Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 18 de 18
1.
Int J Mol Sci ; 25(7)2024 Mar 22.
Article En | MEDLINE | ID: mdl-38612413

Cancers reprogram macrophages (MΦs) to a tumor-growth-promoting TAM (tumor-associated MΦ) phenotype that is similar to the anti-inflammatory M2 phenotype. Poly(ADP-ribose) polymerase (PARP) enzymes regulate various aspects of MΦ biology, but their role in the development of TAM phenotype has not yet been investigated. Here, we show that the multispectral PARP inhibitor (PARPi) PJ34 and the PARP14 specific inhibitor MCD113 suppress the expression of M2 marker genes in IL-4-polarized primary murine MΦs, in THP-1 monocytic human MΦs, and in primary human monocyte-derived MΦs. MΦs isolated from PARP14 knockout mice showed a limited ability to differentiate to M2 cells. In a murine model of TAM polarization (4T1 breast carcinoma cell supernatant transfer to primary MΦs) and in a human TAM model (spheroids formed from JIMT-1 breast carcinoma cells and THP-1-MΦs), both PARPis and the PARP14 KO phenotype caused weaker TAM polarization. Increased JIMT-1 cell apoptosis in co-culture spheroids treated with PARPis suggested reduced functional TAM reprogramming. Protein profiling arrays identified lipocalin-2, macrophage migration inhibitory factor, and plasminogen activator inhibitor-1 as potential (ADP-ribosyl)ation-dependent mediators of TAM differentiation. Our data suggest that PARP14 inhibition might be a viable anticancer strategy with a potential to boost anticancer immune responses by reprogramming TAMs.


Breast Neoplasms , Tumor-Associated Macrophages , Animals , Female , Humans , Mice , Cell Differentiation , Macrophages , Mice, Knockout , Poly(ADP-ribose) Polymerases , Tamoxifen
2.
Biomedicines ; 10(6)2022 Jun 10.
Article En | MEDLINE | ID: mdl-35740393

Acute pancreatitis (AP) poses a worldwide challenge due to the growing incidence and its potentially life-threatening course and complications. Specific targeted therapies are not available, prompting the identification of new pathways and novel therapeutic approaches. Flavonoids comprise several groups of biologically active compounds with wide-ranging effects. The flavone compound, tricetin (TCT), has not yet been investigated in detail but sporadic reports indicate diverse biological activities. In the current study, we evaluated the potential protective effects of TCT in AP. TCT (30 µM) protected isolated primary murine acinar cells from the cytotoxic effects of cerulein, a cholecystokinin analog peptide. The protective effects of TCT were observed in a general viability assay (calcein ester hydrolysis), in an apoptosis assay (caspase activity), and in necrosis assays (propidium iodide uptake and lactate dehydrogenase release). The effects of TCT were not related to its potential antioxidant effects, as TCT did not protect against H2O2-induced acinar cell death despite possessing radical scavenging activity. Cerulein-induced expression of IL1ß, IL6, and matrix metalloproteinase 2 and activation of nuclear factor-κB (NFκB) were reduced by 30 µM TCT. In vivo experiments confirmed the protective effect of TCT in a mouse model of cerulein-induced AP. TCT suppressed edema formation and apoptosis in the pancreas and reduced lipase and amylase levels in the serum. Moreover, TCT inhibited interleukin-1ß (IL1ß), interleukin-6 (IL6), and tumor necrosis factor-α (TNFα) expression in the pancreas and reduced the activation of the oxidative DNA damage sensor enzyme poly(ADP-ribose) polymerase-1 (PARP-1). Our data indicate that TCT can be a potential treatment option for AP.

3.
Cancer Immunol Immunother ; 71(9): 2151-2168, 2022 Sep.
Article En | MEDLINE | ID: mdl-35066605

Despite recent advances in the development of novel personalized therapies, breast cancer continues to challenge physicians with resistance to various advanced therapies. The anticancer action of the anti-HER2 antibody, trastuzumab, involves antibody-dependent cell-mediated cytotoxicity (ADCC) by natural killer (NK) cells. Here, we report a repurposing screen of 774 clinically used compounds on NK-cell + trastuzumab-induced killing of JIMT-1 breast cancer cells. Using a calcein-based high-content screening (HCS) assay for the image-based quantitation of ADCC that we have developed and optimized for this purpose, we have found that the multitargeted tyrosine kinase inhibitor sunitinib inhibits ADCC in this model. The cytoprotective effect of sunitinib was also confirmed with two other assays (lactate dehydrogenase release, and electric cell substrate impedance sensing, ECIS). The drug suppressed NK cell activation as indicated by reduced granzyme B deposition on to the target cells and inhibition of interferon-γ production by the NK cells. Moreover, sunitinib induced downregulation of HER2 on the target cells' surface, changed the morphology and increased adherence of the target cells. Moreover, sunitinib also triggered the autophagy pathway (speckled LC3b) as an additional potential underlying mechanism of the cytoprotective effect of the drug. Sunitinib-induced ADCC resistance has been confirmed in a 3D tumor model revealing the prevention of apoptotic cell death (Annexin V staining) in JIMT-1 spheroids co-incubated with NK cells and trastuzumab. In summary, our HCS assay may be suitable for the facile identification of ADCC boosting compounds. Our data urge caution concerning potential combinations of ADCC-based immunotherapies and sunitinib.


