1
|
Shi K, Li D, Jiang X, Du Y, Yu M. Identification and Characterization of the miRNA Transcriptome Controlling Green Pigmentation of Chicken Eggshells. Genes (Basel) 2024; 15:811. [PMID: 38927746 PMCID: PMC11202967 DOI: 10.3390/genes15060811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/17/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Green eggs are mainly caused by inserting an avian endogenous retrovirus (EVA-HP) fragment into the SLCO1B3 gene. Although the genotypes for this insertion allele are consistent, eggshell color (ESC) may vary after a peak laying period; light-colored eggs are undesired by consumers and farmers and result in financial loss, so it is necessary to resolve this problem. miRNAs are small non-coding RNAs that exert essential functions in animal development and diseases. However, the regulatory miRNAs and detailed molecular mechanisms regulating eggshell greenness remain unclear. In the present study, we determined the genotype of green-eggshell hens through the detection of a homozygous allele insertion in the SLCO1B3 gene. The shell gland epithelium was obtained from green-eggshell hens that produced white and green shell eggs to perform transcriptome sequencing and investigate the important regulatory mechanisms that influence the ESC. Approximately 921 miRNAs were expressed in these two groups, which included 587 known miRNAs and 334 novel miRNAs, among which 44 were differentially expressed. There were 22 miRNAs that were significantly upregulated in the green and white groups, respectively, which targeted hundreds of genes, including KIT, HMOX2, and several solute carrier family genes. A Gene Ontology enrichment analysis of the target genes showed that the differentially expressed miRNA-targeted genes mainly belonged to the functional categories of homophilic cell adhesion, gland development, the Wnt signaling pathway, and epithelial tube morphogenesis. A KEGG enrichment analysis showed that the Hedgehog signaling pathway was significantly transformed in this study. The current study provides an overview of the miRNA expression profiles and the interaction between the miRNAs and their target genes. It provides valuable insights into the molecular mechanisms underlying green eggshell pigmentation, screening more effective hens to produce stable green eggs and obtaining higher economic benefits.
Collapse
Affiliation(s)
| | | | | | | | - Minli Yu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (K.S.); (D.L.)
| |
Collapse
|
2
|
Soubeyrand S, Lau P, McPherson R. Regulation of TRIB1 abundance in hepatocyte models in response to proteasome inhibition. Sci Rep 2023; 13:9320. [PMID: 37291259 PMCID: PMC10250549 DOI: 10.1038/s41598-023-36512-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 06/05/2023] [Indexed: 06/10/2023] Open
Abstract
Tribbles related homolog 1 (TRIB1) contributes to lipid and glucose homeostasis by facilitating the degradation of cognate cargos by the proteasome. In view of the key metabolic role of TRIB1 and the impact of proteasome inhibition on hepatic function, we continue our exploration of TRIB1 regulation in two commonly used human hepatocyte models, transformed cell lines HuH-7 and HepG2. In both models, proteasome inhibitors potently upregulated both endogenous and recombinant TRIB1 mRNA and protein levels. Increased transcript abundance was unaffected by MAPK inhibitors while ER stress was a weaker inducer. Suppressing proteasome function via PSMB3 silencing was sufficient to increase TRIB1 mRNA expression. ATF3 was required to sustain basal TRIB1 expression and support maximal induction. Despite increasing TRIB1 protein abundance and stabilizing bulk ubiquitylation, proteasome inhibition delayed but did not prevent TRIB1 loss upon translation block. Immunoprecipitation experiments indicated that TRIB1 was not ubiquitylated in response to proteasome inhibition. A control bona fide proteasome substrate revealed that high doses of proteasome inhibitors resulted in incomplete proteasome inhibition. Cytoplasm retained TRIB1 was unstable, suggesting that TRIB1 lability is regulated prior to its nuclear import. N-terminal deletion and substitutions were insufficient to stabilize TRIB1. These findings identify transcriptional regulation as a prominent mechanism increasing TRIB1 abundance in transformed hepatocyte cell lines in response to proteasome inhibition and provide evidence of an inhibitor resistant proteasome activity responsible for TRIB1 degradation.
Collapse
Affiliation(s)
- Sébastien Soubeyrand
- Atherogenomics Laboratory, University of Ottawa Heart Institute, Ottawa, Canada.
| | - Paulina Lau
- Atherogenomics Laboratory, University of Ottawa Heart Institute, Ottawa, Canada
| | - Ruth McPherson
- Atherogenomics Laboratory, University of Ottawa Heart Institute, Ottawa, Canada.
- Division of Cardiology, Ruddy Canadian Cardiovascular Genetics Centre, University of Ottawa Heart Institute, Ottawa, Canada.
| |
Collapse
|
3
|
Moyano P, García JM, García J, Anadon MJ, Naval MV, Frejo MT, Sola E, Pelayo A, Pino JD. Manganese increases Aβ and Tau protein levels through proteasome 20S and heat shock proteins 90 and 70 alteration, leading to SN56 cholinergic cell death following single and repeated treatment. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 203:110975. [PMID: 32678756 DOI: 10.1016/j.ecoenv.2020.110975] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 06/10/2020] [Accepted: 06/29/2020] [Indexed: 06/11/2023]
Abstract
Manganese (Mn) produces cholinergic neuronal loss in basal forebrain (BF) region that was related to cognitive dysfunction induced after single and repeated Mn treatment. All processes that generate cholinergic neuronal loss in BF remain to be understood. Mn exposure may produce the reduction of BF cholinergic neurons by increasing amyloid beta (Aβ) and phosphorylated Tau (pTau) protein levels, altering heat shock proteins' (HSPs) expression, disrupting proteasome P20S activity and generating oxidative stress. These mechanisms, described to be altered by Mn in regions different than BF, could lead to the memory and learning process alteration produced after Mn exposure. The research performed shows that single and repeated Mn treatment of SN56 cholinergic neurons from BF induces P20S inhibition, increases Aβ and pTau protein levels, produces HSP90 and HSP70 proteins expression alteration, and oxidative stress generation, being the last two effects mediated by NRF2 pathway alteration. The increment of Aβ and pTau protein levels was mediated by HSPs and proteasome dysfunction. All these mechanisms mediated the cell decline observed after Mn treatment. Our results are relevant because they may assist to reveal the processes leading to the neurotoxicity and cognitive alterations observed after Mn exposure.
Collapse
Affiliation(s)
- Paula Moyano
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040, Madrid, Spain
| | - José Manuel García
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040, Madrid, Spain
| | - Jimena García
- Department of Pharmacolgy, Health Sciences School, Alfonso X University, 28691, Madrid, Spain
| | - María José Anadon
- Department of Legal Medicine, Psychiatry and Pathology, Medicine School, Complutense University of Madrid, 28041, Madrid, Spain
| | - María Victoria Naval
- Department of Pharmacology, Pharmacognosy and Botany, Pharmacy School, Complutense University of Madrid, 28040, Madrid, Spain
| | - María Teresa Frejo
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040, Madrid, Spain
| | - Emma Sola
- Department of Legal Medicine, Psychiatry and Pathology, Medicine School, Complutense University of Madrid, 28041, Madrid, Spain
| | - Adela Pelayo
- Department of Legal Medicine, Psychiatry and Pathology, Medicine School, Complutense University of Madrid, 28041, Madrid, Spain
| | - Javier Del Pino
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040, Madrid, Spain.
| |
Collapse
|
4
|
Khamis I, Heikkila JJ. Effect of isothiocyanates, BITC and PEITC, on stress protein accumulation, protein aggregation and aggresome-like structure formation in Xenopus A6 kidney epithelial cells. Comp Biochem Physiol C Toxicol Pharmacol 2018; 204:1-13. [PMID: 29100952 DOI: 10.1016/j.cbpc.2017.10.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 10/25/2017] [Accepted: 10/27/2017] [Indexed: 01/09/2023]
Abstract
Numerous studies have elucidated the health benefits of organosulfur compounds, known as isothiocyanates (ITCs), derived from cruciferous vegetables. As electrophiles, ITCs have the ability to directly bind and modify thiol-containing compounds such as glutathione and cellular protein, including tubulin. While the biochemical effects of ITCs have been well characterized, less information is available regarding their effects on the accumulation of stress-inducible heme oxygenase-1 (HO-1), heat shock proteins (HSPs) and the possible formation of aggregated protein due to thiol modification. The present study has examined the effect of the ITCs, benzyl isothiocyanate (BITC) and phenethyl isothiocyanate (PEITC), on the accumulation of HO-1, HSP70 and HSP30 in Xenopus laevis A6 kidney epithelial cells. Immunoblot analysis revealed that both BITC and PEITC induced the accumulation of HO-1 and HSP70 whereas HSP30 levels were enhanced only in cells treated with BITC. Immunocytochemistry determined that ITC treatment induced F-actin disorganization and membrane ruffling and enhanced accumulation of HO-1 in the cytoplasm. Additionally, BITC induced enhanced levels of ubiquitinated protein, aggregated protein, and the collapse and fragmentation of microtubules. In comparison, treatment of cells with the proteasomal inhibitor, MG132, induced the accumulation of all three stress proteins, aggregated protein and aggresome-like structures. Finally, cells pretreated with BITC inhibited the formation of MG132-induced aggresome-like structures in the perinuclear region. This latter finding suggests that BITC-induced microtubule fragmentation may impede the movement of aggregated protein via microtubules and their subsequent coalescence into aggresome-like structures in the perinuclear region.
Collapse
Affiliation(s)
- Imran Khamis
- Department of Biology, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - John J Heikkila
- Department of Biology, University of Waterloo, Waterloo, ON N2L 3G1, Canada.
| |
Collapse
|
5
|
Sheng XJ, Tu HJ, Chien WL, Kang KH, Lu DH, Liou HH, Lee MJ, Fu WM. Antagonism of proteasome inhibitor-induced heme oxygenase-1 expression by PINK1 mutation. PLoS One 2017; 12:e0183076. [PMID: 28806787 PMCID: PMC5555616 DOI: 10.1371/journal.pone.0183076] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 07/29/2017] [Indexed: 11/25/2022] Open
Abstract
PTEN-induced putative kinase 1 (PINK1) is an integral protein in the mitochondrial membrane and maintains mitochondrial fidelity. Pathogenic mutations in PINK1 have been identified as a cause of early-onset autosomal recessive familial Parkinson’s disease (PD). The ubiquitin proteasome pathway is associated with neurodegenerative diseases. In this study, we investigated whether mutations of PINK1 affects the cellular stress response following proteasome inhibition. Administration of MG132, a peptide aldehyde proteasome inhibitor, significantly increased the expression of heme oxygenase-1 (HO-1) in rat dopaminergic neurons in the substantia nigra and in the SH-SY5Y neuronal cell line. The induction of HO-1 expression by proteasome inhibition was reduced in PINK1 G309D mutant cells. MG132 increased the levels of HO-1 through the Akt, p38, and Nrf2 signaling pathways. Compared with the cells expressing WT-PINK1, the phosphorylation of Akt and p38 was lower in those cells expressing the PINK1 G309D mutant, which resulted in the inhibition of the nuclear translocation of Nrf2. Furthermore, MG132-induced neuronal death was enhanced by the PINK1 G309D mutation. In this study, we demonstrated that the G309D mutation impairs the neuroprotective function of PINK1 following proteasome inhibition, which may be related to the pathogenesis of PD.
Collapse
Affiliation(s)
- Xiang-Jun Sheng
- Pharmacological Institute, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hunag-Ju Tu
- Pharmacological Institute, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wei-Lin Chien
- Pharmacological Institute, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Kai-Hsiang Kang
- Pharmacological Institute, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Dai-Hua Lu
- Pharmacological Institute, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Horng-Huei Liou
- Pharmacological Institute, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ming-Jen Lee
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
- * E-mail: (WF); (ML)
| | - Wen-Mei Fu
- Pharmacological Institute, College of Medicine, National Taiwan University, Taipei, Taiwan
- * E-mail: (WF); (ML)
| |
Collapse
|
6
|
Shukla N, Somwar R, Smith RS, Ambati S, Munoz S, Merchant M, D'Arcy P, Wang X, Kobos R, Antczak C, Bhinder B, Shum D, Radu C, Yang G, Taylor BS, Ng CKY, Weigelt B, Khodos I, de Stanchina E, Reis-Filho JS, Ouerfelli O, Linder S, Djaballah H, Ladanyi M. Proteasome Addiction Defined in Ewing Sarcoma Is Effectively Targeted by a Novel Class of 19S Proteasome Inhibitors. Cancer Res 2016; 76:4525-34. [PMID: 27256563 DOI: 10.1158/0008-5472.can-16-1040] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 05/09/2016] [Indexed: 01/05/2023]
Abstract
Ewing sarcoma is a primitive round cell sarcoma with a peak incidence in adolescence that is driven by a chimeric oncogene created from the fusion of the EWSR1 gene with a member of the ETS family of genes. Patients with metastatic and recurrent disease have dismal outcomes and need better therapeutic options. We screened a library of 309,989 chemical compounds for growth inhibition of Ewing sarcoma cells to provide the basis for the development of novel therapies and to discover vulnerable pathways that might broaden our understanding of the pathobiology of this aggressive sarcoma. This screening campaign identified a class of benzyl-4-piperidone compounds that selectively inhibit the growth of Ewing sarcoma cell lines by inducing apoptosis. These agents disrupt 19S proteasome function through inhibition of the deubiquitinating enzymes USP14 and UCHL5. Functional genomic data from a genome-wide shRNA screen in Ewing sarcoma cells also identified the proteasome as a node of vulnerability in Ewing sarcoma cells, providing orthologous confirmation of the chemical screen findings. Furthermore, shRNA-mediated silencing of USP14 or UCHL5 in Ewing sarcoma cells produced significant growth inhibition. Finally, treatment of a xenograft mouse model of Ewing sarcoma with VLX1570, a benzyl-4-piperidone compound derivative currently in clinical trials for relapsed multiple myeloma, significantly inhibited in vivo tumor growth. Overall, our results offer a preclinical proof of concept for the use of 19S proteasome inhibitors as a novel therapeutic strategy for Ewing sarcoma. Cancer Res; 76(15); 4525-34. ©2016 AACR.
Collapse
Affiliation(s)
- Neerav Shukla
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Romel Somwar
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Roger S Smith
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sri Ambati
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Stanley Munoz
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Melinda Merchant
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Padraig D'Arcy
- Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Xin Wang
- Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Rachel Kobos
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Christophe Antczak
- High-Throughput Drug Screening Facility, Memorial Sloan Kettering Cancer Center, New YorkNew York
| | - Bhavneet Bhinder
- High-Throughput Drug Screening Facility, Memorial Sloan Kettering Cancer Center, New YorkNew York
| | - David Shum
- High-Throughput Drug Screening Facility, Memorial Sloan Kettering Cancer Center, New YorkNew York
| | - Constantin Radu
- High-Throughput Drug Screening Facility, Memorial Sloan Kettering Cancer Center, New YorkNew York
| | - Guangbin Yang
- Organic Synthesis Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Barry S Taylor
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York. Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Charlotte K Y Ng
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Inna Khodos
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elisa de Stanchina
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ouathek Ouerfelli
- Organic Synthesis Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Stig Linder
- Department of Medical and Health Sciences, Linköping University, Linköping, Sweden. Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden
| | - Hakim Djaballah
- High-Throughput Drug Screening Facility, Memorial Sloan Kettering Cancer Center, New YorkNew York
| | - Marc Ladanyi
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
7
|
Huseby NE, Ravuri C, Moens U. The proteasome inhibitor lactacystin enhances GSH synthesis capacity by increased expression of antioxidant components in an Nrf2-independent, but p38 MAPK-dependent manner in rat colorectal carcinoma cells. Free Radic Res 2015; 50:1-13. [PMID: 26530909 DOI: 10.3109/10715762.2015.1100730] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Proteasome inhibitors may induce ER stress and oxidative stress, disrupt signaling pathways, and trigger apoptosis in several cancer cells. However, they are also reported to increase glutathione (GSH) synthesis and protect cells from oxidative stress. In the present study, we showed that the proteasome inhibitor lactacystin increased reactive oxygen species (ROS) and GSH levels after the treatment of HT-29 colorectal cancer cells. The increased GSH depended upon the activity of glutamate cysteine ligase (GCL), uptake of cystine/cysteine via the cystine/glutamate transporter [Formula: see text], and the activity of γ-glutamyltransferase (GGT). Increased transcription levels of the catalytic subunit of glutamate cysteine ligase (GCLC), the catalytic subunit xCT of [Formula: see text], and GGT were induced by lactacystin, although with different kinetics and stoichiometry. Lactacystin treatment also augmented protein levels of GCLC, xCT, and GGT, but significant levels were not detected until 48 h after initiation of lactacystin treatment. These increases in protein levels were dependent on the p38 MAPK pathway. Studies in cells transfected with siRNA against the transcription factor Nrf2 demonstrated that the promoter activities of xCT and GCLC, but not of GGT, depended on Nrf2. However, depletion of Nrf2 had no effect on lactacystin-induced upregulation of the GGT, GCLC, and xCT mRNA levels. Taken together, our results suggest that oxidative stress provoked by proteasomal inhibition results in the elevation of cellular GSH levels due to increased synthesis of GSH and uptake of cystine/cysteine. Following treatment with lactacystin, enhanced expression of antioxidant components involved in GSH homeostasis is p38 MAPK-dependent, but Nrf2-independent, resulting in increased GSH synthesis capacity.
Collapse
Affiliation(s)
- Nils-Erik Huseby
- a Tumor Biology Research Group, Department of Medical Biology, Faculty of Health Sciences , University of Tromsø , Tromsø , Norway
| | - Chandra Ravuri
- a Tumor Biology Research Group, Department of Medical Biology, Faculty of Health Sciences , University of Tromsø , Tromsø , Norway
| | - Ugo Moens
- b Molecular Inflammation Research Group, Department of Medical Biology, Faculty of Health Sciences , University of Tromsø , Tromsø , Norway
| |
Collapse
|
8
|
Ambegaokar SS, Kolson DL. Heme oxygenase-1 dysregulation in the brain: implications for HIV-associated neurocognitive disorders. Curr HIV Res 2015; 12:174-88. [PMID: 24862327 PMCID: PMC4155834 DOI: 10.2174/1570162x12666140526122709] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Revised: 01/20/2014] [Accepted: 01/27/2014] [Indexed: 12/17/2022]
Abstract
Heme oxygenase-1 (HO-1) is a highly inducible and ubiquitous cellular enzyme that subserves cytoprotective responses to toxic insults, including inflammation and oxidative stress. In neurodegenerative diseases such as Alzheimer’s disease, Parkinson’s disease and multiple sclerosis, HO-1 expression is increased, presumably reflecting an endogenous neuroprotective response against ongoing cellular injury. In contrast, we have found that in human immunodeficiency virus (HIV) infection of the brain, which is also associated with inflammation, oxidative stress and neurodegeneration, HO-1 expression is decreased, likely reflecting a unique role for HO-1 deficiency in neurodegeneration pathways activated by HIV infection. We have also shown that HO-1 expression is significantly suppressed by HIV replication in cultured macrophages which represent the primary cellular reservoir for HIV in the brain. HO-1 deficiency is associated with release of neurotoxic levels of glutamate from both HIV-infected and immune-activated macrophages; this glutamate-mediated neurotoxicity is suppressed by pharmacological induction of HO-1 expression in the macrophages. Thus, HO-1 induction could be a therapeutic strategy for neuroprotection against HIV infection and other neuroinflammatory brain diseases. Here, we review various stimuli and signaling pathways regulating HO-1 expression in macrophages, which could promote neuronal survival through HO-1-modulation of endogenous antioxidant and immune modulatory pathways, thus limiting the oxidative stress that can promote HIV disease progression in the CNS. The use of pharmacological inducers of endogenous HO-1 expression as potential adjunctive neuroprotective therapeutics in HIV infection is also discussed.
Collapse
Affiliation(s)
| | - Dennis L Kolson
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 280 Clinical Research Building, 415 Curie Blvd., Philadelphia, PA 19104, USA.
| |
Collapse
|
9
|
Music E, Khan S, Khamis I, Heikkila JJ. Accumulation of heme oxygenase-1 (HSP32) in Xenopus laevis A6 kidney epithelial cells treated with sodium arsenite, cadmium chloride or proteasomal inhibitors. Comp Biochem Physiol C Toxicol Pharmacol 2014; 166:75-87. [PMID: 25064141 DOI: 10.1016/j.cbpc.2014.07.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 07/09/2014] [Accepted: 07/17/2014] [Indexed: 12/12/2022]
Abstract
The present study examined the effect of sodium arsenite, cadmium chloride, heat shock and the proteasomal inhibitors MG132, withaferin A and celastrol on heme oxygenase-1 (HO-1; also known as HSP32) accumulation in Xenopus laevis A6 kidney epithelial cells. Immunoblot analysis revealed that HO-1 accumulation was not induced by heat shock but was enhanced by sodium arsenite and cadmium chloride in a dose- and time-dependent fashion. Immunocytochemistry revealed that these metals induced HO-1 accumulation in a granular pattern primarily in the cytoplasm. Additionally, in 20% of the cells arsenite induced the formation of large HO-1-containing perinuclear structures. In cells recovering from sodium arsenite or cadmium chloride treatment, HO-1 accumulation initially increased to a maximum at 12h followed by a 50% reduction at 48 h. This initial increase in HO-1 levels was likely the result of new synthesis as it was inhibited by cycloheximide. Interestingly, treatment of cells with a mild heat shock enhanced HO-1 accumulation induced by low concentrations of sodium arsenite and cadmium chloride. Finally, we determined that HO-1 accumulation was induced in A6 cells by the proteasomal inhibitors, MG132, withaferin A and celastrol. An examination of heavy metal and proteasomal inhibitor-induced HO-1 accumulation in amphibians is of importance given the presence of toxic heavy metals in aquatic habitats.
