Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Free Radic Biol Med ; 219: 215-230, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38636715

RESUMO

Selenium (Se) is indispensable in alleviating various types of intestinal injuries. Here, we thoroughly investigated the protective effect of Se on the regulation of the epithelial cell-M2 macrophages pathway in deoxynivalenol (DON)-induced intestinal damage. In the present study, Se has positive impacts on gut health by improving gut barrier function and reducing the levels of serum DON in vivo. Furthermore, our study revealed that Se supplementation increased the abundances of GPX4, p-PI3K, and AKT, decreased the levels of 4-HNE and inhibited ferroptosis. Moreover, when mice were treated with DON and Fer-1(ferroptosis inhibitor), ferroptosis was suppressed and PI3K/AKT pathway was activated. These results indicated that GPX4-PI3K/AKT-ferroptosis was a predominant pathway in DON-induced intestinal inflammation. Interestingly, we discovered that both the number of M2 anti-inflammatory macrophages and the levels of CSF-1 decreased while the pro-inflammatory cytokine IL-6 increased in the intestine and MODE-K cells supernatant. Therefore, Se supplementation activated the CSF-1-M2 macrophages axis, resulting in a decrease in IL-6 expression and an enhancement of the intestinal anti-inflammatory capacity. This study provides novel insights into how intestinal epithelial cells regulate the CSF-1-M2 macrophage pathway, which is essential in maintaining intestinal homeostasis confer to environmental hazardous stimuli.


Assuntos
Células Epiteliais , Mucosa Intestinal , Macrófagos , Selênio , Tricotecenos , Animais , Tricotecenos/toxicidade , Camundongos , Macrófagos/metabolismo , Macrófagos/efeitos dos fármacos , Selênio/farmacologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/patologia , Células Epiteliais/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Ativação de Macrófagos/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Transdução de Sinais/efeitos dos fármacos , Ferroptose/efeitos dos fármacos , Masculino , Fosfatidilinositol 3-Quinases/metabolismo
2.
Cancer Res ; 84(3): 419-433, 2024 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-37991725

RESUMO

Despite the immense success of immune checkpoint blockade (ICB) in cancer treatment, many tumors, including melanoma, exhibit innate or adaptive resistance. Tumor-intrinsic T-cell deficiency and T-cell dysfunction have been identified as essential factors in the emergence of ICB resistance. Here, we found that protein arginine methyltransferase 1 (PRMT1) expression was inversely correlated with the number and activity of CD8+ T cells within melanoma specimen. PRMT1 deficiency or inhibition with DCPT1061 significantly restrained refractory melanoma growth and increased intratumoral CD8+ T cells in vivo. Moreover, PRMT1 deletion in melanoma cells facilitated formation of double-stranded RNA derived from endogenous retroviral elements (ERV) and stimulated an intracellular interferon response. Mechanistically, PRMT1 deficiency repressed the expression of DNA methyltransferase 1 (DNMT1) by attenuating modification of H4R3me2a and H3K27ac at enhancer regions of Dnmt1, and DNMT1 downregulation consequently activated ERV transcription and the interferon signaling. Importantly, PRMT1 inhibition with DCPT1061 synergized with PD-1 blockade to suppress tumor progression and increase the proportion of CD8+ T cells as well as IFNγ+CD8+ T cells in vivo. Together, these results reveal an unrecognized role and mechanism of PRMT1 in regulating antitumor T-cell immunity, suggesting PRMT1 inhibition as a potent strategy to increase the efficacy of ICB. SIGNIFICANCE: Targeting PRMT1 stimulates interferon signaling by increasing expression of endogenous retroviral elements and double-stranded RNA through repression of DNMT1, which induces antitumor immunity and synergizes with immunotherapy to suppress tumor progression.


Assuntos
Interferons , Melanoma , Humanos , Melanoma/metabolismo , RNA de Cadeia Dupla , Linfócitos T CD8-Positivos , Metiltransferases/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo
3.
ACS Biomater Sci Eng ; 10(1): 455-467, 2024 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-38146624

RESUMO

Nanoattapulgite (nano-ATP), a magnesium-aluminum silicate clay, can absorb substances and is a suitable material for bone repair and regeneration. In this study, using three-dimensional printing technology, a nano-ATP/polycaprolactone (PCL) scaffold was fabricated and modified using NaOH to form a rough surface. Biomimetic hydroxyapatite (HA) on nano-ATP/PCL scaffolds was fabricated using a biomineralized approach. The scaffold provided structural support through PCL and was modified with ATP and HA to improve hydrophilicity and promote the delivery of nutrients. The biocompatibility and osteogenic induction of scaffolds were assessed in vitro using mouse bone marrow mesenchymal stem cells. According to the in vitro study results, the nano-ATP/PCL/HA composite scaffold significantly boosted the expression levels of genes related to osteogenesis (p < 0.05), attributed to its superior alkaline phosphatase activity and calcium deposition capabilities. The outcomes of in vivo experimentation demonstrated an augmentation in bone growth at the rat cranial defect site when treated with the ATP/PCL/HA composite scaffold. It can be inferred from the results that the implementation of ATP and HA for the bone tissue engineering repair material displays encouraging prospects.


Assuntos
Durapatita , Alicerces Teciduais , Ratos , Camundongos , Animais , Durapatita/farmacologia , Durapatita/química , Alicerces Teciduais/química , Engenharia Tecidual/métodos , Biomimética , Regeneração Óssea , Impressão Tridimensional , Trifosfato de Adenosina/farmacologia , Crânio
4.
Bioinformatics ; 39(11)2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37934154

RESUMO

MOTIVATION: Recent frameworks based on deep learning have been developed to identify cancer subtypes from high-throughput gene expression profiles. Unfortunately, the performance of deep learning is highly dependent on its neural network architectures which are often hand-crafted with expertise in deep neural networks, meanwhile, the optimization and adjustment of the network are usually costly and time consuming. RESULTS: To address such limitations, we proposed a fully automated deep neural architecture search model for diagnosing consensus molecular subtypes from gene expression data (DNAS). The proposed model uses ant colony algorithm, one of the heuristic swarm intelligence algorithms, to search and optimize neural network architecture, and it can automatically find the optimal deep learning model architecture for cancer diagnosis in its search space. We validated DNAS on eight colorectal cancer datasets, achieving the average accuracy of 95.48%, the average specificity of 98.07%, and the average sensitivity of 96.24%, respectively. Without the loss of generality, we investigated the general applicability of DNAS further on other cancer types from different platforms including lung cancer and breast cancer, and DNAS achieved an area under the curve of 95% and 96%, respectively. In addition, we conducted gene ontology enrichment and pathological analysis to reveal interesting insights into cancer subtype identification and characterization across multiple cancer types. AVAILABILITY AND IMPLEMENTATION: The source code and data can be downloaded from https://github.com/userd113/DNAS-main. And the web server of DNAS is publicly accessible at 119.45.145.120:5001.


Assuntos
Neoplasias da Mama , Aprendizado Profundo , Humanos , Feminino , Redes Neurais de Computação , Algoritmos , Software
5.
Bioorg Med Chem ; 84: 117262, 2023 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-37018878

RESUMO

Autophagy related 4B (ATG4B) which regulates autophagy by promoting the formation of autophagosome through reversible modification of LC3, is closely related to cancer cell growth and drug resistance, and therefore is an attractive therapeutic target. Recently, ATG4B inhibitors have been reported, yet with drawbacks including weak potency. To discover more promising ATG4B inhibitors, we developed a high-throughput screening (HTS) assay and identified a new ATG4B inhibitor named DC-ATG4in. DC-ATG4in directly binds to ATG4B and inhibits its enzyme activity with an IC50 of 3.08 ± 0.47 µM. We further confirmed that DC-ATG4in is an autophagy inhibitor and blocks autophagy induced by Sorafenib in Hepatocellular Carcinoma (HCC) cells. More importantly, combination of DC-ATG4in with Sorafenib synergized the cancer cell killing effect and proliferation inhibition activities on HCC cells. Our data suggested that inactivation of autophagy via ATG4B inhibition may be a viable strategy to sensitize existing targeted therapy such as Sorafenib in the future.


Assuntos
Proteínas Relacionadas à Autofagia , Autofagia , Sorafenibe , Humanos , Autofagia/efeitos dos fármacos , Proteínas Relacionadas à Autofagia/antagonistas & inibidores , Proteínas Relacionadas à Autofagia/metabolismo , Carcinoma Hepatocelular/tratamento farmacológico , Cisteína Endopeptidases/metabolismo , Neoplasias Hepáticas/tratamento farmacológico , Sorafenibe/farmacologia , Sorafenibe/uso terapêutico
6.
Front Immunol ; 13: 1028418, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36569896

RESUMO

The extracellular vesicles (EVs) in edible food have a typical saucer-like structure and are nanoparticles released by numerous cells. They have different components and interact with other biological samples in diverse ways. Therefore, these nanoparticles could be used to develop bioactives delivery nanoplatforms and anti-inflammatory treatments to meet the stringent demands of current clinical challenges. This review aims to summarize current researches into EVs from edible plants, particularly those that can protect siRNAs or facilitate drug transportation. We will discuss their isolation, characterization and functions, their regulatory effects under various physiological and pathological conditions, and their immune regulation, anti-tumor, regeneration, and anti-inflammatory effects. We also review advances in their potential application as bioactives carriers, and medicinal and edible plants that change their EVs compositions during disease to achieve a therapy propose. It is expected that future research on plant-derived EVs will considerably expand their application.


Assuntos
Vesículas Extracelulares , Neoplasias , Plantas Comestíveis , Vesículas Extracelulares/patologia , Sistemas de Liberação de Medicamentos , Neoplasias/patologia , Anti-Inflamatórios
7.
Front Vet Sci ; 9: 1016972, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36419727

RESUMO

Echinococcosis, also known as hydatid disease, is caused by the metacestode stage of the species cluster Echinococcus granulosus sensu lato (E. granulosus s.l.). It is almost widespread worldwide, especially in countries and regions dominated by animal husbandry. It is a major parasitic disease that seriously endangers human health, public health safety, environmental safety, and the development of animal husbandry production in western China. In this study, the mitochondrial cox1 gene was used to analyze the genetic diversity and haplotype of bovine and sheep echinococcal cysts isolated in Tibet. Echinococcus granulosus sensu stricto (E. granulosus s.s., G1, G3) was still the dominant species in the infected samples of yak and sheep in some parts of Tibet. Through haplotype analysis, Hap_1 was deemed the dominant haplotype, 14 of the 20 haplotypes were similar to the reference sequence previously published in Genbank, and the rest of the 6 haplotypes were found for the first time. Through Tajima's D value, neutral test Fu's Fs analysis, and haplotype network map, it can be concluded that Echinococcus population expansion has occurred in Xigaze, Tibet. This study provides basic data for understanding the genetic characteristics, epidemiology, and control of echinococcosis in this area.

8.
Microorganisms ; 10(10)2022 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-36296277

RESUMO

Butyrate provides energy for colonocytes and is a functional metabolite that mitigates weanling piglet stress. However, its effects and mechanisms remain largely unknown. We established a lipopolysaccharide (LPS)-induced inflammatory stress piglet model to examine how butyrate mechanisms impacted piglet intestinal histology, microbiota, and inflammation. We randomly assigned 18 crossbred male piglets to three treatment groups: CON, LPS, and BT-LPS. Coated butyrate was supplemented in the BT-LPS feed for 21 days. On days 19 and 21, piglets in LPS and BT-LPS groups were challenged with LPS at 100 µg/kg body weight. Dietary butyrate improved LPS-injured intestinal histology by significantly increasing jejunal and ileal villus height, villus height to crypt depth ratios, and decreasing histological scores. LPS challenge activated hypoxia-inducible factor 1α and nuclear factor-κB, and enhanced interleukins (IL-1ß, IL-6, IL-12), tumor necrosis factor-α, and also downstream inducible nitric oxide synthase and cyclooxygenase 2, but decreased anti-inflammatory cytokines (IL-10, IL-13). Most molecule levels were significantly reversed by butyrate administration. When compared with the CON or LPS groups, the BT-LPS group had a higher relative abundance of jejunal Firmicutes, Bacteroidetes, Clostridiaceae, Lactobacillus, and Prevotella but a lower abundance of Proteobacteria, Enterobacteriaceae, and Escherichia-Shigella. Phylogenetic investigation of communities by reconstruction of unobserved states and correlation analyses suggested these bacteria contributed to butyrate-alleviating jejunal inflammation and infectious diseases. Butyrate-based diets significantly reduced apoptosis via mitochondrial pathways by downregulating apoptotic caspase 3 mRNA levels. Diets also altered enterocyte metabolism in the jejunum by upregulating peroxisome-proliferator-activated receptor α expression but downregulating carnitine palmitoyltransferase 1 level when compared with CON or LPS groups. Butyrate supplementation improved immunity homeostasis, generated beneficial shifts in microbial communities, improved enterocyte energy metabolism, and prevented apoptosis to protect intestinal histology from LPS-induced injury.

9.
Int J Mol Sci ; 23(13)2022 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-35806389

RESUMO

∆Np63α is a key transcription factor overexpressed in types of squamous cell carcinomas (SCCs), which represses epithelial-mesenchymal transition (EMT) and cell migration. In this study, we found that CDK1 phosphorylates ∆Np63α at the T123 site, impairing its affinity to the target promoters of its downstream genes and its regulation of them in turn. Database analysis revealed that CDK1 is overexpressed in head and neck squamous cell carcinomas (HNSCCs), especially the metastatic HNSCCs, and is negatively correlated with overall survival. We further found that CDK1 promotes the EMT and migration of HNSCC cells by inhibiting ∆Np63α. Altogether, our study identified CDK1 as a novel regulator of ΔNp63α, which can modulate EMT and cell migration in HNSCCs. Our findings will help to elucidate the migration mechanism of HNSCC cells.


Assuntos
Proteína Quinase CDC2 , Neoplasias de Cabeça e Pescoço , Carcinoma de Células Escamosas de Cabeça e Pescoço , Fatores de Transcrição , Proteínas Supressoras de Tumor , Proteína Quinase CDC2/genética , Proteína Quinase CDC2/metabolismo , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Transição Epitelial-Mesenquimal , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/patologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
10.
Microb Pathog ; 168: 105586, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35605738

RESUMO

Cystic Echinococcosis (hydatidosis) is caused by the larval stage of Echinococcus spp. It is an animal-borne zoonotic parasitic disease with local epidemic and natural foci, which is very common in northwest China. In recent years, a considerable attention has been paid to the epidemic investigation of hydatid disease in humans and sheep, but there are few large-scale epidemic investigation and data analysis of bovine hydatid disease. We systematically reviewed and analyzed the prevalence of bovine hydatid disease (2000-2021) in China for the first time. Several databases including CNKI, Wanfang, VIP Chinese periodical database, Baidu Library, PubMed and ScienceDirect were used to search 57 articles and 72 sets of valid data about bovine hydatid disease in China from 2000 to 2021. We used the random effect model in META package of R software, and PAS for rate conversion. The subgroup analysis and univariate meta regression analysis were used to reveal the factors leading to the heterogeneity of the study. The total prevalence rate of bovine hydatid disease in China from 2000 to 2021 is estimated to be 17.27% (10898/63113). According to the analysis of sampling years, the lowest positive rate since 2016 is 7.54% (1503/19929). The highest prevalence rate of bovine hydatid disease is 53.93% (4340/8048). The infection rate of bovine liver accounted for the highest proportion of the total infections, 45.2% (2040/4507). We also assessed the effects of different geographical and climatic factors on the prevalence of bovine hydatid disease. The results showed that the prevalence rate of hydatid disease was higher in cold and humid areas. Although the infection rate of bovine hydatid disease has declined in recent years, it is still necessary to carry out long-term surveillance and control of hydatid disease, cut off the infection route and reduce the risk of infection in high-risk areas.


Assuntos
Doenças dos Bovinos , Equinococose , Epidemias , Animais , Bovinos , Doenças dos Bovinos/epidemiologia , Doenças dos Bovinos/parasitologia , China/epidemiologia , Equinococose/epidemiologia , Equinococose/parasitologia , Equinococose/veterinária , Prevalência , Ovinos
11.
Acta Pharmacol Sin ; 43(2): 457-469, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33850273

RESUMO

Mantle cell lymphoma (MCL) is a lymphoproliferative disorder lacking reliable therapies. PI3K pathway contributes to the pathogenesis of MCL, serving as a potential target. However, idelalisib, an FDA-approved drug targeting PI3Kδ, has shown intrinsic resistance in MCL treatment. Here we report that a p300/CBP inhibitor, A-485, could overcome resistance to idelalisib in MCL cells in vitro and in vivo. A-485 was discovered in a combinational drug screening from an epigenetic compound library containing 45 small molecule modulators. We found that A-485, the highly selective catalytic inhibitor of p300 and CBP, was the most potent compound that enhanced the sensitivity of MCL cell line Z-138 to idelalisib. Combination of A-485 and idelalisib remarkably decreased the viability of three MCL cell lines tested. Co-treatment with A-485 and idelalisib in Maver-1 and Z-138 MCL cell xenograft mice for 3 weeks dramatically suppressed the tumor growth by reversing the unsustained inhibition in PI3K downstream signaling. We further demonstrated that p300/CBP inhibition decreased histone acetylation at RTKs gene promoters and reduced transcriptional upregulation of RTKs, thereby inhibiting the downstream persistent activation of MAPK/ERK signaling, which also contributed to the pathogenesis of MCL. Therefore, additional inhibition of p300/CBP blocked MAPK/ERK signaling, which rendered maintaining activation to PI3K-mTOR downstream signals p-S6 and p-4E-BP1, thus leading to suppression of cell growth and tumor progression and eliminating the intrinsic resistance to idelalisib ultimately. Our results provide a promising combination therapy for MCL and highlight the potential use of epigenetic inhibitors targeting p300/CBP to reverse drug resistance in tumor.


Assuntos
Classe Ia de Fosfatidilinositol 3-Quinase/efeitos dos fármacos , Linfoma de Célula do Manto/tratamento farmacológico , Purinas/uso terapêutico , Quinazolinonas/uso terapêutico , Fatores de Transcrição de p300-CBP/antagonistas & inibidores , Animais , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Sinergismo Farmacológico , Feminino , Compostos Heterocíclicos de 4 ou mais Anéis/uso terapêutico , Humanos , Camundongos , Transplante de Neoplasias
12.
Acta Pharmacol Sin ; 43(4): 941-953, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34341511

RESUMO

Nonalcoholic steatohepatitis (NASH) is a common chronic liver disease that is increasingly prevalent worldwide. Liver inflammation is an important contributor to disease progression from nonalcoholic fatty liver (NAFL) to NASH, but there is a lack of efficient therapies. In the current study we evaluated the therapeutic potential of givinostat, a histone deacetylase (HDAC) inhibitor, in the treatment of NASH in vivo and in vitro. Liver inflammation was induced in mice by feeding a methionine- and choline-deficient diet (MCD) or a fructose, palmitate, cholesterol diet (FPC). The mice were treated with givoinostat (10 mg·kg-1·d-1, ip) for 8 or 10 weeks. At the end of the experiment, the livers were harvested for analysis. We showed that givoinostat administration significantly alleviated inflammation and attenuated hepatic fibrosis in MCD-induced NASH mice. RNA-seq analysis of liver tissues form MCD-fed mice revealed that givinostat potently blocked expression of inflammation-related genes and regulated a broad set of lipid metabolism-related genes. In human hepatocellular carcinoma cell line HepG2 and human derived fetal hepatocyte cell line L02, givinostat significantly decreased palmitic acid-induced intracellular lipid accumulation. The benefit of givinostat was further confirmed in FPC-induced NASH mice. Givinostat administration significantly attenuated hepatic steatosis, inflammation as well as liver injury in this mouse model. In conclusion, givinostat is efficacious in reversing diet-induced NASH, and may serve as a therapeutic agent for the treatment of human NASH.


Assuntos
Hepatopatia Gordurosa não Alcoólica , Animais , Carbamatos , Inibidores de Histona Desacetilases/farmacologia , Inibidores de Histona Desacetilases/uso terapêutico , Fígado/metabolismo , Cirrose Hepática/patologia , Metionina , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/metabolismo
13.
J Med Chem ; 64(12): 8194-8207, 2021 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-34077206

RESUMO

Disruption of EZH2-embryonic ectoderm development (EED) protein-protein interaction (PPI) is a new promising cancer therapeutic strategy. We have previously reported the discovery of astemizole, a small-molecule inhibitor targeting the EZH2-EED PPI. Herein, we report the cocrystal structure of EED in complex with astemizole at 2.15 Å. The structure elucidates the detailed binding mode of astemizole to EED and provides a structure-guided design for the discovery of a novel EZH2-EED interaction inhibitor, DC-PRC2in-01, with an affinity Kd of 4.56 µM. DC-PRC2in-01 destabilizes the PRC2 complex, thereby leading to the degradation of PRC2 core proteins and the decrease of global H3K27me3 levels in cancer cells. The proliferation of PRC2-driven lymphomas cells is effectively inhibited, and the cell cycle is arrested in the G0/G1 phase. Together, these data demonstrate that DC-PRC2in-01 could be an effective chemical probe for investigating the PRC2-related physiology and pathology and providing a promising chemical scaffold for further development.


Assuntos
Astemizol/análogos & derivados , Astemizol/farmacologia , Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Complexo Repressor Polycomb 2/antagonistas & inibidores , Ligação Proteica/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Reposicionamento de Medicamentos , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Inibidores Enzimáticos/síntese química , Humanos , Simulação de Acoplamento Molecular , Estrutura Molecular , Complexo Repressor Polycomb 2/metabolismo , Relação Estrutura-Atividade
14.
Photodermatol Photoimmunol Photomed ; 37(5): 395-403, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33565151

RESUMO

BACKGROUND: ΔNp63α and c-Myc are key transcription factors controlling proliferation and senescence in epithelial cells. We previously reported that the c-Myc modulator MM1 and its E3 ligase, HERC3, together with the transcription factor ΔNp63α, compose a feedback loop, which regulates proliferative senescence in MCF-10A mammary epithelial cells. However, it is unknown whether this loop is involved in skin ageing. On the other hand, ultraviolet B (UVB) rays are assumed to be the main culprits for photoageing of the epidermis, but the underlying mechanisms are obscure. AIMS: To investigate whether MM1/ΔNp63α axis is involved in UVB-induced photoageing of the epidermis. MATERIALS AND METHODS: HaCaT human immortalized keratinocytes overexpressed with MM1, knocked down with c-Myc or irradiated with UVB, were subjected to MTT assays to measure cell proliferation, as well as RT-qPCR or immunoblot to detect the members of MM1/ΔNp63α loop and the cellular senescence markers. Meanwhile, primary normal human keratinocytes (NHKs) or mice were irradiated with UVB, followed by immunoblot analysis, SA-ß-gal, haematoxylin-eosin or immunohistochemistry staining. RESULTS: Overexpression of MM1 down-regulated ΔNp63α and induced proliferative senescence in the HaCaT cells. In the HaCaT cells, NHKs and the mouse epidermis, UVB irradiation increased MM1 mRNA level and led to a down-regulation of ΔNp63α, HERC3 and c-Myc, concomitant with cellular senescence or photoageing. Additionally, knock-down of c-Myc induced proliferative senescence in the HaCaT cells and abrogated UVB-induced cellular senescence. CONCLUSIONS: UVB up-regulates MM1 and consequently modulates ΔNp63α and c-Myc, which may account for the proliferative senescence of keratinocytes and photoageing of the epidermis.


Assuntos
Epiderme , Envelhecimento da Pele , Animais , Linhagem Celular , Humanos , Queratinócitos , Camundongos , Transativadores , Fatores de Transcrição , Proteínas Supressoras de Tumor , Raios Ultravioleta
15.
FEBS Open Bio ; 11(3): 890-897, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33548094

RESUMO

The p63 gene encodes at least 10 isoforms, which can be classified into TA and ∆N isotypes (TAp63 and ∆Np63 proteins) according to their differences at the N termini. TAp63γ is an important transcription factor. We previously reported that peptidyl-prolyl isomerase (PPI) Pin1 directly binds to TAp63γ protein and identified that serine 12 (S12 ) in the transactivation domain (TAD) of TAp63γ is required for regulation of its transcriptional activity. In the present study, we report that Pin1 stimulates transcriptional and pro-apoptotic activities of TAp63γ; this Pin1-mediated stimulation may depend on phosphorylation of S12 mediated by JNK1 and results in striking activation of TAp63γ. JNK1 represses transactivity of TAp63γ in cells without abundant Pin1 proteins and enhances it in the presence of sufficient levels of Pin1. Collectively, our data suggest a novel mechanism for regulation of TAp63γ transactivity: TAp63γ with unphosphorylated S12 is moderately active, phosphorylation at this residue (pS12 ) makes it hypoactive, and Pin1 binds to the pS12 -P13 motif and makes TAp63γ hyperactive. Our findings will aid in the elucidation of the mechanism underlying modulation of TAp63γ.


Assuntos
Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Peptidilprolil Isomerase de Interação com NIMA/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Linhagem Celular Tumoral , Regulação da Expressão Gênica , Células HeLa , Humanos , Mutação , Fosforilação , Domínios Proteicos , Serina/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/genética , Ativação Transcricional , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/genética
16.
Theranostics ; 10(22): 10290-10308, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32929349

RESUMO

Background: Oxidative stress from elevated reactive oxygen species (ROS) has been reported to induce cell apoptosis and may provide a means to target cancer cells. Celastrol is a natural bioactive compound that was recently shown to increase ROS levels and cause apoptosis in cancer cells. However, the underlying mechanism for this cytotoxic action remains unclear and direct molecular targets of Celastrol have not been identified. Methods: Proteome microarray, surface plasmon resonance, isothermal titration calorimetry and molecular simulation were used to identify the molecular target of Celastrol. Binding and activity assays were used to validate the interaction of Celastrol with target protein in cell-free and gastric cancer cell lysates. We then assessed target transcript levels in in biopsy specimens obtained from patients with gastric cancer. Gastric cancer growth-limiting and cytotoxic activity of Celastrol was evaluated in BALB/c nu/nu mice. Results: Our data show that Celastrol directly binds to an antioxidant enzyme, peroxiredoxin-2 (Prdx2), which then inhibits its enzyme activity at both molecular and cellular level. Inhibition of Prdx2 by Celastrol increased cellular ROS levels and led to ROS-dependent endoplasmic reticulum stress, mitochondrial dysfunction, and apoptosis in gastric cancer cells. Functional tests demonstrated that Celastrol limits gastric cancer cells, at least in part, through targeting Prdx2. Celastrol treatment of mice implanted with gastric cancer cells also inhibited tumor growth, associated with Prdx2 inhibition and increased ROS. Analysis of human gastric cancer also showed increased Prdx2 levels and correlation with survival. Conclusion: Our studies have uncovered a potential Celastrol-interacting protein Prdx2 and a ROS-dependent mechanism of its action. The findings also highlight Prdx2 as a potential target for the treatment of gastric cancer.


Assuntos
Apoptose/efeitos dos fármacos , Triterpenos Pentacíclicos/farmacologia , Peroxirredoxinas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/metabolismo , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Estresse Oxidativo/efeitos dos fármacos
17.
Biochem Biophys Res Commun ; 533(3): 613-619, 2020 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-32981679

RESUMO

c-Myc modulator 1 (MM1), also known as PFDN5, is the fifth subunit of prefoldin. It was previously reported that MM1-based prefoldin promotes folding of actin during assembly of cytoskeleton, which plays key roles in cell migration. However, no evidence supports that MM1 affects cell migration. In the present study, we found that MM1 promotes cell migration in multiple cell lines. Further study revealed that MM1 promotes polymerization of ß-actin into filamentous form and increases both density and length of filopodia. Effects of MM1 on filopodia formation and cell migration depend on its prefoldin activity. Though c-Myc is repressed by MM1, simultaneous knock-down of c-Myc fails to rescue migration inhibition induced by MM1 ablation. Taken together, we here, for the first time, report that prefoldin subunit MM1 is involved in cell migration; this involvement of MM1 in cell migration is due to its prefoldin activity to boost polymerization of ß-actin during filopodia formation. Our findings may be helpful to elucidate the mechanism of cell migration and cancer metastasis.


Assuntos
Movimento Celular , Chaperonas Moleculares/fisiologia , Pseudópodes/metabolismo , Actinas/metabolismo , Linhagem Celular , Humanos , Chaperonas Moleculares/metabolismo , Subunidades Proteicas/metabolismo , Subunidades Proteicas/fisiologia
18.
Oncogenesis ; 7(11): 83, 2018 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-30385738

RESUMO

Small molecular inhibitors targeting BRD4 family proteins are emerging as promising therapies in many types of human malignancies. However, whether BRD4, as well as other BET family members, may serve as therapeutic targets in renal cell carcinoma (RCC) remains unknown. In this study, we found that both BRD2 and BRD4 were over-expressed in RCC tissues, knock-down both of which achieved potent anti-proliferative effects in RCC cells. A novel category of BET inhibitors, originated from an approved drug Nitroxoline, were synthesized and evaluated with biochemical and cellular assays, as well as the method of crystallography. The complex crystal structures of several compounds in this category with the first bromodomain of BRD4 (BRD4-BD1) were solved, revealing the binding mechanism and facilitating further structural optimizations. Among them, compound BDF-1253 showed an approximately four-fold improvement in BRD4 inhibition compared with the prototype Nitroxoline and had good selectivity for BET proteins against other bromodomain proteins or epi-enzymes in biochemical assays. Compound BDF-1253 efficiently suppressed the expression of BET downstream genes, impaired RCC cells viability via inducing cell cycle arrest and apoptosis, and decreased tumor growth in RCC xenografts. In summary, these results suggest that inhibition of BET family members has great therapeutic potentials in the treatment of RCC, and the novel series of BET inhibitors reported here are promising to become RCC drug candidates.

19.
Cell Death Differ ; 25(12): 2118-2129, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29880857

RESUMO

p63 and c-Myc are key transcription factors controlling genes involved in the cell cycle and cellular senescence. We previously reported that p63α can destabilize MM1 protein to derepress c-Myc, resulting in cell cycle progress and tumorigenesis. However, how the proteasomal degradation of MM1 is facilitated remains unclear. In the present study, we identified a novel E3 ligase, HERC3, which can mediate ubiquitination of MM1 and promote its proteasome-dependent degradation. We found that ΔNp63α transcriptionally up-regulates HERC3 and knockdown of HERC3 abrogates ΔNp63α-induced down-regulation of MM1. Either overexpression of MM1 or ablation of HERC3 induces cell senescence, while knockdown of MM1 rescues cell senescence induced by deficiency of either ΔNp63α or HERC3, implicating the involvement of the ΔNp63α/HERC3/MM1/c-Myc axis in the modulation of cell senescence. Additionally, our Oncomine analysis indicates activation of the ΔNp63α/HERC3/MM1/c-Myc axis in invasive breast carcinoma. Together, our data illuminate a novel axis regulating cell senescence: ΔNp63α stimulates transcription of E3 ligase HERC3, which mediates ubiquitination of c-Myc modulator MM1 and targets it to proteasomal degradation; subsequently, c-Myc is derepressed by ΔNp63α, thereby cell senescence is modulated by this axis. Our work provides a new interpretation of crosstalk between p63 and c-Myc, and also sheds new light on ΔNp63α-controlled cell senescence and tumorigenesis.


Assuntos
Senescência Celular , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Células HEK293 , Humanos , Ubiquitina-Proteína Ligases
20.
FEBS Lett ; 592(13): 2334-2340, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29782646

RESUMO

Potassium channel tetramerization domain containing 5 (KCTD5) was previously documented as a component of the Cullin3-RING ligase (CRL3). It has been reported that KCTD5 can induce enrichment of polyubiquitinated proteins, and KCTD5-based CRL3 destabilizes several proteins. In our present study, we report that KCTD5 may physically interact with ΔNp63α, which is a member of the p53 family. Our further investigation revealed that Cullin3/KCTD5 can induce monoubiquitination of ΔNp63α. Cullin3/KCTD5 downregulates the DNA-binding affinity of ΔNp63α, impairing either its transactivity or its transinhibitory activity. Functionally, Cullin3/KCTD5 abates the proproliferation activity of ΔNp63α. These findings suggest that KCTD5-based CRL3 may mediate monoubiquitination and is a novel regulator of ΔNp63α.


Assuntos
Proteínas Culina/fisiologia , Canais de Potássio/fisiologia , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Ubiquitinação , Células Cultivadas , Células HEK293 , Humanos , Canais de Potássio/metabolismo , Ligação Proteica , Multimerização Proteica , Ubiquitina-Proteína Ligases/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA