Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Int J Mol Med ; 53(4)2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38426579

RESUMEN

SET domain bifurcated 1 (SETDB1), a pivotal histone lysine methyltransferase, is transported to the cytoplasm via a chromosome region maintenance 1 (CMR1)­dependent pathway, contributing to non­histone methylation. However, the function and underlying mechanism of cytoplasmic SETDB1 in breast cancer remain elusive. In the present study, immunohistochemistry revealed that elevated cytoplasmic SETDB1 was correlated with lymph node metastasis and more aggressive breast cancer subtypes. Functionally, wound healing and Transwell assays showed that cytoplasmic SETDB1 is key for cell migration and invasion, as well as induction of epithelial­mesenchymal transition (EMT), which was reversed by leptomycin B (LMB, a CMR1 inhibitor) treatment. Furthermore, RNA­seq and metabolite detection revealed that cytoplasmic SETDB1 was associated with metabolism pathway and elevated levels of metabolites involved in the Warburg effect, including glucose, pyruvate, lactate and ATP. Immunoblotting and reverse transcription­quantitative PCR verified that elevation of cytoplasmic SETDB1 contributed to elevation of c­MYC expression and subsequent upregulation of lactate dehydrogenase A (LDHA) expression. Notably, gain­ and loss­of­function approaches revealed that LDHA overexpression in T47D cells enhanced migration and invasion by inducing EMT, while its depletion in SETDB1­overexpressing MCF7 cells reversed SETDB1­induced migration and invasion, as well as the Warburg effect and EMT. In conclusion, subcellular localization of cytoplasmic SETDB1 may be a pivotal factor in breast cancer progression. The present study offers valuable insight into the novel functions and mechanisms of cytoplasmic SETDB1.


Asunto(s)
Neoplasias de la Mama , Dominios PR-SET , Femenino , Humanos , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/genética , Citoplasma/metabolismo , Regulación Neoplásica de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Lactato Deshidrogenasa 5/genética , Lactato Deshidrogenasa 5/metabolismo
2.
Pharmacol Res ; 200: 107051, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38190956

RESUMEN

Renal interstitial fibrosis/tubular atrophy (IF/TA) is a prominent pathological feature of chronic allograft dysfunction (CAD). Our previous study has demonstrated that epithelial-mesenchymal transition (EMT) plays a significant role in shaping the development of IF/TA. Nuclear SET domain (NSD2), a histone methyltransferase catalyzing methylation at lysine 36 of histone 3, is crucially involved in the development and progression of solid tumors. But its role in the development of renal allograft interstitial fibrosis has yet to be elucidated. Here, we characterize NSD2 as a crucial mediator in the mouse renal transplantation model in vivo and a model of tumor necrosis factor-α (TNF-α) stimulated-human renal tubular epithelial cells (HK-2) in vitro. Functionally, NSD2 knockdown inhibits EMT, dynamin-related protein 1 (Drp1)-mediated mitochondrial fission in mice. Conversely, NSD2 overexpression exacerbates fibrosis-associated phenotypes and mitochondrial fission in tubular cells. Mechanistically, tubular NSD2 aggravated the Drp-1 mediated mitochondrial fission via STAT1/ERK/PI3K/Akt signaling pathway in TNF-α-induced epithelial cell models. Momentously, mass spectrometry (MS) Analysis and site-directed mutagenesis assays revealed that NSD2 interacted with and induced Mono-methylation of STAT1 on K173, leading to its phosphorylation, IMB1-dependent nuclear translocation and subsequent influence on TNF-α-induced EMT and mitochondrial fission in NSD2-dependent manner. Collectively, these findings shed light on the mechanisms and suggest that targeting NSD2 could be a promising therapeutic approach to enhance tubular cell survival and alleviate interstitial fibrosis in renal allografts during CAD.


Asunto(s)
Enfermedades Renales , Trasplante de Riñón , Humanos , Ratones , Animales , Factor de Necrosis Tumoral alfa/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Dinámicas Mitocondriales , Dominios PR-SET , Fibrosis , Aloinjertos/metabolismo , Transición Epitelial-Mesenquimal , Factor de Transcripción STAT1/metabolismo
3.
J Mol Biol ; 436(7): 168318, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-37863247

RESUMEN

Within the SET domain superfamily of lysine methyltransferases, there is a well-conserved subfamily, frequently referred to as the Set3 SET domain subfamily, which contain noncanonical SET domains carrying divergent amino acid sequences. These proteins are implicated in diverse biological processes including stress responses, cell differentiation, and development, and their disruption is linked to diseases including cancer and neurodevelopmental disorders. Interestingly, biochemical and structural analysis indicates that they do not possess catalytic methyltransferase activity. At the molecular level, Set3 SET domain proteins appear to play critical roles in the regulation of gene expression, particularly repression and heterochromatin maintenance, and in some cases, via scaffolding other histone modifying activities at chromatin. Here, we explore the common and unique functions among Set3 SET domain subfamily proteins and analyze what is known about the specific contribution of the conserved SET domain to functional roles of these proteins, as well as propose areas of investigation to improve understanding of this important, noncanonical subfamily of proteins.


Asunto(s)
N-Metiltransferasa de Histona-Lisina , Dominios PR-SET , Secuencia de Aminoácidos , Cromatina/química , Cromatina/metabolismo , N-Metiltransferasa de Histona-Lisina/química , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Animales
4.
Epigenetics Chromatin ; 16(1): 47, 2023 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-38057834

RESUMEN

Histone methyltransferase SETDB1 (SET domain bifurcated histone lysine methyltransferase 1, also known as ESET or KMT1E) is known to be involved in the deposition of the di- and tri-methyl marks on H3K9 (H3K9me2 and H3K9me3), which are associated with transcription repression. SETDB1 exerts an essential role in the silencing of endogenous retroviruses (ERVs) in embryonic stem cells (mESCs) by tri-methylating H3K9 (H3K9me3) and interacting with DNA methyltransferases (DNMTs). Additionally, SETDB1 is engaged in regulating multiple biological processes and diseases, such as ageing, tumors, and inflammatory bowel disease (IBD), by methylating both histones and non-histone proteins. In this review, we provide an overview of the complex biology of SETDB1, review the upstream regulatory mechanisms of SETDB1 and its partners, discuss the functions and molecular mechanisms of SETDB1 in cell fate determination and stem cell, as well as in tumors and other diseases. Finally, we discuss the current challenges and prospects of targeting SETDB1 for the treatment of different diseases, and we also suggest some future research directions in the field of SETDB1 research.


Asunto(s)
Neoplasias , Dominios PR-SET , Humanos , Histonas/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Metilación de ADN , Neoplasias/genética
5.
ACS Chem Biol ; 18(8): 1846-1853, 2023 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-37556795

RESUMEN

Increased expression and hyperactivation of the methyltransferase SET domain bifurcated 1 (SETDB1) are commonly observed in cancer and central nervous system disorders. However, there are currently no reported SETDB1-specific methyltransferase inhibitors in the literature, suggesting that this is a challenging target. Here, we disclose that the previously reported small-molecule ligand for SETDB1's triple tudor domain, (R,R)-59, is unexpectedly able to increase SETDB1 methyltransferase activity both in vitro and in cells. Specifically, (R,R)-59 promotes in vitro SETDB1-mediated methylation of lysine 64 of the protein kinase Akt1. Treatment with (R,R)-59 also increased Akt1 threonine 308 phosphorylation and activation, a known consequence of Akt1 methylation, resulting in stimulated cell proliferation in a dose-dependent manner. (R,R)-59 is the first SETDB1 small-molecule positive activator for the methyltransferase activity of this protein. Mechanism of action studies show that full-length SETDB1 is required for significant in vitro methylation of an Akt1-K64 peptide and that this activity is stimulated by (R,R)-59 primarily through an increase in catalytic activity rather than a change in S-adenosyl methionine binding.


Asunto(s)
N-Metiltransferasa de Histona-Lisina , Dominios PR-SET , N-Metiltransferasa de Histona-Lisina/metabolismo , Ligandos , Metilación , Dominio Tudor
6.
J Med Chem ; 66(16): 10991-11026, 2023 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-37578463

RESUMEN

Nuclear receptor binding SET domain proteins (NSDs) catalyze the mono- or dimethylation of histone 3 lysine 36 (H3K36me1 and H3K36me2), using S-adenosyl-l-methionine (SAM) as a methyl donor. As a key member of the NSD family of proteins, NSD2 plays an important role in the pathogenesis and progression of various diseases such as cancers, inflammations, and infectious diseases, serving as a promising drug target. Developing potent and specific NSD2 inhibitors may provide potential novel therapeutics. Several NSD2 inhibitors and degraders have been discovered while remaining in the early stage of drug development. Excitingly, KTX-1001, a selective NSD2 inhibitor, has entered clinical trials. In this Perspective, the structures and functions of NSD2, its roles in various human diseases, and the recent advances in drug discovery strategies targeting NSD2 have been summarized. The challenges, opportunities, and future directions for developing NSD2 inhibitors and degraders are also discussed.


Asunto(s)
Neoplasias , Dominios PR-SET , Humanos , Descubrimiento de Drogas , Histonas/metabolismo , Dominios PR-SET/efectos de los fármacos
7.
Chem Biol Drug Des ; 102(3): 500-513, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37072259

RESUMEN

NSD3/WHSC1L1 lysine methyltransferase promotes the transcription of target genes through di- or tri-methylation at histone H3K36 using SAM as a cofactor. Genetic alterations such as amplification and gain-of-function mutation of NSD3 act as oncogenic drivers in several cancers including squamous cell lung cancer and breast cancer. NSD3 is an important therapeutic target for cancers, but the reported NSD3 inhibitors targeting the catalytic SET domain are very rare and show a poor activity. Herein, from a virtual library screening and the subsequent medicinal chemistry optimization, we identified a novel class of NSD3 inhibitors. Our docking analysis and pulldown result suggested that the most potent analogue 13i shows a unique, bivalent binding mode interacting with both SAM-binding site and BT3-bindig site within the SET domain. We found 13i inhibits NSD3 activity with IC50 = 287 µM in vitro and suppresses the proliferation of JIMT1 breast cancer cells with GI50 = 36.5 µM, which express a high level of NSD3. Also, 13i downregulated the levels of H3K36me2/3 in a dose-dependent manner. Our study could provide an insight in designing high-affinity NSD3 inhibitors. Also, as the acrylamide group of 13i was predicted to position near Cys1265 in the BT3-binding site, further optimization would lead to a discovery of novel irreversible NSD3 inhibitors.


Asunto(s)
Neoplasias de la Mama , Dominios PR-SET , Humanos , Femenino , Histonas , Dominios Proteicos , Metilación , Neoplasias de la Mama/tratamiento farmacológico
8.
Cancer Med ; 12(9): 10961-10978, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37062069

RESUMEN

BACKGROUND: Members of the nuclear receptor-binding SET domain (NSD) family of histone H3 lysine 36 methyltransferases comprise NSD1, NSD2 (MMSET/WHSC1), and NSD3 (Wolf-Hirschhorn syndrome candidate 1-like 1, WHSC1L1). While the expression of NSD genes is essential to normal biological processes and cancer, knowledge of their expression levels to prognosticate in cancer remains unclear. METHODS: We analyzed the expression patterns for NSD family genes across multiple cancer types and examined their association with clinical features and patient survival profiles. Next, we explored the association between NSD3 expression and described features of the tumor microenvironment (TME) in PAAD, a severe type of pancreatic cancer. In particular, we correlated promoter methylation levels for NSD3 with patient outcomes in PAAD. Finally, we explored the putative oncogenic roles for NSD3 using a series of experiments with pancreatic cancer cells. RESULTS: We report that the expression of NSD family members is correlated with clinical prognosis across multiple types of cancers. Also, we demonstrate that NSD3 variants are most prevalent among NSD genes across cancers we analyzed. Notably, when compared with NSD1 and NSD2, we find that NSD3 is prominently expressed, and its expression is significantly linked with clinical outcome in pancreatic cancer. Furthermore, NSD3 is frequently amplified, exhibits low promoter methylation, and is correlated with immune cell infiltration and enhanced proliferation of pancreatic cancer. Finally, we demonstrate that knockdown of NSD3 alters H3K36me2 methylation, downstream gene expression and EGFR/ERK signaling in pancreatic cancer cells. CONCLUSIONS: We find that expression levels, the presence of genetic variants of NSD family genes, as well as their promoter methylation are correlated with clinical outcomes in cancer, including pancreatic cancer. Our in vitro experiments suggest that NSD3 may be relevant to gene expression regulation and growth factor signaling in pancreatic cancer.


Asunto(s)
Histonas , Neoplasias Pancreáticas , Humanos , Histonas/metabolismo , Dominios PR-SET , Pronóstico , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Histona Metiltransferasas/metabolismo , Neoplasias Pancreáticas/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Biomarcadores , Microambiente Tumoral , Neoplasias Pancreáticas
9.
Eur J Med Chem ; 250: 115232, 2023 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-36863225

RESUMEN

Nuclear receptor-binding SET domain 2 (NSD2) is a histone lysine methyltransferase (HKMTase), which is mainly responsible for the di-methylation of lysine residues on histones, which are involved in the regulation of various biological pathways. The amplification, mutation, translocation, or overexpression of NSD2 can be linked to various diseases. NSD2 has been identified as a promising drug target for cancer therapy. However, relatively few inhibitors have been discovered and this field still needs further exploration. This review provides a detailed summary of the biological studies related to NSD2 and the current progress of inhibitors, research, and describes the challenges in the development of NSD2 inhibitors, including SET (su(var), enhancer-of-zeste, trithorax) domain inhibitors and PWWP1 (proline-tryptophan-tryptophan-proline 1) domain inhibitors. Through analysis and discussion of the NSD2-related crystal complexes and the biological evaluation of related small molecules, we hope to provide insights for future drug design and optimization methods that will stimulate the development of novel NSD2 inhibitors.


Asunto(s)
Dominios PR-SET , Proteínas Represoras , Proteínas Represoras/metabolismo , Histonas/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo
10.
Mol Med Rep ; 27(3)2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36799151

RESUMEN

The present study aimed to identify the function and expression of trimethylated protein histone H3 lysine 36 (H3K36)me3 and the upstream specific enzyme histone methyltransferase SET domain containing 2 (SETD2), during the differentiation of hepatic oval cells (HOCs) into cholangiocytes in mice following partial liver resection and fed with 2­acetamidofluorene. HOCs were isolated from Kunming male mice fed with 2­acetamidofluorene for 10 days. Their liver tissues were then isolated following partial liver resection and another week of 2­acetamidofluorene treatment. HOCs were collected following a two­step enzyme digestion procedure involving protease E and collagenase 4. The target cells were cultured in DMEM/F12 supplemented with 10 µg/ml EGF, 5 µg/ml stem cell growth factor and 5 µg/ml leukemia inhibitory factor. Target cells using the markers OV­6, CK­19, SETD2, H3K36me3, were detected with flow cytometry and immunofluorescence microscopy; reverse transcription­quantitative PCR and western blotting were used to quantify the protein levels of SETD2 and H3K36me3. The retrieved primary hepatocytes developed into cholangiocytes with increasing CK­19 and decreasing OV­6 expression in each subsequent passage, whereas the SETD2 and H3K36me3 levels gradually increased, suggesting the possible involvement of both of these factors in differentiation.


Asunto(s)
Histonas , Lisina , Ratones , Masculino , Animales , Histonas/metabolismo , Histona Metiltransferasas/metabolismo , Lisina/metabolismo , Dominios PR-SET , Diferenciación Celular , Células Epiteliales/metabolismo , Conductos Biliares/metabolismo
11.
Int Immunopharmacol ; 115: 109554, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36580757

RESUMEN

Macrophages exhibit distinct phenotypes that are pro-inflammatory (M1) or anti-inflammatory (M2) in response to inflammation. In this study, we tried to identify the roles and mechanisms of interferon regulatory factor 7 (IRF7) in modulating the phenotypes of macrophages in lipopolysaccharide (LPS)-induced intestinal inflammation. The mouse model of intestinal inflammation was induced by lipopolysaccharide (LPS), and mouse bone marrow-derived macrophages (BMDMs) and mouse intestinal epithelial cells were selected for experimental verification in vitro. Results demonstrated that IRF7 was highly expressed in the mouse model of intestinal inflammation, while IRF7 deficiency repressed macrophage M1 polarization and attenuated intestinal inflammation in mice. p65 and SET domain bifurcated 1 (SETDB1) synergistically promoted histone 3 lysine 4 trimethylation (H3K4me3) methylation to elevate IRF7 expression, which activated the Nod-like receptor (NLR) pathway to induce macrophage M1 polarization. Through this mechanism, IRF7 in BMDMs functioned to accelerate intestinal epithelial cell apoptosis and their release of pro-inflammatory proteins. Furthermore, the promoting effect of p65 and SETDB1 on LPS-induced intestinal inflammation was validated in vivo. To sum up, NF-κB p65 and SETDB1 facilitated IRF7-mediated macrophage M1 polarization, thereby aggravating the LPS-induced intestinal inflammation. Hence, this study highlights the appealing value of these factors as anti-inflammatory targets.


Asunto(s)
Lipopolisacáridos , FN-kappa B , Ratones , Animales , FN-kappa B/metabolismo , Lipopolisacáridos/farmacología , Factor 7 Regulador del Interferón/metabolismo , Dominios PR-SET , Macrófagos , Inflamación/inducido químicamente , Inflamación/metabolismo , Antiinflamatorios/farmacología , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo
12.
Immunology ; 169(1): 3-12, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36524435

RESUMEN

Oncogene SET Domain Bifurcated 1 (SETDB1)/ESET, an H3K9 methyltransferase, was originally discovered over two decades ago; however, its function in the immune response was not first reported until 2011. SETDB1 immune functions include B cell maturation, T cell activity regulation, and immune escape in cancer cells. In B lymphocytes, SETDB1 mediates the transition from pro-B to pre-B cells and represses endogenous retroviruses (ERV) to encourage B cell lineage differentiation and maturation. SETDB1 alters T cell function by methylating IL-2 and IL-17 promoters and mediating T cell lineage commitment and development. In addition, SETDB1 plays a critical role in ERV silencing within a variety of immune cells, which can indirectly weaken the immune response. Although SETDB1 is critical for normal immune cell function, overexpression in cancer cells negatively impacts immune cell fights against cancer through decreased tumour immunogenicity. Within cancer cells, SETDB1 overexpression represses production and infiltration of antitumour immune cells, mediates immune escape through TE and ERV silencing, represses the type I interferon pathway, and interferes in immune checkpoint blockade (ICB) outcomes by regulation of PD-L1 expression and IFN signalling. In this review, we further discuss the immunological mechanisms of SETDB1 in normal and cancerous cells and its implications in cancer immunotherapy.


Asunto(s)
Retrovirus Endógenos , Inmunoterapia , Neoplasias , Retrovirus Endógenos/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Neoplasias/inmunología , Neoplasias/terapia , Dominios PR-SET , Humanos
13.
Cell Cycle ; 22(3): 269-275, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36101480

RESUMEN

SET domain containing lysine methyltransferase 7 (SETD7) belongs to the protein lysine methyltransferase family and can catalyze the monomethylation of histone H3K4, which plays a vital role in the regulation of cell cycle, cell differentiation, DNA damage response and chromatin remodeling through K/R-S/T-K (K is lysine residue) sites and the recognition of substrates mediated by SET, i-SET, and n-SET domains and electrostatic action. SETD7 also can regulate the transcription of several genes including ß-catenin, Cullin l and lin-28 homolog A (LIN28A), etc. In addition, the abnormal expression of SETD7 can promote the proliferation, migration, invasion of tumor cells, predict the poor prognosis of tumor patients, and may be a potential target for tumor therapy. This paper reviews the structure of SETD7, its role in tumor genesis and development, and the current research progress of relevant targeted drugs to explore its regulatory mechanism in tumor genesis and development and the prospect of targeted therapy.


Asunto(s)
Transformación Celular Neoplásica , N-Metiltransferasa de Histona-Lisina , Humanos , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Dominios PR-SET
14.
J Med Chem ; 65(15): 10611-10625, 2022 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-35895319

RESUMEN

Overexpression of nuclear receptor binding SET domain protein 2 (NSD2) is frequent in multiple myeloma (MM). However, existing NSD2 inhibitors are largely ineffective in suppressing MM cell proliferation. Here, we report the discovery of a first-in-class NSD2 proteolysis targeting chimera (PROTAC) degrader, 9 (MS159), and two structurally similar controls, 17 (MS159N1) and 18 (MS159N2), with diminished binding to the cereblon (CRBN) E3 ligase and NSD2, respectively. Compound 9, but not 17 and 18, effectively degraded NSD2 in a concentration-, time-, CRBN-, and proteasome-dependent manner. Compound 9 also effectively degraded CRBN neo-substrates IKZF1 and IKZF3, but not GSPT1. Importantly, compound 9 was much more effective in suppressing the growth in cancer cells than the parent NSD2 binder. Moreover, compound 9 was bioavailable in mice. Altogether, compound 9 and its two controls 17 and 18 are valuable chemical tools for exploring the roles of NSD2 in health and disease.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/metabolismo , Factor de Transcripción Ikaros/metabolismo , Dominios PR-SET , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Ratones , Proteolisis , Receptores Citoplasmáticos y Nucleares/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
15.
Methods Mol Biol ; 2529: 297-311, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35733021

RESUMEN

Identification of histone lysine methyltransferase (HKMT) substrates has recently benefited from chemical-biology-based strategies in which artificial S-adenosyl-L-methionine (SAM) cofactors are engineered to allow substrate labeling using either the wild-type target enzyme or designed mutants. Once labeled, substrates can be selectively functionalized with an affinity tag, using a bioorthogonal ligation reaction, to allow their recovery from cell extracts and subsequent identification. In this chapter, we describe steps on how to proceed to set up such an approach to characterize substrates of specific HKMTs of the SET domain superfamily, from the characterization of the HKMT able to accommodate a SAM surrogate containing a bioorthogonal moiety, to the proteomic analysis conducted on a cell extract. We focus in particular on the controls that are necessary to ensure reliable proteomic data analysis. The example of PR-Set7 on which we have implemented this approach is shown.


Asunto(s)
Metionina , S-Adenosilmetionina , N-Metiltransferasa de Histona-Lisina/química , Dominios PR-SET , Proteómica , S-Adenosilmetionina/química
16.
Biomed Res Int ; 2022: 3307873, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35372573

RESUMEN

Several genes on chromosome 1q21 region predict a high risk of multiple myeloma (MM); however, the underlying molecular pathology remains elusive. Overexpression, amplification, or activation of SET Domain Bifurcated 1 (SETDB1), which is located on 1q21, is closely associated with poor prognosis of many human solid malignancies. In our study, upregulation of SETDB1 might indicate an unfavorable prognosis of MM using bioinformatics analysis from GEO databases and MMRF-CoMMpass. Here, increased SETDB1 expression was observed in the plasma cells from newly diagnosed multiple myeloma patients compared to those from the normal controls. Meanwhile, SETDB1 overexpression was the result of increased copy numbers of SETDB1 gene. In MM patients, the Kaplan-Meier analysis was employed to demonstrate that increased SETDB1 expression was associated with shorter overall survival (OS) and event-free survival (EFS). Besides, we conducted multifactorial cox regression analysis to state that SETDB1 expression was an independent biomarker for OS and EFS. MM patients with higher SETDB1 expression showed higher levels of beta-2 microglobulin (ß2M), lactate dehydrogenase (LDH), and bone marrow biopsy plasma cells (BMPC) and lower levels of haemoglobin (HGB). Functional enrichment analysis suggested that SETDB1 could promote cell cycle progression in myeloma. Finally, we observed that SETDB1 was distinctly correlated with tumor immunity in MM. SETDB1 expression in myeloma cells was positively correlated with CD56dim natural killer cells but negatively correlated with infiltrating levels of type17 T helper cells, effector memory CD8 T cells, activated dendritic cells, and natural killer T cells from whole bone marrow (WBM) biopsies. Taken together, these results indicated that SETDB1 could be used as a novel biomarker for predicting the prognosis of MM patients.


Asunto(s)
Mieloma Múltiple , Biomarcadores , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Estimación de Kaplan-Meier , Mieloma Múltiple/patología , Dominios PR-SET , Pronóstico
17.
Bioengineered ; 13(3): 7293-7302, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35260047

RESUMEN

Liver cancer is a common malignant tumor with high incidence and mortality rates. However, a reliable prognostic signature has not yet been confirmed. Circular RNAs (circRNAs) play a role in the development and prognosis of numerous malignancies as well as liver cancer. Therefore, identifying abnormally expressed circRNAs in liver cancer tissue is essential for early diagnosis and treatment. This study found that circular RNA circ SET domain containing 2 (circSETD2) is abnormally expressed in liver cancer tissues, but the role and molecular mechanismsin the occurrence and development of liver cancer are still unclear. The expression level of circSETD2 was evaluated through Quantitative Real-time Polymerase chain reaction (qRT-PCR) in cancerous liver tissues (30 cases), liver cancer cell lines and para-cancerous tissues. Knockdown and overexpression circSETD2 lentiviral vector was constructed and applied to transfect hepatoma cells. Cell Counting Kit-8 (CCK-8), colony formation assay, flow cytometry and Transwell assay were used to examine the effects of circSETD2 overexpression or knockdown on liver cancer migration, invasion, cell cycle and cell proliferation. The tumourigenicity in vivo was utilized to assess the effect of circSETD2 on the proliferation of liver cancer cells. circSETD2 expression is lower in cell lines and liver cancer tissues. circSETD2 knockdown can considerably increase liver cancer cells' invasion, proliferation and colony formation. While In vitro and in vivo, circSETD2 overexpression shows opposite effect. Western blot showed that circSETD2 knockdown can considerably promote E-cadherin expression and inhibit Vimentin, N-cadherin, matrix metallopeptidase-9 (MMP-9) and MMP-2 expression. These findings improve our understanding of the mechanisms of liver cancer progression and will guide future development of therapeutic strategies against the disease by targeting circ-SETD2.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , MicroARNs , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hepáticas/metabolismo , MicroARNs/metabolismo , Dominios PR-SET , ARN Circular/genética
18.
Bioengineered ; 13(3): 7253-7261, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35259059

RESUMEN

Oxidative stress and inflammation are implicated in the pathogenesis of cerebral ischemia-reperfusion (I/R) injury. SETD7 (SET Domain Containing 7) functions as a histone lysine methyltransferase, participates in cardiac lineage commitment, and silence of SETD7 exerts anti-inflammatory or antioxidant capacities. The effect of SETD7 in in vitro cell model of cerebral I/R injury was investigated in this study. Firstly, adrenal pheochromocytoma cell (PC12) was conducted with oxygen-glucose deprivation/reoxygenation (OGD/R) to establish cell model of cerebral I/R injury. OGD/R-enhanced SETD7 expression in PC12 cells. Cell viability of OGD/R-induced PC12 was reduced, while the apoptosis was promoted. Secondly, knockdown of SETD7 reversed the effect of OGD/R on cell viability and apoptosis of PC12. Moreover, OGD/R-induced inflammation in PC12 with decreased interleukin (IL)-10, increased IL-6, IL-1ß, tumor necrosis factor-α (TNF-α), and cyclooxygenase 2 (COX-2) were restored by knockdown of SETD7. Thirdly, knockdown of SETD7 attenuated OGD/R-induced decrease of superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), and catalase (CAT), as well as increase of malondialdehyde (MDA) and reactive oxygen species (ROS) in PC12. Lastly, OGD/R-induced decrease of NF-κB inhibitor α (IκBα), increase of phosphorylated (p)-p65, p-IκBα, and Keap1 (Kelch-like ECH-associated protein 1) were reversed by silence of SETD7. Silence of SETD7 increased heme oxygenase-1 (HO-1) and nuclear factor erythroid 2-related factor 2 (Nrf2) expression in OGD/R-induced PC12. In conclusion, suppression of SETD7 ameliorated OGD/R-induced inflammation and oxidative stress in PC12 cell through inactivation of NF-κB and activation of Keap1/Nrf2/ARE pathway.


Asunto(s)
Factor 2 Relacionado con NF-E2 , Daño por Reperfusión , Animales , Apoptosis/genética , Glucosa/metabolismo , N-Metiltransferasa de Histona-Lisina , Inflamación/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/genética , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Inhibidor NF-kappaB alfa/metabolismo , FN-kappa B/metabolismo , Estrés Oxidativo , Oxígeno/farmacología , Células PC12 , Dominios PR-SET , Ratas , Especies Reactivas de Oxígeno/metabolismo , Daño por Reperfusión/genética , Daño por Reperfusión/metabolismo , Transducción de Señal
19.
Hum Exp Toxicol ; 41: 9603271211061497, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35187972