Collapse
Affiliation(s)
- Ena Music
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | - Saad Khan
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | - Imran Khamis
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | - John J Heikkila
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada.
| |
Collapse
|
10
|
Skalniak L, Koj A, Jura J. Proteasome inhibitor MG-132 induces MCPIP1 expression. FEBS J 2013; 280:2665-74. [PMID: 23551903 PMCID: PMC3806276 DOI: 10.1111/febs.12264] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Revised: 03/19/2013] [Accepted: 03/22/2013] [Indexed: 01/14/2023]
Abstract
The proteasome is a protein complex responsible for the degradation of polyubiquitin-tagged proteins. Besides the removal of target proteins, the proteasome also participates in the regulation of gene transcription in both proteolytic and non-proteolytic fashion. In this study the effect of proteasome inhibition on the basal expression of monocyte chemotactic protein-1 induced protein 1 (MCPIP1) was examined. Treatment of HepG2 or HeLa cells with proteasome inhibitor MG-132 resulted in a significant increase of MCPIP1 expression, both at mRNA and protein level. Interestingly, MG-132 did not alter MCPIP1 stability. Instead, the observed protein increase was blocked by actinomycin D, suggesting the involvement of de novo mRNA synthesis in the increase of MCPIP1 protein following MG-132 treatment. Using several inhibitors we determined the participation of extracellular-signal-regulated kinase 1/2 and p38 kinases in MCPIP1 upregulation by MG-132. Our findings show for the first time the impact of proteasome inhibition on MCPIP1 protein expression by modulation of the activity of intracellular signaling pathways. Overexpression of MCPIP1-myc protein decreased the viability of HeLa cells but not HepG2 cells, which correlates with the increased susceptibility of HeLa cells to MG-132 toxicity. Notably, both MG-132 treatment and MCPIP1-myc overexpression led to the activation of apoptosis, as revealed by the induction of caspases 3/7 in both types of cell lines. This suggests the involvement of MCPIP1 upregulation in toxic properties of proteasome inhibition, which is an acknowledged approach to the treatment of several cancer types.
Collapse
Affiliation(s)
- Lukasz Skalniak
- Department of General Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
| | | | | |
Collapse
|
11
|
Kästle M, Woschee E, Grune T. Histone deacetylase 6 (HDAC6) plays a crucial role in p38MAPK-dependent induction of heme oxygenase-1 (HO-1) in response to proteasome inhibition. Free Radic Biol Med 2012; 53:2092-101. [PMID: 23010497 DOI: 10.1016/j.freeradbiomed.2012.09.023] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2012] [Revised: 08/30/2012] [Accepted: 09/17/2012] [Indexed: 12/26/2022]
Abstract
The proteasome is responsible for the degradation of polyubiquitinated proteins. Inhibition of the proteasome leads to an accumulation of polyubiquitinated proteins and thus to an impairment of the cellular protein homeostasis. To prevent cellular damage on proteasome inhibition there is an up-regulation of several heat shock proteins (Hsps), including Hsp27, Hsp70, and heme oxygenase-1 (HO-1). It was demonstrated that the induction of classical Hsps, such as Hsp27 and Hsp70, is dependent on a HDAC6-dependent mechanism which releases HSF-1 and induces the expression of newly synthesized Hsps. In this study we demonstrate that the up-regulation of HO-1 on proteasome inhibition is mediated by p38MAPK and Nrf-2. Interestingly we found additional evidence, proving the involvement of HDAC6 in the up-regulation of HO-1. By using RNAi technologies against HDAC6 we demonstrate that there is a lack of the expected induction of HO-1, Nrf-2, and phosphorylated p38 (pp38) after proteasome inhibition. Furthermore, we can show that p38 is acetylated in unstressed cells and is a good substrate for HDAC6-mediated deacetylation. Therefore, we propose that on proteasome inhibition HDAC6 deacetylates p38, allowing the subsequent phosphorylation of p38 and resultant activation of NRF-2. NRF-2 enters the nucleus and functions as a transcription factor for HO-1.
Collapse
Affiliation(s)
- Marc Kästle
- Department of Nutritional Toxicology, Institute of Nutrition, Friedrich-Schiller University Jena, Dornburger-Strasse 24, 07743 Jena, Germany
| | | | | |
Collapse
|
12
|
Gambogic acid is cytotoxic to cancer cells through inhibition of the ubiquitin-proteasome system. Invest New Drugs 2012. [DOI: 10.1007/s10637-012-9902-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
13
|
Alexander HA, Sooranna SR, Myatt L, Johnson MR. Myometrial tumor necrosis factor-α receptors increase with gestation and labor and modulate gene expression through mitogen-activated kinase and nuclear factor-κB. Reprod Sci 2012; 19:43-54. [PMID: 22228740 DOI: 10.1177/1933719111413297] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Previously, we found that myometrial tumor necrosis factor-α (TNF-α) messenger RNA (mRNA) expression did not increase with preterm or term labor. To further investigate the role of TNF-α in human labor, we studied TNF-α receptor (TNFR1A and B) expression, regulation, and associated intracellular signaling pathways in human myometrial samples obtained both before and after the onset of labor and in primary cultures of uterine smooth muscle cells (USMCs). We found that the mRNA expression of both receptors increased with advancing gestation and labor and protein levels of TNFR1B were significantly higher in term laboring myometrial samples than in nonlabor controls. Tumor necrosis factor- treatment of USMCs activated all mitogen-activated protein kinase (MAPK) subtypes and nuclear factor κ-B (NF-κB). The TNF-α induced increases in the expression of TNFR1B and prostaglandin H synthase type 2 were reduced by inhibitors of NF-κB and MAPKs, respectively. The TNF-α induced increase in interleukin 8 (IL-8) appeared to be independent of MAPK and NF-κB pathway. These data suggest that the uterus may become more sensitive to the action of TNF-α with advancing gestation and labor and that TNF-α acts via MAPK and NF-κB to promote labor-associated gene expression.
Collapse
Affiliation(s)
- Helen A Alexander
- Imperial College Parturition Research Group, Academic Department of Obstetrics & Gynaecology, Imperial College School of Medicine Chelsea and Westminster Hospital, London, UK
| | | | | | | |
Collapse
|
14
|
Han D, Spengler BA, Ross RA. Increased wild-type N-ras activation by neurofibromin down-regulation increases human neuroblastoma stem cell malignancy. Genes Cancer 2012; 2:1034-43. [PMID: 22737269 DOI: 10.1177/1947601912443127] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 02/20/2012] [Indexed: 12/30/2022] Open
Abstract
Cellular heterogeneity is a well-known feature of human neuroblastoma tumors and cell lines. Of the 3 phenotypes (N-, I-, and S-type) isolated and characterized, the I-type cancer stem cell of neuroblastoma is the most malignant. Here, we report that, although wild-type N-Ras protein is expressed at the same level in all 3 neuroblastoma cell phenotypes, activated N-Ras-GTP level is significantly higher in I-type cancer stem cells. When activated N-Ras levels were decreased by transfection of a dominant-negative N-Ras construct, the malignant potential of I-type cancer stem cells decreased significantly. Conversely, when weakly malignant N-type cells were transfected with a constitutively active N-Ras construct, activated N-Ras levels, and malignant potential, were significantly increased. Thus, high levels of N-Ras-GTP are required for the increased malignancy of I-type neuroblastoma cancer stem cells. Moreover, increased activation of N-Ras results from significant down-regulation of neurofibromin (NF1), an important RasGAP. This specific down-regulation is mediated by an ubiquitin-proteasome-dependent pathway. Thus, decreased expression of NF1 in I-type neuroblastoma cancer stem cells causes a high level of activated N-Ras that is, at least in part, responsible for their higher tumorigenic potential.
Collapse
Affiliation(s)
- Dan Han
- Department of Biological Sciences, Fordham University, Bronx, NY, USA
| | | | | |
Collapse
|
15
|
Wu WT. Evaluation of anti-inflammatory effects of Broussonetia papyrifera stem bark. Indian J Pharmacol 2012; 44:26-30. [PMID: 22345865 PMCID: PMC3271534 DOI: 10.4103/0253-7613.91862] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Revised: 07/06/2011] [Accepted: 10/18/2011] [Indexed: 11/09/2022] Open
Abstract
Objective: Broussonetia papyrifera is used as a traditional medicine to treat few diseases. However, the antiinflammatory effect of B. papyrifera stem bark has not been evaluated. The aim of this study is to investigate the effects of n-hexane fraction from methanol extract of B. papyrifera stem bark on lipopolysaccharide (LPS)-stimulated inflammation using RAW 264.7 cells. Materials and Methods: Methanol extract was obtained from B. papyrifera stem bark and its sequential fractions (hexane, dichloromathane, ethyl acetate, butanol, and aqueous) were obtained by extraction in solvents with increasing polarity and were examined in RAW 264.7 cells. Results: The secretion profiles of pro-inflammatory parameters, including nitric oxide (NO), tumor necrosis factor-α (TNF-α), and interleukin-1β (IL-1β) were found to be significantly reduced in 10-80 μg/ml dose ranges of n-hexane fraction (BP-H) from methanol extract of B. papyrifera stem bark. The expressions of inducible NO synthase (iNOS) was also significantly inhibited by BP-H. Reverse transcription-polymerase chain reaction (RT-PCR) analysis showed that BP-H treatment decreased LPS-induced iNOS mRNA expression in RAW 264.7 cells. Conclusion: The results suggest that the B. papyrifera stem bark has anti-inflammatory activity which inhibits the NO production and proinflammatory cytokines in RAW 264.7 cells. B. papyrifera stem bark might act as a potential therapeutic agent for inflammatory diseases.
Collapse
Affiliation(s)
- Wen-Tung Wu
- Department of Biotechnology, Yung-Ta Institute of Technology and Commerce, Pingtung, Taiwan.
| |
Collapse
|
16
|
Prchal-Murphy M, Semper C, Lassnig C, Wallner B, Gausterer C, Teppner-Klymiuk I, Kobolak J, Müller S, Kolbe T, Karaghiosoff M, Dinnyés A, Rülicke T, Leitner NR, Strobl B, Müller M. TYK2 kinase activity is required for functional type I interferon responses in vivo. PLoS One 2012; 7:e39141. [PMID: 22723949 PMCID: PMC3377589 DOI: 10.1371/journal.pone.0039141] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 05/20/2012] [Indexed: 01/25/2023] Open
Abstract
Tyrosine kinase 2 (TYK2) is a member of the Janus kinase (JAK) family and is involved in cytokine signalling. In vitro analyses suggest that TYK2 also has kinase-independent, i.e., non-canonical, functions. We have generated gene-targeted mice harbouring a mutation in the ATP-binding pocket of the kinase domain. The Tyk2 kinase-inactive (Tyk2K923E) mice are viable and show no gross abnormalities. We show that kinase-active TYK2 is required for full-fledged type I interferon- (IFN) induced activation of the transcription factors STAT1-4 and for the in vivo antiviral defence against viruses primarily controlled through type I IFN actions. In addition, TYK2 kinase activity was found to be required for the protein’s stability. An inhibitory function was only observed upon over-expression of TYK2K923Ein vitro. Tyk2K923E mice represent the first model for studying the kinase-independent function of a JAK in vivo and for assessing the consequences of side effects of JAK inhibitors.
Collapse
Affiliation(s)
- Michaela Prchal-Murphy
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Vienna, Austria
| | - Christian Semper
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Vienna, Austria
| | - Caroline Lassnig
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Vienna, Austria
- Biomodels Austria, University of Veterinary Medicine, Vienna, Austria
| | - Barbara Wallner
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Vienna, Austria
| | - Christian Gausterer
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Vienna, Austria
| | | | - Julianna Kobolak
- Genetic Reprogramming Group Agricultural Biotechnology Center, Gödöllö, Hungary
| | - Simone Müller
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Vienna, Austria
| | - Thomas Kolbe
- Biomodels Austria, University of Veterinary Medicine, Vienna, Austria
- Department for Agrobiotechnology IFA Tulln, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Marina Karaghiosoff
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Vienna, Austria
| | - Andras Dinnyés
- Genetic Reprogramming Group Agricultural Biotechnology Center, Gödöllö, Hungary
- Molecular Animal Biotechnology Laboratory, Szent Istvan University, Gödöllö, Hungary
- BioTalentum Ltd., Gödöllö, Hungary
| | - Thomas Rülicke
- Biomodels Austria, University of Veterinary Medicine, Vienna, Austria
- Institute of Laboratory Animal Science, University of Veterinary Medicine, Vienna, Austria
| | - Nicole R. Leitner
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Vienna, Austria
| | - Birgit Strobl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Vienna, Austria
| | - Mathias Müller
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Vienna, Austria
- Biomodels Austria, University of Veterinary Medicine, Vienna, Austria
- * E-mail:
| |
Collapse
|
17
|
Kästle M, Grune T. Interactions of the Proteasomal System with Chaperones. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2012; 109:113-60. [DOI: 10.1016/b978-0-12-397863-9.00004-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
18
|
An IκBα phosphorylation inhibitor induces heme oxygenase-1(HO-1) expression through the activation of reactive oxygen species (ROS)–Nrf2–ARE signaling and ROS–PI3K/Akt signaling in an NF-κB-independent mechanism. Cell Signal 2011; 23:1505-13. [DOI: 10.1016/j.cellsig.2011.05.013] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Accepted: 05/12/2011] [Indexed: 12/22/2022]
|
19
|
Miller CP, Manton CA, Hale R, Debose L, Macherla VR, Potts BC, Palladino MA, Chandra J. Specific and prolonged proteasome inhibition dictates apoptosis induction by marizomib and its analogs. Chem Biol Interact 2011; 194:58-68. [PMID: 21864512 DOI: 10.1016/j.cbi.2011.08.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Revised: 07/30/2011] [Accepted: 08/08/2011] [Indexed: 01/25/2023]
Abstract
Marizomib (NPI-0052) is a naturally derived irreversible proteasome inhibitor that potently induces apoptosis via a caspase-8 and ROS-dependent mechanism in leukemia cells. We aim to understand the relationship between the irreversible inhibition of the proteasome and induction of cell death in leukemia cells by using analogs of marizomib that display reversible and irreversible properties. We highlight the importance of sustained inhibition of at least two proteasome activities as being key permissive events for the induction of the apoptotic process in leukemia cells. These data provide the basis for the development of new approaches to generate more effective anti-proteasome therapies.
Collapse
Affiliation(s)
- Claudia P Miller
- Department of Pediatrics Research, Children's Cancer Hospital at M.D. Anderson, University of Texas M.D. Anderson Cancer Center, Houston, United States
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Kaitsuka T, Tomizawa K, Matsushita M. Transformation of eEF1Bδ into heat-shock response transcription factor by alternative splicing. EMBO Rep 2011; 12:673-81. [PMID: 21597468 DOI: 10.1038/embor.2011.82] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2010] [Revised: 03/13/2011] [Accepted: 04/07/2011] [Indexed: 12/24/2022] Open
Abstract
Protein translation factors have crucial roles in a variety of stress responses. Here, we show that eukaryotic elongation factor 1Bδ (eEF1Bδ) changes its structure and function from a translation factor into a heat-shock response transcription factor by alternative splicing. The long isoform of eEF1Bδ (eEF1BδL) is localized in the nucleus and induces heat-shock element (HSE)-containing genes in cooperation with heat-shock transcription factor 1 (HSF1). Moreover, the amino-terminal domain of eEF1BδL binds to NF-E2-related factor 2 (Nrf2) and induces stress response haem oxygenase 1 (HO1). Specific inhibition of eEF1BδL with small-interfering RNA completely inhibits Nrf2-dependent HO1 induction. In addition, eEF1BδL directly binds to HSE oligo DNA in vitro and associates with the HSE consensus in the HO1 promoter region in vivo. Thus, the transcriptional role of eEF1BδL could provide new insights into the molecular mechanism of stress responses.
Collapse
Affiliation(s)
- Taku Kaitsuka
- Mitsubishi Kagaku Institute of Life Sciences, 11 Minami-ooya, Machida, Tokyo 194-8511, Japan; Department of Molecular Physiology, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | | | | |
Collapse
|
21
|
Abstract
AIM Proteasome inhibitors have been found to suppress glioma cell proliferation and induce apoptosis, but the mechanisms are not fully elucidated. In this study we investigated the mechanisms underlying the apoptosis induced by the proteasome inhibitor MG-132 in glioma cells. METHODS C6 glioma cells were used. MTT assay was used to analyze cell proliferation. Proteasome activity was assayed using Succinyl-LLVY-AMC, and intracellular ROS level was evaluated with the redox-sensitive dye DCFH-DA. Apoptosis was detected using fluorescence and transmission electron microscopy as well as flow cytometry. The expression of apoptosis-related proteins was investigated using Western blot analysis. RESULTS MG-132 inhibited C6 glioma cell proliferation in a time- and dose-dependent manner (the IC(50) value at 24 h was 18.5 μmol/L). MG-132 (18.5 μmol/L) suppressed the proteasome activity by about 70% at 3 h. It induced apoptosis via down-regulation of antiapoptotic proteins Bcl-2 and XIAP, up-regulation of pro-apoptotic protein Bax and caspase-3, and production of cleaved C-terminal 85 kDa PARP). It also caused a more than 5-fold increase of reactive oxygen species. Tiron (1 mmol/L) effectively blocked oxidative stress induced by MG-132 (18.5 μmol/L), attenuated proliferation inhibition and apoptosis in C6 glioma cells, and reversed the expression pattern of apoptosis-related proteins. CONCLUSION MG-132 induced apoptosis of C6 glioma cells via the oxidative stress.
Collapse
|
22
|
Yerlikaya A, Dokudur H. Investigation of the eIF2α phosphorylation mechanism in response to proteasome inhibition in melanoma and breast cancer cells. Mol Biol 2010. [DOI: 10.1134/s0026893310050122] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
23
|
Yamamoto N, Izumi Y, Matsuo T, Wakita S, Kume T, Takada-Takatori Y, Sawada H, Akaike A. Elevation of heme oxygenase-1 by proteasome inhibition affords dopaminergic neuroprotection. J Neurosci Res 2010; 88:1934-42. [PMID: 20155807 DOI: 10.1002/jnr.22363] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Postmortem studies have shown that heme oxygenase-1 (HO-1) immunoreactivity is increased in patients with Parkinson disease. HO-1 expression is highly upregulated by a variety of stress. Since the proteasome activity is decreased in patients with Parkinson disease, we investigated whether proteasome activity regulates HO-1 content. MG-132, a proteasome inhibitor, increased the amount of HO-1 protein mainly in astrocytes of primary mesencephalic cultures. Quantitative RT-PCR analysis revealed that lactacystin upregulated HO-1 mRNA expression. Proteasome inhibition with MG132 also increased the cytomegalovirus promoter-driven expression of Flag-HO-1 protein and resulted in an accumulation of ubiquitinated Flag-HO-1 in Flag-HO-1-overexpressing PC12 cells. In addition, a cycloheximide chase assay demonstrated that the degradation of Flag-HO-1 protein was slowed by MG-132. Next, the function of HO-1 which was upregulated by proteasome inhibitors was examined. Proteasome inhibitors protected dopaminergic neurons from 6-hydroxydopamine (6-OHDA)-induced toxicity and this neuroprotection was abrogated by co-treatment with zinc protoporphyrin IX, a HO-1 inhibitor. Furthermore, 6-OHDA-induced toxicity was blocked by bilirubin and carbon monoxide, products of the HO-1-catalyzed degradation of heme. These results suggest that mesencephalic HO-1 protein level is regulated by proteasome activity and the elevation by proteasome inhibition affords neuroprotection.
Collapse
Affiliation(s)
- Noriyuki Yamamoto
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Ebert B, Kisiela M, Malátková P, El-Hawari Y, Maser E. Regulation of human carbonyl reductase 3 (CBR3; SDR21C2) expression by Nrf2 in cultured cancer cells. Biochemistry 2010; 49:8499-511. [PMID: 20806931 DOI: 10.1021/bi100814d] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Carbonyl reduction is a central metabolic process that controls the level of key regulatory molecules as well as xenobiotics. Carbonyl reductase 3 (CBR3; SDR21C2), a member of the short-chain dehydrogenase/reductase (SDR) superfamily, has been poorly characterized so far, and the regulation of its expression is a complete mystery. Here, we show that CBR3 expression is regulated via Nrf2, a key regulator in response to oxidative stress. In human cancer cell lines, CBR3 mRNA was expressed differentially, ranging from very high (A549, lung) to very low (HT-29, colon; HepG2, liver) levels. CBR3 protein was highly expressed in SW-480 (colon) cells but was absent in HCT116 (colon) and HepG2 cells. CBR3 mRNA could be induced in HT-29 cells by Nrf2 agonists [sulforaphane (SUL, 7-fold) and diethyl maleate (DEM, 4-fold)] or hormone receptor ligand Z-guggulsterone (5-fold). Aryl hydrocarbon receptor agonist B[k]F failed to induce CBR3 mRNA after incubation for 8 h but elevated CBR3 levels after 24 h, most likely mediated by B[k]F metabolites that can activate Nrf2 signaling. Inhibition of Nrf2-activating upstream kinase MEK/ERK by PD98059 weakened DEM-mediated induction of CBR3 mRNA. Proteasome inhibitors MG-132 (5 μM) and bortezomib (50 nM) dramatically increased the level of CBR3 mRNA, obviously because of the increase in the level of Nrf2 protein. While siRNA-mediated knockdown of Nrf2 led to a decrease in the level of CBR3 mRNA in A549 cells (30% of control), Keap1 knockdown increased the level of CBR3 mRNA expression in HepG2 (9.3-fold) and HT-29 (2.7-fold) cells. Here, we provide for the first time evidence that human CBR3 is a new member of the Nrf2 gene battery.
Collapse
Affiliation(s)
- Bettina Ebert
- Institute of Toxicology and Pharmacology for Natural Scientists, University Medical School Schleswig-Holstein, Kiel, Germany
| | | | | | | | | |
Collapse
|
25
|
Liu HT, Li WM, Li XY, Xu QS, Liu QS, Bai XF, Yu C, Du YG. Chitosan oligosaccharides inhibit the expression of interleukin-6 in lipopolysaccharide-induced human umbilical vein endothelial cells through p38 and ERK1/2 protein kinases. Basic Clin Pharmacol Toxicol 2009; 106:362-71. [PMID: 19929981 DOI: 10.1111/j.1742-7843.2009.00493.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Chitosan oligosaccharides (COS) have been reported to exert anti-fungal activities, antitumour activities and immuno-enhancing effects. However, the potential roles of COS in the treatment of vascular inflammations remain unknown. In the present study, we examined the effects of COS on interleukin-6 (IL-6) production in human umbilical vein endothelial cells (HUVECs) induced by lipopolysaccharide (LPS). Induction of HUVECs with LPS (100 ng/ml) increased the mRNA expression and protein secretion of IL-6 (versus the vehicle-treated group, p < 0.01), which were significantly reverted by the pre-treatment with COS (50-200 microg/ml) for 24 hr before LPS exposure (versus the LPS-treated group, p < 0.05 or 0.01). Signal transduction studies showed that the pre-treatment of HUVECs with COS (50-200 microg/ml) for 24 hr markedly inhibited the LPS-induced over-expression of phosphorylated p38 mitogen-activated protein kinase (MAPK), phosphorylated ERK1/2 and nuclear factor kappaB (NF-kappaB). Moreover, the LPS-induced NF-kappaB activation was suppressed by the specific ERK1/2 inhibitor PD98059 (30 microM) (versus the LPS-treated group, p < 0.01), but not by the specific p38 MAPK inhibitor SB203580 (25 microM). Additionally, both MAPK inhibitors markedly suppressed LPS-induced IL-6 mRNA expression in HUVECs (versus the LPS-treated group, p < 0.01). In conclusion, our results suggest that COS inhibit LPS-induced up-regulation of IL-6 in HUVECs, and this can be regulated by at least two parallel signalling pathways: one via p38 MAPK pathway independent of NF-kappaB activation and one via ERK1/2 pathway dependent on NF-kappaB activation.
Collapse
Affiliation(s)
- Hong-Tao Liu
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Rottlerin induces heme oxygenase-1 (HO-1) up-regulation through reactive oxygen species (ROS) dependent and PKC delta-independent pathway in human colon cancer HT29 cells. Biochimie 2009; 92:110-5. [PMID: 19833168 DOI: 10.1016/j.biochi.2009.10.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2009] [Accepted: 10/04/2009] [Indexed: 01/18/2023]
Abstract
Heme oxygenase-1 (HO-1) is a cytoprotective enzyme activated by its substrate heme and diverse stimuli. The induction of HO-1 gene expression is one of the important events in cellular response to pro-oxidative and pro-inflammatory insults. In this study, the effect of rottlerin, a putative PKC delta inhibitor, on HO-1 expression in HT29 human colon cancer cells was investigated. Rottlerin-induced HO-1 at both protein and mRNA levels in a dose- and time-dependent manner. Rottlerin-mediated HO-1 induction was abrogated in the presence of N-acetylcysteine (NAC) or glutathione (GSH). Rottlerin induced nuclear translocation of NF-E2-related factor 2 (Nrf2) and increased antioxidant response element (ARE)-driven transcriptional activity. Additionally, rottlerin activated p38 mitogen-activated protein kinase (MAPK) and ERK. The pharmacological inhibition of ERK and p38 MAPK inhibited rottlerin-induced HO-1 up-regulation. However, suppression of protein kinase C delta (PKC delta) expression by siRNA or overexpression of WT-PKC delta did not abrogate the rottlerin-mediated induction of HO-1. These results suggest that rottlerin induces up-regulation of HO-1 via PKC delta-independent pathway. Taken together, the present study identified rottlerin as a novel inducer of HO-1 expression and identified the mechanisms involved in this process.
Collapse
|
27
|
Du ZX, Zhang HY, Meng X, Guan Y, Wang HQ. Role of oxidative stress and intracellular glutathione in the sensitivity to apoptosis induced by proteasome inhibitor in thyroid cancer cells. BMC Cancer 2009; 9:56. [PMID: 19216805 PMCID: PMC2666756 DOI: 10.1186/1471-2407-9-56] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2008] [Accepted: 02/16/2009] [Indexed: 01/01/2023] Open
Abstract
Background The proteasome inhibitor bortezomib has shown impressive clinical activity alone and in combination with conventional and other novel agents for the treatment of multiple myeloma (MM) and some solid cancers. Although bortezomib is known to be a selective proteasome inhibitor, the downstream mechanisms of cytotoxicity and drug resistance are poorly understood. Methods Proteasome activity, intracellular glutathione (GSH) and ROS levels, as well as activities of GSH synthesis enzymes were measured using spectrophotometric methods. Cell death was analyzed using flow cytometry and caspase activity assay. The expression level of GSH synthesis enzymes were measured using real-time RT-PCR. Results At concentrations that effectively inhibited proteasome activity, bortezomib induced apoptosis in FRO cells, but not in ARO cells. Bortezomib elevated the amount of glutathione (GSH) and the treatment with bortezomib increased the level of mRNA for GCL, a rate-limiting enzyme in glutathione synthesis. Furthermore, depletion of GSH increases apoptosis induced by bortezomib, in contrast, repletion of GSH decreases bortezomib-mediated cell death. Conclusion GSH protects cells from proteasome inhibition-induced oxidative stress and glutathione-dependent redox system might play an important role in the sensitivity to proteasome inhibition-induced apoptosis.
Collapse
Affiliation(s)
- Zhen-Xian Du
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang, PR China.
| | | | | | | | | |
Collapse
|
28
|
Singal M, Finkelstein JN. Use of Indicator Cell Lines for Determining Inflammatory Gene Changes and Screening the Inflammatory Potential of Particulate and Non-Particulate Stimuli. Inhal Toxicol 2008; 17:415-25. [PMID: 16020038 DOI: 10.1080/08958370591002021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Ultrafine particulate matter, from environmental or industrial exposure, can induce the expression of inflammatory mediators and promote the production of reactive oxygen species (ROS), which can damage the alveolar epithelium of the lung. Previous studies have shown that various cellular stresses can activate signaling pathways that operate through the specific transcription factors (TF), AP-1, and nuclear factor (NF)-kappaB that are known to regulate inflammatory gene expression. Persistent inflammation can induce a cascade of events that precedes the development of both acute and chronic fibrosis. From a murine Type II epithelial cell line, MLE15, a stable luciferase-transfected line, MLE15Luc2, was created. The luciferase reporter, operating under the guidance of a truncated human interleukin (IL)-8 promoter, contains NF-kappaB and AP-1 DNA binding sites. MLE15Luc2 cells were exposed to inflammatory or particulate stimuli, of varying size fractions and composition, under standard culture conditions, and inflammatory gene transcription, represented by luciferase enzyme activity, was determined. IkappaBalpha degradation appeared to be incongruent to changes in luciferase activity. The results were compared to those obtained using a stable luciferase-transfected human cell line, A549Luc1. Time-course data demonstrated increased luciferase enzyme activity, peaking by 6 h postexposure, and returning to baseline by 24 h, regardless of stimulus, in the absence of enhanced cytotoxicity. This suggests that key regulatory functions in these transfected cell lines are not clearly understood. These transfected cell lines may be useful for determining the inflammatory potential of various types of particulate and/or non-particulate stimuli; however, conclusive signaling information cannot be gained from their use alone.
Collapse
Affiliation(s)
- Madhuri Singal
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester, Rochester, New York 14642, USA
| | | |
Collapse
|
29
|
Rajan S, Ye J, Bai S, Huang F, Guo YL. NF-kappaB, but not p38 MAP kinase, is required for TNF-alpha-induced expression of cell adhesion molecules in endothelial cells. J Cell Biochem 2008; 105:477-86. [PMID: 18613029 PMCID: PMC4422387 DOI: 10.1002/jcb.21845] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In response to inflammation stimuli, tumor necrosis factor-alpha (TNF-alpha) induces expression of cell adhesion molecules (CAMs) in endothelial cells (ECs). Studies have suggested that the nuclear factor-kappaB (NF-kappaB) and the p38 MAP kinase (p38) signaling pathways play central roles in this process, but conflicting results have been reported. The objective of this study is to determine the relative contributions of the two pathways to the effect of TNF-alpha. Our initial data indicated that blockade of p38 activity by chemical inhibitor SB203580 (SB) at 10 microM moderately inhibited TNF-alpha-induced expression of three types of CAMs; ICAM-1, VCAM-1 and E-selectin, indicating that p38 may be involved in the process. However, subsequent analysis revealed that neither 1 microM SB that could completely inhibit p38 nor specific knockdown of p38alpha and p38beta with small interference RNA (siRNA) had an apparent effect, indicating that p38 activity is not essential for TNF-alpha-induced CAMs. The most definitive evidence to support this conclusion was from the experiments using cells differentiated from p38alpha knockout embryonic stem cells. We could show that deletion of p38alpha gene did not affect TNF-alpha-induced ICAM-1 and VCAM-1 expression when compared with wild-type cells. We further demonstrated that inhibition of NF-kappaB completely blocked TNF-alpha-induced expression of ICAM-1, VCAM-1 and E-selectin. Taken together, our results clearly demonstrate that NF-kappaB, but not p38, is critical for TNF-alpha-induced CAM expression. The inhibition of SB at 10 microM on TNF-alpha-induced ICAM-1, VCAM-1 and E-selectin is likely due to the nonspecific effect of SB.
