Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Cancer ; 15(9): 2746-2758, 2024.
Article in English | MEDLINE | ID: mdl-38577602

ABSTRACT

Purpose: To explore the effects of miR-383-5p and serine hydroxymethyltransferase 2 (SHMT2) on the proliferation and migration of lung adenocarcinoma cells. Methods: SHMT2 expression in lung adenocarcinoma and normal tissues was investigated using The Cancer Genome Atlas database. Immunohistochemical analysis was performed to confirm SHMT2 expression in lung adenocarcinoma and adjacent normal lung tissues. Bioinformatics analysis and luciferase reporter assays were used to analyze the relationship between miR-383-5p and SHMT2 expression. The protein expression levels of SHMT2, vimentin, N-cadherin, E-cadherin, Bcl-2, and cyclinD1 were analyzed using western blotting. The reverse transcription-quantitative polymerase chain reaction was used to detect SHMT2 knockdown efficiency, miR-383-5p overexpression, and inhibition efficiency. The proliferative ability of cells was detected using the Cell Counting Kit-8 assay. The Transwell assay was used to detect the migration ability of cells. Results: SHMT2 expression was significantly increased in patients with lung adenocarcinoma compared to that in control patients; the higher the SHMT2 expression the worse the outcomes were in patients with lung adenocarcinoma. SHMT2 knockdown inhibited the proliferation, migration, and epithelial-mesenchymal transition of lung adenocarcinoma A549 and H1299 cells. MiR-383-5p directly targeted and downregulated SHMT2 in A549 and H1299 cells. The effects of miRNA-383-5p on the proliferation and migration of these cells differed from those of SHMT2. Exogenous overexpression of SHMT2 reversed the miR-383-5p-induced proliferation and migration inhibition in A549 and H1299 cells. Conclusion: MiR-383-5p inhibits the proliferation and migration of lung adenocarcinoma cells by targeting and downregulating SHMT2.

2.
Cancer Res ; 84(5): 652-658, 2024 03 04.
Article in English | MEDLINE | ID: mdl-38194336

ABSTRACT

Mammalian members of the lysyl oxidase (LOX) family of proteins carry a copper-dependent monoamine oxidase domain exclusively within the C-terminal region, which catalyzes ε-amine oxidation of lysine residues of various proteins. However, recent studies have demonstrated that in LOX-like (LOXL) 2-4 the C-terminal canonical catalytic domain and N-terminal scavenger receptor cysteine-rich (SRCR) repeats domain exhibit lysine deacetylation and deacetylimination catalytic activities. Moreover, the N-terminal SRCR repeats domain is more catalytically active than the C-terminal oxidase domain. Thus, LOX is the third family of lysine deacetylases in addition to histone deacetylase and sirtuin families. In this review, we discuss how the LOX family targets different cellular proteins for deacetylation and deacetylimination to control the development and metastasis of cancer.


Subject(s)
Neoplasms , Protein-Lysine 6-Oxidase , Animals , Humans , Protein-Lysine 6-Oxidase/metabolism , Amino Acid Oxidoreductases/metabolism , Lysine , Protein Domains , Neoplasms/genetics , Mammals/metabolism
3.
Int Immunopharmacol ; 124(Pt B): 111031, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37827058

ABSTRACT

Abnormalities in the IL-6/STAT3 signaling pathway can sometimes result in extremely high levels of IgE concentration in serum, however, the regulatory role of STAT3 in IgE production is elusive. We used several genetically modified mice with distinctive germline Stat3 transcriptional activity to assess the influence of Stat3 on the biochemical characteristics of IgE and found that the IgE concentration in serum is inversely proportional to Stat3 transcriptional activity. Intriguingly, the serum IgE concentration is directly proportional to IgE-producing B cells in Stat3-GOF mice but inversely proportional in mice carrying Stat3 mutations with reduced transcriptional activity. For reduced Stat3 transcriptional activity induced high levels of IgE in the mice, IL-4/Stat6 signaling is indispensable for IgE production, but it was observed that an increased IgE concentration was accompanied by reduced IL-4/Stat6 signaling and lessened IgE-producing B cells, which implies that an increase in IgE concentration may result from an extended half-life of IgE but not an increasing number of IgE-producing cells.


Subject(s)
Immunoglobulin E , Interleukin-4 , Mice , Animals , Interleukin-4/genetics , Interleukin-4/metabolism , B-Lymphocytes/metabolism , Signal Transduction , Mutation , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
4.
Mol Breed ; 43(8): 62, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37521314

ABSTRACT

Heading date is a critical agronomic trait that determines crop yield. Although numerous genes associated with heading date have been identified in rice, the mechanisms involving Small Auxin Up RNA (SAUR) family have not been elucidated. In this study, the biological function of several SAUR genes was initially investigated using the CRISPR-Cas9 technology in the Japonica cultivar Zhonghua11 (ZH11) background. Further analysis revealed that the loss-of-function of OsSAUR56 affected heading date in both NLD (natural long-day) and ASD (artificial short-day). OsSAUR56 exhibited predominant expression in the anther, with its protein localized in both the cytoplasm and nucleus. OsSAUR56 regulated flowering time and heading date by modulating the expression of the clock gene OsGI, as well as two repressors Ghd7 and DTH8. Furthermore, haplotype-phenotype association analysis revealed a strong correlation between OsSAUR56 and heading date, suggesting its role in selection during the domestication of rice. In summary, these findings highlights the importance of OsSAUR56 in the regulation of heading date for further potential facilitating genetic engineering for flowering time during rice breeding. Supplementary Information: The online version contains supplementary material available at 10.1007/s11032-023-01409-w.

5.
Adv Sci (Weinh) ; 10(3): e2205462, 2023 01.
Article in English | MEDLINE | ID: mdl-36453571

ABSTRACT

Acetylation of extracellular proteins has been observed in many independent studies where particular attention has been given to the dynamic change of the microenvironmental protein post-translational modifications. While extracellular proteins can be acetylated within the cells prior to their micro-environmental distribution, their deacetylation in a tumor microenvironment remains elusive. Here it is described that multiple acetyl-vWA domain-carrying proteins including integrin ß3 (ITGB3) and collagen 6A (COL6A) are deacetylated by Sirtuin family member SIRT2 in extracellular space. SIRT2 is secreted by macrophages following toll-like receptor (TLR) family member TLR4 or TLR2 activation. TLR-activated SIRT2 undergoes autophagosome translocation. TNF receptor associated factor 6 (TRAF6)-mediated autophagy flux in response to TLR2/4 activation can then pump SIRT2 into the microenvironment to function as extracellular SIRT2 (eSIRT2). In the extracellular space, eSIRT2 deacetylates ITGB3 on aK416 involved in cell attachment and migration, leading to a promotion of cancer cell metastasis. In lung cancer patients, significantly increased serum eSIRT2 level correlates with dramatically decreased ITGB3-K416 acetylation in cancer cells. Thus, the extracellular space is a subcellular organelle-like arena where eSIRT2 promotes cancer cell metastasis via catalyzing extracellular protein deacetylation.


Subject(s)
Lung Neoplasms , Sirtuin 2 , Humans , Sirtuin 2/genetics , Sirtuin 2/metabolism , Toll-Like Receptor 2/metabolism , Protein Processing, Post-Translational , Acetylation , Tumor Microenvironment
6.
Int J Mol Sci ; 23(23)2022 Nov 23.
Article in English | MEDLINE | ID: mdl-36498964

ABSTRACT

The loss of the shattering ability is one of the key events in rice domestication. The strength of the seed shattering ability is closely related to the harvest yield and the adaptability of modern mechanical harvesting methods. In this study, using a population of 587 natural rice cultivars, quantitative trait loci associated with seed shattering were detected by genome-wide association studies (GWASs). We consider the quantitative trait loci (QTLs) qBTS1 and qBTS3 to be the key loci for seed shattering in rice. Additionally, the abscission zone (AZ) and nonabscission zone (NAZ) of materials with a loss of shattering (DZ129) and easy shattering (W517) were subjected to RNA-Seq, and high-quality differential expression profiles were obtained. The AZ-specific differentially expressed genes (DEGs) of W517 were significantly enriched in plant hormone signal transduction, while the AZ-specific DEGs of DZ129 were enriched in phenylpropanoid biosynthesis. We identified candidate genes for the lignin-associated laccase precursor protein (LOC_Os01g63180) and the glycoside hydrolase family (LOC_Os03g14210) in the QTLs qBTS1 (chromosome 1) and qBTS3 (chromosome 3), respectively. In summary, our findings lay the foundation for the further cloning of qBTS1 and qBTS3, which would provide new insights into seed shattering in rice.


Subject(s)
Oryza , Oryza/genetics , Genome-Wide Association Study , RNA-Seq , Phenotype , Seeds/genetics
7.
Cancer Res ; 82(23): 4400-4413, 2022 12 02.
Article in English | MEDLINE | ID: mdl-36197797

ABSTRACT

Lysyl oxidase-like 2 (LOXL2) is a member of the scavenger receptor cysteine-rich (SRCR) repeat carrying LOX family. Although LOXL2 is suspected to be involved in histone association and chromatin modification, the role of LOXL2 in epigenetic regulation during tumorigenesis and cancer progression remains unclear. Here, we report that nuclear LOXL2 associates with histone H3 and catalyzes H3K36ac deacetylation and deacetylimination. Both the N-terminal SRCR repeats and the C-terminal catalytic domain of LOXL2 carry redundant deacetylase catalytic activity. Overexpression of LOXL2 markedly reduced H3K36 acetylation and blocked H3K36ac-dependent transcription of genes, including c-MYC, CCND1, HIF1A, and CD44. Consequently, LOXL2 overexpression reduced cancer cell proliferation in vitro and inhibited xenograft tumor growth in vivo. In contrast, LOXL2 deficiency resulted in increased H3K36 acetylation and aberrant expression of H3K36ac-dependent genes involved in multiple oncogenic signaling pathways. Female LOXL2-deficient mice spontaneously developed uterine hypertrophy and uterine carcinoma. Moreover, silencing LOXL2 in cancer cells enhanced tumor progression and reduced the efficacy of cisplatin and anti-programmed cell death 1 (PD-1) combination therapy. Clinically, low nuclear LOXL2 expression and high H3K36ac levels corresponded to poor prognosis in uterine endometrial carcinoma patients. These results suggest that nuclear LOXL2 restricts cancer development in the female reproductive system via the regulation of H3K36ac deacetylation. SIGNIFICANCE: LOXL2 loss reprograms the epigenetic landscape to promote uterine cancer initiation and progression and repress the efficacy of anti-PD-1 immunotherapy, indicating that LOXL2 is a tumor suppressor.


Subject(s)
Amino Acid Oxidoreductases , Epigenesis, Genetic , Humans , Mice , Female , Animals , Amino Acid Oxidoreductases/genetics , Amino Acid Oxidoreductases/metabolism , Acetylation , Histones/metabolism , Hypertrophy/genetics , Gene Expression
8.
Front Oncol ; 12: 872444, 2022.
Article in English | MEDLINE | ID: mdl-35707358

ABSTRACT

Axin1 is a fundamental scaffolding protein of the destruction complex in the canonical Wnt signaling pathway, which plays a critical role in various biological processes. However, how Axin1 is regulated in the activation of the canonical Wnt signaling pathway remains elusive. Here, we report that Axin1 is constitutively acetylated in resting cells. Upon stimulation with Wnt, SIRT4 translocates from mitochondria to the cytoplasm and catalyzes Axin1 deacetylation, thus turning off the destruction complex. In this process, Lys147, a residue in the RGS domain of Axin1, plays a key role. We proved that the Axin1-K147R mutant impairs the assembly of ß-TrCP to the destruction complex, which leads to ß-catenin accumulation even without Wnt stimulation. In summary, our work proposes a new model for better understanding the initial stage of the canonical Wnt signaling pathway in which SIRT4 translocates from mitochondria into the cytoplasm to deacetylate Axin1-K147 after Wnt stimulation, which results in reduced assembly of ß-TrCP to the destruction complex.

9.
Cell Death Differ ; 29(11): 2303-2315, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35614130

ABSTRACT

T helper 1 (Th1) immunity is typically viewed as a critical adaptation by vertebrates against intracellular pathogens. Identifying novel targets to enhance Th1 cell differentiation and function is increasingly important for anti-infection immunity. Here, through small-molecule screening focusing on epigenetic modifiers during the in vitro Th1 cell differentiation process, we identified that the selective histone deacetylase 6 (HDAC6) inhibitors ricolinostat and nexturastat A (Nex A) promoted Th1 cell differentiation. HDAC6-depleted mice exhibit elevation of Th1 cell differentiation, and decreased severity of Listeria monocytogenes infection. Mechanistically, HDAC6 directly deacetylated CBP-catalyzed acetylation of signal transducer and activator of transcription 4 (STAT4)-lysine (K) 667 via its enzymatic activity. Acetylation of STAT4-K667 is required for JAK2-mediated phosphorylation and activation of STAT4. Stat4K667R mutant mice lost the ability to normally differentiate into Th1 cells and developed severe Listeria infection. Our study identifies acetylation of STAT4-K667 as an essential signaling event for Th1 cell differentiation and defense against intracellular pathogen infections, and highlights the therapeutic potential of HDAC6 inhibitors for controlling intracellular pathogen infections.


Subject(s)
Listeria monocytogenes , Listeriosis , Mice , Animals , Acetylation , Th1 Cells , STAT4 Transcription Factor , Signal Transduction , Cell Differentiation
10.
Front Immunol ; 13: 855645, 2022.
Article in English | MEDLINE | ID: mdl-35392084

ABSTRACT

Background: Ulcerative colitis (UC) is an inflammatory disease of the intestinal mucosa, and its incidence is steadily increasing worldwide. Intestinal immune dysfunction has been identified as a central event in UC pathogenesis. However, the underlying mechanisms that regulate dysfunctional immune cells and inflammatory phenotype remain to be fully elucidated. Methods: Transcriptome profiling of intestinal mucosa biopsies were downloaded from the GEO database. Robust Rank Aggregation (RRA) analysis was performed to identify statistically changed genes and differentially expressed genes (DEGs). Gene Set Enrichment Analysis (GSEA), Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) were used to explore potential biological mechanisms. CIBERSORT was used to evaluate the proportion of 22 immune cells in biopsies. Weighted co-expression network analysis (WGCNA) was used to determine key module-related clinical traits. Protein-Protein Interaction (PPI) network and Cytoscape were performed to explore protein interaction network and screen hub genes. We used a validation cohort and colitis mouse model to validate hub genes. Several online websites were used to predict competing endogenous RNA (ceRNA) network. Results: RRA integrated analysis revealed 1838 statistically changed genes from four training cohorts (adj. p-value < 0.05). GSEA showed that statistically changed genes were enriched in the innate immune system. CIBERSORT analysis uncovered an increase in activated dendritic cells (DCs) and M1 macrophages. The red module of WGCNA was considered the most critical module related to active UC. Based on the results of the PPI network and Cytoscape analyses, we identified six critical genes and transcription factor NF-κB. RT-PCR revealed that andrographolide (AGP) significantly inhibited the expression of hub genes. Finally, we identified XIST and three miRNAs (miR-9-5p, miR-129-5p, and miR-340-5p) as therapeutic targets. Conclusions: Our integrated analysis identified four hub genes (CXCL1, IL1B, MMP1, and MMP10) regulated by NF-κB. We further revealed that AGP decreased the expression of hub genes by inhibiting NF-κB activation. Lastly, we predicted the involvement of ceRNA network in the regulation of NF-κB expression. Collectively, our results provide valuable information in understanding the molecular mechanisms of active UC. Furthermore, we predict the use of AGP and small RNA combination for the treatment of UC.


Subject(s)
Colitis, Ulcerative , MicroRNAs , Animals , Colitis, Ulcerative/genetics , Computational Biology/methods , Gene Regulatory Networks , Humans , Mice , MicroRNAs/genetics , NF-kappa B/genetics
11.
J Cancer ; 12(19): 5888-5894, 2021.
Article in English | MEDLINE | ID: mdl-34476002

ABSTRACT

In the last few years, cellular metabolic reprogramming has been acknowledged as a hallmark of human cancer and evaluated for its crucial role in supporting the proliferation and survival of human cancer cells. In a variety of human tumours, including hepatocellular carcinoma (HCC), breast cancer and non-small-cell lung cancer (NSCLC), a large amount of carbon is reused in serine/glycine biosynthesis, accompanied by higher expression of the key glycine synthetic enzyme mitochondrial serine hydroxymethyltransferase 2 (SHMT2). This enzyme can convert serine into glycine and a tetrahydrofolate-bound one-carbon unit, ultimately supporting thymidine synthesis and purine synthesis and promoting tumour growth. In tumour samples, elevated expression of SHMT2 was found to be associated with poor prognosis. In this review, the pivotal roles of SHMT2 in human carcinogenesis are described, highlighting the underlying regulatory mechanisms through promotion of tumour progression. In conclusion, SHMT2 may serve as a prognostic marker and a target for anticancer therapies.

12.
Int J Mol Sci ; 21(7)2020 Mar 28.
Article in English | MEDLINE | ID: mdl-32231094

ABSTRACT

MAPK (mitogen-activated protein kinase) signaling pathways regulate a variety of biological processes through multiple cellular mechanisms. In most of these processes, such as apoptosis, MAPKs have a dual role since they can act as activators or inhibitors, depending on the cell type and the stimulus. In this review, we present the main pro- and anti-apoptotic mechanisms regulated by MAPKs, as well as the crosstalk observed between some MAPKs. We also describe the basic signaling properties of MAPKs (ultrasensitivity, hysteresis, digital response), and the presence of different positive feedback loops in apoptosis. We provide a simple guide to predict MAPKs' behavior, based on the intensity and duration of the stimulus. Finally, we consider the role of MAPKs in osmostress-induced apoptosis by using Xenopus oocytes as a cell model. As we will see, apoptosis is plagued with multiple positive feedback loops. We hope this review will help to understand how MAPK signaling pathways engage irreversible cellular decisions.


Subject(s)
Apoptosis , MAP Kinase Signaling System , Animals , Enzyme Activation , Humans , Mitogen-Activated Protein Kinases/metabolism , Osmotic Pressure
13.
Dev Biol ; 416(1): 42-51, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27297883

ABSTRACT

Xenopus ERK2, also known as Xp42 MAPK, is activated by progesterone and regulates meiotic progression in the oocytes through activation of the phosphatase Cdc25C and inhibition of the protein kinase Myt1, thus promoting dephosphorylation and activation of cyclinB/Cdc2 (MPF). Indeed, it has been reported that stress protein kinases p38 and JNK are activated during meiotic progression and, more specifically, that p38γ regulates meiosis through activation of Cdc25C. However, the role of JNK in meiotic progression is not so clear, and despite a 42kDa protein is detected with pJNK antibodies (XpJNK-p42), the specific isoform activated by progesterone has not been characterized in detail. The serine/threonine kinase MEKK1, an upstream activator of JNK and p38, is activated during stress conditions and regulates apoptosis in different cell types. Here we show that ectopic expression of a constitutively active MEKK1 in Xenopus oocytes induces phosphorylation of p38, JNK and ERK and accelerates meiotic progression induced by progesterone. Inhibition of each individual pathway reduces the acceleration of meiosis induced by MEKK1. However, constitutively active MEKK1 induces phosphorylation of two JNK isoforms (p40 and p49, corresponding to JNK1-1 and JNK1-2 respectively) distinct to the p42 protein detected with pJNK antibodies during meiotic progression (XpJNK-p42). Moreover, a constitutively active MKK7, which specifically activates the JNK signaling pathway and induces phosphorylation of the p40 and p49 isoforms, does not accelerate meiotic progression. Immunoprecipitation of the p42 protein with pJNK antibodies and subsequent analysis by mass spectrometry shows that XpJNK-p42 is, in fact, pERK2. Ectopic expression of ERK2 in oocytes treated with progesterone or hyperosmotic shock indicates that ERK2 is phosphorylated in both conditions but is only detected with pJNK antibodies in progesterone-treated oocytes. In addition, mature oocytes only present a moderate increase of Jun kinase activity, which is not inhibited by SP600125. In conclusion, JNKs are not activated during meiotic progression and XpJNK-p42 is a post-translational modification of pERK induced by progesterone.


Subject(s)
MAP Kinase Kinase 4/metabolism , MAP Kinase Kinase Kinase 1/metabolism , Meiosis , Oocytes/cytology , eIF-2 Kinase/metabolism , Animals , Female , MAP Kinase Kinase 7/metabolism , MAP Kinase Signaling System , Mesothelin , Mice , Mitogen-Activated Protein Kinase 1 , Oocytes/enzymology , Progesterone/metabolism , Xenopus , Xenopus Proteins , p38 Mitogen-Activated Protein Kinases/metabolism
14.
J Biol Chem ; 290(51): 30375-89, 2015 Dec 18.
Article in English | MEDLINE | ID: mdl-26511318

ABSTRACT

Hyperosmotic shock induces early calpain activation, Smac/DIABLO release from the mitochondria, and p38/JNK activation in Xenopus oocytes. These pathways regulate late cytochrome c release and caspase-3 activation. Here, we show that JNK1-1 and JNK1-2 are activated early by osmostress, and sustained activation of both isoforms accelerates the apoptotic program. When caspase-3 is activated, JNK1-2 is proteolyzed at Asp-385 increasing the release of cytochrome c and caspase-3 activity, thereby creating a positive feedback loop. Expression of Bcl-xL markedly reduces hyperosmotic shock-induced apoptosis. In contrast, expression of Bid induces rapid caspase-3 activation, even in the absence of osmostress, which is blocked by Bcl-xL co-expression. In these conditions a significant amount of Bid in the cytosol is mono- and bi-ubiquitinated. Caspase-3 activation by hyperosmotic shock induces proteolysis of Bid and mono-ubiquitinated Bid at Asp-52 increasing the release of cytochrome c and caspase-3 activation, and thus creating a second positive feedback loop. Revealing the JNK isoforms and the loops activated by osmostress could help to design better treatments for human diseases caused by perturbations in fluid osmolarity.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Caspase 3/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Oocytes/metabolism , Osmotic Pressure/physiology , Proteolysis , Signal Transduction , Xenopus Proteins/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Caspase 3/genetics , Enzyme Activation , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Mitogen-Activated Protein Kinase 8/genetics , Oocytes/cytology , Xenopus Proteins/genetics , Xenopus laevis
15.
PLoS One ; 10(4): e0124482, 2015.
Article in English | MEDLINE | ID: mdl-25866890

ABSTRACT

Hyperosmotic shock induces cytochrome c release and caspase-3 activation in Xenopus oocytes, but the regulators and signaling pathways involved are not well characterized. Here we show that hyperosmotic shock induces rapid calpain activation and high levels of Smac/DIABLO release from the mitochondria before significant amounts of cytochrome c are released to promote caspase-3 activation. Calpain inhibitors or EGTA microinjection delays osmostress-induced apoptosis, and blockage of Smac/DIABLO with antibodies markedly reduces cytochrome c release and caspase-3 activation. Hyperosmotic shock also activates the p38 and JNK signaling pathways very quickly. Simultaneous inhibition of both p38 and JNK pathways reduces osmostress-induced apoptosis, while sustained activation of these kinases accelerates the release of cytochrome c and caspase-3 activation. Therefore, at least four different pathways early induced by osmostress converge on the mitochondria to trigger apoptosis. Deciphering the mechanisms of hyperosmotic shock-induced apoptosis gives insight for potential treatments of human diseases that are caused by perturbations in fluid osmolarity.


Subject(s)
Apoptosis , Calpain/metabolism , MAP Kinase Kinase 4/metabolism , Mitochondrial Proteins/metabolism , Oocytes/cytology , Xenopus Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Female , Osmotic Pressure , Signal Transduction , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...