RESUMEN

BACKGROUND: As a common postoperative complication to elderly patients, postoperative cognitive dysfunction (POCD) is a central nervous system complication, often taking place after anesthesia and surgery. (Su(var)3-9, enhancer-of-zeste, and trithorax) domain-containing protein 7 (SETD7) plays important roles in metabolic-related diseases, viral infections, tumor formation, and some inflammatory reactions. However, the role and mechanism of SETD7 in POCD have not been previously studied. METHODS: RT-PCR and Western blot were performed to evaluate the efficiency of knockdown of SETD7. The pathological changes of hippocampal neurons in isoflurane-anesthetized mice were detected by HE staining, and the Morris water maze experiment was performed to evaluate the learning and memory abilities of mice. The effect of SETD7 on the hippocampus in isoflurane-induced aged mice was examined by Western blot and TUNEL assay. Then ELISA assay was applied to determine the expression of some inflammatory cytokines, followed by the detection of expression of NOD-like receptor protein 3 (NLRP3) inflammasome through Western blot. RESULTS: The data of this research revealed that SETD7 knockdown improved cognitive impairment in isoflurane-anesthetized mice, ameliorated cell pyroptosis, inhibited the release of inflammatory cytokines, and suppressed the activation of NLRP3 inflammasome in the hippocampus in isoflurane-induced aged mice. CONCLUSION: Collectively, these results provided evidence that the inhibition of SETD7 could alleviate neuroinflammation, pyroptosis, and cognitive impairment by suppressing the activation of the NLRP3 inflammasome in isoflurane-induced aged mice.


Asunto(s)
Anestésicos por Inhalación/efectos adversos , Técnicas de Silenciamiento del Gen , Inflamasomas/metabolismo , Metiltransferasas/metabolismo , Proteínas NLR/metabolismo , Dominios PR-SET/genética , Complicaciones Cognitivas Postoperatorias/inducido químicamente , Envejecimiento/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Inflamasomas/genética , Isoflurano/efectos adversos , Masculino , Metiltransferasas/genética , Ratones , Proteínas NLR/genética
20.
Arch Oral Biol ; 131: 105251, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34521010

RESUMEN

OBJECTIVE: The aim of this study is to show that the proliferation of chondrocytes is regulated by SET domain bifurcated 1 (SETDB1) along with the downregulation of parathyroid hormone (PTH)/parathyroid hormone-related peptide (PTHrP) receptor in Meckel's cartilage. DESIGN: Setdb1 was knocked down or overexpressed in a mouse chondrogenic ATDC5 cells, by transfecting the cells with short interfering RNA against Setdb1 or wild-type Setdb1 expression vector, respectively. Cell proliferation was detected by bromodeoxyuridine incorporation. Setdb1 was conditionally deleted in neural crest cells with Wnt1-Cre (Setdb1 conditional knockout mice). Immunofluorescence staining of paraffin sections of embryonic days 13.5 and 14.5 Setdb1 conditional knockout mice or transfected ATDC5 cells was performed to detect PTH/PTHrP receptor. Protein kinase B (AKT) phosphorylation inhibitor was added to both siRNA-transfected ATDC5 cultures to determine whether AKT activation induces PTH/PTHrP receptor expression after Setdb1 knockdown or vice versa. RESULTS: Setdb1 knockdown in ATDC5 cells showed increased cell proliferation and parathyroid hormone receptor 1 expression. Contrasting results were observed in the Setdb1-overexpressed wild-type cells. Immunofluorescence staining showed the highly expressed PTH/PTHrP receptor in Setdb1-knocked down ATDC5 cells and in the chondrocytes of Setdb1 conditional knockout embryonic Meckel's cartilage, indicating the negative regulation of SETDB1 on PTH/PTHrP receptor. Strong staining of phosphorylated AKT was observed in Setdb1-knocked down ATDC5 cells. However, the inhibition of AKT phosphorylation significantly reduced both the PTH/PTHrP receptor staining and the Setdb1-knockdown-induced increase in ATDC5 cell proliferation. CONCLUSIONS: Our findings contribute new insights on SETDB1 function in relation with AKT and PTH/PTHrP receptor during chondrocyte proliferation.


Asunto(s)
Condrocitos , N-Metiltransferasa de Histona-Lisina/genética , Receptor de Hormona Paratiroídea Tipo 1 , Animales , Cartílago , Proliferación Celular , Ratones , Dominios PR-SET , Hormona Paratiroidea , Proteína Relacionada con la Hormona Paratiroidea
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA