Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.711
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(31): e2123467119, 2022 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-35881788

RESUMO

Capicua (Cic) proteins are conserved HMG-box transcriptional repressors that control receptor tyrosine kinase (RTK) signaling responses and are implicated in human neurological syndromes and cancer. While Cic is known to exist as short (Cic-S) and long (Cic-L) isoforms with identical HMG-box and associated core regions but distinct N termini, most previous studies have focused on Cic-S, leaving the function of Cic-L unexplored. Here we show that Cic-L acts in two capacities during Drosophila oogenesis: 1) as a canonical sensor of RTK signaling in somatic follicle cells, and 2) as a regulator of postmitotic growth in germline nurse cells. In these latter cells, Cic-L behaves as a temporal signal that terminates endoreplicative growth before they dump their contents into the oocyte. We show that Cic-L is necessary and sufficient for nurse cell endoreplication arrest and induces both stabilization of CycE and down-regulation of Myc. Surprisingly, this function depends mainly on the Cic-L-specific N-terminal module, which is capable of acting independently of the Cic HMG-box-containing core. Mirroring these observations, basal metazoans possess truncated Cic-like proteins composed only of Cic-L N-terminal sequences, suggesting that this module plays unique, ancient roles unrelated to the canonical function of Cic.


Assuntos
Proteínas de Drosophila , Drosophila melanogaster , Proteínas HMGB , Oogênese , Proteínas Repressoras , Animais , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/fisiologia , Proteínas HMGB/genética , Proteínas HMGB/fisiologia , Oogênese/genética , Receptores Proteína Tirosina Quinases/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/fisiologia
2.
Sci Rep ; 12(1): 3011, 2022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35194062

RESUMO

High-grade serous ovarian cancer (HGSOC) is an aggressive malignancy often diagnosed at an advanced stage. Although most HGSOC patients respond initially to debulking surgery combined with cytotoxic chemotherapy, many ultimately relapse with platinum-resistant disease. Thus, improving outcomes requires new ways of limiting metastasis and eradicating residual disease. We identified previously that Liver kinase B1 (LKB1) and its substrate NUAK1 are implicated in EOC spheroid cell viability and are required for efficient metastasis in orthotopic mouse models. Here, we sought to identify additional signalling pathways altered in EOC cells due to LKB1 or NUAK1 loss-of-function. Transcriptome analysis revealed that inflammatory signalling mediated by NF-κB transcription factors is hyperactive due to LKB1-NUAK1 loss in HGSOC cells and spheroids. Upregulated NF-κB signalling due to NUAK1 loss suppresses reactive oxygen species (ROS) production and sustains cell survival in spheroids. NF-κB signalling is also activated in HGSOC precursor fallopian tube secretory epithelial cell spheroids, and is further enhanced by NUAK1 loss. Finally, immunohistochemical analysis of OVCAR8 xenograft tumors lacking NUAK1 displayed increased RelB expression and nuclear staining. Our results support the idea that NUAK1 and NF-κB signalling pathways together regulate ROS and inflammatory signalling, supporting cell survival during each step of HGSOC pathogenesis. We propose that their combined inhibition may be efficacious as a novel therapeutic strategy for advanced HGSOC.


Assuntos
Quinases Proteína-Quinases Ativadas por AMP/genética , Quinases Proteína-Quinases Ativadas por AMP/fisiologia , Mutação com Perda de Função , NF-kappa B/metabolismo , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Proteínas Quinases/genética , Proteínas Quinases/fisiologia , Proteínas Repressoras/genética , Proteínas Repressoras/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Esferoides Celulares , Animais , Sobrevivência Celular , Modelos Animais de Doenças , Feminino , Perfilação da Expressão Gênica , Humanos , Camundongos , Terapia de Alvo Molecular , Transplante de Neoplasias , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/terapia , Espécies Reativas de Oxigênio/metabolismo , Esferoides Celulares/metabolismo , Esferoides Celulares/fisiologia , Transcriptoma/genética , Células Tumorais Cultivadas
3.
Cells ; 11(4)2022 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-35203395

RESUMO

Among the first discovered and most prominent cellular oncogenes is MYC, which encodes a bHLH-ZIP transcription factor (Myc) that both activates and suppresses numerous genes involved in proliferation, energy production, metabolism and translation. Myc belongs to a small group of bHLH-ZIP transcriptional regulators (the Myc Network) that includes its obligate heterodimerization partner Max and six "Mxd proteins" (Mxd1-4, Mnt and Mga), each of which heterodimerizes with Max and largely opposes Myc's functions. More recently, a second group of bHLH-ZIP proteins (the Mlx Network) has emerged that bears many parallels with the Myc Network. It is comprised of the Myc-like factors ChREBP and MondoA, which, in association with the Max-like member Mlx, regulate smaller and more functionally restricted repertoires of target genes, some of which are shared with Myc. Opposing ChREBP and MondoA are heterodimers comprised of Mlx and Mxd1, Mxd4 and Mnt, which also structurally and operationally link the two Networks. We discuss here the functions of these "Extended Myc Network" members, with particular emphasis on their roles in suppressing normal and neoplastic growth. These roles are complex due to the temporal- and tissue-restricted expression of Extended Myc Network proteins in normal cells, their regulation of both common and unique target genes and, in some cases, their functional redundancy.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Neoplasias , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Humanos , Neoplasias/genética , Proteínas Repressoras/fisiologia , Fatores de Transcrição/metabolismo
4.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35217604

RESUMO

BEN domain-containing proteins are emerging rapidly as an important class of factors involved in modulating gene expression, yet the molecular basis of how they regulate chromatin function and transcription remains to be established. BEND3 is a quadruple BEN domain-containing protein that associates with heterochromatin and functions as a transcriptional repressor. We find that BEND3 is highly expressed in pluripotent cells, and the induction of differentiation results in the down-regulation of BEND3. The removal of BEND3 from pluripotent cells results in cells exhibiting upregulation of the differentiation-inducing gene expression signature. We find that BEND3 binds to the promoters of differentiation-associated factors and key cell cycle regulators, including CDKN1A, encoding the cell cycle inhibitor p21, and represses the expression of differentiation-associated genes by enhancing H3K27me3 decoration at these promoters. Our results support a model in which transcription repression mediated by BEND3 is essential for normal development and to prevent differentiation.


Assuntos
Diferenciação Celular/genética , Células-Tronco Pluripotentes/citologia , Proteínas Repressoras/fisiologia , Quadruplex G , Regulação da Expressão Gênica , Humanos , Regiões Promotoras Genéticas
5.
Biochem Biophys Res Commun ; 594: 131-138, 2022 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-35081502

RESUMO

Diabetic nephropathy (DN) is one of the most common causes for end-stage renal disease without effective therapies available. NLR family, pyrin domain-containing 3 (NLRP3) inflammasome possesses a fundamental effect to facilitate the pathogenesis of DN. Unfortunately, how NLRP3 inflammasome is mediated still remains largely unclear. In the present study, an E3 ubiquitin ligase Speckle-type BTB-POZ protein (Spop) was identified as a suppressor of NLRP3 inflammasome. We first showed that Spop expression was extensively down-regulated in kidney of DN patients, which was confirmed in kidney of streptozotocin (STZ)-challenged mice and in high glucose (HG)-stimulated podocytes. Intriguingly, we showed that conditional knockout (cKO) of Spop in podocytes considerably accelerated renal dysfunction and pathological changes in the glomerulus of STZ-induced mice with DN, along with severe podocyte injury. Furthermore, Spop specific ablation in podocytes dramatically facilitated inflammatory response in glomeruli of DN mice via enhancing NLRP3 inflammasome and nuclear factor κB (NF-κB) signaling pathways, which were confirmed in HG-cultured podocytes. Notably, our findings indicated that Spop directly interacted with NLRP3. More importantly, Spop promoted NLRP3 degradation via elevating K48-linked polyubiquitination of NLRP3. Collectively, our findings disclosed a mechanisms through which Spop limited NLRP3 inflammasome under HG condition, and illustrated that Spop may be a novel therapeutic target to suppress NLRP3 inflammasome, contributing to the DN management.


Assuntos
Nefropatias Diabéticas/metabolismo , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Proteínas Nucleares/fisiologia , Proteínas Repressoras/fisiologia , Complexos Ubiquitina-Proteína Ligase/fisiologia , Animais , Glicemia/metabolismo , Regulação para Baixo , Células HEK293 , Humanos , Inflamação , Rim/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Proteínas Nucleares/metabolismo , Podócitos/metabolismo , Transdução de Sinais , Estreptozocina , Ubiquitina/química , Ubiquitina-Proteína Ligases/química
6.
Bioengineered ; 13(2): 2917-2926, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35043753

RESUMO

Transcription factor forkhead box protein 1 (FOXP1) has been shown cardiovascular protection. We aimed to analyze the role of FOXP1 in oxidized low-density lipoprotein (ox-LDL)-induced macrophages and its possible regulatory effect on sestrin1 (SESN1) expression. After stimulation with ox-LDL, FOXP1 expression in RAW264.7 cells was evaluated with RT-qPCR and Western blotting. Then, FOXP1 was overexpressed, followed by detection of inflammatory mediator levels using ELISA kits and RT-qPCR. Lipid accumulation was detected with oil red O staining. Additionally, the JASPAR database was used to predict the potential genes that could be transcriptionally regulated by FOXP1. ChIP and luciferase reporter assays were used to verify this combination. To further clarify the regulatory effects of FOXP1 on SESN1 in damage of macrophages triggered by ox-LDL, SESN1 was silenced to determine the inflammation and lipid accumulation under the condition of FOXP1 overexpression. Results indicated that ox-LDL stimulation led to a significant decrease in FOXP1 expression. FOXP1 overexpression notably reduced the levels of tumor necrosis factor (TNF)-α, interleukin (IL)-1ß and IL-6, accompanied by a decreased in phosphorylated NF-κB p65 expression. Besides, FOXP1-upregulation inhibited lipid accumulation and reduced CD36 expression level in RAW264.7 cells upon ox-LDL stimulation. Moreover, results of ChIP and luciferase reporter assays suggested that FOXP1 could transcriptionally regulate SESN1 expression. Further experiments supported that SESN1 silencing restored the inhibitory effects of FOXP1 overexpression on the inflammation and lipid accumulation in RAW264.7 cells exposed to ox-LDL. Collectively, FOXP1 transcriptionally activates SESN1 for the alleviation of ox-LDL-induced inflammation and lipid accumulation in macrophages.


Assuntos
Proteínas de Ciclo Celular/genética , Fatores de Transcrição Forkhead/fisiologia , Inflamação/genética , Metabolismo dos Lipídeos/genética , Macrófagos/metabolismo , Proteínas Repressoras/fisiologia , Animais , Proteínas de Ciclo Celular/metabolismo , Células Espumosas/metabolismo , Células Espumosas/patologia , Inflamação/induzido quimicamente , Inflamação/metabolismo , Inflamação/patologia , Lipoproteínas LDL , Camundongos , Células RAW 264.7 , Ativação Transcricional/genética
7.
Genes (Basel) ; 13(1)2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-35052467

RESUMO

Reduced cognitive flexibility, characterized by restricted interests and repetitive behavior, is associated with atypical memory performance in autism spectrum disorder (ASD), suggesting hippocampal dysfunction. FOXP1 syndrome is a neurodevelopmental disorder characterized by ASD, language deficits, global developmental delay, and mild to moderate intellectual disability. Strongly reduced Foxp1 expression has been detected in the hippocampus of Foxp1+/- mice, a brain region required for learning and memory. To investigate learning and memory performance in these animals, fear conditioning tests were carried out, which showed impaired associative learning compared with wild type (WT) animals. To shed light on the underlying mechanism, we analyzed various components of the mitochondrial network in the hippocampus. Several proteins regulating mitochondrial biogenesis (e.g., Foxo1, Pgc-1α, Tfam) and dynamics (Mfn1, Opa1, Drp1 and Fis1) were significantly dysregulated, which may explain the increased mitophagy observed in the Foxp1+/- hippocampus. The reduced activity of complex I and decreased expression of Sod2 most likely increase the production of reactive oxygen species and the expression of the pre-apoptotic proteins Bcl-2 and Bax in this tissue. In conclusion, we provide evidence that a disrupted mitochondrial network and the resulting oxidative stress in the hippocampus contribute to the altered learning and cognitive impairment in Foxp1+/- mice, suggesting that similar alterations also play a major role in patients with FOXP1 syndrome.


Assuntos
Modelos Animais de Doenças , Fatores de Transcrição Forkhead/fisiologia , Haploinsuficiência , Aprendizagem/fisiologia , Transtornos da Memória/patologia , Mitocôndrias/patologia , Mitofagia , Proteínas Repressoras/fisiologia , Animais , Hipocampo/metabolismo , Hipocampo/patologia , Transtornos da Memória/etiologia , Transtornos da Memória/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/genética , Mitocôndrias/metabolismo
8.
Mol Med Rep ; 25(1)2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34779499

RESUMO

Our previous study revealed that metastasis­associated protein 1 (MTA1), which is expressed in vascular endothelial cells, acts as a tube formation promoting factor. The present study aimed to clarify the importance of MTA1 expression in tube formation using MTA1­knockout (KO) endothelial cells (MTA1­KO MSS31 cells). Tube formation was significantly suppressed in MTA1­KO MSS31 cells, whereas MTA1­overexpression MTA1­KO MSS31 cells regained the ability to form tube­like structures. In addition, western blotting analysis revealed that MTA1­KO MSS31 cells showed significantly higher levels of phosphorylation of non­muscle myosin heavy chain IIa, which resulted in suppression of tube formation. This effect was attributed to a decrease of MTA1/S100 calcium­binding protein A4 complex formation. Moreover, inhibition of tube formation in MTA1­KO MSS31 cells could not be rescued by stimulation with vascular endothelial growth factor (VEGF). These results demonstrated that MTA1 may serve as an essential molecule for angiogenesis in endothelial cells and be involved in different steps of the angiogenic process compared with the VEGF/VEGF receptor 2 pathway. The findings showed that endothelial MTA1 and its pathway may serve as promising targets for inhibiting tumor angiogenesis, further supporting the development of MTA1­based antiangiogenic therapies.


Assuntos
Células Endoteliais/metabolismo , Neovascularização Patológica/metabolismo , Proteínas Repressoras/metabolismo , Transativadores/metabolismo , Inibidores da Angiogênese/farmacologia , Animais , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Células Cultivadas , China , Camundongos , Morfogênese/efeitos dos fármacos , Cadeias Pesadas de Miosina/metabolismo , Metástase Neoplásica/genética , Fosforilação , Proteínas Repressoras/fisiologia , Proteína A4 de Ligação a Cálcio da Família S100/metabolismo , Transativadores/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Fatores de Crescimento do Endotélio Vascular/metabolismo
9.
Prog Neurobiol ; 208: 102177, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34582949

RESUMO

The tangential migration of immature neurons in the postnatal brain involves consecutive migration cycles and depends on constant remodeling of the cell cytoskeleton, particularly in the leading process (LP). Despite the identification of several proteins with permissive and empowering functions, the mechanisms that specify the direction of migration remain largely unknown. Here, we report that planar cell polarity protein Celsr3 orients neuroblasts migration from the subventricular zone (SVZ) to olfactory bulb (OB). In Celsr3-forebrain conditional knockout mice, neuroblasts loose directionality and few can reach the OB. Celsr3-deficient neuroblasts exhibit aberrant branching of LP, de novo LP formation, and decreased growth rate of microtubules (MT). Mechanistically, we show that Celsr3 interacts physically with Kif2a, a MT depolymerizing protein and that conditional inactivation of Kif2a in the forebrain recapitulates the Celsr3 knockout phenotype. Our findings provide evidence that Celsr3 and Kif2a cooperatively specify the directionality of neuroblasts tangential migration in the postnatal brain.


Assuntos
Caderinas , Cinesinas , Células-Tronco Neurais , Neurogênese , Receptores de Superfície Celular , Proteínas Repressoras , Animais , Caderinas/fisiologia , Movimento Celular/fisiologia , Cinesinas/fisiologia , Ventrículos Laterais/metabolismo , Camundongos , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Bulbo Olfatório/fisiologia , Receptores de Superfície Celular/fisiologia , Proteínas Repressoras/fisiologia
10.
Pathol Res Pract ; 229: 153693, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34826740

RESUMO

BACKGROUND: Small cell lung cancer (SCLC) is a malignant lung neuroendocrine tumor with early metastasis, rapid progression, and poor outcomes. Insulinoma-associated protein 1 (INSM1) has been an excellent marker for neuroendocrine (NE) differentiation and widely used in the diagnosis of NE neoplasms, including SCLC. However, its role beyond NE diagnostic marker remained little reported. METHODS: We examined immunohistochemical expression of INSM1 in 73 surgically resected SCLC, analyzed its prognostic value by Kaplan-Meier method, and investigated clinical-pathological features of INSM1 high SCLC. In vitro, We assessed INSM1 function on glucose intake, tumor migration, and Cisplatin resistance by 2-NBDG glucose uptake fluorescent assay, transwell assay, and ANNEXIN V/PI assay, respectively. In vivo, we evaluated the therapeutic value of metformin on reversing INSM1 induced chemoresistance by BALB/c nude mice xenograft tumor model. RESULTS: High INSM1 expression was correlated with lymph node metastasis (LNM) (p = 0.0005), later TNM stages (p = 0.0003), and predicted poor survival (Log-rank p = 0.038). Multivariate Cox analysis confirmed INSM1 as an independent prognostic factor in SCLC (p = 0.012, HR:3.195, 95%CI:1.288-7.927). Interestingly, LNM was correlated with worse prognosis only in patients received chemotherapy (Log-rank p = 0.027) rather than the others (Log-rank p = 0.40). In patients having LNM and treated with chemotherapy, high INSM1 was correlated with worse clinic outcome (Log-rank p = 0.009). In vitro, overexpression of INSM1 decreased AMPK-α expression as well as glucose intake, promoted tumor cell migration, and limited the apoptosis induced by Cisplatin, which all could be reversed by Metformin. In vivo, INSM1 overexpression also contributed to tumor growth beyond inducing Cisplatin resistance. CONCLUSION: Our finding suggested INSM1 played more role than a NE marker, partly through down-regulating AMPK signal. INSM1 may serve as a novel prognostic marker and therapeutic target in SCLC.


Assuntos
Neoplasias Pulmonares/etiologia , Proteínas Repressoras/fisiologia , Carcinoma de Pequenas Células do Pulmão/etiologia , Animais , Biomarcadores Tumorais , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Prognóstico
11.
Clin Transl Oncol ; 24(1): 145-153, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34216368

RESUMO

PURPOSE: Vascular mimicry (VM) tubules are lumen structures comprised of malignant tumor cells without the participation of endothelial cells. VM simulates blood vessel function in tumors to deliver a sufficient blood supply for proliferation, invasion, and metastasis of malignant tumors, thereby reducing the clinical effects of anti-angiogenic treatments. The elimination or prevention of malignant tumor VM development therefore represents an urgent research goal as a therapeutic strategy to and cut off nutrients required for tumor growth. The GATA transcription factor TRPS1 is abnormally up-regulated in breast cancer, osteosarcoma, prostate cancer, and other tumor tissues, and is instrumental in regulating cell proliferation, differentiation, and tissue growth and development. METHODS: Here, we explored the effects of TRPS1 knockdown on VM and the proteins underlying its development in triple-negative breast cancer cell line MDA-MB-231. RESULTS: We found that TRPS1 knockdown resulted in obvious inhibition of VM development. Fluorescence microscopy of F-actin and tubulin revealed that loss of TRPS1 function resulted in disruption of cytoskeleton and microtubule formation, respectively. In addition, TRPS1-suppressed cells exhibited reduced accumulation of VM-associated proteins EphA2, MMP-2, MMP-9, VEGF, and VE-cadherin. Moreover, it is interesting to know that the capacity for migration and invasion were limited in MDA-MB-231cells after TRPS1 knockdown and that the average number of VM tubules, their length, and number of intersections were also significantly decreased. CONCLUSIONS: Based on our results, and in light of previous studies, we thus proposed that TRPS1 suppression negatively affects vascular mimicry possibly through reduced TRPS1-mediated transcriptional regulation of VM-related protein VEGF-A.


Assuntos
Neovascularização Patológica , Proteínas Repressoras/fisiologia , Neoplasias de Mama Triplo Negativas/irrigação sanguínea , Neoplasias de Mama Triplo Negativas/patologia , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Humanos , Proteínas Repressoras/genética
12.
Leukemia ; 36(2): 370-382, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34465864

RESUMO

T-cell acute lymphoblastic leukemia (T-ALL) is a malignant hematologic disease caused by gene mutations in T-cell progenitors. As an important epigenetic regulator, PHF6 mutations frequently coexist with JAK3 mutations in T-ALL patients. However, the role(s) of PHF6 mutations in JAK3-driven leukemia remain unclear. Here, the cooperation between JAK3 activation and PHF6 inactivation is examined in leukemia patients and in mice models. We found that the average survival time is shorter in patients with JAK/STAT and PHF6 comutation than that in other patients, suggesting a potential role of PHF6 in leukemia progression. We subsequently found that Phf6 deficiency promotes JAK3M511I-induced T-ALL progression in mice by inhibiting the Bai1-Mdm2-P53 signaling pathway, which is independent of the JAK3/STAT5 signaling pathway. Furthermore, combination therapy with a JAK3 inhibitor (tofacitinib) and a MDM2 inhibitor (idasanutlin) reduces the Phf6 KO and JAK3M511I leukemia burden in vivo. Taken together, our study suggests that combined treatment with JAK3 and MDM2 inhibitors may potentially increase the drug benefit for T-ALL patients with PHF6 and JAK3 comutation.


Assuntos
Transformação Celular Neoplásica/patologia , Janus Quinase 3/metabolismo , Mutação , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patologia , Proteínas Repressoras/fisiologia , Animais , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Humanos , Janus Quinase 3/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Leucemia-Linfoma Linfoblástico de Células T Precursoras/etiologia , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Transdução de Sinais
13.
Mediators Inflamm ; 2021: 6359652, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34924813

RESUMO

Ellagic acid (EA) was reported to play protective roles in rheumatoid arthritis (RA). It was found that the level of metastasis-associated gene 1 (MTA1)/histone deacetylase 1 (HDAC1) protein complex was downregulated by polyphenols in several human disorders. Notably, inhibition of MTA1 or HDAC1 has anti-inflammatory effects on RA. Therefore, our study is aimed at investigating whether EA prevents RA progression through regulating the MTA1/HDAC1 complex. Herein, the human fibroblast-like synoviocyte (FLS) cell line MH7A was treated with TNF-α to induce an inflammation model in vitro and then incubated with different concentrations of EA. Western blot analysis showed that EA reduced MTA1 expression in a dose-dependent manner in MH7A cells. Then, TNF-α-treated MH7A cells were incubated with EA alone or together with MTA1 overexpression plasmid (pcDNA-MTA1), and we found that EA inhibited proliferation, inflammation cytokine levels, and oxidative stress marker protein levels and promoted apoptosis in MH7A cells, while MTA1 overexpression abolished these effects. Moreover, coimmunoprecipitation assay verified the interaction between MTA1 and HDAC1. EA downregulated the MTA1/HDAC1 complex in MH7A cells. MTA1 knockdown inhibited proliferation, inflammation, and oxidative stress and promoted apoptosis in MH7A cells, while HDAC1 overexpression reversed these effects. Moreover, chromatin immunoprecipitation assay indicated that EA inhibited HDAC1-mediated Nur77 deacetylation. Rescue experiments demonstrated that Nur77 knockdown reversed the effects of EA on MH7A cell biological behaviors. Additionally, EA treatment attenuated arthritis index, paw swelling, synovial hyperplasia, and inflammation in collagen-induced arthritis (CIA) rats. In conclusion, EA inhibited proliferation, inflammation, and oxidative stress and promoted apoptosis in MH7A cells and alleviated the severity of RA in CIA rats though downregulating MTA1/HDAC1 complex and promoting HDAC1 deacetylation-mediated Nur77 expression.


Assuntos
Artrite Reumatoide/tratamento farmacológico , Ácido Elágico/farmacologia , Histona Desacetilase 1/antagonistas & inibidores , Proteínas Repressoras/antagonistas & inibidores , Transativadores/antagonistas & inibidores , Acetilação , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Histona Desacetilase 1/fisiologia , Humanos , Masculino , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/fisiologia , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Wistar , Proteínas Repressoras/fisiologia , Transativadores/fisiologia
14.
Int J Mol Sci ; 22(23)2021 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-34884676

RESUMO

Indirect neurogenesis, during which neural stem cells generate neurons through intermediate progenitors, drives the evolution of lissencephalic brains to gyrencephalic brains. The mechanisms that specify intermediate progenitor identity and that regulate stem cell competency to generate intermediate progenitors remain poorly understood despite their roles in indirect neurogenesis. Well-characterized lineage hierarchy and available powerful genetic tools for manipulating gene functions make fruit fly neural stem cell (neuroblast) lineages an excellent in vivo paradigm for investigating the mechanisms that regulate neurogenesis. Type II neuroblasts in fly larval brains repeatedly undergo asymmetric divisions to generate intermediate neural progenitors (INPs) that undergo limited proliferation to increase the number of neurons generated per stem cell division. Here, we review key regulatory genes and the mechanisms by which they promote the specification and generation of INPs, safeguarding the indirect generation of neurons during fly larval brain neurogenesis. Homologs of these regulators of INPs have been shown to play important roles in regulating brain development in vertebrates. Insight into the precise regulation of intermediate progenitors will likely improve our understanding of the control of indirect neurogenesis during brain development and brain evolution.


Assuntos
Células-Tronco Neurais/fisiologia , Neurogênese/genética , Animais , Proteínas de Drosophila/fisiologia , Humanos , Proteínas Repressoras/fisiologia
15.
Thorac Cancer ; 12(24): 3356-3369, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34741437

RESUMO

BACKGROUND: Lung cancer is the most prevalent malignancy in adults. Circular RNA (circRNA) circCPA4 (hsa_circ_0082374) is highly expressed in non-small cell lung cancer (NSCLC). The purpose of this study was to explore the role and mechanism of circCPA4 in lung cancer. METHODS: CircCPA4, linear CPA4, TGF-ß-induced factor homeobox 2 (TGIF2), and microRNA-214-3p (miR-214-3p) levels were measured by real-time quantitative polymerase chain reaction (RT-qPCR). The protein levels of TGIF2, Beclin1, and p62 were assessed by western blot assay. Colony numbers, migration, invasion, apoptosis, and cell cycle progression were examined by colony formation, wound-healing, transwell, and flow cytometry assays, respectively. The binding relationship between miR-214-3p and circCPA4 or TGIF2 was predicted by StarBase or TargetScan and then verified by a dual-luciferase reporter, RNA immunoprecipitation (RIP), and RNA pulldown assays. The biological role of circCPA4 on lung tumor growth was assessed by a xenograft tumor model in vivo, and TGIF2 and ki-67 expression was assessed by immunohistochemistry. RESULTS: We determined that CircCPA4 and TGIF2 were increased, and miR-214-3p was decreased in lung cancer tissues and cells. Functionally, circCPA4 knockdown could suppress colony formation, migration, invasion, cell cycle progression, and expedite apoptosis of lung cancer cells in vitro. Mechanically, circCPA4 could regulate TGIF2 expression by sponging miR-214-3p. In addition, circCPA4 deficiency inhibited the tumor growth in lung cancer in the mouse model. CONCLUSIONS: CircCPA4 could act as a sponge of miR-214-3p to upregulate TGIF2 expression, thereby promoting the progression of lung cancer cells. These findings suggested underlying therapeutic targets for the treatment of lung cancer.


Assuntos
Carcinogênese/genética , Proteínas de Homeodomínio/genética , Neoplasias Pulmonares/genética , MicroRNAs/genética , RNA Circular/genética , Proteínas Repressoras/genética , Idoso , Animais , Apoptose/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Regulação para Baixo , Feminino , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , MicroRNAs/fisiologia , Pessoa de Meia-Idade , Proteínas Repressoras/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto
16.
J Biol Chem ; 297(6): 101372, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34756885

RESUMO

Chronic exposure to high levels of manganese (Mn) leads to manganism, a neurological disorder with similar symptoms to those inherent to Parkinson's disease. However, the underlying mechanisms of this pathological condition have yet to be established. Since the human excitatory amino acid transporter 2 (EAAT2) (glutamate transporter 1 in rodents) is predominantly expressed in astrocytes and its dysregulation is involved in Mn-induced excitotoxic neuronal injury, characterization of the mechanisms that mediate the Mn-induced impairment in EAAT2 function is crucial for the development of novel therapeutics against Mn neurotoxicity. Repressor element 1-silencing transcription factor (REST) exerts protective effects in many neurodegenerative diseases. But the effects of REST on EAAT2 expression and ensuing neuroprotection are unknown. Given that the EAAT2 promoter contains REST binding sites, the present study investigated the role of REST in EAAT2 expression at the transcriptional level in astrocytes and Mn-induced neurotoxicity in an astrocyte-neuron coculture system. The results reveal that astrocytic REST positively regulates EAAT2 expression with the recruitment of an epigenetic modifier, cAMP response element-binding protein-binding protein/p300, to its consensus binding sites in the EAAT2 promoter. Moreover, astrocytic overexpression of REST attenuates Mn-induced reduction in EAAT2 expression, leading to attenuation of glutamate-induced neurotoxicity in the astrocyte-neuron coculture system. Our findings demonstrate that astrocytic REST plays a critical role in protection against Mn-induced neurotoxicity by attenuating Mn-induced EAAT2 repression and the ensuing excitotoxic dopaminergic neuronal injury. This indicates that astrocytic REST could be a potential molecular target for the treatment of Mn toxicity and other neurological disorders associated with EAAT2 dysregulation.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Transportador 2 de Aminoácido Excitatório/metabolismo , Manganês/farmacologia , Proteínas Repressoras/fisiologia , Regulação para Cima/fisiologia , Animais , Astrócitos/metabolismo , Linhagem Celular , Neurônios Dopaminérgicos/efeitos dos fármacos , Transportador 2 de Aminoácido Excitatório/genética , Ácido Glutâmico/metabolismo , Humanos , Camundongos , Regiões Promotoras Genéticas , Sequências Reguladoras de Ácido Nucleico , Transcrição Gênica/fisiologia
17.
Dev Cell ; 56(22): 3128-3145.e15, 2021 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-34762852

RESUMO

Identification of physiological modulators of nuclear hormone receptor (NHR) activity is paramount for understanding the link between metabolism and transcriptional networks that orchestrate development and cellular physiology. Using libraries of metabolic enzymes alongside their substrates and products, we identify 1-deoxysphingosines as modulators of the activity of NR2F1 and 2 (COUP-TFs), which are orphan NHRs that are critical for development of the nervous system, heart, veins, and lymphatic vessels. We show that these non-canonical alanine-based sphingolipids bind to the NR2F1/2 ligand-binding domains (LBDs) and modulate their transcriptional activity in cell-based assays at physiological concentrations. Furthermore, inhibition of sphingolipid biosynthesis phenocopies NR2F1/2 deficiency in endothelium and cardiomyocytes, and increases in 1-deoxysphingosine levels activate NR2F1/2-dependent differentiation programs. Our findings suggest that 1-deoxysphingosines are physiological regulators of NR2F1/2-mediated transcription.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Organogênese/efeitos dos fármacos , Esfingolipídeos/farmacologia , Animais , Diferenciação Celular/fisiologia , Regulação da Expressão Gênica/fisiologia , Humanos , Vasos Linfáticos/efeitos dos fármacos , Camundongos , Organogênese/fisiologia , Proteínas Repressoras/fisiologia
18.
Cancer Res ; 81(24): 6183-6195, 2021 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-34666993

RESUMO

Neoadjuvant immunotherapy, given before surgical resection, is a promising approach to develop systemic antitumor immunity for the treatment of high-risk resectable disease. Here, using syngeneic and orthotopic mouse models of triple-negative breast cancer, we have tested the hypothesis that generation of tumor-specific T-cell responses by induction and activation of tumor-residing Batf3-dependent conventional type 1 dendritic cells (cDC1) before resection improves control of distant metastatic disease and survival. Mice bearing highly metastatic orthotopic tumors were treated with a combinatorial in situ immunomodulation (ISIM) regimen comprised of intratumoral administration of Flt3L, local radiotherapy, and in situ TLR3/CD40 stimulations, followed by surgical resection. Neoadjuvant ISIM (neo-ISIM) generated tumor-specific CD8+ T cells that infiltrated into distant nonirradiated metastatic sites, which delayed the progression of lung metastases and improved survival after the resection of primary tumors. The efficacy of neo-ISIM was dependent on de novo adaptive T-cell immunity elicited by Batf3-dependent dendritic cells and was enhanced by increasing dose and fractionation of radiotherapy, and early surgical resection after the completion of neo-ISIM. Importantly, neo-ISIM synergized with programmed cell death protein-1 ligand-1 (PD-L1) blockade to improve control of distant metastases and prolong survival, while removal of tumor-draining lymph nodes abrogated the antimetastatic efficacy of neo-ISIM. Our findings illustrate the therapeutic potential of neoadjuvant multimodal intralesional therapy for the treatment of resectable tumors with high risk of relapse. SIGNIFICANCE: Neoadjuvant induction and activation of cDC1s in primary tumors enhances systemic antitumor immunity, suppresses metastatic progression, improves survival, and synergizes with anti-PD-L1 therapy.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/fisiologia , Neoplasias da Mama/terapia , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Imunomodulação , Neoplasias Pulmonares/terapia , Terapia Neoadjuvante/métodos , Proteínas Repressoras/fisiologia , Animais , Apoptose , Antígeno B7-H1/antagonistas & inibidores , Neoplasias da Mama/imunologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proliferação de Células , Terapia Combinada , Feminino , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Imunoterapia , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Mastectomia , Proteínas de Membrana/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Radioterapia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Biochemistry ; 60(44): 3315-3322, 2021 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-34714626

RESUMO

Cellular hypoxia plays a crucial role in tissue development and adaptation to pO2. Central to cellular oxygen sensing is factor-inhibiting HIF-1α (FIH), an α-ketoglutarate (αKG)/non-heme iron(II)-dependent dioxygenase that hydroxylates a specific asparagine residue of hypoxia inducible factor-1α (HIF-1α). The high KM(O2) and rate-limiting decarboxylation step upon O2 activation are key features of the enzyme that classify it as an oxygen sensor and set it apart from other αKG/Fe(II)-dependent dioxygenases. Although the chemical intermediates following decarboxylation are presumed to follow the consensus mechanism of other αKG/Fe(II)-dependent dioxygenases, experiments have not previously demonstrated these canonical steps in FIH. In this work, a deuterated peptide substrate was used as a mechanistic probe for the canonical hydrogen atom transfer (HAT). Our data show a large kinetic isotope effect (KIE) in steady-state kinetics (Dkcat = 10 ± 1), revealing that the HAT occurs and is partially rate limiting on kcat. Kinetic studies showed that the deuterated peptide led FIH to uncouple O2 activation and provided the opportunity to spectroscopically observe the ferryl intermediate. This enzyme uncoupling was used as an internal competition with respect to the fate of the ferryl intermediate, demonstrating a large observed KIE on the uncoupling (Dk5 = 1.147 ± 0.005) and an intrinsic KIE on the HAT step (Dk > 15). The close energy barrier between αKG decarboxylation and HAT distinguishes FIH as an O2-sensing enzyme and is crucial for ensuring substrate specificity in the regulation of cellular O2 homeostasis.


Assuntos
Hipóxia Celular/fisiologia , Hidrogênio/metabolismo , Oxigenases de Função Mista/metabolismo , Proteínas Repressoras/metabolismo , Aclimatação , Adaptação Fisiológica , Dioxigenases/metabolismo , Metabolismo Energético/fisiologia , Homeostase , Humanos , Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Isótopos , Cinética , Oxigenases de Função Mista/fisiologia , Proteínas Repressoras/fisiologia , Especificidade por Substrato
20.
Eur Rev Med Pharmacol Sci ; 25(18): 5610-5618, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34604953

RESUMO

OBJECTIVE: This study aims to explore the biological roles of long non-coding RNA (lncRNA) ZNF281 and KLF15 in regulating cervical carcinoma progression. PATIENTS AND METHODS: Differential expressions of ZNF281 in 58 collected cervical carcinoma and normal tissues were detected by quantitative real-time polymerase chain reaction (qRT-PCR). The relationship between ZNF281 and clinicopathologic characteristics in cervical carcinoma patients was analyzed. By generating ZNF281 knockdown model in HeLa and SiHa cells through the transfection of shZNF281, migratory ability changes were examined via transwell and wound healing assay. The role of ZNF281 in in vivo tumorgenicity of cervical carcinoma was examined by implanting xenografted cancers in nude mice. The downstream target of ZNF281 and their interaction were assessed by bioinformatics tool and Dual-Luciferase reporter assay, respectively. Finally, co-regulations of ZNF281 and KLF15 on cervical carcinoma progression were elucidated. RESULTS: ZNF281 was upregulated in cervical carcinoma tissues and cell lines. It was correlated to TNM staging, and incidences of lymphatic metastasis and distant metastasis in cervical carcinoma patients, while it was unrelated to age and tumor size. The knockdown of ZNF281 effectively attenuated migratory ability in HeLa and SiHa cells. Besides, knockdown of ZNF281 also reduced tumorigenicity of cervical carcinoma in nude mice. KLF15 was the downstream gene binding ZNF281, and they were negatively correlated to each other in cervical carcinoma tissues. Notably, KLF15 was responsible for ZNF281-induced regulation on cervical carcinoma migration. CONCLUSIONS: LncRNA ZNF281 is upregulated in cervical carcinoma samples, and it is correlated to lymphatic metastasis, distant metastasis, and poor prognosis in cervical carcinoma patients. By targeting KLF15, ZNF281 triggers migratory potential in cervical carcinoma. We believed that ZNF281 is a promising biomarker for cervical carcinoma.


Assuntos
Regulação Neoplásica da Expressão Gênica/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Fatores de Transcrição Kruppel-Like/fisiologia , RNA Longo não Codificante/metabolismo , RNA Longo não Codificante/fisiologia , Proteínas Repressoras/metabolismo , Proteínas Repressoras/fisiologia , Neoplasias do Colo do Útero/genética , Animais , Carcinogênese/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Progressão da Doença , Feminino , Células HeLa , Humanos , Metástase Linfática/genética , Camundongos Nus , Ligação Proteica/genética , Neoplasias do Colo do Útero/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...