo , Heterocromatina/metabolismo , Proteínas de Neoplasias/fisiologia , Proteínas Repressoras/fisiologia , Processamento Alternativo , Linhagem Celular Tumoral , DNA de Neoplasias/genética , DNA Satélite/genética , Repressão Epigenética , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Instabilidade Genômica , Humanos , Melanoma/patologia , Proteínas de Neoplasias/genética , Mutação Puntual , Complexo Repressor Polycomb 1/antagonistas & inibidores , Complexo Repressor Polycomb 1/genética , Domínios Proteicos , Dobramento de Proteína , RNA Interferente Pequeno/genética , Proteínas Recombinantes/metabolismo , Proteínas Repressoras/genética , Deleção de Sequência , Transcrição Gênica , Dedos de Zinco/fisiologia
15.
Comput Biol Med ; 109: 218-225, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31078126

RESUMO

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.


Assuntos
Aprendizado Profundo , Genoma Humano , Glioma , Processamento de Imagem Assistida por Computador , Modelos Biológicos , Sequência de Bases , Deleção Cromossômica , Cromossomos Humanos Par 1/genética , Cromossomos Humanos Par 1/metabolismo , Cromossomos Humanos Par 19/genética , Cromossomos Humanos Par 19/metabolismo , Metilação de DNA , DNA de Neoplasias/genética , DNA de Neoplasias/metabolismo , Regulação Neoplásica da Expressão Gênica , Glioma/diagnóstico por imagem , Glioma/genética , Glioma/metabolismo , Humanos , MicroRNAs/biossíntese , MicroRNAs/genética , RNA Neoplásico/biossíntese , RNA Neoplásico/genética
16.
Gene ; 701: 161-168, 2019 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-30849537

RESUMO

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.


Assuntos
Biomarcadores Tumorais , Neoplasias Encefálicas , Aberrações Cromossômicas , Cromossomos Humanos Par 1 , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias , Oligodendroglioma , Adulto , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Cromossomos Humanos Par 1/genética , Cromossomos Humanos Par 1/metabolismo , Intervalo Livre de Doença , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Oligodendroglioma/genética , Oligodendroglioma/metabolismo , Oligodendroglioma/mortalidade , Oligodendroglioma/patologia , Estudos Retrospectivos , Taxa de Sobrevida
17.
Mol Hum Reprod ; 25(4): 194-205, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30770928

RESUMO

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.


Assuntos
Endometriose/genética , Efeito Fundador , Genótipo , Células Estromais/metabolismo , Telomerase/genética , Adulto , Biomarcadores/metabolismo , Linhagem Celular Transformada , Proliferação de Células , Cromossomos Humanos Par 1/química , Cromossomos Humanos Par 1/metabolismo , Endometriose/metabolismo , Endometriose/patologia , Endométrio/metabolismo , Endométrio/patologia , Feminino , Expressão Gênica , Homozigoto , Humanos , Queratinas/genética , Queratinas/metabolismo , Repetições de Microssatélites , Polimorfismo de Nucleotídeo Único , RNA Longo não Codificante , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Risco , Células Estromais/patologia , Telomerase/metabolismo , Vimentina/genética , Vimentina/metabolismo
18.
Proc Natl Acad Sci U S A ; 115(27): E6283-E6290, 2018 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-29915083

RESUMO

Renal oncocytomas are benign tumors characterized by a marked accumulation of mitochondria. We report a combined exome, transcriptome, and metabolome analysis of these tumors. Joint analysis of the nuclear and mitochondrial (mtDNA) genomes reveals loss-of-function mtDNA mutations occurring at high variant allele fractions, consistent with positive selection, in genes encoding complex I as the most frequent genetic events. A subset of these tumors also exhibits chromosome 1 loss and/or cyclin D1 overexpression, suggesting they follow complex I loss. Transcriptome data revealed that many pathways previously reported to be altered in renal oncocytoma were simply differentially expressed in the tumor's cell of origin, the distal nephron, compared with other nephron segments. Using a heuristic approach to account for cell-of-origin bias we uncovered strong expression alterations in the gamma-glutamyl cycle, including glutathione synthesis (increased GCLC) and glutathione degradation. Moreover, the most striking changes in metabolite profiling were elevations in oxidized and reduced glutathione as well as γ-glutamyl-cysteine and cysteinyl-glycine, dipeptide intermediates in glutathione biosynthesis, and recycling, respectively. Biosynthesis of glutathione appears adaptive as blockade of GCLC impairs viability in cells cultured with a complex I inhibitor. Our data suggest that loss-of-function mutations in complex I are a candidate driver event in renal oncocytoma that is followed by frequent loss of chromosome 1, cyclin D1 overexpression, and adaptive up-regulation of glutathione biosynthesis.


Assuntos
Adenoma Oxífilo , Complexo I de Transporte de Elétrons/deficiência , Glutationa , Neoplasias Renais , Mitocôndrias , Proteínas de Neoplasias/deficiência , Adenoma Oxífilo/genética , Adenoma Oxífilo/metabolismo , Adenoma Oxífilo/patologia , Sobrevivência Celular/genética , Cromossomos Humanos Par 1/genética , Cromossomos Humanos Par 1/metabolismo , Ciclina D1/genética , Ciclina D1/metabolismo , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , DNA de Neoplasias/genética , DNA de Neoplasias/metabolismo , Feminino , Perfilação da Expressão Gênica , Glutationa/genética , Glutationa/metabolismo , Humanos , Neoplasias Renais/genética , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Masculino , Mitocôndrias/genética , Mitocôndrias/metabolismo , Mitocôndrias/patologia
19.
Blood Cancer J ; 7(9): e607, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-29016570
20.
Pediatr Blood Cancer ; 64(10)2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28436581

RESUMO

BACKGROUND: In childhood acute lymphoblastic leukemia (ALL), t(1;19)(q23;p13.3) with TCF3-PBX1 fusion is one of the most frequent translocations. Historically, it has been associated with poor prognosis. Intensive treatment, however, has improved its outcome. We determined the outcome of children with this genotype treated with contemporary intensive chemotherapy in Taiwan. PROCEDURE: In Taiwan Pediatric Oncology Group 2002 ALL studies, genotypes were determined by cytogenetic analysis and/or reverse transcriptase polymerase chain reaction assay. Based on presenting features, immunophenotype and genotype, patients were assigned to one of the three risk groups: standard risk (SR), high risk (HR), or very high risk (VHR). The patients with t(1;19)/TCF3-PBX1 were treated in the HR arm receiving more intensive chemotherapy. The outcomes of patients with t(1;19)/TCF3-PBX1 were compared to that of patients with other subtypes of B-precursor ALL (B-ALL). RESULTS: Of the 1,129 patients with B-ALL, 64 (5.7%) had t(1;19)/TCF3-PBX1; 51 of whom were treated in the HR arm, but 11 were treated in the VHR and 2 in the SR arm because of physician's preference. As a group, 64 patients with t(1;19)/TCF3-PBX1 had similar 5-year event-free survival (83.3 ± 4.8%) as those with TEL-AML1 (85.2 ± 3.4%, P = 0.984) or those with hyperdiploidy >50 (84.0 ± 3.1%, P = 0.748). The cumulative risk of any (isolated plus combined) central nervous system relapse among patients with t(1;19)/TCF3-PBX1 (8.7 ± 3.8%) tended to be higher than that of patients with TEL-AML1 (5.8 ± 2.3%, P = 0.749) or those with hyperdiploidy (4.1 ± 1.8%, P = 0.135), albeit the differences did not reach statistical significance. CONCLUSIONS: With contemporary intensive chemotherapy, children with t(1;19)/TCF3-PBX1 fared as well as those with favorable genotypes (TEL-AML1 or hyperdiploidy).


Assuntos
Cromossomos Humanos Par 19 , Cromossomos Humanos Par 1 , Leucemia-Linfoma Linfoblástico de Células Precursoras B , Translocação Genética , Adolescente , Criança , Pré-Escolar , Cromossomos Humanos Par 1/genética , Cromossomos Humanos Par 1/metabolismo , Cromossomos Humanos Par 19/genética , Cromossomos Humanos Par 19/metabolismo , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Taiwan
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA