Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 32(4): 1063-1070, 2024 Aug.
Artigo em Chinês | MEDLINE | ID: mdl-39192399

RESUMO

OBJECTIVE: To investigate the incidence of PTPN11 gene mutation and its associated gene mutations in adult patients with acute myeloid leukemia (AML), and analyze its clinical characteristics. METHODS: Second-generation sequencing and Sanger sequencing were used to detect 51 gene mutations, and multiplex-PCR was used to detect 41 fusion genes from 451 newly diagnosed adult AML patients admitted to Affiliated Hospital of Jiangnan University, Changzhou Second People's Hospital, Wuxi People's Hospital and Wuxi Second People's Hospital from January 2017 to July 2022. RESULTS: Among 451 primary adult AML patients, the PTPN11 gene mutation was detected in 34 cases, and the mutation rate was 7.5%. In the 34 patients, 37 PTPN11 alterations were found, which were exclusively missense mutations affecting residues located within the N-SH2 (31 cases) and PTP (6 cases) domains and clustered overwhelmingly in exon 3. The platelet count of PTPN11 mutation patients was 76.5(23.5, 119.0)×109/L, which was significantly higher than 41.0(22.0, 82.5)×109/L of wild-type patients (P < 0.05). While, there were no significant differences in sex, age, peripheral white blood cell count, hemoglobin, and bone marrow blast between PTPN11 mutation and wild-type patients (P >0.05). In FAB subtypes, PTPN11 mutations were mainly distributed in M5, followed by M2 and M4, less frequently in M3 and M6. There was no significant difference in the distribution of FAB subtypes between PTPN11 mutation and wild-type patients (P >0.05). A total of 118 AML patients were detected positive fusion gene, among which patients with PTPN11 mutations had a higher incidence of positive MLL-AF6 than wild-type ones (P < 0.01). 97.1% of 34 patients with PTPN11 mutations were accompanied by other mutations, in descending order, they were respectively NPM1 (38.2%), NRAS (32.4%), FLT3-ITD (32.4%), DNMT3A (32.4%) and KRAS (23.5%), etc . CONCLUSION: PTPN11 mutation has a certain incidence in AML patients and is clustered overwhelmingly in exon 3. ALL of them are exclusively missense mutations, and most often present in conjunction with NPM1 mutations. FAB typing of PTPN11 mutation is mostly manifested as M5 subtype, which is associated with higher platelet counts.


Assuntos
Leucemia Mieloide Aguda , Mutação , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Humanos , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Leucemia Mieloide Aguda/genética , Adulto , Nucleofosmina , Éxons , Masculino , Feminino
2.
Free Radic Biol Med ; 224: 130-143, 2024 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-39182738

RESUMO

Oxidative signaling plays a dual role in tumor initiation and progression to malignancy; however, the regulatory mechanisms of oxidative stress in gastric cancer remain to be explored. In this study, we discovered that Prohibitin 2 (PHB2) specifically regulates cytosolic reactive oxygen species production in gastric cancer and facilitates its malignant progression. Previously, we found that PHB2 is upregulated in gastric cancer, correlating with increased tumorigenicity of gastric cancer cells and poor patient prognosis. Here, we discovered that PHB2 expression correlates with the activation of the ERK/MAPK cascade, positively regulating the top gene NADPH oxidase 1 (NOX1) within this pathway. Further mechanistic investigation reveals that PHB2 enhances NOX1 transcription by interacting with the transcription factor C/EBP-beta and promoting its translocation into the nucleus, resulting in elevated intracellular oxidative signaling driven by NOX1, which subsequently activates ERK. Therefore, we propose that targeting PHB2-C/EBP-beta-NOX1-mediated cytosolic oxidative stress could offer a promising therapeutic avenue for combating gastric cancer malignant progression.

3.
Front Immunol ; 15: 1391504, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38887292

RESUMO

Diabetes is a prevalent chronic disease that traditionally requires severe reliance on medication for treatment. Oral medication and exogenous insulin can only temporarily maintain blood glucose levels and do not cure the disease. Most patients need life-long injections of exogenous insulin. In recent years, advances in islet transplantation have significantly advanced the treatment of diabetes, allowing patients to discontinue exogenous insulin and avoid complications.Long-term follow-up results from recent reports on islet transplantation suggest that they provide significant therapeutic benefit although patients still require immunotherapy, suggesting the importance of future transplantation strategies. Although organ shortage remains the primary obstacle for the development of islet transplantation, new sources of islet cells, such as stem cells and porcine islet cells, have been proposed, and are gradually being incorporated into clinical research. Further research on new transplantation sites, such as the subcutaneous space and mesenteric fat, may eventually replace the traditional portal vein intra-islet cell infusion. Additionally, the immunological rejection reaction in islet transplantation will be resolved through the combined application of immunosuppressant agents, islet encapsulation technology, and the most promising mesenchymal stem cells/regulatory T cell and islet cell combined transplantation cell therapy. This review summarizes the progress achieved in islet transplantation, and discusses the research progress and potential solutions to the challenges faced.


Assuntos
Transplante das Ilhotas Pancreáticas , Transplante das Ilhotas Pancreáticas/métodos , Humanos , Animais , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/prevenção & controle , Diabetes Mellitus Tipo 1/terapia , Diabetes Mellitus Tipo 1/cirurgia , Diabetes Mellitus Tipo 1/imunologia
4.
Clin Transl Med ; 14(6): e1734, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38888967

RESUMO

BACKGROUND: Sporadic parathyroid adenoma (PA) is the most common cause of hyperparathyroidism, yet the mechanisms involved in its pathogenesis remain incompletely understood. METHODS: Surgically removed PA samples, along with normal parathyroid gland (PG) tissues that were incidentally dissected during total thyroidectomy, were analysed using single-cell RNA-sequencing with the 10× Genomics Chromium Droplet platform and Cell Ranger software. Gene set variation analysis was conducted to characterise hallmark pathway gene signatures, and single-cell regulatory network inference and clustering were utilised to analyse transcription factor regulons. Immunohistochemistry and immunofluorescence were performed to validate cellular components of PA tissues. siRNA knockdown and gene overexpression, alongside quantitative polymerase chain reaction, Western blotting and cell proliferation assays, were conducted for functional investigations. RESULTS: There was a pervasive increase in gene transcription in PA cells (PACs) compared with PG cells. This is associated with high expression of histone-lysine N-methyltransferase 2A (KMT2A). High KMT2A levels potentially contribute to promoting PAC proliferation through upregulation of the proto-oncogene CCND2, which is mediated by the transcription factors signal transducer and activator of transcription 3 (STAT3) and GATA binding protein 3 (GATA3). PA tissues are heavily infiltrated with myeloid cells, while fibroblasts, endothelial cells and macrophages in PA tissues are commonly enriched with proinflammatory gene signatures relative to their counterparts in PG tissues. CONCLUSIONS: We revealed the previously underappreciated involvement of the KMT2A‒STAT3/GATA3‒CCND2 axis and chronic inflammation in the pathogenesis of PA. These findings underscore the therapeutic promise of KMT2A inhibition and anti-inflammatory strategies, highlighting the need for future investigations to translate these molecular insights into practical applications. HIGHLIGHTS: Single-cell RNA-sequencing reveals a transcriptome catalogue comparing sporadic parathyroid adenomas (PAs) with normal parathyroid glands. PA cells show a pervasive increase in gene expression linked to KMT2A upregulation. KMT2A-mediated STAT3 and GATA3 upregulation is key to promoting PA cell proliferation via cyclin D2. PAs exhibit a proinflammatory microenvironment, suggesting a potential role of chronic inflammation in PA pathogenesis.


Assuntos
Adenoma , Histona-Lisina N-Metiltransferase , Inflamação , Neoplasias das Paratireoides , Humanos , Neoplasias das Paratireoides/genética , Neoplasias das Paratireoides/metabolismo , Neoplasias das Paratireoides/patologia , Adenoma/genética , Adenoma/metabolismo , Adenoma/patologia , Inflamação/genética , Inflamação/metabolismo , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Proteína de Leucina Linfoide-Mieloide/genética , Proteína de Leucina Linfoide-Mieloide/metabolismo , Proto-Oncogene Mas , Proliferação de Células/genética
5.
Cancer Res ; 84(9): 1460-1474, 2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38593213

RESUMO

Patients with triple-negative breast cancer (TNBC) have a poor prognosis due to the lack of effective molecular targets for therapeutic intervention. Here we found that the long noncoding RNA (lncRNA) MILIP supports TNBC cell survival, proliferation, and tumorigenicity by complexing with transfer RNAs (tRNA) to promote protein production, thus representing a potential therapeutic target in TNBC. MILIP was expressed at high levels in TNBC cells that commonly harbor loss-of-function mutations of the tumor suppressor p53, and MILIP silencing suppressed TNBC cell viability and xenograft growth, indicating that MILIP functions distinctively in TNBC beyond its established role in repressing p53 in other types of cancers. Mechanistic investigations revealed that MILIP interacted with eukaryotic translation elongation factor 1 alpha 1 (eEF1α1) and formed an RNA-RNA duplex with the type II tRNAs tRNALeu and tRNASer through their variable loops, which facilitated the binding of eEF1α1 to these tRNAs. Disrupting the interaction between MILIP and eEF1α1 or tRNAs diminished protein synthesis and cell viability. Targeting MILIP inhibited TNBC growth and cooperated with the clinically available protein synthesis inhibitor omacetaxine mepesuccinate in vivo. Collectively, these results identify MILIP as an RNA translation elongation factor that promotes protein production in TNBC cells and reveal the therapeutic potential of targeting MILIP, alone and in combination with other types of protein synthesis inhibitors, for TNBC treatment. SIGNIFICANCE: LncRNA MILIP plays a key role in supporting protein production in TNBC by forming complexes with tRNAs and eEF1α1, which confers sensitivity to combined MILIP targeting and protein synthesis inhibitors.


Assuntos
Proliferação de Células , Fator 1 de Elongação de Peptídeos , Biossíntese de Proteínas , RNA Longo não Codificante , RNA de Transferência , Neoplasias de Mama Triplo Negativas , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia , Neoplasias de Mama Triplo Negativas/metabolismo , Humanos , Feminino , RNA de Transferência/genética , RNA de Transferência/metabolismo , Animais , Camundongos , Fator 1 de Elongação de Peptídeos/metabolismo , Fator 1 de Elongação de Peptídeos/genética , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Linhagem Celular Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto , Camundongos Nus , Regulação Neoplásica da Expressão Gênica
6.
FASEB J ; 38(2): e23387, 2024 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-38193649

RESUMO

Human brain microvascular endothelial cells (HBMVECs) and microglia play critical roles in regulating cerebral homeostasis during ischemic stroke. However, the role of HBMVECs-derived exosomes in microglia polarization after stroke remains unknown. We isolated exosomes (Exos) from oxygen glucose deprivation (OGD)-exposed HBMVECs, before added them into microglia. Microglia polarization markers were tested using RT-qPCR or flow cytometry. Inflammatory cytokines were measured with ELISA. Endothelial cell damage was assessed by cell viability, apoptosis, apoptosis-related proteins, oxidative stress, and angiogenic activity using CCK-8, flow cytometry, western blot, ELISA, and endothelial tube formation assay, respectively. We also established middle cerebral artery occlusion (MCAO) mice model to examine the function of circ_0000495 on stroke in vivo. Our study found that HBMVECs-Exos reduced M2 markers (IL-10, CD163, and CD206), increased M1 markers (TNF-α, IL-1ß, and IL-12), CD86-positive cells, and inflammatory cytokines (TNF-α and IL-1ß), indicating the promotion of microglial M1-polarization. Microglial M1-polarization induced by HBMVECs-Exos reduced viability and promoted apoptosis and oxidative stress, revealing the aggravation of endothelial cell damage. However, circ_0000495 silencing inhibited HBMVECs-Exos-induced alterations. Mechanistically, circ_0000495 adsorbed miR-579-3p to upregulate toll-like receptor 4 (TLR4) in microglia; miR-579-3p suppressed HBMVECs-Exos-induced alterations via declining TLR4; furthermore, Yin Yang 1 (YY1) transcriptionally activated circ_0000495 in HBMVECs. Importantly, circ_0000495 aggravated ischemic brain injury in vivo via activating TLR4/nuclear factor-κB (NF-κB) pathway. Collectively, OGD-treated HBMVECs-Exos transmitted circ_0000495 to regulate miR-579-3p/TLR4/NF-κB axis in microglia, thereby facilitating microglial M1-polarization and endothelial cell damage.


Assuntos
Exossomos , MicroRNAs , Acidente Vascular Cerebral , Animais , Camundongos , Humanos , Células Endoteliais , Microglia , Receptor 4 Toll-Like/genética , NF-kappa B , Fator de Necrose Tumoral alfa , Encéfalo , Hipóxia , Oxigênio , Citocinas , MicroRNAs/genética
7.
J Exp Clin Cancer Res ; 43(1): 17, 2024 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-38200519

RESUMO

BACKGROUND: Prohibitin 2 (PHB2) exhibits opposite functions of promoting or inhibiting tumour across various cancer types. In this study, we aim to investigate its functions and underlying mechanisms in the context of gastric cancer (GC). METHODS: PHB2 protein expression levels in GC and normal tissues were examined using western blot and immunohistochemistry. PHB2 expression level associations with patient outcomes were examined through Kaplan-Meier plotter analysis utilizing GEO datasets (GSE14210 and GSE29272). The biological role of PHB2 and its subsequent regulatory mechanisms were elucidated in vitro and in vivo. GC cell viability and proliferation were assessed using MTT cell viability analysis, clonogenic assays, and BrdU incorporation assays, while the growth of GC xenografted tumours was measured via IHC staining of Ki67. The interaction among PHB2 and SHIP2, as well as between SHIP2 and NEDD4, was identified through co-immunoprecipitation, GST pull-down assays, and deletion-mapping experiments. SHIP2 ubiquitination and degradation were assessed using cycloheximide treatment, plasmid transfection and co-immunoprecipitation, followed by western blot analysis. RESULTS: Our analysis revealed a substantial increase in PHB2 expression in GC tissues compared to adjacent normal tissues. Notably, higher PHB2 levels correlated with poorer patient outcomes, suggesting its clinical relevance. Functionally, silencing PHB2 in GC cells significantly reduced cell proliferation and retarded GC tumour growth, whereas overexpression of PHB2 further enhanced GC cell proliferation. Mechanistically, PHB2 physically interacted with Src homology 2-containing inositol 5-phosphatase 2 (SHIP2) in the cytoplasm of GC cells, thus leading to SHIP2 degradation via its novel E3 ligase NEDD4. It subsequently activated the PI3K/Akt signaling pathway and thus promoted GC cell proliferation. CONCLUSIONS: Our findings highlight the importance of PHB2 upregulation in driving GC progression and its association with adverse patient outcomes. Understanding the functional impact of PHB2 on GC growth contributes valuable insights into the molecular underpinnings of GC and may pave the way for the development of targeted therapies to improve patient outcomes.


Assuntos
Neoplasias Gástricas , Ubiquitina-Proteína Ligases , Humanos , Inositol Polifosfato 5-Fosfatases , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais , Neoplasias Gástricas/genética , Ubiquitinação
8.
Theranostics ; 13(3): 1109-1129, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36793869

RESUMO

While psychological factors have long been linked to breast cancer pathogenesis and outcomes, accumulating evidence is revealing how the nervous system contributes to breast cancer development, progression, and treatment resistance. Central to the psychological-neurological nexus are interactions between neurotransmitters and their receptors expressed on breast cancer cells and other types of cells in the tumor microenvironment, which activate various intracellular signaling pathways. Importantly, the manipulation of these interactions is emerging as a potential avenue for breast cancer prevention and treatment. However, an important caveat is that the same neurotransmitter can exert multiple and sometimes opposing effects. In addition, certain neurotransmitters can be produced and secreted by non-neuronal cells including breast cancer cells that similarly activate intracellular signaling upon binding to their receptors. In this review we dissect the evidence for the emerging paradigm linking neurotransmitters and their receptors with breast cancer. Foremost, we explore the intricacies of such neurotransmitter-receptor interactions, including those that impinge on other cellular components of the tumor microenvironment, such as endothelial cells and immune cells. Moreover, we discuss findings where clinical agents used to treat neurological and/or psychological disorders have exhibited preventive/therapeutic effects against breast cancer in either associative or pre-clinical studies. Further, we elaborate on the current progress to identify druggable components of the psychological-neurological nexus that can be exploited for the prevention and treatment of breast cancer as well as other tumor types. We also provide our perspectives regarding future challenges in this field where multidisciplinary cooperation is a paramount requirement.


Assuntos
Neoplasias da Mama , Humanos , Feminino , Neoplasias da Mama/prevenção & controle , Células Endoteliais/metabolismo , Neurotransmissores , Transdução de Sinais , Microambiente Tumoral
9.
Adv Sci (Weinh) ; 10(7): e2204599, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36638271

RESUMO

P53 inactivation occurs in about 50% of human cancers, where p53-driven p21 activity is devoid and p27 becomes essential for the establishment of the G1/S checkpoint upon DNA damage. Here, this work shows that the E2F1-responsive lncRNA LIMp27 selectively represses p27 expression and contributes to proliferation, tumorigenicity, and treatment resistance in p53-defective colon adenocarcinoma (COAD) cells. LIMp27 competes with p27 mRNA for binding to cytoplasmically localized hnRNA0, which otherwise stabilizes p27 mRNA leading to cell cycle arrest at the G0/G1 phase. In response to DNA damage, LIMp27 is upregulated in both wild-type and p53-mutant COAD cells, whereas cytoplasmic hnRNPA0 is only increased in p53-mutant COAD cells due to translocation from the nucleus. Moreover, high LIMp27 expression is associated with poor survival of p53-mutant but not wild-type p53 COAD patients. These results uncover an lncRNA mechanism that promotes p53-defective cancer pathogenesis and suggest that LIMp27 may constitute a target for the treatment of such cancers.


Assuntos
Adenocarcinoma , Neoplasias do Colo , Inibidor de Quinase Dependente de Ciclina p27 , RNA Longo não Codificante , Humanos , Dano ao DNA/genética , RNA Longo não Codificante/genética , RNA Mensageiro/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo
10.
Proc Natl Acad Sci U S A ; 119(49): e2208904119, 2022 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-36445966

RESUMO

The protooncoprotein N-Myc, which is overexpressed in approximately 25% of neuroblastomas as the consequence of MYCN gene amplification, has long been postulated to regulate DNA double-strand break (DSB) repair in neuroblastoma cells, but experimental evidence of this function is presently scant. Here, we show that N-Myc transcriptionally activates the long noncoding RNA MILIP to promote nonhomologous end-joining (NHEJ) DNA repair through facilitating Ku70-Ku80 heterodimerization in neuroblastoma cells. High MILIP expression was associated with poor outcome and appeared as an independent prognostic factor in neuroblastoma patients. Knockdown of MILIP reduced neuroblastoma cell viability through the induction of apoptosis and inhibition of proliferation, retarded neuroblastoma xenograft growth, and sensitized neuroblastoma cells to DNA-damaging therapeutics. The effect of MILIP knockdown was associated with the accumulation of DNA DSBs in neuroblastoma cells largely due to decreased activity of the NHEJ DNA repair pathway. Mechanistical investigations revealed that binding of MILIP to Ku70 and Ku80 increased their heterodimerization, and this was required for MILIP-mediated promotion of NHEJ DNA repair. Disrupting the interaction between MILIP and Ku70 or Ku80 increased DNA DSBs and reduced cell viability with therapeutic potential revealed where targeting MILIP using Gapmers cooperated with the DNA-damaging drug cisplatin to inhibit neuroblastoma growth in vivo. Collectively, our findings identify MILIP as an N-Myc downstream effector critical for activation of the NHEJ DNA repair pathway in neuroblastoma cells, with practical implications of MILIP targeting, alone and in combination with DNA-damaging therapeutics, for neuroblastoma treatment.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades , Neuroblastoma , RNA Longo não Codificante , Humanos , DNA/genética , Reparo do DNA por Junção de Extremidades/genética , Reparo do DNA/genética , Neuroblastoma/tratamento farmacológico , Neuroblastoma/genética , RNA Longo não Codificante/genética
11.
Mikrochim Acta ; 189(1): 17, 2021 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-34873664

RESUMO

As well known, the electrochemiluminescence (ECL) of tris(2,2'-bipyridine)ruthenium(II) (Ru(bpy)32+) heavily relies on highly positive or negative triggered voltage, prejudicing the detection toward the bio-molecules. In this work, Ru(bpy)32+ could generate enhanced and stable ECL at a low potential of 0.05 V (vs. Ag/AgCl) on graphene-PtPd hybrid, attributing to its excellent electrocatalysis from the synergistic effect between Pt and Pd. The obtained low-potential-driven ECL could be quenched by MoS2 nanoflowers. Based on the quenching effect, a sandwich "signal-off" ECL immunosensor was fabricated to sensitively detect carcinoembryonic antigen (CEA). A linear calibration curve from 1 fg mL-1 to 1 ng mL-1 was obtained along with a low detection limit of 0.54 fg mL-1 (S/N = 3) under optimal conditions. The sensor showed satisfactory specificity, stability, and reproducibility and was successfully applied to determine CEA in actual samples. The recoveries ranged from 98.80 to 100.23%, and the relative standard deviation (RSD) was lower than 5%. Above all, this work explored new materials in low-potential-driven ECL system and provided a reliable sensing strategy for clinical applications.


Assuntos
Antígeno Carcinoembrionário/sangue , Técnicas Eletroquímicas/métodos , Imunoensaio/métodos , Substâncias Luminescentes/química , Nanocompostos/química , Compostos Organometálicos/química , Anticorpos Imobilizados/imunologia , Antígeno Carcinoembrionário/imunologia , Dissulfetos/química , Grafite/química , Humanos , Limite de Detecção , Molibdênio/química , Paládio/química , Platina/química , Reprodutibilidade dos Testes
12.
Theranostics ; 11(19): 9605-9622, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34646389

RESUMO

Rationale: Recurrent and metastatic cancers often undergo a period of dormancy, which is closely associated with cellular quiescence, a state whereby cells exit the cell cycle and are reversibly arrested in G0 phase. Curative cancer treatment thus requires therapies that either sustain the dormant state of quiescent cancer cells, or preferentially, eliminate them. However, the mechanisms responsible for the survival of quiescent cancer cells remain obscure. Methods: Dual genome-editing was carried out using a CRISPR/Cas9-based system to label endogenous p27 and Ki67 with the green and red fluorescent proteins EGFP and mCherry, respectively, in melanoma cells. Analysis of transcriptomes of isolated EGFP-p27highmCherry-Ki67low quiescent cells was conducted at bulk and single cell levels using RNA-sequencing. The extracellular acidification rate and oxygen consumption rate were measured to define metabolic phenotypes. SiRNA and inducible shRNA knockdown, chromatin immunoprecipitation and luciferase reporter assays were employed to elucidate mechanisms of the metabolic switch in quiescent cells. Results: Dual labelling of endogenous p27 and Ki67 with differentiable fluorescent probes allowed for visualization, isolation, and analysis of viable p27highKi67low quiescent cells. Paradoxically, the proto-oncoprotein c-Myc, which commonly drives malignant cell cycle progression, was expressed at relatively high levels in p27highKi67low quiescent cells and supported their survival through promoting mitochondrial oxidative phosphorylation (OXPHOS). In this context, c-Myc selectively transactivated genes encoding OXPHOS enzymes, including subunits of isocitric dehydrogenase 3 (IDH3), whereas its binding to cell cycle progression gene promoters was decreased in quiescent cells. Silencing of c-Myc or the catalytic subunit of IDH3, IDH3α, preferentially killed quiescent cells, recapitulating the effect of treatment with OXPHOS inhibitors. Conclusion: These results establish a rigorous experimental system for investigating cellular quiescence, uncover the high selectivity of c-Myc in activating OXPHOS genes in quiescent cells, and propose OXPHOS targeting as a potential therapeutic avenue to counter cancer cells in quiescence.


Assuntos
Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Antígeno Ki-67/metabolismo , Melanoma/metabolismo , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Senescência Celular , Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Isocitrato Desidrogenase/metabolismo , Neoplasias/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fase de Repouso do Ciclo Celular , Transcriptoma/genética
13.
Nat Commun ; 12(1): 3734, 2021 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-34145290

RESUMO

Genomic amplification of the distal portion of chromosome 3q, which encodes a number of oncogenic proteins, is one of the most frequent chromosomal abnormalities in malignancy. Here we functionally characterise a non-protein product of the 3q region, the long noncoding RNA (lncRNA) PLANE, which is upregulated in diverse cancer types through copy number gain as well as E2F1-mediated transcriptional activation. PLANE forms an RNA-RNA duplex with the nuclear receptor co-repressor 2 (NCOR2) pre-mRNA at intron 45, binds to heterogeneous ribonucleoprotein M (hnRNPM) and facilitates the association of hnRNPM with the intron, thus leading to repression of the alternative splicing (AS) event generating NCOR2-202, a major protein-coding NCOR2 AS variant. This is, at least in part, responsible for PLANE-mediated promotion of cancer cell proliferation and tumorigenicity. These results uncover the function and regulation of PLANE and suggest that PLANE may constitute a therapeutic target in the pan-cancer context.


Assuntos
Processamento Alternativo/genética , Regulação Neoplásica da Expressão Gênica/genética , Neoplasias/genética , RNA Longo não Codificante/genética , Células A549 , Linhagem Celular Tumoral , Proliferação de Células/genética , Cromossomos Humanos Par 3/genética , Variações do Número de Cópias de DNA/genética , Fator de Transcrição E2F1/metabolismo , Células HCT116 , Ribonucleoproteínas Nucleares Heterogêneas Grupo M/genética , Humanos , Células MCF-7 , Neoplasias/patologia , Correpressor 2 de Receptor Nuclear/genética , Interferência de RNA , RNA Interferente Pequeno/genética
14.
J Cancer ; 11(7): 1940-1948, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32194805

RESUMO

Objective: To identify a multi-gene prognostic factor in patients with lung adenocarcinoma (LUAD). Materials and methods Prognosis-related genes were screened in the TCGA-LUAD cohort. Then, patients in this cohort were randomly separated into training set and test set. Least absolute shrinkage and selection operator (LASSO) regression was performed to the penalized the Cox proportional hazards regression (CPH) model on the training set, and a prognostication combination based on the result of LASSO analysis was developed. By performing Kaplan-Meier curve analysis, univariate and multivariable CPH model on the overall survival (OS) as well as recurrence free survival (RFS), the prognostication performance of the multigene combination were evaluated. Moreover, we constructed a nomogram and performed decision curve analysis to evaluate the clinical application of the multigene combination. Results We obtained 99 prognosis-related genes and screened out a 4-gene combination (including CIDEC, ZFP3, DKK1, and USP4) according to the LASSO analysis. The results of survival analyses suggested that patients in the 4-gene combination low-risk group had better OS and RFS than those in the 4-gene combination high-risk group. The 4-gene mentioned was demonstrated to be independent prognostic factor of patients with LUAD in the training set(OS, HR=11.962, P<0.001; RFS, HR=9.281, P<0.001) and test set (OS, HR=5.377, P=0.003; RFS, HR=2.949, P=0.104). More importantly, its prognosis performance was well in the validation set (OS, HR=0.955, P=0.002; RFS, HR=1.042, P<0.001). Conclusions We introduced a 4-gene combination which could predict the survival of LUAD patients and might be an independent prognostic factor in LUAD.

15.
Mol Cell Oncol ; 8(1): 1842714, 2020 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-33553602

RESUMO

We have recently identified the MYC proto-oncogene, bHLH transcription factor (MYC, best known as c-Myc)-responsive pan-cancer lncRNA c-Myc-Inducible Long noncoding RNA Inactivating P53 (MILIP) as an oncogenic driver. Our studies show that MILIP facilitates tumor protein p53 (TP53, best known as p53) turnover by reducing its SUMOylation through suppressing tripartite-motif family-like 2 (TRIML2), thus promoting cell survival, proliferation, and tumorigenicity. MILIP may thus represent an anti-cancer target for counteracting the c-Myc axis.

16.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 24(5): 1484-1488, 2016 Oct.
Artigo em Chinês | MEDLINE | ID: mdl-27784379

RESUMO

OBJECTIVE: To investigate the clinical efficacy and safety of "3+7" regimen and decitabine + HAG pre-excitation regimen in the treatment of elderly MDS patients with AML transformed by abnormal proliferation of bone marrow. METHODS: Fifty elderly patients with AML transformed by abnormal proliferation of bone marrow in the period from March 2012 to May 2014 were chosen and randomly divided into 2 groups including A group (25 elderly patients treated with "3+7" regimen) and B group (25 elderly patients treated with decitabine+HAG pre-excitation regimen), and the clinical effeicacy, the median survival time and the incidence of drug adverse effects in 2 groups were compared. RESULTS: The clinical total remission rate in B group was significantly higher than that in A group(P<0.05). The leukopenia time, neutropenia time and recovery time of leukocyte and neutrophilia in B group were significantly shorter than those in A group, the differences were statistically significant (P<0.05). The rate of lung infection in B group was significantly lower than that in A group. There was no significant difference in the incidence of other drug adverse effects between 2 groups(P>0.05). The life quality of patients in B group was significantly prior to patients in A group, and the difference was statistically significant (P<0.05). The average survival time and the median survival time of patients in B group was significantly longer than those in A group(P<0.05). CONCLUSION: Compared with "3+7" regimen, the decitabine+HAG pre-excitation regimen in the treatment of elderly patients with AML transformed by abnormal proliferation of bone marrow can efficiently delay the disease progression process, shorten the time of bone marrow suppression, decrease the rate of lung infection and prolong the survival time.


Assuntos
Medula Óssea , Síndromes Mielodisplásicas , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica , Azacitidina/análogos & derivados , Proliferação de Células , Decitabina , Humanos , Leucemia Mieloide Aguda , Neutropenia , Qualidade de Vida , Resultado do Tratamento
17.
Can J Cardiol ; 32(9): 1166.e21-8, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27166075

RESUMO

BACKGROUND: The number of elderly people receiving treatment for coronary artery disease (CAD) is increasing, and there are few studies that compared the outcomes of coronary artery bypass grafting (CABG) and percutaneous coronary intervention (PCI) in the elderly. The objective of this study was to compare outcomes of CABG and PCI in octogenarians with CAD. METHODS: We conducted a search to identify articles that reported the results of 2-arm studies that compared CABG with PCI in octogenarians. The primary outcomes were short-term mortality and overall survival, and secondary outcomes included length of hospital stay and cerebrovascular accident (CVA) and myocardial infarction (MI) rates. RESULTS: Seven studies that enrolled 1879 patients who received CABG and 1432 treated with PCI were included. Short-term mortality was significantly less for patients in the PCI arms (odds ratio [OR], 1.47; 95% confidence interval [CI], 1.05-2.06; P = 0.02), as was duration of hospital stay (difference in means, 6.07; 95% CI, 2.81-9.34; P < 0.001). Patients in the CABG arms had longer overall survival (hazard ratio, 0.81; 95% CI, 0.73-0.89; P < 0.001). CVA and MI rates were similar (CVA: OR, 1.06; 95% CI, 0.57-1.95; P = 0.86; MI: OR, 0.70; 95% CI, 0.42-1.17; P = 0.17). CONCLUSIONS: The results suggest that physicians should consider not only the clinical features of CAD, but also the elderly patients future health outlook when choosing a revascularization procedure.


Assuntos
Ponte de Artéria Coronária , Doença da Artéria Coronariana/cirurgia , Intervenção Coronária Percutânea , Idoso de 80 Anos ou mais , Doença da Artéria Coronariana/mortalidade , Humanos , Tempo de Internação , Infarto do Miocárdio/epidemiologia , Acidente Vascular Cerebral/epidemiologia , Análise de Sobrevida
18.
Int J Clin Exp Pathol ; 8(10): 12845-52, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26722475

RESUMO

MicroRNAs are small noncoding RNA molecules that regulate gene expression at the post-transcriptional level. Compelling evidence reveals that there is a causative link between microRNAs deregulation and lung cancer development and metastasis. The aim of present study was to explore the function of miR-140-3p in the development and metastasis of lung cancer cell. Using real-time PCR, we detected the miR-140-3p expression of lung cancer tissues and its pared non-lung cancer tissue. Then, we evaluated the role of miR-140-3p in cell proliferation, invasion and migration using MTT, colony formation assay, Transwell invasion and Transwell migration assay in lung cancer cell lines. As a result, miR-140-3p expression level was lower in lung cancer tissues compared to adjacent normal lung cancer tissue. After miR-140-3p was upregulated in A549 or H1299 cells, cell proliferation, invasion and migration was notably attenuated. Furthermore, we identified ATP6AP2, which is associated with adenosine triphosphatases (ATPases), was a directly target of miR-140-3p in lung cancer cells. In conclusion, our data suggest miR-140-3p/ATP6AP2 axis might act as a potential therapeutic biomarker for lung cancer.


Assuntos
Movimento Celular/genética , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/metabolismo , MicroRNAs/metabolismo , Invasividade Neoplásica/patologia , Receptores de Superfície Celular/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo , Linhagem Celular Tumoral , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , MicroRNAs/genética , Invasividade Neoplásica/genética , Receptores de Superfície Celular/genética , Regulação para Cima , ATPases Vacuolares Próton-Translocadoras/genética
19.
Bing Du Xue Bao ; 30(1): 25-32, 2014 Jan.
Artigo em Chinês | MEDLINE | ID: mdl-24772894

RESUMO

This study aims to analyze the epidemiological features and pathogenic characteristics of hand, foot and mouth disease (HFMD) in Gansu Province, China and to provide a basis for the development of effective prevention and control measures. The descriptive epidemiological analysis was used to analyse the data of HFMD cases in Gansu. The specimens collected from hospitals were subjected to RT-PCR or real-time PCR to detect human enterovirus (HEV) nucleic acid, and HEV strains were isolated using human rhabdomyosarcoma cells and human laryngeal carcinoma cells. The complete VP1-encoding region of several identified enterovirus A71 (EV71) and coxsackievirus A16 (CVA16) was subjected to full-length amplification by RT-PCR and then to sequencing and analysis. A total of 52 550 HFMD cases were reported in Gansu from 2008 to 2012, including 205 severe cases and 27 deaths. The incidence rates in the whole province from 2008 to 2012 were 22.42/10(5), 49.29/10(5), 47.20/10(5), 27.27/10(5), and 55.84/10(5), respectively. There were cases in all the 14 cities or prefectures in Gansu, and Lanzhou had the largest number of cases (16 001 cases), accounting for 30.45% of all cases in the province. HFMD cases were mostly reported during May to July, accounting for 51.69% of all cases throughout the year. The male-to-female ratio was 1.69:1. Of all the cases, 87.59% were under the age of five. Of the 5 416 cases for laboratory tests, 3 322 (61.34%) were positive for HEV nucleic acid, including EV71 (46.96%), CVA16 (41.57%), and other HEVs (11.47%). Among the 186 severe cases, 114 (61.29%) were positive for HEV nucleic acid, and 82.46% of the positive cases for EV71. All the 25 dead cases were infected with EV71. A total of 402 strains were isolated from 3 111 specimens collected from hospitals (2 123 throat swab specimens, 705 stool specimens, and 705 herpes specimens), including EV71 (70.15%), CVA16 (27.11), other coxsackievirus A (3.98%), coxsackievirus B (2.49%), echovirus (1.74%), and adenovirus (1.99%). The genotyping of VP1- encoding region showed that all the 194 EV71 strains isolated during 2008-2012 belonged to the C4a evolutionary branch of C4 subtype; among the 45 CVA16 strains, 12 belonged to the Bla evolutionary branch of B1 subtype and 33 to the B1b evolutionary branch, and B1b became the predominant subtype in 2012. In conclusion, in Gansu Province, HFMD occurs mostly in children under the age of five; EV71 and CVA16 are the main pathogens of this disease, and the two are predominant alternately from 2008 to 2012; the severe and dead cases of HFMD are closely related to infection with EV71; the types of pathogens varied across different regions in the same year during 2008-2012.


Assuntos
Doença de Mão, Pé e Boca/epidemiologia , Doença de Mão, Pé e Boca/virologia , Adolescente , Criança , Pré-Escolar , China/epidemiologia , Análise por Conglomerados , Enterovirus/patogenicidade , Enterovirus/fisiologia , Evolução Molecular , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Adulto Jovem
20.
Mol Med Rep ; 5(2): 483-8, 2012 02.
Artigo em Inglês | MEDLINE | ID: mdl-22012179

RESUMO

Apoptosis of osteoblasts has been proposed as the common basis of osteoporosis, with oxidative stress as the major cause. This study was performed to investigate the protective effect of simvastatin (0.001-0.1 µM) on 100 µM hydrogen peroxide (H2O2)-mediated oxidative stress-induced apoptosis in human osteosarcoma (MG63) cells and the molecular mechanisms involved. Cell apoptosis was evaluated by observation of morphological changes and Annexin V-propidium iodide double staining followed by flow cytometric analysis. MTS assays were used to evaluate cell viability. To investigate the underlying molecular mechanisms, the expression of caspase-3, caspase-9 and Bcl-2 were analyzed by Western blotting. Following stimulation with H2O2 for 24 h, a high proportion of MG63 cells underwent apoptosis, while a dose-dependent inhibition of apoptosis was observed in the presence of simvastatin. A significant, dose-dependent reduction in the expression of caspase-3 and caspase-9 protein induced by H2O2 in MG63 cells was observed in response to simvastatin and the Bcl-2 levels were increased. In conclusion, simvastatin protects MG63 cells from oxidative stress-induced apoptosis through downregulation of caspase-3 and caspase-9 activation and upregulation of Bcl-2 expression, suggesting a protective effect in osteoporosis.


Assuntos
Anticolesterolemiantes/farmacologia , Apoptose/efeitos dos fármacos , Peróxido de Hidrogênio/farmacologia , Mitocôndrias/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais , Sinvastatina/farmacologia , Caspase 3/metabolismo , Caspase 9/metabolismo , Linhagem Celular Tumoral , Regulação para Baixo , Humanos , Osteossarcoma/fisiopatologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA