Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 341
Filter
1.
BMC Cancer ; 23(1): 18, 2023 Jan 06.
Article in English | MEDLINE | ID: mdl-36604642

ABSTRACT

BACKGROUND: SET domain containing 6 (SETD6) has been shown to be upregulated in multiple human cancers and can promote malignant cell survival. However, expression and function of SETD6 in lung adenocarcinoma (LUAD) remains unaddressed. This study aimed to demonstrate the expression pattern, biological roles and potential mechanisms by which SETD6 dysregulation is associated with LUAD. METHODS: The expression level of SETD6 was evaluated in LUAD clinical specimens and its correlation with clinical parameters were analyzed. In vitro, gain-of-function and loss-of-function experiments were performed to evaluate the effects of SETD6 on cell proliferation, apoptosis, migration, and colony formation of LUAD cell line A549. Western-blot was performed to investigate the involvement of nuclear factor-κB (NF-κB) and nuclear factor erythroid 2-related factor 2 (Nrf2) pathways as downstream signaling of SETD6 in LUAD cells. RESULTS: Compared with non-tumorous tissues, SETD6 was overexpressed in tumor tissues, and its overexpression significantly correlates with higher rates of regional lymph node metastasis and poor prognosis in patients with LUAD. In A549 cell line, SETD6 overexpression could promote cell proliferation, migration, colony formation and inhibit cell apoptosis, whereas SETD6 knockdown caused the opposite effects. Furthermore, we demonstrated that the mechanisms underlying the effect of SETD6 on LUAD biological behaviors may be through its interaction with NF-κB and Nrf2 signaling pathways. CONCLUSIONS: SETD6, which is highly expressed in LUAD tumor tissues, plays an important role in promoting the malignant behaviors of LUAD via likely the NF-κB and Nrf2 signaling pathways.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Humans , Lung Neoplasms/pathology , NF-kappa B/genetics , NF-kappa B/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Cell Line, Tumor , Adenocarcinoma of Lung/pathology , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Cell Movement/genetics , Protein Methyltransferases/genetics
2.
Clin Immunol ; 243: 109105, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36055572

ABSTRACT

Epigenetic modifications contribute to lymphomagenesis. Here, we performed an expression clustering analysis and identified two epigenetic-related clusters (EC1 and EC2). EC1 presented abundant TP53, MYD88, HIST1H1D, HIST1H1C, KMT2D and EZH2 mutations and an inferior prognosis. Pathways involved in the regulation of DNA methylation/demethylation, histone methyltransferase activity, and protein methyltransferase activity were significantly enriched in EC1. However, EC2 was frequently accompanied by B2M, CD70 and MEF2B mutations, which presented with enrichments in DNA damage repair, cytokine-mediated and B-cell activated immune signaling, increased levels of CD8+ T-, γδT- and T helper-cells, as well as immune scores and immunogenic cell death (ICD) modulators. According to the prediction, EC1 was more sensitive to vorinostat, serdemetan and navitoclax. However, ruxolitinib, cytarabine and CP466722 were more suitable treatments for EC2. The novel immune-related epigenetic signature exhibits promising clinical predictive value for diffuse large B-cell lymphoma (DLBCL), particularly for guiding epigenetic therapeutic regimens. R-CHOP (rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone) based combination treatment regimens are suggested.


Subject(s)
Epigenesis, Genetic , Lymphoma, Large B-Cell, Diffuse , Transcriptome , Antibodies, Monoclonal, Murine-Derived/genetics , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cyclophosphamide/therapeutic use , Cytarabine/therapeutic use , Cytokines/genetics , Doxorubicin/therapeutic use , Epigenesis, Genetic/immunology , Histone Methyltransferases/genetics , Humans , Lymphoma, Large B-Cell, Diffuse/diagnosis , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/genetics , Myeloid Differentiation Factor 88/genetics , Prednisone/therapeutic use , Prognosis , Protein Methyltransferases/genetics , Rituximab/therapeutic use , Vincristine/therapeutic use , Vorinostat/therapeutic use
3.
Methods Mol Biol ; 2529: 3-40, 2022.
Article in English | MEDLINE | ID: mdl-35733008

ABSTRACT

Dynamic posttranslational modifications to canonical histones that constitute the nucleosome (H2A, H2B, H3, and H4) control all aspects of enzymatic transactions with DNA. Histone methylation has been studied heavily for the past 20 years, and our mechanistic understanding of the control and function of individual methylation events on specific histone arginine and lysine residues has been greatly improved over the past decade, driven by excellent new tools and methods. Here, we will summarize what is known about the distribution and some of the functions of protein methyltransferases from all major eukaryotic supergroups. The main conclusion is that protein, and specifically histone, methylation is an ancient process. Many taxa in all supergroups have lost some subfamilies of both protein arginine methyltransferases (PRMT) and the heavily studied SET domain lysine methyltransferases (KMT). Over time, novel subfamilies, especially of SET domain proteins, arose. We use the interactions between H3K27 and H3K36 methylation as one example for the complex circuitry of histone modifications that make up the "histone code," and we discuss one recent example (Paramecium Ezl1) for how extant enzymes that may resemble more ancient SET domain KMTs are able to modify two lysine residues that have divergent functions in plants, fungi, and animals. Complexity of SET domain KMT function in the well-studied plant and animal lineages arose not only by gene duplication but also acquisition of novel DNA- and histone-binding domains in certain subfamilies.


Subject(s)
Histones , Protein Methyltransferases , Animals , Arginine/metabolism , Eukaryota/genetics , Eukaryota/metabolism , Histone-Lysine N-Methyltransferase/chemistry , Histones/metabolism , Lysine/metabolism , Methylation , Methyltransferases/genetics , Methyltransferases/metabolism , Protein Methyltransferases/genetics , Protein Methyltransferases/metabolism , Protein Processing, Post-Translational
4.
Biochimie ; 200: 27-35, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35550916

ABSTRACT

Among the protein lysine methyltransferases family members, it appears that SETD6 is highly similar and closely related to SETD3. The two methyltransferases show high similarity in their structure, which raised the hypothesis that they share cellular functions. Using a proteomic screen, we identified 52 shared interacting-proteins. Gene Ontology (GO) analysis of the shared proteins revealed significant enrichment of proteins involved in transcription. Our RNA-seq data of SETD6 KO and SETD3 KO HeLa cells identified ∼100 up-regulated and down-regulated shared genes. We have also identified a substantial number of genes that changed dramatically in the double KO cells but did not significantly change in the single KO cells. GO analysis of these genes revealed enrichment of apoptotic genes. Accordingly, we show that the double KO cells displayed high apoptotic levels, suggesting that SETD6 and SETD3 inhibit apoptosis. Collectively, our data strongly suggest a functional link between SETD6 and SETD3 in the regulation of apoptosis.


Subject(s)
Histone Methyltransferases , Protein Methyltransferases , Proteomics , Apoptosis/genetics , HeLa Cells , Histone Methyltransferases/genetics , Histone Methyltransferases/metabolism , Humans , Protein Methyltransferases/genetics , Protein Methyltransferases/metabolism , Structure-Activity Relationship
5.
J Clin Lab Anal ; 36(5): e24348, 2022 May.
Article in English | MEDLINE | ID: mdl-35312113

ABSTRACT

BACKGROUND: circRNA hsa_circ_0018289-mediated growth and metastasis of CC cells were investigated, as well as the mechanistic pathway. METHODS: Quantitative real-time reverse transcription-polymerase chain reaction (qRT-PCR) was carried out to examine the expression of hsa_circ_0018289, microRNA (miR)-1294, and isoprenylcysteine carboxyl methyltransferase (ICMT). CC cell proliferation, migration, and invasion were evaluated by 5-ethynyl-2'-deoxyuridine (EdU) incorporation, colony formation, transwell assays, Western blot analysis of ICMT, and glycolysis-associated proteins. Dual-luciferase reporter or RNA pull-down analysis of the target interaction between miR-1294 and hsa_hsa_circ_0018289 or ICMT. Xenograft model assay was implemented to assess the role of hsa_circ_0018289 in vivo. Immunofluorescence (IHC) was employed to detect the level of Ki-67. RESULTS: Hsa_circ_0018289 was elevated in CC tissues and cells, its deficiency could repress growth, metastasis, and glycolysis of CC cells in vitro, as well as hamper tumor growth in vivo. Hsa_circ_0018289 sponged miR-1294 while miR-1294 bound with ICMT, and the inhibition of miR-1294 or addition of ICMT could partially relieve the effect caused by hsa_circ_0018289 depletion. CONCLUSION: Hsa_circ_0018289 contributes to malignant development by regulating the miR-1294/ICMT axis, affording novel insight into CC therapy.


Subject(s)
MicroRNAs , Protein Methyltransferases , RNA, Circular , Uterine Cervical Neoplasms , Carcinogenesis , Cell Proliferation/genetics , Female , Humans , MicroRNAs/genetics , Protein Methyltransferases/genetics , RNA, Circular/genetics , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology
6.
FEBS J ; 289(5): 1256-1275, 2022 03.
Article in English | MEDLINE | ID: mdl-33774927

ABSTRACT

PRDF1 and RIZ1 homology domain containing (PRDMs) are a subfamily of Krüppel-like zinc finger proteins controlling key processes in metazoan development and in cancer. PRDMs exhibit unique dualities: (a) PR domain/ZNF arrays-their structure combines a SET-like domain known as a PR domain, typically found in methyltransferases, with a variable array of C2H2 zinc fingers (ZNF) characteristic of DNA-binding transcription factors; (b) transcriptional activators/repressors-their physiological function is context- and cell-dependent; mechanistically, some PRDMs have a PKMT activity and directly catalyze histone lysine methylation, while others are rather pseudomethyltransferases and act by recruiting transcriptional cofactors; (c) oncogenes/tumor suppressors-their pathological function depends on the specific PRDM isoform expressed during tumorigenesis. This duality is well known as the 'Yin and Yang' of PRDMs and involves a complex regulation of alternative splicing or alternative promoter usage, to generate full-length or PR-deficient isoforms with opposing functions in cancer. In conclusion, once their dualities are fully appreciated, PRDMs represent a promising class of targets in oncology by virtue of their widespread upregulation across multiple tumor types and their somatic dispensability, conferring a broad therapeutic window and limited toxic side effects. The recent discovery of a first-in-class compound able to inhibit PRDM9 activity has paved the way for the identification of further small molecular inhibitors able to counteract PRDM oncogenic activity.


Subject(s)
Epigenesis, Genetic , Protein Methyltransferases/metabolism , Amino Acid Sequence , Carcinogenesis , Crystallization , DNA/metabolism , Meiosis , Neoplasms/enzymology , Neoplasms/pathology , Oncogenes , Protein Binding , Protein Conformation , Protein Domains , Protein Methyltransferases/chemistry , Protein Methyltransferases/genetics , Sequence Alignment , Signal Transduction
7.
Life Sci Alliance ; 4(12)2021 12.
Article in English | MEDLINE | ID: mdl-34610973

ABSTRACT

DNA damage is a double-edged sword for cancer cells. On the one hand, DNA damage-induced genomic instability contributes to cancer development; on the other hand, accumulating damage compromises proliferation and survival of cancer cells. Understanding the key regulators of DNA damage repair machinery would benefit the development of cancer therapies that induce DNA damage and apoptosis. In this study, we found that isoprenylcysteine carboxylmethyltransferase (ICMT), a posttranslational modification enzyme, plays an important role in DNA damage repair. We found that ICMT suppression consistently reduces the activity of MAPK signaling, which compromises the expression of key proteins in the DNA damage repair machinery. The ensuing accumulation of DNA damage leads to cell cycle arrest and apoptosis in multiple breast cancer cells. Interestingly, these observations are more pronounced in cells grown under anchorage-independent conditions or grown in vivo. Consistent with the negative impact on DNA repair, ICMT inhibition transforms the cancer cells into a "BRCA-like" state, hence sensitizing cancer cells to the treatment of PARP inhibitor and other DNA damage-inducing agents.


Subject(s)
Breast Neoplasms/metabolism , DNA Damage/genetics , DNA Repair/genetics , MAP Kinase Signaling System/genetics , Protein Methyltransferases/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Benzamides/pharmacology , Breast Neoplasms/pathology , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , DNA Damage/drug effects , DNA Repair/drug effects , Female , Genetic Vectors , HEK293 Cells , Humans , Indazoles/pharmacology , MAP Kinase Signaling System/drug effects , Mice , Mice, SCID , Piperidines/pharmacology , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Protein Methyltransferases/genetics , RNA, Small Interfering/genetics , Ribonucleosides/pharmacology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
8.
Molecules ; 26(16)2021 Aug 21.
Article in English | MEDLINE | ID: mdl-34443661

ABSTRACT

Protein methyltransferases are vital to the epigenetic modification of gene expression. Thus, obtaining a better understanding of and control over the regulation of these crucial proteins has significant implications for the study and treatment of numerous diseases. One ideal mechanism of protein regulation is the specific installation of a photolabile-protecting group through the use of photocaged non-canonical amino acids. Consequently, PRMT1 was caged at a key tyrosine residue with a nitrobenzyl-protected Schultz amino acid to modulate protein function. Subsequent irradiation with UV light removes the caging group and restores normal methyltransferase activity, facilitating the spatial and temporal control of PRMT1 activity. Ultimately, this caged PRMT1 affords the ability to better understand the protein's mechanism of action and potentially regulate the epigenetic impacts of this vital protein.


Subject(s)
Epigenesis, Genetic/radiation effects , Protein Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/genetics , Repressor Proteins/genetics , Amino Acid Sequence/genetics , Amino Acids , Epigenesis, Genetic/genetics , Gene Expression/radiation effects , Humans , Methylation/radiation effects , Protein Methyltransferases/radiation effects , Protein-Arginine N-Methyltransferases/radiation effects , Repressor Proteins/radiation effects , Transcription Factors/genetics , Tyrosine/chemistry , Ultraviolet Rays
9.
Sci Adv ; 7(22)2021 05.
Article in English | MEDLINE | ID: mdl-34039605

ABSTRACT

The transcriptional coactivator BRD4 has a fundamental role in transcription regulation and thus became a promising epigenetic therapeutic candidate to target diverse pathologies. However, the regulation of BRD4 by posttranslational modifications has been largely unexplored. Here, we show that BRD4 is methylated on chromatin at lysine-99 by the protein lysine methyltransferase SETD6. BRD4 methylation negatively regulates the expression of genes that are involved in translation and inhibits total mRNA translation in cells. Mechanistically, we provide evidence that supports a model where BRD4 methylation by SETD6 does not have a direct role in the association with acetylated histone H4 at chromatin. However, this methylation specifically determines the recruitment of the transcription factor E2F1 to selected target genes that are involved in mRNA translation. Together, our findings reveal a previously unknown molecular mechanism for BRD4 methylation-dependent gene-specific targeting, which may serve as a new direction for the development of therapeutic applications.


Subject(s)
Cell Cycle Proteins , Nuclear Proteins , Protein Methyltransferases , Transcription Factors , Cell Cycle Proteins/genetics , Chromatin , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Methylation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Biosynthesis , Protein Methyltransferases/genetics , Protein Processing, Post-Translational , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Oncogene ; 40(20): 3548-3563, 2021 05.
Article in English | MEDLINE | ID: mdl-33927350

ABSTRACT

Mitochondrial oxidative phosphorylation (OXPHOS) is a vital regulator of tumor metastasis. However, the mechanisms governing OXPHOS to facilitate tumor metastasis remain unclear. In this study, we discovered that arginine 21(R21) and lysine 108 (K108) of mitochondrial ribosomal protein S23 (MRPS23) was methylated by the protein arginine methyltransferase 7 (PRMT7) and SET-domain-containing protein 6 (SETD6), respectively. R21 methylation accelerated the poly-ubiquitin-dependent degradation of MRPS23 to a low level. The MRPS23 degradation inhibited OXPHOS with elevated mtROS level, which consequently increased breast cancer cell invasion and metastasis. In contrast, K108 methylation increased MRPS23 stability, and K108 methylation coordinated with R21 methylation to maintain a low level of MRPS23, which was in favor of supporting breast cancer cell survival through regulating OXPHOS. Consistently, R21 and K108 methylation was correlated with malignant breast carcinoma. Significantly, our findings unveil a unique mechanism of controlling OXPHOS by arginine and lysine methylation and point to the impact of the PRMT7-SETD6-MRPS23 axis during breast cancer metastasis.


Subject(s)
Arginine/chemistry , Breast Neoplasms/metabolism , Lysine/chemistry , Mitochondria/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/physiology , Female , Humans , Methylation , Mitochondria/pathology , Neoplasm Metastasis , Organoids , Oxidative Phosphorylation , Protein Methyltransferases/genetics , Protein Methyltransferases/metabolism , Protein Processing, Post-Translational , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism
11.
Histol Histopathol ; 36(2): 229-237, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33710605

ABSTRACT

BACKGROUND: Oral squamous cell carcinoma (OSCC) is one of the most comment types of oral malignancies. SET-domain-containing protein 6 (SETD6) was recently identified as an important regulator of multiple signaling pathways through methylating protein substrates. Meanwhile, SETD6 is known to participate in multiple cancers. However, the role of SETD6 in OSCC remains unclear. METHODS: Gene and protein expressions in OSCC cells or tissues were detected by RT-qPCR and western blot, respectively. In addition, CCK-8 assay was used to test the cell viability. A transwell assay was performed to measure cell migration and invasion. Flow cytometry was used to test cell apoptosis and cycle. Meanwhile, methylation-specific PCR (MSP) was used to detect the status of promoter methylation. RESULTS: SETD6 was significantly upregulated in OSCC tissues. In addition, knockdown of SETD6 notably inhibited the proliferation and induced the apoptosis of OSCC cells. Furthermore, silencing of SETD6 notably suppressed the migration and invasion of OSCC cells. Meanwhile, SETD6 siRNA significantly inhibited the promoter methylation of RelA (NF-κB p65) and PAK4. Furthermore, SETD6 siRNA induced G1 arrest in OSCC cells. CONCLUSION: Knockdown of SETD6 inhibits the tumorigenesis of OSCC by suppressing promoter methylation of PAK4 and RelA. Therefore, our study might shed new light on exploring strategies for the treatment of OSCC.


Subject(s)
Carcinogenesis , Carcinoma, Squamous Cell/metabolism , Gene Silencing , Mouth Neoplasms/metabolism , Protein Methyltransferases/genetics , Transcription Factor RelA/metabolism , p21-Activated Kinases/metabolism , Apoptosis , Carcinoma, Squamous Cell/genetics , Cell Cycle , Cell Line, Tumor , Cell Proliferation , DNA Methylation , Gene Expression Regulation, Neoplastic , Genome, Human , Humans , Immunohistochemistry , Mouth Neoplasms/genetics , Neoplasms/metabolism , Polymerase Chain Reaction , Protein Domains , Protein Methyltransferases/metabolism , RNA, Small Interfering/metabolism , Signal Transduction
12.
PLoS Negl Trop Dis ; 15(3): e0009230, 2021 03.
Article in English | MEDLINE | ID: mdl-33651805

ABSTRACT

Leishmania major is the main causative agent of cutaneous leishmaniasis in the Old World. In Leishmania parasites, the lack of transcriptional control is mostly compensated by post-transcriptional mechanisms. Methylation of arginine is a conserved post-translational modification executed by Protein Arginine Methyltransferase (PRMTs). The genome from L. major encodes five PRMT homologs, including the cytosolic protein associated with several RNA-binding proteins, LmjPRMT7. It has been previously reported that LmjPRMT7 could impact parasite infectivity. In addition, a more recent work has clearly shown the importance of LmjPRMT7 in RNA-binding capacity and protein stability of methylation targets, demonstrating the role of this enzyme as an important epigenetic regulator of mRNA metabolism. In this study, we unveil the impact of PRMT7-mediated methylation on parasite development and virulence. Our data reveals that higher levels of LmjPRMT7 can impair parasite pathogenicity, and that deletion of this enzyme rescues the pathogenic phenotype of an attenuated strain of L. major. Interestingly, lesion formation caused by LmjPRMT7 knockout parasites is associated with an exacerbated inflammatory reaction in the tissue correlated with an excessive neutrophil recruitment. Moreover, the absence of LmjPRMT7 also impairs parasite development within the sand fly vector Phlebotomus duboscqi. Finally, a transcriptome analysis shed light onto possible genes affected by depletion of this enzyme. Taken together, this study highlights how post-transcriptional regulation can affect different aspects of the parasite biology.


Subject(s)
Leishmania major/enzymology , Leishmaniasis, Cutaneous/pathology , Neutrophils/physiology , Protein Methyltransferases/metabolism , Protozoan Proteins/metabolism , Animals , Gene Deletion , Gene Expression Regulation, Enzymologic , Leishmania major/genetics , Leishmania major/metabolism , Leishmaniasis, Cutaneous/parasitology , Mice , Protein Methyltransferases/genetics
13.
Elife ; 102021 02 02.
Article in English | MEDLINE | ID: mdl-33526168

ABSTRACT

A farnesylated and methylated form of prelamin A called progerin causes Hutchinson-Gilford progeria syndrome (HGPS). Inhibiting progerin methylation by inactivating the isoprenylcysteine carboxylmethyltransferase (ICMT) gene stimulates proliferation of HGPS cells and improves survival of Zmpste24-deficient mice. However, we don't know whether Icmt inactivation improves phenotypes in an authentic HGPS mouse model. Moreover, it is unknown whether pharmacologic targeting of ICMT would be tolerated by cells and produce similar cellular effects as genetic inactivation. Here, we show that knockout of Icmt improves survival of HGPS mice and restores vascular smooth muscle cell numbers in the aorta. We also synthesized a potent ICMT inhibitor called C75 and found that it delays senescence and stimulates proliferation of late-passage HGPS cells and Zmpste24-deficient mouse fibroblasts. Importantly, C75 did not influence proliferation of wild-type human cells or Zmpste24-deficient mouse cells lacking Icmt, indicating drug specificity. These results raise hopes that ICMT inhibitors could be useful for treating children with HGPS.


Subject(s)
Cellular Senescence/drug effects , Progeria/drug therapy , Protein Methyltransferases/drug effects , Pyrans/pharmacology , Animals , Aorta/pathology , Cell Line , Cell Proliferation/drug effects , Disease Models, Animal , Humans , Lamin Type A/metabolism , Mice , Mice, Knockout , Myocytes, Smooth Muscle , Progeria/genetics , Progeria/pathology , Protein Methyltransferases/genetics , Protein Methyltransferases/metabolism
14.
Sci Rep ; 10(1): 17068, 2020 10 13.
Article in English | MEDLINE | ID: mdl-33051544

ABSTRACT

P21-activated kinase 4 (PAK4), a member of serine/threonine kinases family is over-expressed in numerous cancer tumors and is associated with oncogenic cell proliferation, migration and invasion. Our recent work demonstrated that the SET-domain containing protein 6 (SETD6) interacts with and methylates PAK4 at chromatin in mammalian cells, leading to activation of the Wnt/ß-catenin signaling pathway. In our current work, we identified lysine 473 (K473) on PAK4 as the primary methylation site by SETD6. Methylation of PAK4 at K473 activates ß-catenin transcriptional activity and inhibits cell adhesion. Specific methylation of PAK4 at K473 also attenuates paxillin localization to focal adhesions leading to overall reduction in adhesion-related features, such as filopodia and actin structures. The altered adhesion of the PAK4 wild-type cells is accompanied with a decrease in the migrative and invasive characteristics of the cells. Taken together, our results suggest that methylation of PAK4 at K473 plays a vital role in the regulation of cell adhesion and migration.


Subject(s)
Cell Adhesion/physiology , Protein Methyltransferases/metabolism , p21-Activated Kinases/metabolism , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Amino Acid Sequence , Animals , Binding Sites/genetics , Cell Adhesion/genetics , Cell Line , Cell Movement/genetics , Cell Movement/physiology , Conserved Sequence , Drosophila melanogaster , Focal Adhesions/genetics , Focal Adhesions/physiology , HEK293 Cells , Humans , MCF-7 Cells , Methylation , Mice , Paxillin/metabolism , Protein Methyltransferases/genetics , Pseudopodia/metabolism , Pseudopodia/ultrastructure , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Up-Regulation , Wnt Signaling Pathway/genetics , Zebrafish , beta Catenin/metabolism , p21-Activated Kinases/chemistry , p21-Activated Kinases/genetics
15.
J Mol Histol ; 51(5): 549-558, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32803470

ABSTRACT

Diabetic nephropathy (DN), a serious complication of hyperglycemia, is one of the most common causes of end-stage renal disease (ESRD). Glomerular podocyte injury is a major mechanism that leads to DN. However, the mechanisms underlying podocyte injury are ambiguous. In this study, we sought to investigate the contribution of SET domain-containing protein 6 (SETD6) to the pathogenesis of podocyte injury induced by glucose (GLU) and palmitic acid (PA), as well as the underlying mechanisms. Our results showed that GLU and PA treatment significantly decreased SETD6 expression in mouse podocytes. Besides, Cell Counting Kit-8 (CCK-8) and flow cytometry assay demonstrated that silencing of SETD6 silence obviously enhanced cell viability, and suppressed apoptosis in GLU and PA-induced podocytes. We also discovered that downregulation of SETD6 suppressed GLU and PA-induced ROS generation and podocyte mitochondrial dysfunction. Nrf2-Keap1 signaling pathway was involved in the effect of SETD6 on mitochondrial dysfunction. Taken together, silencing of SETD6 protected mouse podocyte against apoptosis and mitochondrial dysfunction through activating Nrf2-Keap1 signaling pathway. Therefore these data provide new insights into new potential therapeutic targets for DN treatment.


Subject(s)
Diabetic Nephropathies/etiology , Diabetic Nephropathies/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Palmitic Acid/pharmacology , Podocytes/drug effects , Podocytes/metabolism , Protein Methyltransferases/genetics , Animals , Apoptosis/drug effects , Biomarkers , Blood Glucose , Diabetic Nephropathies/pathology , Gene Expression Regulation , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Protein Methyltransferases/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
16.
Microbiology (Reading) ; 166(9): 837-848, 2020 09.
Article in English | MEDLINE | ID: mdl-32639227

ABSTRACT

Bacterial soft rot caused by the bacteria Dickeya and Pectobacterium is a destructive disease of vegetables, as well as ornamental plants. Several management options exist to help control these pathogens. Because of the limited success of these approaches, there is a need for the development of alternative methods to reduce losses. In this study, we evaluated the effect of potassium tetraborate tetrahydrate (PTB) on the growth of six Dickeya and Pectobacterium spp. Disc diffusion assays showed that Dickeya spp. and Pectobacterium spp. differ in their sensitivity to PTB. Spontaneous PTB-resistant mutants of Pectobacterium were identified and further investigation of the mechanism of PTB resistance was conducted by full genome sequencing. Point mutations in genes cpdB and supK were found in a single Pectobacterium atrosepticum PTB-resistant mutant. Additionally, point mutations in genes prfB (synonym supK) and prmC were found in two independent Pectobacterium brasiliense PTB-resistant mutants. prfB and prmC encode peptide chain release factor 2 and its methyltransferase, respectively. We propose the disruption of translation activity due to PTB leads to Pectobacterium growth inhibition. The P. atrosepticum PTB-resistant mutant showed altered swimming motility. Disease severity was reduced for P. atrosepticum-inoculated potato stems sprayed with PTB. We discuss the potential risk of selecting for bacterial resistance to this chemical.


Subject(s)
Anti-Bacterial Agents/pharmacology , Borates/pharmacology , Dickeya/drug effects , Pectobacterium/drug effects , Solanum tuberosum/microbiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Dickeya/genetics , Dickeya/growth & development , Dickeya/physiology , Disk Diffusion Antimicrobial Tests , Drug Resistance, Bacterial/genetics , Genes, Bacterial , Genetic Complementation Test , Movement , Pectobacterium/genetics , Pectobacterium/growth & development , Pectobacterium/physiology , Peptide Termination Factors/genetics , Peptide Termination Factors/metabolism , Plant Diseases/microbiology , Point Mutation , Protein Methyltransferases/genetics , Protein Methyltransferases/metabolism
17.
Oncogene ; 39(31): 5373-5389, 2020 07.
Article in English | MEDLINE | ID: mdl-32561852

ABSTRACT

Cancer stem cells possess the capacity for self-renewal and resistance to chemotherapy. It is therefore crucial to understand the molecular regulators of stemness in the quest to develop effective cancer therapies. TAZ is a transcription activator that promotes stem cell functions in post-development mammalian cells; suppression of TAZ activity reduces or eliminates cancer stemness in select cancers. Isoprenylcysteine carboxylmethyltransferase (ICMT) is the unique enzyme of the last step of posttranslational prenylation processing pathway that modifies several oncogenic proteins, including RAS. We found that suppression of ICMT results in reduced self-renewal/stemness in KRAS-driven pancreatic and breast cancer cells. Silencing of ICMT led to significant reduction of TAZ protein levels and loss of self-renewal ability, which could be reversed by overexpressing mutant KRAS, demonstrating the functional impact of ICMT modification on the ability of KRAS to control TAZ stability and function. Contrary to expectation, YAP protein levels appear to be much less susceptible than TAZ to the regulation by ICMT and KRAS, and YAP is less consequential in regulating stemness characteristics in these cells. Further, we found that the ICMT-dependent KRAS regulation of TAZ was mediated through RAF, but not PI3K, signaling. Functionally, we demonstrate that a signaling cascade from ICMT modification of KRAS to TAZ protein stability supports cancer cell self-renewal abilities in both in vitro and in vivo settings. In addition, studies using the proof-of-concept small molecule inhibitors of ICMT confirmed its role in regulating TAZ and self-renewal, demonstrating the potential utility of targeting ICMT to control aggressive KRAS-driven cancers.


Subject(s)
Breast Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms/metabolism , Protein Methyltransferases/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Trans-Activators/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Self Renewal/physiology , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Doxorubicin/pharmacology , Female , HEK293 Cells , Heterografts , Humans , MAP Kinase Signaling System , Mice , Mice, Inbred NOD , Mice, SCID , Mutation , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Protein Methyltransferases/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Trans-Activators/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Gemcitabine
18.
Aging (Albany NY) ; 12(12): 11500-11516, 2020 06 17.
Article in English | MEDLINE | ID: mdl-32554858

ABSTRACT

Metastatic disease caused by castration-resistant prostate cancer (CRPC) is the principal cause of prostate cancer (PCa)-related mortality. CRPC occurs within 2-3 years of initiation of androgen deprivation therapy (ADT), which is an important factor of influencing PCa metastasis. Recent studies have revealed that non-coding RNAs in PCa can enhance metastasis and progression, while the mechanisms are still unclear. In this study, we reported that the long noncoding RNA-LINC00963 was increased in CRPC tissues and promoted migration of PCa cells in vitro and their metastasis in vivo. High levels of LINC00963 significantly decreased tumor suppressor miR-542-3p, whose levels in metastasis tissues were low compared to those in non-metastasis tissues. LINC00963 promotes and miR-542-3p inhibits metastasis. Furthermore, the expression levels of LINC00963 and miR-542-3p were positively and negatively associated with the expression of NOP2. We demonstrated that NOP2 promoted PCa by activating the epithelial-mesenchymal transition (EMT) pathway. For specific mechanism, dual luciferase reporter assays showed that miR-542-3p directly binds to both 3'-untranslated region (UTR) of LINC00963 and NOP2 mRNA. Taken together, our results show that LINC00963 acts as an inducer of PCa metastasis by binding miR-542-3p, thereby promoting NOP2. This axis may have diagnostic and therapeutic potential for advanced PCa.


Subject(s)
MicroRNAs/metabolism , Nuclear Proteins/genetics , Prostatic Neoplasms, Castration-Resistant/genetics , RNA, Long Noncoding/metabolism , tRNA Methyltransferases/genetics , Animals , Cell Line, Tumor , Disease Models, Animal , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Male , Mice , Mice, Transgenic , MicroRNAs/agonists , MicroRNAs/antagonists & inhibitors , Neoplasm Metastasis/genetics , Prostate/pathology , Prostatic Neoplasms, Castration-Resistant/pathology , Protein Methyltransferases/genetics , RNA, Long Noncoding/genetics , RNA-Seq , Xenograft Model Antitumor Assays
19.
Curr Protein Pept Sci ; 21(7): 675-689, 2020.
Article in English | MEDLINE | ID: mdl-32188384

ABSTRACT

Protein histidine methylation is a rarely studied posttranslational modification in eukaryotes. Although the presence of N-methylhistidine was demonstrated in actin in the early 1960s, so far, only a limited number of proteins containing N-methylhistidine have been reported, including S100A9, myosin, skeletal muscle myosin light chain kinase (MLCK 2), and ribosomal protein Rpl3. Furthermore, the role of histidine methylation in the functioning of the protein and in cell physiology remains unclear due to a shortage of studies focusing on this topic. However, the molecular identification of the first two distinct histidine-specific protein methyltransferases has been established in yeast (Hpm1) and in metazoan species (actin-histidine N-methyltransferase), giving new insights into the phenomenon of protein methylation at histidine sites. As a result, we are now beginning to recognize protein histidine methylation as an important regulatory mechanism of protein functioning whose loss may have deleterious consequences in both cells and in organisms. In this review, we aim to summarize the recent advances in the understanding of the chemical, enzymological, and physiological aspects of protein histidine methylation.


Subject(s)
Actins/metabolism , Histidine/metabolism , Myosin-Light-Chain Kinase/metabolism , Protein Methyltransferases/metabolism , Protein Processing, Post-Translational , Ribosomal Proteins/metabolism , Actins/genetics , Animals , Calgranulin B/genetics , Calgranulin B/metabolism , Histone Methyltransferases/genetics , Histone Methyltransferases/metabolism , Humans , Methylation , Methylhistidines/metabolism , Methyltransferases/genetics , Methyltransferases/metabolism , Myosin-Light-Chain Kinase/genetics , Protein Methyltransferases/genetics , Ribosomal Protein L3 , Ribosomal Proteins/genetics , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction
20.
Proc Natl Acad Sci U S A ; 117(3): 1485-1495, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31911473

ABSTRACT

Many large proteins suffer from slow or inefficient folding in vitro. It has long been known that this problem can be alleviated in vivo if proteins start folding cotranslationally. However, the molecular mechanisms underlying this improvement have not been well established. To address this question, we use an all-atom simulation-based algorithm to compute the folding properties of various large protein domains as a function of nascent chain length. We find that for certain proteins, there exists a narrow window of lengths that confers both thermodynamic stability and fast folding kinetics. Beyond these lengths, folding is drastically slowed by nonnative interactions involving C-terminal residues. Thus, cotranslational folding is predicted to be beneficial because it allows proteins to take advantage of this optimal window of lengths and thus avoid kinetic traps. Interestingly, many of these proteins' sequences contain conserved rare codons that may slow down synthesis at this optimal window, suggesting that synthesis rates may be evolutionarily tuned to optimize folding. Using kinetic modeling, we show that under certain conditions, such a slowdown indeed improves cotranslational folding efficiency by giving these nascent chains more time to fold. In contrast, other proteins are predicted not to benefit from cotranslational folding due to a lack of significant nonnative interactions, and indeed these proteins' sequences lack conserved C-terminal rare codons. Together, these results shed light on the factors that promote proper protein folding in the cell and how biomolecular self-assembly may be optimized evolutionarily.


Subject(s)
Escherichia coli Proteins/chemistry , Intrinsically Disordered Proteins/chemistry , Protein Folding , Alcohol Oxidoreductases/chemistry , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/metabolism , Kinetics , Molecular Dynamics Simulation , Phosphotransferases/chemistry , Phosphotransferases/genetics , Phosphotransferases/metabolism , Protein Biosynthesis , Protein Methyltransferases/chemistry , Protein Methyltransferases/genetics , Protein Methyltransferases/metabolism , Repressor Proteins/chemistry , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tetrahydrofolate Dehydrogenase/chemistry , Tetrahydrofolate Dehydrogenase/genetics , Tetrahydrofolate Dehydrogenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...