Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cell Signal ; 119: 111156, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38574938

ABSTRACT

In the seemingly well-researched field of vascular research, there are still many underestimated factors and molecular mechanisms. In recent years, SUMOylation has become increasingly important. SUMOylation is a post-translational modification in which small ubiquitin-related modifiers (SUMO) are covalently attached to target proteins. Sites where these SUMO modification processes take place in the cell nucleus are PML nuclear bodies (PML-NBs) - multiprotein complexes with their essential main component and organizer, the PML protein. PML and SUMO, either alone or as partners, influence a variety of cellular processes, including regulation of transcription, senescence, DNA damage response and defence against microorganisms, and are involved in innate immunity and inflammatory responses. They also play an important role in maintaining homeostasis in the vascular system and in pathological processes leading to the development and progression of cardiovascular diseases. This review summarizes information about the function of SUMO(ylation) and PML(-NBs) in the human vasculature from angiogenesis to disease and highlights their clinical potential as drug targets.


Subject(s)
Nuclear Proteins , Promyelocytic Leukemia Protein , Sumoylation , Transcription Factors , Humans , Promyelocytic Leukemia Protein/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Animals , Tumor Suppressor Proteins/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology
2.
Exp Cell Res ; 434(2): 113863, 2024 01 15.
Article in English | MEDLINE | ID: mdl-38097153

ABSTRACT

Rhabdomyosarcoma (RMS), a tumor that consists of poorly differentiated skeletal muscle cells, is the most common soft-tissue sarcoma in children. Despite considerable progress within the last decades, therapeutic options are still limited, warranting the need for novel approaches. Recent data suggest deregulation of the Smyd1 protein, a sumoylation target as well as H3K4me2/3 methyltransferase and transcriptional regulator in myogenesis, and its binding partner skNAC, in RMS cells. Here, we show that despite the fact that most RMS cells express at least low levels of Smyd1 and skNAC, failure to upregulate expression of these genes in reaction to differentiation-promoting signals can always be observed. While overexpression of the Smyd1 gene enhances many aspects of RMS cell differentiation and inhibits proliferation rate and metastatic potential of these cells, functional integrity of the putative Smyd1 sumoylation motif and its SET domain, the latter being crucial for HMT activity, appear to be prerequisites for most of these effects. Based on these findings, we explored the potential for novel RMS therapeutic strategies, employing small-molecule compounds to enhance Smyd1 activity. In particular, we tested manipulation of (a) Smyd1 sumoylation, (b) stability of H3K4me2/3 marks, and (c) calpain activity, with calpains being important targets of Smyd1 in myogenesis. We found that specifically the last strategy might represent a promising approach, given that suitable small-molecule compounds will be available for clinical use in the future.


Subject(s)
Rhabdomyosarcoma , Transcription Factors , Child , Humans , Transcription Factors/metabolism , DNA-Binding Proteins/metabolism , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/therapy , Rhabdomyosarcoma/pathology , Muscle Fibers, Skeletal/metabolism , Cell Differentiation/genetics , Cell Line, Tumor
3.
Int J Mol Sci ; 24(8)2023 Apr 15.
Article in English | MEDLINE | ID: mdl-37108469

ABSTRACT

In addition to its function as an intravascular lipid transporter, LDL also triggers signal transduction in endothelial cells (ECs), which, among other things, trigger immunomodulatory cascades, e.g., IL-6 upregulation. However, the molecular mechanisms of how these LDL-triggered immunological responses in ECs are realized are not fully understood. Since promyelocytic leukemia protein (PML) plays a role in promoting inflammatory processes, we examined the relationship between LDL, PML, and IL-6 in human ECs (HUVECs and EA.hy926 cells). RT-qPCR, immunoblotting, and immunofluorescence analyses showed that LDL but not HDL induced higher PML expression and higher numbers of PML-nuclear bodies (PML-NBs). Transfection of the ECs with a PML gene-encoding vector or PML-specific siRNAs demonstrated PML-regulated IL-6 and IL-8 expression and secretion after LDL exposure. Moreover, incubation with the PKC inhibitor sc-3088 or the PKC activator PMA showed that LDL-induced PKC activity leads to the upregulation of PML mRNA and PML protein. In summary, our experimental data suggest that high LDL concentrations trigger PKC activity in ECs to upregulate PML expression, which then increases production and secretion of IL-6 and IL-8. This molecular cascade represents a novel cellular signaling pathway with immunomodulatory effects in ECs in response to LDL exposure.


Subject(s)
Endothelial Cells , Interleukin-6 , Humans , Endothelial Cells/metabolism , Interleukin-6/genetics , Interleukin-8 , Nuclear Proteins/genetics , Promyelocytic Leukemia Protein/genetics , Promyelocytic Leukemia Protein/metabolism , Transcription Factors/metabolism , Lipoproteins, LDL/metabolism , Protein Kinase C/metabolism
4.
Cells ; 10(12)2021 12 13.
Article in English | MEDLINE | ID: mdl-34944023

ABSTRACT

The lysine methyltransferase Smyd1 with its characteristic catalytic SET-domain is highly enriched in the embryonic heart and skeletal muscles, participating in cardiomyogenesis, sarcomere assembly and chromatin remodeling. Recently, significant Smyd1 levels were discovered in endothelial cells (ECs) that responded to inflammatory cytokines. Based on these biochemical properties, we hypothesized that Smyd1 is involved in inflammation-triggered signaling in ECs and therefore, investigated its role within the LPS-induced signaling cascade. Human endothelial cells (HUVECs and EA.hy926 cells) responded to LPS stimulation with higher intrinsic Smyd1 expression. By transfection with expression vectors containing gene inserts encoding either intact Smyd1, a catalytically inactive Smyd1-mutant or Smyd1-specific siRNAs, we show that Smyd1 contributes to LPS-triggered expression and secretion of IL-6 in EA.hy926 cells. Further molecular analysis revealed this process to be based on two signaling pathways: Smyd1 increased the activity of NF-κB and promoted the trimethylation of lysine-4 of histone-3 (H3K4me3) within the IL-6 promoter, as shown by ChIP-RT-qPCR combined with IL-6-promoter-driven luciferase reporter gene assays. In summary, our experimental analysis revealed that LPS-binding to ECs leads to the up-regulation of Smyd1 expression to transduce the signal for IL-6 up-regulation via activation of the established NF-κB pathway as well as via epigenetic trimethylation of H3K4.


Subject(s)
DNA Methylation/genetics , DNA-Binding Proteins/genetics , Endothelial Cells/metabolism , Interleukin-6/genetics , Muscle Proteins/genetics , Transcription Factors/genetics , DNA Methylation/drug effects , DNA-Binding Proteins/antagonists & inhibitors , Endothelial Cells/drug effects , Epigenesis, Genetic , Gene Expression Regulation, Developmental/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Inflammation/chemically induced , Lipopolysaccharides/pharmacology , Muscle Proteins/antagonists & inhibitors , NF-kappa B/genetics , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Transcription Factors/antagonists & inhibitors
5.
Clin Sci (Lond) ; 135(7): 887-905, 2021 04 16.
Article in English | MEDLINE | ID: mdl-33764440

ABSTRACT

Promyelocytic leukemia protein (PML) is a constitutive component of PML nuclear bodies (PML-NBs), which function as stress-regulated SUMOylation factories. Since PML can also act as a regulator of the inflammatory and fibroproliferative responses characteristic of atherosclerosis, we investigated whether PML is implicated in this disease. Immunoblotting, ELISA and immunohistochemistry showed a stronger expression of PML in segments of human atherosclerotic coronary arteries and sections compared with non-atherosclerotic ones. In particular, PML was concentrated in PML-NBs from α-smooth muscle actin (α-SMA)-immunoreactive cells in plaque areas. To identify possible functional consequences of PML-accumulation in this cell type, differentiated human coronary artery smooth muscle cells (dHCASMCs) were transfected with a vector containing the intact PML-gene. These PML-transfected dHCASMCs showed higher levels of small ubiquitin-like modifier (SUMO)-1-dependent SUMOylated proteins, but lower levels of markers for smooth muscle cell (SMC) differentiation and revealed more proliferation and migration activities than dHCASMCs transfected with the vector lacking a specific gene insert or with the vector containing a mutated PML-gene coding for a PML-form without SUMOylation activity. When dHCASMCs were incubated with different cytokines, higher PML-levels were observed only after interferon γ (IFN-γ) stimulation, while the expression of differentiation markers was lower. However, these phenotypic changes were not observed in dHCASMCs treated with small interfering RNA (siRNA) suppressing PML-expression prior to IFN-γ stimulation. Taken together, our results imply that PML is a previously unknown functional factor in the molecular cascades associated with the pathogenesis of atherosclerosis and is positioned in vascular SMCs (VSMCs) between upstream IFN-γ activation and downstream SUMOylation.


Subject(s)
Myocytes, Smooth Muscle/pathology , Plaque, Atherosclerotic/metabolism , Promyelocytic Leukemia Protein/genetics , Promyelocytic Leukemia Protein/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Coronary Vessels/metabolism , Female , Humans , Interferon-gamma , Male , Peptide Fragments , Phenotype , Plaque, Atherosclerotic/pathology , Sumoylation
6.
Biochem J ; 478(1): 217-234, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33241844

ABSTRACT

Smyd1 is an epigenetic modulator of gene expression that has been well-characterized in muscle cells. It was recently reported that Smyd1 levels are modulated by inflammatory processes. Since inflammation affects the vascular endothelium, this study aimed to characterize Smyd1 expression in endothelial cells. We detected Smyd1 in human endothelial cells (HUVEC and EA.hy926 cells), where the protein was largely localized in PML nuclear bodies (PML-NBs). By transfection of EA.hy926 cells with expression vectors encoding Smyd1, PML, SUMO1, active or mutant forms of the SUMO protease SuPr1 and/or the SUMO-conjugation enzyme UBC9, as well as Smyd1- or PML-specific siRNAs, in the presence or absence of the translation blocker cycloheximide or the proteasome-inhibitor MG132, and supported by computational modeling, we show that Smyd1 is SUMOylated in a PML-dependent manner and thereby addressed for degradation in proteasomes. Furthermore, transfection with Smyd1-encoding vectors led to PML up-regulation at the mRNA level, while PML transfection lowered Smyd1 protein stability. Incubation of EA.hy926 cells with the pro-inflammatory cytokine TNF-α resulted in a constant increase in Smyd1 mRNA and protein over 24 h, while incubation with IFN-γ induced a transient increase in Smyd1 expression, which peaked at 6 h and decreased to control values within 24 h. The IFN-γ-induced increase in Smyd1 was accompanied by more Smyd1 SUMOylation and more/larger PML-NBs. In conclusion, our data indicate that in endothelial cells, Smyd1 levels are regulated through a negative feedback mechanism based on SUMOylation and PML availability. This molecular control loop is stimulated by various cytokines.


Subject(s)
Cytokines/pharmacology , DNA-Binding Proteins/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Muscle Proteins/metabolism , Promyelocytic Leukemia Protein/metabolism , Sumoylation/drug effects , Transcription Factors/metabolism , Cell Nucleus/metabolism , Cycloheximide/pharmacology , DNA-Binding Proteins/genetics , Gene Expression , Human Umbilical Vein Endothelial Cells , Humans , Interferon-gamma/pharmacology , Leupeptins/pharmacology , Muscle Proteins/genetics , Promyelocytic Leukemia Protein/genetics , Proteasome Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/genetics , RNA, Small Interfering , SUMO-1 Protein/genetics , SUMO-1 Protein/metabolism , Sumoylation/genetics , Transcription Factors/genetics , Transfection , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation
7.
Front Physiol ; 11: 28, 2020.
Article in English | MEDLINE | ID: mdl-32116748

ABSTRACT

It is unclear how microangiopathic changes in skeletal muscle vary among systemic vascular pathologies. We therefore analyzed the capillary fine structure in skeletal muscle from patients with arterial hypertension (HYPT), diabetes mellitus type 2 (T2DM) or intermittent claudication - peripheral arterial disease (IC/PAD). Tablet-based image analysis (TBIA) was carried out to largely re-evaluate 5,000 transmission electron micrographs of capillaries from 126 vastus lateralis biopsies of 75 individuals (HYPT, T2DM or IC/PAD patients as well as healthy individuals before and after endurance exercise training) used in previous morphometric studies, but assessed using stereological counting grids of different sizes. Serial block-face scanning electron microscopy (SBFSEM) of mouse skeletal muscle was used for validation of the particular fine structural events observed in human biopsies. The peri-capillary basement membrane (BM) was 38.5 and 45.5% thicker (P < 0.05) in T2DM and IC/PAD patients than in the other groups. A 17.7-39.6% lower (P < 0.05) index for intraluminal endothelial cell (EC) surface enlargement by projections was exclusively found in T2DM patients by TBIA morphometry. The proportion of capillaries with disrupted BM between pericytes (PC) and EC was higher (P < 0.05) in HYPT (33.2%) and T2DM (38.7%) patients than in the control group. Empty EC-sockets were more frequent (P < 0.05) in the three patient groups (20.6% in HYPT, 27.1% in T2DM, 30.0% in IC/PAD) than in the healthy individuals. SBFSEM confirmed that EC-sockets may exhibit close proximity to the capillary lumen. Our comparative morphometric analysis demonstrated that structural arrangement of skeletal muscle capillaries is more affected in T2DM than in HYPT or IC/PAD, although some similar elements of remodeling were found. The increased frequency of empty EC-sockets in the three patient groups indicates that the PC-EC interaction is commonly disturbed in these systemic vascular pathologies.

8.
J Inflamm (Lond) ; 15: 20, 2018.
Article in English | MEDLINE | ID: mdl-30386182

ABSTRACT

BACKGROUND: Zinc finger protein 580 (ZNF580) was reported to modulate angiogenesis, endothelial homeostasis and blood pressure control. ZNF580 regulated genes include VEGF-A and IL-8. However, it is unknown if ZNF580 could play a role during inflammation. The aim of this study was to find out if ZNF580 affects the expression of IL-6, if it occurs in monocytic cells and responds to inflammatory mediators. RESULTS: Overexpression of ZNF580 reduced LPS-induced promotor activity of IL-6. Consistently, overexpression of ZNF580 reduced by half the LPS-induced expression of IL-6. ZNF580 was strongly expressed in the nucleus of MonoMac6, a human monocytic cell line. LPS-stimulated IL-6 secretion increased when ZNF580 was suppressed with siRNA. After stimulation of MonoMac6 with LPS for 24 h, ZNF580 negatively correlated with the amount of secreted IL-6. In response to LPS, ZNF580 was increased within the first 8 h, followed by a marked decrease after 16 h. This decrease coincided with sustained IL-6 production. CONCLUSION: This study demonstrated that ZNF580 inhibits LPS-induced expression of IL-6. ZNF580 was highly expressed in monocytic cells and therefore may contribute to the modulation of its IL-6 production, at least in response to LPS. This suggests cooperation between ZNF580 and NFκB, which could play a role during sepsis.

9.
Int J Mol Med ; 42(1): 534-546, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29693115

ABSTRACT

Photodynamic therapy (PDT) has emerged as an effective and minimally invasive cancer treatment modality. In the present study, two novel phthalocyanines, tetra­triethyleneoxysulfonyl substituted zinc phthalocyanine (ZnPc) and dihydroxy­2,9(10),16(17),23(24)­tetrakis(4,7,10­trioxaundecan­1­sulfonyl) silicon phthalocyanine (Pc32), were investigated as photosensitizers (PS) for PDT of cholangiocarcinoma (CC). ZnPc showed a pronounced dose­dependent and predominantly cytoplasmic accumulation in EGI­1 and TFK­1 CC cell lines. Pc32 also accumulated in the CC cells, but this was less pronounced. Without photoactivation, the PS did not exhibit any antiproliferative or cytotoxic effects. Upon photoactivation, ZnPc induced the formation of reactive oxygen species (ROS) and immediate phototoxicity, leading to a dose­dependent decrease in cell proliferation, and an induction of mitochondria­driven apoptosis and cell cycle arrest of EGI­1 and TFK­1 cells. Although photoactivated Pc32 also induced ROS formation in the two cell lines, the extent was less marked, compared with that induced by ZnPc­PDT, and pronounced antipoliferative effects occurred only in the less differentiated EGI­1 cells, whereas the more differentiated TFK­1 cells did not show sustained growth inhibition upon Pc32­PDT induction. In vivo examinations on the antiangiogenic potency of the novel PS were performed using chorioallantoic membrane (CAM) assays, which revealed reduced angiogenic sprouting with a concomitant increase in nonperfused regions and degeneration of the vascular network of the CAM following induction with ZnPc­PDT only. The study demonstrated the pronounced antiproliferative and antiangiogenic potency of ZnPc as a novel PS for PDT, meriting further elucidation as a promising PS for the photodynamic treatment of CC.


Subject(s)
Cholangiocarcinoma/drug therapy , Indoles/therapeutic use , Photochemotherapy , Photosensitizing Agents/therapeutic use , Silicon/therapeutic use , Zinc/therapeutic use , Angiogenesis Inhibitors/pharmacology , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chickens , Cholangiocarcinoma/pathology , Chorioallantoic Membrane/drug effects , Chorioallantoic Membrane/metabolism , Humans , Indoles/pharmacology , Isoindoles , Photosensitizing Agents/pharmacology , Reactive Oxygen Species/metabolism , Silicon/pharmacology , Zinc/pharmacology
10.
Micron ; 108: 6-10, 2018 05.
Article in English | MEDLINE | ID: mdl-29499397

ABSTRACT

skNAC (skeletal and heart muscle-specific variant of nascent polypeptide-associated complex) and Smyd1 (SET and MYND domain-containing 1) form a protein dimer which is specific for striated muscle cells. Its function is largely unknown. On the one hand, skNAC-Smyd1 appears to control transcriptional processes in the nucleus, on the other hand, specifically at later stages of myogenic differentiation, both proteins translocate to the sarcoplasm and at least Smyd1 specifically associates with sarcomeric structures and might control myofibrillogenesis and/or sarcomere architecture. Here, using immunofluorescence and electron microscopy, we analyzed sarcomere formation and myofibril organization after siRNA-mediated knockdown of skNAC or Smyd1 expression in murine C2C12 skeletal muscle cells. We found that inhibition of skNAC or Smyd1 expression indeed prevents myofibrillogenesis and sarcomere formation, leading to a disorganized array of myofilaments predominantly within the region immediately beneath the plasma membrane.


Subject(s)
DNA-Binding Proteins/biosynthesis , Molecular Chaperones/biosynthesis , Muscle Development/genetics , Muscle Proteins/biosynthesis , Myofibrils/metabolism , Sarcomeres/metabolism , Transcription Factors/biosynthesis , Animals , Cell Line , DNA-Binding Proteins/genetics , Fluorescent Antibody Technique , Mice , Microscopy, Electron , Molecular Chaperones/genetics , Muscle Proteins/genetics , Muscle, Striated/cytology , Myofibrils/genetics , RNA Interference , RNA, Small Interfering/genetics , Sarcomeres/genetics , Transcription Factors/genetics
11.
Eur J Pharmacol ; 810: 1-8, 2017 Sep 05.
Article in English | MEDLINE | ID: mdl-28606850

ABSTRACT

Human podocytes (hPC) are essential for maintaining normal kidney function and dysfunction or loss of hPC play a pivotal role in the manifestation and progression of chronic kidney diseases including diabetic nephropathy. Previously, α-Lipoic acid (α-LA), a licensed drug for treatment of diabetic neuropathy, was shown to exhibit protective effects on diabetic nephropathy in vivo. However, the effect of α-LA on hPC under non-diabetic conditions is unknown. Therefore, we analyzed the impact of α-LA on cell viability and expression of nephrin and zinc finger protein 580 (ZNF580) in normal hPC in vitro. Protein analyses were done via Western blot techniques. Cell viability was determined using a functional assay. hPC viability was dynamically modulated via α-LA stimulation in a concentration-dependent manner. This was associated with reduced nephrin and ZNF580 expression and increased nephrin phosphorylation in normal hPC. Moreover, α-LA reduced nephrin and ZNF580 protein expression via 'kappa-light-chain-enhancer' of activated B-cells (NF-κB) inhibition. These data demonstrate that low α-LA had no negative influence on hPC viability, whereas, high α-LA concentrations induced cytotoxic effects on normal hPC and reduced nephrin and ZNF580 expression via NF-κB inhibition. These data provide first novel information about potential cytotoxic effects of α-LA on hPC under non-diabetic conditions.


Subject(s)
Cell Survival/drug effects , Gene Expression Regulation/drug effects , Membrane Proteins/metabolism , Podocytes/drug effects , Podocytes/metabolism , Thioctic Acid/pharmacology , Transcription Factors/metabolism , Dose-Response Relationship, Drug , Humans , Membrane Proteins/genetics , Podocytes/cytology , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factor RelA/metabolism , Transcription Factors/genetics
12.
Int J Oncol ; 50(3): 953-963, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28098886

ABSTRACT

Photodynamic therapy (PDT) has gathered much attention in the field of cancer treatment and is increasingly used as an alternative solution for esophageal cancer therapy. However, there is a constant need for improving the effectiveness and tolerability of the applied photosensitizers (PS). Here, we propose tetra-triethyleneoxysulfonyl substituted zinc phthalocyanine (ZnPc) as a promising PS for photodynamic treatment of esophageal cancer. ZnPc-induced phototoxicity was studied in two human esophageal cancer cell lines: OE-33 (adenocarcinoma) and Kyse-140 (squamous cell carcinoma). In vitro studies focused on the uptake and intracellular distribution of the novel ZnPc as well as on its growth inhibitory potential, reactive oxygen species (ROS) formation and the induction of apoptosis. The chicken chorioallantoic membrane assay (CAM assay) and studies on native Wistar rats were employed to determine the antineoplastic and antiangiogenic activity of ZnPc-PDT as well as the tolerability and safety of non-photoactivated ZnPc in vivo. ZnPc was taken up by cancer cells in a dose- and time-dependent manner and showed a homogeneous cytoplasmic distribution. Photoactivation of ZnPc-loaded (1-10 µM) cells led to a dose-dependent growth inhibition of esophageal adenocarcinoma and squamous cell carcinoma cells of >90%. The antiproliferative effect was based on ROS-induced cytotoxicity and the induction of mitochondria-driven apoptosis. In vivo studies on esophageal tumor plaques grown on the CAM revealed pronounced antiangiogenic and antineoplastic effects. ZnPc-PDT caused long-lasting changes in the vascular architecture and a marked reduction of tumor feeding blood vessels. Animal studies confirmed the good tolerability and systemic safety of ZnPc, as no changes in immunological, behavioral and organic parameters could be detected upon treatment with the non-photoactivated ZnPc. Our findings show the extraordinary photoactive potential of the novel ZnPc as a photosensitizer for PDT of esophageal cancer.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Esophageal Neoplasms/therapy , Indoles/pharmacology , Organometallic Compounds/pharmacology , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Angiogenesis Inhibitors/adverse effects , Angiogenesis Inhibitors/pharmacokinetics , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chick Embryo , Chorioallantoic Membrane/blood supply , Chorioallantoic Membrane/drug effects , Humans , Indoles/adverse effects , Indoles/pharmacokinetics , Isoindoles , Organometallic Compounds/adverse effects , Organometallic Compounds/pharmacokinetics , Photosensitizing Agents/adverse effects , Photosensitizing Agents/pharmacokinetics , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Zinc Compounds
13.
Exp Cell Res ; 336(2): 182-91, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26162853

ABSTRACT

Skeletal and heart muscle-specific variant of the alpha subunit of nascent polypeptide associated complex (skNAC) is exclusively found in striated muscle cells. Its function, however, is largely unknown. Previous reports could demonstrate that skNAC binds to Smyd1 (SET and MYND domain containing protein 1). The facts that (a) SET domains have histone methyltransferase activity, and (b) MYND domains are known recruiters of histone deacetylases (HDACs), implicate the skNAC-Smyd1 complex in transcriptional control. To study potential target genes, we carried out cDNA microarray analysis on differentiating C2C12 myoblasts in which expression of the skNAC gene had been knocked down. We found and confirmed a series of targets, specifically genes encoding regulators of inflammation, cellular metabolism, and cell migration. Mechanistically, as shown by Western blot, ELISA, and ChIP analysis at selected promoter regions, transcriptional control by skNAC-Smyd1 appears to be exerted at least in part by affecting a series of histone modifications, specifically H3K4 di- and trimethylation and potentially also histone acetylation. Taken together, our data suggest that the skNAC-Smyd1 complex is involved in transcriptional regulation both via the control of histone methylation and histone (de)acetylation.


Subject(s)
DNA-Binding Proteins/genetics , Histones/metabolism , Molecular Chaperones/genetics , Muscle Proteins/genetics , Transcription Factors/genetics , Transcription, Genetic/genetics , Acetylation , Animals , Cell Differentiation , Cell Line , Cell Movement/genetics , Energy Metabolism/genetics , Gene Expression Regulation , Histone Deacetylases/metabolism , Inflammation/genetics , Methylation , Mice , Muscle, Skeletal/metabolism , Myoblasts, Cardiac/cytology , Myoblasts, Skeletal/cytology , Myocardium/metabolism , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics , RNA Interference , RNA, Small Interfering , Succinate Dehydrogenase/metabolism , ras Guanine Nucleotide Exchange Factors/biosynthesis
14.
Clin Exp Metastasis ; 31(8): 869-79, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25209525

ABSTRACT

Skeletal and heart muscle-specific variant of the alpha subunit of nascent polypeptide complex (skNAC) is exclusively present in striated muscle cells. During skeletal muscle cell differentiation, skNAC expression is strongly induced, suggesting that the protein might be a regulator of the differentiation process. Rhabdomyosarcoma is a tumor of skeletal muscle origin. Since there is a strong inverse correlation between rhabdomyosarcoma cell differentiation status and metastatic potential, we analyzed skNAC expression patterns in a set of rhabdomyosarcoma cell lines: Whereas RD/12 and RD/18 cells showed a marked induction of skNAC gene expression upon the induction of differentiation-similarly as the one seen in nontransformed myoblasts-skNAC was not induced in CCA or Rh30 cells. Overexpressing skNAC in CCA and Rh30 cells led to a reduction in cell cycle progression and cell proliferation accompanied by an upregulation of specific myogenic differentiation markers, such as Myogenin or Myosin Heavy Chain. Furthermore, in contrast to vector-transfected controls, a high percentage of the cells formed long, Myosin Heavy Chain-positive, multinucleate myotubes. Consistently, soft agar assays revealed a drop in the metastatic potential of skNAC-overexpressing cells. Taken together, these data indicate that reconstitution of skNAC expression can enhance the differentiation potential of rhabdomyosarcoma cells and reduces their metastatic potential, a finding which might have important therapeutic implications.


Subject(s)
Apoptosis , Cell Differentiation , Cell Proliferation , Molecular Chaperones/genetics , Myoblasts/pathology , Rhabdomyosarcoma/genetics , Blotting, Northern , Blotting, Western , Fluorescent Antibody Technique , Gene Expression , Humans , Immunoenzyme Techniques , Myoblasts/metabolism , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Rhabdomyosarcoma/pathology
15.
J Cell Sci ; 127(Pt 17): 3794-804, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25002400

ABSTRACT

Skeletal and heart muscle-specific variant of the α subunit of nascent polypeptide associated complex (skNAC; encoded by NACA) is exclusively found in striated muscle cells. Its function, however, is largely unknown. Previous reports have demonstrated that skNAC binds to m-Bop/Smyd1, a multi-functional protein that regulates myogenesis both through the control of transcription and the modulation of sarcomerogenesis, and that both proteins undergo nuclear-to-cytoplasmic translocation at the later stages of myogenic differentiation. Here, we show that skNAC binds to the E3 SUMO ligase mammalian Mms21/Nse2 and that knockdown of Nse2 expression inhibits specific aspects of myogenic differentiation, accompanied by a partial blockade of the nuclear-to-cytoplasmic translocation of the skNAC-Smyd1 complex, retention of the complex in promyelocytic leukemia (PML)-like nuclear bodies and disturbed sarcomerogenesis. In addition, we show that the skNAC interaction partner Smyd1 contains a putative sumoylation motif and is sumoylated in muscle cells, with depletion of Mms21/Nse2 leading to reduced concentrations of sumoylated Smyd1. Taken together, our data suggest that the function, specifically the balance between the nuclear and cytosolic roles, of the skNAC-Smyd1 complex might be regulated by sumoylation.


Subject(s)
DNA-Binding Proteins/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Morphogenesis/genetics , Muscle Development/genetics , Muscle Proteins/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Differentiation/physiology , Cell Line , Mice , Muscle, Skeletal/metabolism , Myocardium/metabolism , Sumoylation/genetics
16.
Biochem J ; 453(2): 303-10, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23662692

ABSTRACT

skNAC (skeletal and heart muscle specific variant of nascent polypeptide-associated complex α) is a skeletal and heart muscle-specific protein known to be involved in the regulation of sarcomerogenesis. The respective mechanism, however, is largely unknown. In the present paper, we demonstrate that skNAC regulates calpain activity. Specifically, we show that inhibition of skNAC gene expression leads to enhanced, and overexpression of the skNAC gene to repressed, activity of calpain 1 and, to a lesser extent, calpain 3 in myoblasts. In skNAC siRNA-treated cells, enhanced calpain activity is associated with increased migration rates, as well as with perturbed sarcomere architecture. Treatment of skNAC-knockdown cells with the calpain inhibitor ALLN (N-acetyl-leucyl-leucyl-norleucinal) reverts both the positive effect on myoblast migration and the negative effect on sarcomere architecture. Taken together, our data suggest that skNAC controls myoblast migration and sarcomere architecture in a calpain-dependent manner.


Subject(s)
Calpain/metabolism , Molecular Chaperones/genetics , Muscle Proteins/metabolism , Myoblasts/metabolism , Sarcomeres , Animals , Base Sequence , Cell Line , DNA Primers , Mice , Polymerase Chain Reaction
17.
Mol Microbiol ; 84(1): 51-65, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22356413

ABSTRACT

RprA is a small regulatory RNA known to weakly affect the translation of σ(S) (RpoS) in Escherichia coli. Here we demonstrate that csgD, which encodes a stationary phase-induced biofilm regulator, as well as ydaM, which encodes a diguanylate cyclase involved in activating csgD transcription, are novel negatively controlled RprA targets. As shown by extensive mutational analysis, direct binding of RprA to the 5'-untranslated and translational initiation regions of csgD mRNA inhibits translation and reduces csgD mRNA levels. In the case of ydaM mRNA, RprA base-pairs directly downstream of the translational start codon. In a feedforward loop, RprA can thus downregulate > 30 YdaM/CsgD-activated genes including those for adhesive curli fimbriae. However, during early stationary phase, when csgD transcription is strongly activated, the synthesis of csgD mRNA exceeds that of RprA, which allows the accumulation of CsgD protein. This situation is reversed when csgD transcription is shut off - for instance, later in stationary phase or during biofilm formation - or by conditions that further activate RprA expression via the Rcs two-component system. Thus, antagonistic regulation of csgD and RprA at the mRNA level integrates cell envelope stress signals with global gene expression during stationary phase and biofilm formation.


Subject(s)
Biofilms/growth & development , Cell Wall/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/physiology , RNA, Bacterial/metabolism , Trans-Activators/metabolism , Bacterial Proteins/metabolism , Binding Sites , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial , Phosphorus-Oxygen Lyases/genetics , Phosphorus-Oxygen Lyases/metabolism , Protein Biosynthesis , RNA, Bacterial/genetics , RNA, Messenger/biosynthesis , Regulon , Sigma Factor/metabolism , Trans-Activators/genetics
18.
Eur J Cell Biol ; 91(2): 150-5, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22154550

ABSTRACT

NAC (nascent polypeptide associated complex) is a heterodimer consisting of an α and a ß subunit. skNAC (skeletal and heart muscle-specific form of αNAC) is a variant of αNAC, which is induced in muscle differentiation and regeneration. We show here that skNAC expression is regulated by p38 MAPK, which plays a crucial role in myogenesis. Furthermore, inhibition of skNAC expression in myoblasts via specific siRNAs might lead to disturbed incorporation of myosin heavy chain (MyHC) into sarcomeres, however, it has no inhibitory effect on absolute MyHC protein levels. Taken together, our data suggest that skNAC regulates specific aspects of myogenesis.


Subject(s)
Molecular Chaperones/metabolism , Muscle, Skeletal/metabolism , Myoblasts/metabolism , Animals , Cell Differentiation , Cells, Cultured , Cycloheximide/pharmacology , Dactinomycin/pharmacology , Mice , Molecular Chaperones/antagonists & inhibitors , Molecular Chaperones/genetics , Muscle Development , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Myoblasts/cytology , Myoblasts/drug effects , Myosin Heavy Chains/metabolism , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/genetics , Regeneration , Sarcomeres/metabolism , Signal Transduction , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...