Breast Neoplasms , Antibody-Dependent Cell Cytotoxicity , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Protein Kinase Inhibitors/therapeutic use , Receptor, ErbB-2/metabolism , Sunitinib/pharmacology , Sunitinib/therapeutic use , Trastuzumab/pharmacology
4.
Int J Mol Sci ; 22(16)2021 Aug 18.
Article En | MEDLINE | ID: mdl-34445574

Osteosarcoma is a frequent and extremely aggressive type of pediatric cancer. New therapeutic approaches are needed to improve the overall survival of osteosarcoma patients. Our previous results suggest that NMNAT1, a key enzyme in nuclear NAD+ synthesis, facilitates the survival of cisplatin-treated osteosarcoma cells. A high-throughput cytotoxicity screening was performed to identify novel pathways or compounds linked to the cancer-promoting role of NMNAT1. Nine compounds caused higher toxicity in the NMNAT1 KO U2OS cells compared to their wild type counterparts, and actinomycin D (ActD) was the most potent. ActD-treatment of NMNAT1 KO cells increased caspase activity and secondary necrosis. The reduced NAD+ content in NMNAT1 KO cells was further decreased by ActD, which partially inhibited NAD+-dependent enzymes, including the DNA nick sensor enzyme PARP1 and the NAD+-dependent deacetylase SIRT1. Impaired PARP1 activity increased DNA damage in ActD-treated NMNAT1 knockout cells, while SIRT1 impairment increased acetylation of the p53 protein, causing the upregulation of pro-apoptotic proteins (NOXA, BAX). Proliferation was decreased through both PARP- and SIRT-dependent pathways. On the one hand, PARP inhibitors sensitized wild type but not NMNAT1 KO cells to ActD-induced anti-clonogenic effects; on the other hand, over-acetylated p53 induced the expression of the anti-proliferative p21 protein leading to cell cycle arrest. Based on our results, NMNAT1 acts as a survival factor in ActD-treated osteosarcoma cells. By inhibiting both PARP1- and SIRT1-dependent cellular pathways, NMNAT1 inhibition can be a promising new tool in osteosarcoma chemotherapy.


Biomarkers, Tumor/metabolism , Bone Neoplasms/prevention & control , Dactinomycin/pharmacology , Gene Expression Regulation, Neoplastic , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Osteosarcoma/prevention & control , Antibiotics, Antineoplastic/pharmacology , Apoptosis , Biomarkers, Tumor/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Proliferation , Humans , Nicotinamide-Nucleotide Adenylyltransferase/antagonists & inhibitors , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Osteosarcoma/metabolism , Osteosarcoma/pathology , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism , Tumor Cells, Cultured
5.
Int J Mol Sci ; 22(7)2021 Mar 30.
Article En | MEDLINE | ID: mdl-33808340

Chronic pancreatitis (CP) is an inflammatory disease of the pancreas characterized by ductal obstructions, tissue fibrosis, atrophy and exocrine and endocrine pancreatic insufficiency. However, our understanding is very limited concerning the disease's progression from a single acute inflammation, via recurrent acute pancreatitis (AP) and early CP, to the late stage CP. Poly(ADP-ribose) polymerase 1 (PARP1) is a DNA damage sensor enzyme activated mostly by oxidative DNA damage. As a co-activator of inflammatory transcription factors, PARP1 is a central mediator of the inflammatory response and it has also been implicated in acute pancreatitis. Here, we set out to investigate whether PARP1 contributed to the pathogenesis of CP. We found that the clinically used PARP inhibitor olaparib (OLA) had protective effects in a murine model of CP induced by multiple cerulein injections. OLA reduced pancreas atrophy and expression of the inflammatory mediators TNFα and interleukin-6 (IL-6), both in the pancreas and in the lungs. Moreover, there was significantly less fibrosis (Masson's trichrome staining) in the pancreatic sections of OLA-treated mice compared to the cerulein-only group. mRNA expression of the fibrosis markers TGFß, smooth muscle actin (SMA), and collagen-1 were markedly reduced by OLA. CP was also induced in PARP1 knockout (KO) mice and their wild-type (WT) counterparts. Inflammation and fibrosis markers showed lower expression in the KO compared to the WT mice. Moreover, reduced granulocyte infiltration (tissue myeloperoxidase activity) and a lower elevation of serum amylase and lipase activity could also be detected in the KO mice. Furthermore, primary acinar cells isolated from KO mice were also protected from cerulein-induced toxicity compared to WT cells. In summary, our data suggest that PARP inhibitors may be promising candidates for repurposing to treat not only acute but chronic pancreatitis as well.


Pancreatitis/physiopathology , Poly (ADP-Ribose) Polymerase-1/metabolism , Acinar Cells/metabolism , Acute Disease , Animals , Ceruletide/pharmacology , Disease Models, Animal , Fibrosis , Inflammation/pathology , Interleukin-6/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreas/metabolism , Pancreatitis/immunology , Pancreatitis, Chronic/pathology , Poly (ADP-Ribose) Polymerase-1/physiology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/metabolism
6.
Int J Mol Sci ; 22(4)2021 Feb 05.
Article En | MEDLINE | ID: mdl-33562813

The conserved Blm10/PA200 proteins are proteasome activators. Previously, we identified PA200-enriched regions in the genome of SH-SY5Y neuroblastoma cells by chromatin immunoprecipitation (ChIP) and ChIP-seq analysis. We also found that selective mitochondrial inhibitors induced PA200 redistribution in the genome. Collectively, our data indicated that PA200 regulates cellular homeostasis at the transcriptional level. In the present study, our aim is to investigate the impact of stable PA200 depletion (shPA200) on the overall transcriptome of SH-SY5Y cells. RNA-seq data analysis reveals that the genetic ablation of PA200 leads to overall changes in the transcriptional landscape of SH-SY5Y neuroblastoma cells. PA200 activates and represses genes regulating metabolic processes, such as the glycolysis and mitochondrial function. Using metabolic assays in live cells, we showed that stable knockdown of PA200 does not change basal respiration. Spare respiratory capacity and proton leak however are slightly, yet significantly, reduced in PA200-deficient cells by 99.834% and 84.147%, respectively, compared to control. Glycolysis and glycolytic capacity show a 42.186% and 26.104% increase in shPA200 cells, respectively, compared to control. These data suggest a shift from oxidative phosphorylation to glycolysis especially when cells are exposed to oligomycin-induced stress. Furthermore, we observed a preserved long and compact tubular mitochondrial morphology after inhibition of ATP synthase by oligomycin, which might be associated with the glycolytic change of shPA200 cells. The present study also demonstrates that the proteolytic cleavage of Opa1 is affected, and that the level of OMA1 is significantly reduced in shPA200 cells upon oligomycin-induced mitochondrial insult. Together, these findings suggest a role for PA200 in the regulation of metabolic changes in response to selective inhibition of ATP synthase in an in vitro cellular model.


GTP Phosphohydrolases/genetics , Gene Expression Profiling/methods , Neuroblastoma/genetics , Nuclear Proteins/genetics , RNA, Small Interfering/pharmacology , Cell Line, Tumor , Chromatin Immunoprecipitation , Gene Deletion , Gene Expression Regulation, Neoplastic/drug effects , Gene Regulatory Networks , Glycolysis/drug effects , Humans , Mitochondria/drug effects , Mitochondria/genetics , Nuclear Proteins/antagonists & inhibitors , Oligomycins/pharmacology , Oxidative Phosphorylation/drug effects , Sequence Analysis, RNA
7.
Sports Med Health Sci ; 3(1): 40-45, 2021 Mar.
Article En | MEDLINE | ID: mdl-35782677

Blood flow restriction (BFR) during exercise bouts has been used to induce hypertrophy of skeletal muscle, even with low loads. However, the effects of BFR during the rest periods between sets are not known. We have tested the hypothesis that BFR during rest periods between sets of high-intensity resistance training would enhance performance. Twenty-two young adult male university students were recruited for the current study, with n = 11 assigned to BFR and n = 11 to a control group. The results revealed that four weeks training at 70% of 1 RM, five sets and 10 repetitions, three times a week with and without BFR, resulted in similar progress in maximal strength and in the number of maximal repetitions. The miR-1 and miR-133a decreased significantly in the vastus lateralis muscle of BFR group compared to the group without BFR, while no significant differences in the levels of miR133b, miR206, miR486, and miR499 were found between groups. In conclusion, it seems that BFR restrictions during rest periods of high-intensity resistance training, do not provide benefit for enhanced performance after a four-week training program. However, BFR-induced downregulation of miR-1 and miR-133a might cause different adaptive responses of skeletal muscle to high intensity resistance training.

8.
Br J Pharmacol ; 178(5): 1095-1113, 2021 03.
Article En | MEDLINE | ID: mdl-33332573

BACKGROUND AND PURPOSE: Excessive oxidative stress can induce PARP1-mediated programmed necrotic cell death, termed parthanatos. Inhibition of parthanatos may be therapeutically beneficial in a wide array of diseases associated with tissue injury and inflammation. Our goal was to identify novel molecules inhibiting parthanatos. EXPERIMENTAL APPROACH: A small library of 774 pharmacologically active compounds was screened in a Sytox Green uptake assay, which identified 20 hits that reduced hydrogen-peroxide-induced parthanatos with an efficiency comparable to the benchmark PARP inhibitor, PJ34. KEY RESULTS: Of these hits, two compounds, antifungal ciclopirox and dopamine receptor agonist apomorphine, inhibited PAR polymer synthesis. These two compounds prevented the binding of PARP1 to oxidatively damaged DNA but did not directly interfere with the interaction between DNA and PARP1. Both compounds inhibited mitochondrial superoxide and H2 O2 production and suppressed DNA breakage. Since H2 O2 -induced damage is dependent on Fe2+ -catalysed hydroxyl radical production (Fenton chemistry), we determined the iron chelation activity of the two test compounds and found that ciclopirox and, to a lesser extent, apomorphine act as iron chelators. We also show that the Fe2+ chelation and indirect PARP inhibitory effects of ciclopirox translate to anti-inflammatory actions as demonstrated in a mouse dermatitis model, where ciclopirox reduced ear swelling, inflammatory cell recruitment and poly(ADP-ribosyl)ation. CONCLUSION AND IMPLICATIONS: Our findings indicate that the antimycotic drug, ciclopirox, acts as an iron chelator and thus targets an early event in hydrogen-peroxide-induced parthanatos. Ciclopirox has the potential to be repurposed as a cytoprotective and anti-inflammatory agent.


Parthanatos , Animals , Ciclopirox/pharmacology , Hydrogen Peroxide/pharmacology , Mice , Oxidative Stress , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology
9.
Cancers (Basel) ; 12(5)2020 May 07.
Article En | MEDLINE | ID: mdl-32392755

Osteosarcoma (OS) is the most common bone tumor in children and adolescents. Modern OS treatment, based on the combination of neoadjuvant chemotherapy (cisplatin + doxorubicin + methotrexate) with subsequent surgical removal of the primary tumor and metastases, has dramatically improved overall survival of OS patients. However, further research is needed to identify new therapeutic targets. Here we report that expression level of the nuclear NAD synthesis enzyme, nicotinamide mononucleotide adenylyltransferase-1 (NMNAT1), increases in U-2OS cells upon exposure to DNA damaging agents, suggesting the involvement of the enzyme in the DNA damage response. Moreover, genetic inactivation of NMNAT1 sensitizes U-2OS osteosarcoma cells to cisplatin, doxorubicin, or a combination of these two treatments. Increased cisplatin-induced cell death of NMNAT1-/- cells showed features of both apoptosis and necroptosis, as indicated by the protective effect of the caspase-3 inhibitor z-DEVD-FMK and the necroptosis inhibitor necrostatin-1. Activation of the DNA damage sensor enzyme poly(ADP-ribose) polymerase 1 (PARP1), a major consumer of NAD+ in the nucleus, was fully blocked by NMNAT1 inactivation, leading to increased DNA damage (phospho-H2AX foci). The PARP inhibitor, olaparib, sensitized wild type but not NMNAT1-/- cells to cisplatin-induced anti-clonogenic effects, suggesting that impaired PARP1 activity is important for chemosensitization. Cisplatin-induced cell death of NMNAT1-/- cells was also characterized by a marked drop in cellular ATP levels and impaired mitochondrial respiratory reserve capacity, highlighting the central role of compromised cellular bioenergetics in chemosensitization by NMNAT1 inactivation. Moreover, NMNAT1 cells also displayed markedly higher sensitivity to cisplatin when grown as spheroids in 3D culture. In summary, our work provides the first evidence that NMNAT1 is a promising therapeutic target for osteosarcoma and possibly other tumors as well.

10.
Int J Mol Sci ; 20(21)2019 Oct 26.
Article En | MEDLINE | ID: mdl-31717806

Huntington's disease (HD) is an inherited neurodegenerative disorder, caused by an abnormal polyglutamine (polyQ) expansion in the huntingtin protein (Htt). Mitochondrial dysfunction and impairment of the ubiquitin-proteasome system (UPS) are hallmarks of HD neurons. The extraneural manifestations of HD are still unclear. We investigated the crosstalk between mitochondria and proteolytic function in skin fibroblasts from juvenile HD patients. We found reduced mitosis, increased cell size, elevated ROS and increased mitochondrial membrane potential in juvenile HD fibroblasts, while cellular viability was maintained. Mitochondrial OXPHOS analysis did not reveal significant differences compared to control. However, the level of mitochondrial fusion and fission proteins was significantly lower and branching in the mitochondria network was reduced. We hypothesized that juvenile HD fibroblasts counterbalance cellular damage and mitochondrial network deficit with altered proteasome activity to promote cell survival. Our data reveal that juvenile HD fibroblasts exhibit higher proteasome activity, which was associated with elevated gene and protein expression of parkin. Moreover, we demonstrate elevated proteasomal degradation of the mitochondrial fusion protein Mfn1 in diseased cells compared to control cells. Our data suggest that juvenile HD fibroblasts respond to mutant polyQ expansion of Htt with enhanced proteasome activity and faster turnover of specific UPS substrates to protect cells.


Fibroblasts/metabolism , Huntingtin Protein/genetics , Huntington Disease/genetics , Proteasome Endopeptidase Complex/metabolism , Ubiquitin-Protein Ligases/metabolism , Cell Proliferation , Cells, Cultured , Fibroblasts/cytology , GTP Phosphohydrolases/metabolism , Glycolysis , Humans , Huntingtin Protein/metabolism , Huntington Disease/metabolism , Membrane Potential, Mitochondrial/physiology , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mutation , Neurons/metabolism , Peptides/metabolism , Reactive Oxygen Species/metabolism , Skin/cytology , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics
11.
Int J Mol Sci ; 20(17)2019 Sep 03.
Article En | MEDLINE | ID: mdl-31484391

Activated macrophages upregulate inducible nitric oxide synthase (iNOS) leading to the profuse production of nitric oxide (NO) and, eventually, tissue damage. Using macrophage NO production as a biochemical marker of inflammation, we tested different parts (flower, leaf, and stem) of the medicinal plant, Spilanthes acmella. We found that extracts prepared from all three parts, especially the flowers, suppressed NO production in RAW macrophages in response to interferon-γ and lipopolysaccharide. Follow up experiments with selected bioactive molecules from the plant (α-amyrin, ß-caryophylline, scopoletin, vanillic acid, trans-ferulic acid, and spilanthol) indicated that the N-alkamide, spilanthol, is responsible for the NO-suppressive effects and provides protection from NO-dependent cell death. Spilanthol reduced the expression of iNOS mRNA and protein and, as a possible underlying mechanism, inhibited the activation of several transcription factors (NFκB, ATF4, FOXO1, IRF1, ETS, and AP1) and sensitized cells to downregulation of Smad (TF array experiments). The iNOS inhibitory effect translated into an anti-inflammatory effect, as demonstrated in a phorbol 12-myristate 13-acetate-induced dermatitis and, to a smaller extent, in cerulein-induced pancreatitis. In summary, we demonstrate that spilanthol inhibits iNOS expression, NO production and suppresses inflammatory TFs. These events likely contribute to the observed anti-inflammatory actions of spilanthol in dermatitis and pancreatitis.


Dermatitis/drug therapy , Dermatitis/metabolism , Macrophages/metabolism , Nitric Oxide Synthase Type II/metabolism , Pancreatitis/drug therapy , Pancreatitis/metabolism , Polyunsaturated Alkamides/therapeutic use , Animals , Cell Survival/drug effects , Dermatitis/genetics , Forkhead Box Protein O1/metabolism , Mice , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Pancreatitis/genetics , Peroxidase/metabolism , RAW 264.7 Cells , Signal Transduction/drug effects , Signal Transduction/genetics
12.
Redox Biol ; 26: 101261, 2019 09.
Article En | MEDLINE | ID: mdl-31279985

Activated macrophages play a central role in both the development and resolution of inflammation. These immune cells need to be functional in harmful conditions with high levels of reactive oxygen and nitrogen species that can damage their basic cell components, which may alter their metabolism. An excessive accumulation of these cell alterations drives macrophages inexorably to cell death, which has been associated to the development of several inflammatory diseases and even with aging in a process termed as "immunosenescence". Macrophages, however, exhibit a prolonged survival in this hostile environment because they equip themselves with a complex network of protective mechanisms. Here we provide an overview of these self-defense mechanisms with special attention being paid to bioactive lipid mediators, NRF2 signaling and metabolic reprogramming.


Immunosenescence , Macrophages/metabolism , Oxidation-Reduction , Oxidative Stress , Animals , Biomarkers , Disease Susceptibility , Humans , Immunity, Innate , Inflammation/etiology , Inflammation/metabolism , Lipid Metabolism , Macrophages/immunology , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction
13.
Free Radic Biol Med ; 131: 184-196, 2019 02 01.
Article En | MEDLINE | ID: mdl-30502458

In inflamed tissues or during ischemia-reperfusion episodes, activated macrophages produce large amounts of reactive species and are, thus, exposed to the damaging effects of reactive species. Here, our goal was to investigate the mechanism whereby activated macrophages protect themselves from oxidant stress-induced cell death. Hydrogen peroxide-treated mouse bone marrow-derived macrophages (BMDM) and THP-1 human monocyte-derived cells were chosen as models. We found a gradual development of resistance: first in monocyte-to-macrophage differentiation, and subsequently after lipopolysaccharide (LPS) exposure. Investigating the mechanism of the latter, we found that exposure to intense hydrogen peroxide stress causes poly(ADP-ribose) polymerase-1 (PARP-1) dependent programmed necrotic cell death, also known as parthanatos, as indicated by the protected status of PARP-1 knockout BMDMs and the protective effect of the PARP inhibitor PJ-34. In hydrogen peroxide-treated macrophages, however, apoptosis inducing factor (AIF) proved dispensable for parthanatos; nuclear translocation of AIF was not observed. A key event in LPS-mediated protection against the hydrogen peroxide-induced AIF independent parthanatos was downregulation of PARP1 mRNA and protein. The importance of this event was confirmed by overexpression of PARP1 in THP1 cells using a viral promoter, which lead to stable PARP1 levels even after LPS treatment and unresponsiveness to LPS-induced cytoprotection. In BMDMs, LPS-induced PARP1 suppression lead to prevention of NAD+ depletion. Moreover, LPS also induced expression of antioxidant proteins (superoxide dismutase-2, thioredoxin reductase 1 and peroxiredoxin) and triggered a metabolic shift to aerobic glycolysis, also known as the Warburg effect. In summary, we provide evidence that in macrophages intense hydrogen peroxide stress causes AIF-independent parthanatos from which LPS provides protection. The mechanism of LPS-mediated cytoprotection involves downregulation of PARP1, spared NAD+ and ATP pools, upregulation of antioxidant proteins, and a metabolic shift from mitochondrial respiration to aerobic glycolysis.


Apoptosis Inducing Factor/genetics , Hydrogen Peroxide/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Poly (ADP-Ribose) Polymerase-1/genetics , Superoxide Dismutase/genetics , Animals , Apoptosis Inducing Factor/metabolism , Gene Expression Regulation , Glycolysis/drug effects , Glycolysis/genetics , Humans , Hydrogen Peroxide/antagonists & inhibitors , Macrophage Activation/drug effects , Macrophages/cytology , Macrophages/metabolism , Mice , Mitochondria/drug effects , Mitochondria/metabolism , NAD/metabolism , Oxidative Phosphorylation/drug effects , Parthanatos/drug effects , Parthanatos/genetics , Peroxiredoxins/genetics , Peroxiredoxins/metabolism , Phenanthrenes/pharmacology , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly (ADP-Ribose) Polymerase-1/metabolism , Primary Cell Culture , Promoter Regions, Genetic , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Superoxide Dismutase/metabolism , THP-1 Cells , Thioredoxin Reductase 1/genetics , Thioredoxin Reductase 1/metabolism
14.
Free Radic Biol Med ; 131: 218-224, 2019 02 01.
Article En | MEDLINE | ID: mdl-30529301

The function of macrophages makes them vulnerable to several sources of stress and damage, and thus there is a considerable requirement for some form of resilient molecular defence. Differentiation of human macrophages and their further pro-inflammatory (M1) polarization with bacterial endotoxin is associated with increased transcription of PARP1 and SOD2. The latter gene responded immediately to LPS with high NFκB-dependent expression rate, and the resulting enzyme made M1 macrophages resistant to hydrogen peroxide-induced oxidative stress and associated cell death. LPS-induced recruitment of RELA to SOD2 promoter was accompanied by release of PARP1 and LSD1 from chromatin and increased H3K4 di- and tri-methylation. PARP1 dissociation from SOD2 promoter occurred at an early stage of SOD2 transcriptional activation. This event contributed to the termination of mRNA synthesis at a later stage of macrophage polarization by allowing LSD1 to rebind to the SOD2 promoter. LSD1 removed transcription-promoting methylation of H3K4 and led to displacement of RELA. Analysis of temporal changes at the SOD2 promoter indicated a direct mutual interdependence between PARP1, LSD1, H3K4 methylation and the ongoing SOD2 transcription, which correlated positively with both PARP1 abundance on the chromatin and dimethylation of H3K4, but negatively with LSD1 and chromatin compaction in LPS-treated macrophages. Deficiency of LSD1 activity and maintenance of PARP1 at the SOD2 promoter substantially upregulated SOD2 level, thereby further increasing resistance of M1 macrophages to hydrogen peroxide. Inhibitors of LSD1 and PARP1 poisons that capture the latter enzyme on the chromatin seem to be prosurvival molecular tools protecting polarized macrophages from certain pro-oxidative conditions.


Cell Death/drug effects , Histone Demethylases/genetics , Hydrogen Peroxide/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Poly (ADP-Ribose) Polymerase-1/genetics , Superoxide Dismutase/genetics , Cell Death/genetics , Chromatin/chemistry , Chromatin/metabolism , Gene Expression Regulation , Histone Demethylases/metabolism , Histones/genetics , Histones/metabolism , Humans , Hydrogen Peroxide/pharmacology , Macrophages/cytology , Macrophages/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Oxidative Stress/drug effects , Poly (ADP-Ribose) Polymerase-1/metabolism , Primary Cell Culture , Promoter Regions, Genetic , Signal Transduction , Superoxide Dismutase/metabolism , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Transcription, Genetic
15.
Oxid Med Cell Longev ; 2018: 5286785, 2018.
Article En | MEDLINE | ID: mdl-30581533

Wound healing is a complex multiphase process which can be hampered by many factors including impaired local circulation, hypoxia, infection, malnutrition, immunosuppression, and metabolic dysregulation in diabetes. Redox dysregulation is a common feature of many skin diseases demonstrated by virtually all cell types in the skin with overproduction of reactive oxygen and nitrogen species. The objective of this study was to characterize the redox environment in wound fluids and sera from patients suffering from chronic leg ulcers (n = 19) and acute wounds (bulla fluids from second degree burns; n = 11) with serum data also compared to those from healthy volunteers (n = 7). Significantly higher concentrations of TNF-α, interleukine-8, vascular endothelial growth factor, and lactate dehydrogenase (measure of cell damage) were found in fluids from chronic wounds compared to acute ones. The extent of protein carbonylation (measure of protein oxidation), lipid peroxidation, and tyrosine nitration (indicator of peroxynitrite production) was similar in acute and chronic wound fluids, while radical scavenging activity and glutathione (GSH) levels were elevated in chronic wound fluids compared to acute wounds. Sera were also assessed for the same set of parameters with no significant differences detected. Nitrotyrosine (the footprint of the potent oxidant peroxynitrite) and poly(ADP-ribose) (the product of the DNA damage sensor enzyme PARP-1) could be detected in wound biopsies. Our data identify multiple signs of redox stress in chronic wounds with notable differences. In chronic wounds, elevations in antioxidant levels/activities may indicate compensatory mechanisms against inflammation. The presence of nitrotyrosine and poly(ADP-ribose) in tissues from venous leg ulcers indicate peroxynitrite production and PARP activation in chronic wounds.


Wound Healing/physiology , Aged , Aged, 80 and over , Female , Glutathione/metabolism , Humans , Interleukin-8/metabolism , L-Lactate Dehydrogenase/metabolism , Lipid Peroxidation/physiology , Male , Middle Aged , Oxidation-Reduction , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Protein Carbonylation/physiology , Thiobarbituric Acid Reactive Substances/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Vascular Endothelial Growth Factor A/metabolism
16.
Redox Biol ; 16: 59-74, 2018 06.
Article En | MEDLINE | ID: mdl-29477046

Redox regulation has been proposed to control various aspects of carcinogenesis, cancer cell growth, metabolism, migration, invasion, metastasis and cancer vascularization. As cancer has many faces, the role of redox control in different cancers and in the numerous cancer-related processes often point in different directions. In this review, we focus on the redox control mechanisms of tumor cell destruction. The review covers the tumor-intrinsic role of oxidants derived from the reduction of oxygen and nitrogen in the control of tumor cell proliferation as well as the roles of oxidants and antioxidant systems in cancer cell death caused by traditional anticancer weapons (chemotherapeutic agents, radiotherapy, photodynamic therapy). Emphasis is also put on the role of oxidants and redox status in the outcome following interactions between cancer cells, cytotoxic lymphocytes and tumor infiltrating macrophages.


Antioxidants/therapeutic use , Neoplasms/drug therapy , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Antineoplastic Agents/therapeutic use , Humans , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Oxidative Stress/genetics , Signal Transduction/drug effects
17.
Article En | MEDLINE | ID: mdl-29313809

Differentiation of human monocytes is associated with proliferation arrest resulting from activation of the inter alia retinoblastoma protein family of gene repressors, which target gene promoters in an E2F-dependent manner. To investigate RBL2 contribution to defining monocyte phenotype and function, we used primer libraries. We identified genes encoding two surface receptors (CXCR1 and IL17RE) and two TLR signaling mediators (CD86 and NFKB2) that are repressed by the RBL2-E2F4-HDAC1-BRM complex. Surprisingly, PARP1 co-regulated 24 out of the 28 identified genes controlled by RBL2. Upon RBL2 silencing, PARP1 was recruited to one subset of RBL2-dependent genes, represented by MAP2K6 and MAPK3. RBL2 silencing also restored PARP1 transcription. Gene promoters enriched in PARP1 were characterized by increased histone acetylation and the replacement of HDAC1 with EP300. While PARP1 was dispensable for HDAC1 dissociation, EP300 was found only at gene promoters enriched in PARP1. EP300 activated transcription of PARP1/RBL2 co-regulated genes, but not genes solely controlled by RBL2. DNA was a prerequisite to the formation of an immunoprecipitated PARP1-EP300 complex, suggesting that PARP1 enabled EP300 binding, which in turn activated gene transcription. Notably, PARP1 overexpression failed to overcome the inhibitory effect of RBL2 on MAP2K6 and MAPK3 transcription. The same interdependence was observed in proliferating cancer cells; the low abundance of RBL2 resulted in PARP1-mediated EP300 recruitment to promoters of the MAP2K6 and MAPK3 genes. We conclude that RBL2 may indirectly regulate transcription of some genes by controlling PARP1-mediated EP300 recruitment.

18.
Free Radic Biol Med ; 53(9): 1680-8, 2012 Nov 01.
Article En | MEDLINE | ID: mdl-22964577

Cigarette smoking can contribute to the development of many human diseases such as cardiovascular disease, lung cancer, asthma, and chronic obstructive pulmonary disease. Thousands of compounds are present in cigarette smoke, including a large number of reactive oxygen species that can cause DNA damage, leading to the activation of poly(ADP-ribose) polymerase (PARP) enzymes. The PAR polymer is degraded by poly(ADP-ribose) glycohydrolase (PARG). Here we have investigated the effects of cigarette smoke extract (CSE) on A549 human lung epithelial cells. CSE induced DNA damage (comet assay), PAR accumulation (immunofluorescence and immunoblotting), impaired proliferation (clonogenic survival assay and electric cell-substrate impedance sensing measurement), and cell death (MTT reduction, propidium iodide uptake, lactate dehydrogenase release). CSE-induced cell death was also characterized by mitochondrial depolarization but massive translocation of apoptosis-inducing factor could not be observed. To investigate the role of PARylation in CSE-induced oxidative stress, PARP-1- and PARG-silenced A549 cells were used. Silencing of both PARP-1 and PARG sensitized cells to CSE-induced toxicity: PARP-1- and PARG-silenced cell lines exhibited reduced clonogenic survival, displayed a delayed repair of DNA breaks, and showed higher levels of cytotoxicity. CSE triggered the production of mitochondrial superoxide and hydrogen peroxide. Addition of superoxide dismutase increased, whereas catalase abolished, CSE-induced PAR formation. In summary, our data show that the superoxide-hydrogen peroxide-DNA breakage pathway activates the PAR cycle by PARP-1 and PARG, which serves as a survival mechanism in CSE-exposed cells. Our data also raise the possibility that the PARP-1/PARG status of smokers may be an important determinant of the efficiency of DNA repair in their lungs and of their susceptibility to CS-induced carcinogenesis.


Cell Survival/drug effects , Hydrogen Peroxide/pharmacology , Nicotiana/chemistry , Oxidants/pharmacology , Poly Adenosine Diphosphate Ribose/metabolism , Smoke , Cell Line, Tumor , DNA Damage , Gene Knockdown Techniques , Glycoside Hydrolases/genetics , Glycoside Hydrolases/metabolism , Humans , L-Lactate Dehydrogenase/metabolism , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Protein Processing, Post-Translational/drug effects , Proteins/metabolism , RNA Interference , Reactive Oxygen Species/metabolism
...