Collapse
Affiliation(s)
- Suja Rajan
- Department of Biological Sciences, The University of Southern Mississippi, Hattiesburg, MS 39406
| | - Jianming Ye
- Department of Biological Sciences, The University of Southern Mississippi, Hattiesburg, MS 39406
| | - Shanshan Bai
- Department of Biological Sciences, The University of Southern Mississippi, Hattiesburg, MS 39406
| | - Faqing Huang
- Department of Chemistry and Biochemistry, The University of Southern Mississippi, Hattiesburg, MS 39406
| | - Yan-Lin Guo
- Department of Biological Sciences, The University of Southern Mississippi, Hattiesburg, MS 39406
| |
Collapse
|
30
|
Naidu S, Wijayanti N, Santoso S, Kietzmann T, Immenschuh S. An Atypical NF-κB-Regulated Pathway Mediates Phorbol Ester-Dependent Heme Oxygenase-1 Gene Activation in Monocytes. THE JOURNAL OF IMMUNOLOGY 2008; 181:4113-23. [DOI: 10.4049/jimmunol.181.6.4113] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
31
|
Fernandes AF, Zhou J, Zhang X, Bian Q, Sparrow J, Taylor A, Pereira P, Shang F. Oxidative inactivation of the proteasome in retinal pigment epithelial cells. A potential link between oxidative stress and up-regulation of interleukin-8. J Biol Chem 2008; 283:20745-53. [PMID: 18502748 PMCID: PMC2475710 DOI: 10.1074/jbc.m800268200] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2008] [Revised: 04/04/2008] [Indexed: 12/19/2022] Open
Abstract
Oxidative stress and inflammation are implicated in the pathogenesis of many age-related diseases. Stress-induced overproduction of inflammatory cytokines, such as interleukin-8 (IL-8), is one of the early events of inflammation. The objective of this study was to elucidate mechanistic links between oxidative stress and overproduction of IL-8 in retinal pigment epithelial (RPE) cells. We found that exposure of RPE cells to H(2)O(2), paraquat, or A2E-mediated photooxidation resulted in increased expression and secretion of IL-8. All of these oxidative stressors also inactivated the proteasome in RPE cells. In contrast, tert-butylhydroperoxide (TBH), a lipophilic oxidant that did not stimulate IL-8 production, also did not inactivate the proteasome. Moreover, prolonged treatment of RPE cells with proteasome-specific inhibitors recapitulated the stimulation of IL-8 production. These data suggest that oxidative inactivation of the proteasome is a potential mechanistic link between oxidative stress and up-regulation of the proinflammatory IL-8. The downstream signaling pathways that govern the production of IL-8 include NF-kappaB and p38 MAPK. Proteasome inhibition both attenuated the activation and delayed the turnoff of NF-kappaB, resulting in biphasic effects on the production of IL-8. Prolonged proteasome inhibition (>2 h) resulted in activation of p38 MAPK via activation of MKK3/6 and increased the production of IL-8. Chemically inhibiting the p38 MAPK blocked the proteasome inhibition-induced up-regulation of IL-8. Together, these data indicate that oxidative inactivation of the proteasome and the related activation of the p38 MAPK pathway provide a potential link between oxidative stress and overproduction of proinflammatory cytokines, such as IL-8.
Collapse
Affiliation(s)
- Alexandre F. Fernandes
- Jean Mayer United States Department of
Agriculture Human Nutrition Research Center on Aging, Tufts University,
Boston, Massachusetts 02111, the Center of
Ophthalmology, IBILI, Faculty of Medicine, University of Coimbra,
3000–354 Portugal, and the Department of
Ophthalmology, Columbia University, New York, New York 10032
| | - Jilin Zhou
- Jean Mayer United States Department of
Agriculture Human Nutrition Research Center on Aging, Tufts University,
Boston, Massachusetts 02111, the Center of
Ophthalmology, IBILI, Faculty of Medicine, University of Coimbra,
3000–354 Portugal, and the Department of
Ophthalmology, Columbia University, New York, New York 10032
| | - Xinyu Zhang
- Jean Mayer United States Department of
Agriculture Human Nutrition Research Center on Aging, Tufts University,
Boston, Massachusetts 02111, the Center of
Ophthalmology, IBILI, Faculty of Medicine, University of Coimbra,
3000–354 Portugal, and the Department of
Ophthalmology, Columbia University, New York, New York 10032
| | - Qingning Bian
- Jean Mayer United States Department of
Agriculture Human Nutrition Research Center on Aging, Tufts University,
Boston, Massachusetts 02111, the Center of
Ophthalmology, IBILI, Faculty of Medicine, University of Coimbra,
3000–354 Portugal, and the Department of
Ophthalmology, Columbia University, New York, New York 10032
| | - Janet Sparrow
- Jean Mayer United States Department of
Agriculture Human Nutrition Research Center on Aging, Tufts University,
Boston, Massachusetts 02111, the Center of
Ophthalmology, IBILI, Faculty of Medicine, University of Coimbra,
3000–354 Portugal, and the Department of
Ophthalmology, Columbia University, New York, New York 10032
| | - Allen Taylor
- Jean Mayer United States Department of
Agriculture Human Nutrition Research Center on Aging, Tufts University,
Boston, Massachusetts 02111, the Center of
Ophthalmology, IBILI, Faculty of Medicine, University of Coimbra,
3000–354 Portugal, and the Department of
Ophthalmology, Columbia University, New York, New York 10032
| | - Paulo Pereira
- Jean Mayer United States Department of
Agriculture Human Nutrition Research Center on Aging, Tufts University,
Boston, Massachusetts 02111, the Center of
Ophthalmology, IBILI, Faculty of Medicine, University of Coimbra,
3000–354 Portugal, and the Department of
Ophthalmology, Columbia University, New York, New York 10032
| | - Fu Shang
- Jean Mayer United States Department of
Agriculture Human Nutrition Research Center on Aging, Tufts University,
Boston, Massachusetts 02111, the Center of
Ophthalmology, IBILI, Faculty of Medicine, University of Coimbra,
3000–354 Portugal, and the Department of
Ophthalmology, Columbia University, New York, New York 10032
| |
Collapse
|
32
|
Derecka K, Sheldrick EL, Wathes DC, Abayasekara DRE, Flint APF. A PPAR-independent pathway to PUFA-induced COX-2 expression. Mol Cell Endocrinol 2008; 287:65-71. [PMID: 18395968 DOI: 10.1016/j.mce.2008.02.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2007] [Revised: 02/15/2008] [Accepted: 02/16/2008] [Indexed: 11/29/2022]
Abstract
Polyunsaturated fatty acids (PUFAs) induce COX-2 in bovine endometrial stromal cells through activation of peroxisome-proliferator-activated receptor alpha (PPARalpha). We have investigated alternative (PPAR-independent) pathways to COX-2 induction using a reporter construct driven by a COX-2 gene promoter sequence lacking a PPAR response element. This construct was induced by PUFAs, but not by PPAR agonists. PPAR-independent reporter gene expression occurred 6h after PPAR-dependent induction of the endogenous COX-2 gene. In contrast to PPAR-dependent COX-2 induction, which is not affected by NF-kappaB inhibitors, the PPAR-independent pathway was blocked by the NF-kappaB inhibitor MG132 or following deletion of NF-kappaB sites in the COX-2 promoter. The PPAR-independent effect of PUFA was mimicked by the PKC activators 4beta-PMA and prostaglandin F(2alpha), but was not blocked by the PKC inhibitor RO318425. The results demonstrate a pathway to the induction of COX-2 by PUFAs requiring NF-kappaB but not PPAR or PKC.
Collapse
Affiliation(s)
- K Derecka
- Division of Animal Physiology, University of Nottingham, Sutton Bonington Campus, Loughborough, Leics LE12 5RD, UK
| | | | | | | | | |
Collapse
|
33
|
Bianchetti CM, Yi L, Ragsdale SW, Phillips GN. Comparison of apo- and heme-bound crystal structures of a truncated human heme oxygenase-2. J Biol Chem 2007; 282:37624-31. [PMID: 17965015 PMCID: PMC2896506 DOI: 10.1074/jbc.m707396200] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Heme oxygenase (HO) catalyzes the first step in the heme degradation pathway. The crystal structures of apo- and heme-bound truncated human HO-2 reveal a primarily alpha-helical architecture similar to that of human HO-1 and other known HOs. Proper orientation of heme in HO-2 is required for the regioselective oxidation of the alpha-mesocarbon. This is accomplished by interactions within the heme binding pocket, which is made up of two helices. The iron coordinating residue, His(45), resides on the proximal helix. The distal helix contains highly conserved glycine residues that allow the helix to flex and interact with the bound heme. Tyr(154), Lys(199), and Arg(203) orient the heme through direct interactions with the heme propionates. The rearrangements of side chains in heme-bound HO-2 compared with apoHO-2 further elucidate HO-2 heme interactions.
Collapse
Affiliation(s)
- Christopher M. Bianchetti
- Graduate Program in Biophysics, University of Wisconsin, and the Madison, Wisconsin 53706
- Department of Biochemistry, University of Wisconsin, and the Madison, Wisconsin 53706
- Center for Eukaryotic Structural Genomics, University of Wisconsin, and the Madison, Wisconsin 53706
| | - Li Yi
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Stephen W. Ragsdale
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - George N. Phillips
- Department of Biochemistry, University of Wisconsin, and the Madison, Wisconsin 53706
- Center for Eukaryotic Structural Genomics, University of Wisconsin, and the Madison, Wisconsin 53706
| |
Collapse
|
34
|
Kolachala VL, Bajaj R, Wang L, Yan Y, Ritzenthaler JD, Gewirtz AT, Roman J, Merlin D, Sitaraman SV. Epithelial-derived fibronectin expression, signaling, and function in intestinal inflammation. J Biol Chem 2007; 282:32965-73. [PMID: 17855340 DOI: 10.1074/jbc.m704388200] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Fibronectin (FN) is a multifunctional extracellular matrix protein that plays an important role in cell proliferation, adhesion, and migration. FN expression or its role in colitis is not known. The goal of this study is to characterize FN expression, regulation, and role during intestinal inflammation. Wild-type and transgenic mice expressing luciferase under the control of the human FN promoter, given water or 3% dextran sodium sulfate, were used as animal models of colitis. The Caco2-BBE model intestinal epithelial cell line was used for in vitro studies. FN protein is abundantly expressed by surface epithelial cells in the normal colon. Immunohistochemistry and luciferase assay in mice expressing the FN promoter linked to luciferase demonstrated that FN synthesis was up-regulated during colitis, during both the acute phase and the healing phase. In vitro experiments demonstrated that FN increased the expression of the FN integrin receptor alpha5beta1 in a dose- and time-dependent manner. FN also induced the expression and activation of NF-kappaB. Further, FN potentiated Caco2-BBE cell attachment and wound healing, which was inhibited by RGD peptide as well as NF-kappaB inhibitors MG-132 and 1-pyrrolidinecarbodithioic acid, ammonium salt. In conclusion, FN is abundantly expressed and synthesized by colonic epithelial cells. FN is transcriptionally up-regulated in epithelial cells during both the dextran sodium sulfate-induced colitic and the recovery phase. FN enhances cell attachment and wound healing, which is dependent on binding to the integrin receptor and the NF-kappaB signaling. Together our data show that epithelial-derived FN potentiates cell attachment and wound healing through epithelial-matrix interactions and that FN expression may have important implications for maintaining normal epithelial integrity as well as regulating epithelial response to injury during colitis.
Collapse
Affiliation(s)
- Vasantha L Kolachala
- Division of Digestive Diseases, Department of Medicine, Emory University, 615 Michael Street, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Sheldrick E, Derecka K, Marshall E, Chin E, Hodges L, Wathes D, Abayasekara D, Flint A. Peroxisome-proliferator-activated receptors and the control of levels of prostaglandin-endoperoxide synthase 2 by arachidonic acid in the bovine uterus. Biochem J 2007; 406:175-83. [PMID: 17516915 PMCID: PMC1948980 DOI: 10.1042/bj20070089] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Arachidonic acid is a potential paracrine agent released by the uterine endometrial epithelium to induce PTGS2 [PG (prostaglandin)-endoperoxide synthase 2] in the stroma. In the present study, bovine endometrial stromal cells were used to determine whether PTGS2 is induced by arachidonic acid in stromal cells, and to investigate the potential role of PPARs (peroxisome-proliferator-activated receptors) in this effect. Arachidonic acid increased PTGS2 levels up to 7.5-fold within 6 h. The cells expressed PPARalpha and PPARdelta (also known as PPARbeta) (but not PPARgamma). PTGS2 protein level was increased by PPAR agonists, including polyunsaturated fatty acids, synthetic PPAR ligands, PGA1 and NSAIDs (non-steroidal anti-inflammatory drugs) with a time course resembling that of arachidonic acid. Use of agonists and antagonists indicated PPARalpha (but not PPARdelta or PPARgamma) was responsible for PTGS2 induction. PTGS2 induction by arachidonic acid did not require PG synthesis. PTGS2 levels were increased by the PKC (protein kinase C) activators 4beta-PMA and PGF(2alpha), and the effects of arachidonic acid, NSAIDs, synthetic PPAR ligands and 4beta-PMA were blocked by PKC inhibitors. This is consistent with PPAR phosphorylation by PKC. Induction of PTGS2 protein by 4beta-PMA in the absence of a PPAR ligand was decreased by the NF-kappaB (nuclear factor kappaB) inhibitors MG132 and parthenolide, suggesting that PKC acted through NF-kappaB in addition to PPAR phosphorylation. Use of NF-kappaB inhibitors allowed the action of arachidonic acid as a PPAR agonist to be dissociated from an effect through PKC. The results are consistent with the hypothesis that arachidonic acid acts via PPARalpha to increase PTGS2 levels in bovine endometrial stromal cells.
Collapse
Affiliation(s)
- E. Linda R. Sheldrick
- *Division of Animal Physiology, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, U.K
| | - Kamila Derecka
- *Division of Animal Physiology, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, U.K
| | - Elaine Marshall
- *Division of Animal Physiology, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, U.K
| | - Evonne C. Chin
- †Reproduction and Development Group, Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield AL9 7TA, U.K
| | - Louise Hodges
- †Reproduction and Development Group, Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield AL9 7TA, U.K
| | - D. Claire Wathes
- †Reproduction and Development Group, Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield AL9 7TA, U.K
| | - D. Robert E. Abayasekara
- †Reproduction and Development Group, Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield AL9 7TA, U.K
| | - Anthony P. F. Flint
- *Division of Animal Physiology, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, U.K
- To whom correspondence should be addressed (email )
| |
Collapse
|
36
|
Li X, Chen D, Yin S, Meng Y, Yang H, Landis-Piwowar KR, Li Y, Sarkar FH, Reddy GPV, Dou QP, Sheng S. Maspin augments proteasome inhibitor-induced apoptosis in prostate cancer cells. J Cell Physiol 2007; 212:298-306. [PMID: 17458898 DOI: 10.1002/jcp.21102] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Proteasome inhibitors are known to induce apoptosis in a variety of cancer cells. On the other hand, maspin, a non-inhibitory serine protease inhibitor, is shown to sensitize cancer cells to therapeutic agents that induce apoptosis. We examined the consequence of maspin expression in prostate cancer cells targeted for treatment with various proteasome inhibitors. We observed that proteasome inhibitors induced apoptosis more effectively in maspin transfected human prostate cancer DU145 cells than in control cells. Interestingly, increased apoptosis in these cells was associated with a significant induction of maspin expression. MG-132, a proteasome inhibitor, induced endogenous and ectopic [cytomegalovirus promoter (CMV)-driven] maspin expression, and maspin siRNA attenuated MG-132-induced apoptosis. Proteasome inhibitor-induced maspin expression was inhibited by actinomycin D (Act D) and cyclohexamide (CHX), and by the inhibitors of p38MAPK, but not ERK1/2 or NF-kappaB. Electrophoretic mobility-shift assay (EMSA) and promoter-reporter activity analyses suggested that p38MAPK activated transcription factor AP-1 is responsible for proteasome inhibitor-induced maspin expression. Taken together, these observations demonstrate that proteasome inhibitors induce maspin expression by activating p38MAPK pathway, and that maspin thus expressed, in turn, augments proteasome inhibitor-induced apoptosis in prostate cancer cells. Our results suggest that gene therapy involving ectopic maspin expression may dramatically improve the efficacy of proteasome inhibitors for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Xiaohua Li
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Cuadrado A, Lafarga V, Cheung PCF, Dolado I, Llanos S, Cohen P, Nebreda AR. A new p38 MAP kinase-regulated transcriptional coactivator that stimulates p53-dependent apoptosis. EMBO J 2007; 26:2115-26. [PMID: 17380123 PMCID: PMC1852783 DOI: 10.1038/sj.emboj.7601657] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2006] [Accepted: 02/26/2007] [Indexed: 01/29/2023] Open
Abstract
The p38 mitogen-activated protein kinase (MAPK) signaling pathway plays an important role in stress-induced cell-fate decisions by orchestrating responses that go from cell-cycle arrest to apoptosis. We have identified a new p38 MAPK-regulated protein that we named p18(Hamlet), which becomes stabilized and accumulates in response to certain genotoxic stresses such as UV or cisplatin treatment. Overexpression of p18(Hamlet) is sufficient to induce apoptosis, whereas its downregulation reduces the apoptotic response to these DNA damage-inducing agents. We show that p18(Hamlet) interacts with p53 and stimulates the transcription of several proapoptotic p53 target genes such as PUMA and NOXA. This correlates with enhanced p18(Hamlet)-induced recruitment of p53 to the promoters. In proliferating cells, low steady-state levels of p18(Hamlet) are probably maintained by a p53-dependent negative feedback loop. Therefore, p18(Hamlet) is a new cell-fate regulator that links the p38 MAPK and p53 pathways and contributes to the establishment of p53-regulated stress responses.
Collapse
Affiliation(s)
- Ana Cuadrado
- CNIO (Spanish National Cancer Center), Madrid, Spain
| | | | - Peter C F Cheung
- MRC Protein Phosphorylation Unit, University of Dundee, Dundee, UK
| | | | - Susana Llanos
- CNIO (Spanish National Cancer Center), Madrid, Spain
| | - Philip Cohen
- MRC Protein Phosphorylation Unit, University of Dundee, Dundee, UK
| | | |
Collapse
|
38
|
Pascal T, Debacq-Chainiaux F, Boilan E, Ninane N, Raes M, Toussaint O. Heme oxygenase-1 and interleukin-11 are overexpressed in stress-induced premature senescence of human WI-38 fibroblasts induced by tert-butylhydroperoxide and ethanol. Biogerontology 2007; 8:409-22. [PMID: 17295091 DOI: 10.1007/s10522-007-9084-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2007] [Accepted: 01/12/2007] [Indexed: 01/25/2023]
Abstract
Acute repeated exposures to subcytotoxic concentrations of tert-butylhydroperoxide and ethanol trigger premature senescence of human diploid fibroblasts. In the present work we found an increased mRNA and protein level of interleukin-11 and heme oxygenase-1 in premature senescence of WI-38 human diploid foetal lung fibroblasts induced by both tert-butylhydroperoxide and ethanol. We tested whether interleukin-11 and heme oxygenase-1 could protect against tert-butylhydroperoxide- or ethanol-induced premature senescence when stable overexpression was established using a retroviral vector-based transduction. No protective effect was found against the decrease of the proliferative potential, the increase of the proportion of senescence-associated ss-galactosidase positive cells and the increase of the mRNA levels of six senescence-associated genes.
Collapse
Affiliation(s)
- Thierry Pascal
- Unit of Research on Cellular Biology (URBC), Department of Biology, University of Namur (FUNDP), Rue de Bruxelles, 5000, Namur, Belgium
| | | | | | | | | | | |
Collapse
|
39
|
Yamamoto N, Sawada H, Izumi Y, Kume T, Katsuki H, Shimohama S, Akaike A. Proteasome Inhibition Induces Glutathione Synthesis and Protects Cells from Oxidative Stress. J Biol Chem 2007; 282:4364-4372. [PMID: 17158454 DOI: 10.1074/jbc.m603712200] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The cause of selective dopaminergic neuronal degeneration in Parkinson disease has still not been resolved, but it has been hypothesized that oxidative stress and the ubiquitin-proteasome system are important in the pathogenesis. In this report, we investigated the effect of proteasome inhibition on oxidative stress-induced cytotoxicity in PC12 cells, an in vitro model of Parkinson disease. Treatment with proteasome inhibitors provided significant protection against toxicity by 6-hydroxydopamine and H(2)O(2) in a concentration-dependent manner. The measurement of intracellular reactive oxygen species using 2',7'-dichlorofluorescein diacetate demonstrated that lactacystin, a proteasome inhibitor, significantly reduced 6-hydroxydopamineand H(2)O(2)-induced reactive oxygen species production. Proteasome inhibitors elevated the amount of glutathione and phosphorylated p38 mitogen-activated protein kinase (MAPK) prior to glutathione elevation. The treatment with lactacystin induced the nuclear translocation of NF-E2-related factor 2 (Nrf2) and increased the level of mRNA for gamma-glutamylcysteine synthetase, a rate-limiting enzyme in glutathione synthesis. Furthermore, SB203580, an inhibitor of p38 MAPK, abolished glutathione elevation and cytoprotection by lactacystin. These data suggest that proteasome inhibition afforded cytoprotection against oxidative stress by the elevation of glutathione content, and its elevation was mediated by p38 MAPK phosphorylation.
Collapse
Affiliation(s)
- Noriyuki Yamamoto
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501
| | - Hideyuki Sawada
- Department of Neurology and Clinical Research Center, Center for Neurological Diseases, Utano National Hospital, 8 Ondoyama-cho, Narutaki, Ukyo-ku, Kyoto 616-5152, and the
| | - Yasuhiko Izumi
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501
| | - Toshiaki Kume
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501
| | - Hiroshi Katsuki
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501
| | - Shun Shimohama
- Department of Neurology, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Akinori Akaike
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501.
| |
Collapse
|
40
|
Kirino Y, Takeno M, Murakami S, Kobayashi M, Kobayashi H, Miura K, Ideguchi H, Ohno S, Ueda A, Ishigatsubo Y. Tumor necrosis factor α acceleration of inflammatory responses by down-regulating heme oxygenase 1 in human peripheral monocytes. ACTA ACUST UNITED AC 2007; 56:464-75. [PMID: 17265482 DOI: 10.1002/art.22370] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE To examine the interaction between heme oxygenase 1 (HO-1), a stress-induced antiinflammatory protein, and tumor necrosis factor alpha (TNFalpha) in human peripheral blood monocytes. METHODS Peripheral blood mononuclear cells (PBMCs) were obtained from healthy donors or from patients with rheumatoid arthritis (RA) receiving the anti-tumor necrosis factor alpha (anti-TNFalpha) monoclonal antibody infliximab. CD14+ cells were isolated by magnetic cell sorting, cultured with TNFalpha or auranofin, and transfected with a plasmid encoding HO-1 or an HO-1-specific small interfering RNA vector. Protein and messenger RNA (mRNA) levels were examined by immunoblotting and real-time polymerase chain reaction. Cytokine levels in culture supernatants were measured by enzyme-linked immunosorbent assay. HO-1 gene transcription was evaluated using a luciferase reporter gene assay. Actinomycin D and cycloheximide were used to monitor the stability of mRNA and protein. RESULTS HO-1 is constitutively expressed by CD14+ PBMCs from healthy donors. TNFalpha suppressed HO-1 expression by accelerating the decay of mRNA without affecting gene transcription or protein stability. Forced expression or selective knock-down of the HO-1 gene expression resulted in down-regulation or up-regulation, respectively, of proinflammatory cytokine synthesis by monocytes. Treatment with infliximab significantly increased HO-1 mRNA levels and reduced TNFalpha synthesis by PBMCs from RA patients. CONCLUSION TNFalpha accelerated inflammatory responses by down-regulating HO-1 expression in human monocytes. TNF antagonists may block this TNF-dependent suppression of HO-1 expression, resulting in an amelioration of inflammation.
Collapse
Affiliation(s)
- Yohei Kirino
- Yokohama City University, Graduate School of Medicine, Yokohama, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Keum YS, Yu S, Chang PPJ, Yuan X, Kim JH, Xu C, Han J, Agarwal A, Kong ANT. Mechanism of action of sulforaphane: inhibition of p38 mitogen-activated protein kinase isoforms contributing to the induction of antioxidant response element-mediated heme oxygenase-1 in human hepatoma HepG2 cells. Cancer Res 2006; 66:8804-13. [PMID: 16951197 DOI: 10.1158/0008-5472.can-05-3513] [Citation(s) in RCA: 230] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Exposure of sulforaphane to HepG2 cells increased heme oxygenase-1 (HO-1) expression by activating antioxidant response element (ARE) through induction of Nrf2 and suppression of Kelch-like ECH-associated protein 1 (Keap1). Using human HO-1 promoter reporter plasmids and ChIP assay, we have identified that sulforaphane transcriptionally activated the upstream ARE-rich enhancer region, located at -9.0 kb upstream human HO-1 promoter. Induction of HO-1 by sulforaphane was attenuated by overexpression of mutant Nrf2 plasmid in HepG2 cells and totally abolished in Nrf2 knockout mouse embryonic keratinocytes and fibroblasts. Overexpression of individual p38 mitogen-activated protein (MAP) kinase (MAPK) isoforms also suppressed constitutive as well as sulforaphane- or Nrf2-induced ARE-dependent gene expression. Among the upstream kinases, although MKK3 was not involved in suppression of ARE by any of p38 MAPK isoforms, MKK6 selectively suppressed ARE by p38 gamma or p38 delta, but not by p38 alpha or p38 beta. Importantly, sulforaphane not only activated MAP/extracellular signal-regulated kinase (ERK) kinases 1/2 and ERK1/2, but also strongly suppressed anisomycin-induced activation of p38 MAPK isoforms by blocking phosphorylation of upstream kinases, MKK3/6. Finally, we found that stimulation of p38 MAPK isoforms phosphorylated purified Nrf2 protein and caused an increase in the interaction between Nrf2 and Keap1 in vitro and the suppression of Nrf2 translocation into the nucleus. Collectively, our results indicate that transcriptional activation of Nrf2/ARE is critical in sulforaphane-mediated induction of HO-1, which can be modulated in part by the blockade of p38 MAPK signaling pathway. In addition, our study shows that p38 MAPK can phosphorylate Nrf2 and promotes the association between Nrf2 and Keap1 proteins, thereby potentially inhibiting nuclear translocation of Nrf2.
Collapse
Affiliation(s)
- Young-Sam Keum
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway, NJ 08854, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Meiners S, Ludwig A, Lorenz M, Dreger H, Baumann G, Stangl V, Stangl K. Nontoxic proteasome inhibition activates a protective antioxidant defense response in endothelial cells. Free Radic Biol Med 2006; 40:2232-41. [PMID: 16785037 DOI: 10.1016/j.freeradbiomed.2006.03.003] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2005] [Revised: 02/07/2006] [Accepted: 03/02/2006] [Indexed: 11/29/2022]
Abstract
Inhibitors of the ubiquitin-proteasome system offer a new and promising approach in the therapy of proliferative and inflammatory diseases. In order to narrow the therapeutic window for cytotoxic effects on the one hand and nontoxic, anti-inflammatory effects on the other hand, we elucidated the complex cellular effects of toxic versus nontoxic proteasome inhibition in human endothelial cells by expressional profiling. Nontoxic doses of proteasome inhibitors induced a defined, dose-dependent transcriptional response that was markedly attenuated in terms of gene number and amplitude of regulation compared to toxic doses. In particular, we observed uniform upregulation of several antioxidative enzymes and differential regulation of genes involved in endothelial function. This adaptive transcriptional pattern was translated into a protective response of endothelial cells against H(2)O(2)-induced oxidative stress and into improvement of endothelial function of rat aortic rings. Our data thus suggest that nontoxic proteasome inhibition might offer a new therapeutic approach for the treatment of endothelial dysfunction in cardiovascular disorders.
Collapse
Affiliation(s)
- Silke Meiners
- Universitätsmedizin Berlin, Charité, Medizinische Klinik und Poliklinik mit Schwerpunkt Kardiologie, Angiologie, Pneumologie, Schumannstrasse 20/21, D - 10117 Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
43
|
Ryter SW, Alam J, Choi AMK. Heme oxygenase-1/carbon monoxide: from basic science to therapeutic applications. Physiol Rev 2006; 86:583-650. [PMID: 16601269 DOI: 10.1152/physrev.00011.2005] [Citation(s) in RCA: 1797] [Impact Index Per Article: 94.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The heme oxygenases, which consist of constitutive and inducible isozymes (HO-1, HO-2), catalyze the rate-limiting step in the metabolic conversion of heme to the bile pigments (i.e., biliverdin and bilirubin) and thus constitute a major intracellular source of iron and carbon monoxide (CO). In recent years, endogenously produced CO has been shown to possess intriguing signaling properties affecting numerous critical cellular functions including but not limited to inflammation, cellular proliferation, and apoptotic cell death. The era of gaseous molecules in biomedical research and human diseases initiated with the discovery that the endothelial cell-derived relaxing factor was identical to the gaseous molecule nitric oxide (NO). The discovery that endogenously produced gaseous molecules such as NO and now CO can impart potent physiological and biological effector functions truly represented a paradigm shift and unraveled new avenues of intense investigations. This review covers the molecular and biochemical characterization of HOs, with a discussion on the mechanisms of signal transduction and gene regulation that mediate the induction of HO-1 by environmental stress. Furthermore, the current understanding of the functional significance of HO shall be discussed from the perspective of each of the metabolic by-products, with a special emphasis on CO. Finally, this presentation aspires to lay a foundation for potential future clinical applications of these systems.
Collapse
Affiliation(s)
- Stefan W Ryter
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, The University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| | | | | |
Collapse
|
44
|
Lee CJ, Lee SS, Chen SC, Ho FM, Lin WW. Oregonin inhibits lipopolysaccharide-induced iNOS gene transcription and upregulates HO-1 expression in macrophages and microglia. Br J Pharmacol 2006; 146:378-88. [PMID: 16025135 PMCID: PMC1576284 DOI: 10.1038/sj.bjp.0706336] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Oregonin isolated from Alnus formosana is a diarylheptanoid derivative, which appears to have antioxidative and anti-inflammatory activities. In this study, our data demonstrated inhibitory actions of oregonin on the LPS-induced iNOS protein in RAW264.7 macrophages and BV-2 microglial cells. We also suggested that HO-1 induction by oregonin might contribute to this action. Oregonin is able to dose-dependently reduce NO production, iNOS protein and iNOS promoter activity stimulated by LPS in RAW264.7 and BV-2 cells. Oregonin also showed inhibition of LPS-mediated NF-kappaB promoter activity and DNA-binding ability, as well as p65 nuclear translocation and phosphorylation. However, oregonin had no effect on IKK activity. AP-1 promoter activity and p38 MAPK activation but not PKC, ERK and JNK activation induced by LPS were attenuated by oregonin. Accompanying with iNOS protein reduction, moreover, we found that oregonin was able to induce HO-1 protein level. Results using a CO donor, [Ru(CO)(3)Cl(2)](2) further showed the ability of CO in reduction of iNOS protein level induced by LPS through the blockade of NF-kappaB and AP-1. Taken together, these results provide new evidences into the anti-inflammatory actions of oregonin, which include the inhibition of iNOS gene transcription via suppressing transcriptional activity of NF-kappaB and AP-1, as well as the upregulation of anti-inflammatory molecule HO-1. The HO-1-derived CO may also be involved in the suppressive effect on iNOS gene regulation.
Collapse
Affiliation(s)
- Cheng-Jui Lee
- Department of Pharmacology, College of Medicine, National Taiwan University, No. 1, Sec 1, Jen-Ai road, Taipei 100, Taiwan
| | - Shoei-Sheng Lee
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Su-Chung Chen
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Feng-Ming Ho
- Department of Internal Medicine, Tao-Yuan General Hospital, Department of Health, the Executive Yuan, Taiwan
| | - Wan-Wan Lin
- Department of Pharmacology, College of Medicine, National Taiwan University, No. 1, Sec 1, Jen-Ai road, Taipei 100, Taiwan
- Author for correspondence:
| |
Collapse
|
45
|
Abstract
Many biological functions of heme oxygenase (HO), such as cytoprotection against oxidative stress, vasodilation, neurotransmission in the central or peripheral nervous systems, and anti-inflammatory, anti-apoptotic, or anti-proliferative potential, have been attributed to its enzymatic byproduct carbon monoxide (CO), although roles for biliverdin/bilirubin and iron have also been proposed. In addition to these well-characterized effects, recent findings reveal that HO-derived CO may act as an oxygen sensor and circadian modulator of heme biosynthesis. In lymphocytes, CO may participate in regulatory T cell function. A number of the known signaling effects of CO depend on stimulation of soluble guanylate cyclase and/or activation of mitogen-activated protein kinases (MAPK). Furthermore, modulation of caveolin-1 status may serve as an essential component of certain aspects of CO action, such as growth control. In this review, we summarize recent findings of the beneficial or detrimental effects of endogenous CO with an emphasis on the signaling pathways and downstream targets that trigger the action of this gas.
Collapse
Affiliation(s)
- Hong Pyo Kim
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| | | | | |
Collapse
|
46
|
Prawan A, Kundu JK, Surh YJ. Molecular basis of heme oxygenase-1 induction: implications for chemoprevention and chemoprotection. Antioxid Redox Signal 2005; 7:1688-703. [PMID: 16356130 DOI: 10.1089/ars.2005.7.1688] [Citation(s) in RCA: 165] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Heme oxygenase (HO)-1, involved in the heme degradation process, is an important antioxidant enzyme. The induction of HO-1 gene expression, in response to diverse oxidative stimuli, represents a critical event in adaptive cellular response. Experimental models of various diseases, including acute inflammation, atherosclerosis, degenerative diseases, and carcinogenesis, have demonstrated that the induction of HO-1 can prevent or mitigate the symptoms associated with these ailments. Recent progress in our understanding of cellular signaling networks as critical modulators of gene transcription sheds light on the molecular basis of HO-1 gene expression. A panel of redox-sensitive transcription factors such as activator protein-1, nuclear factor- kappaB, and nuclear factor E2-related factor-2, and some of the upstream kinases have been identified as regulators of HO-1 gene induction. The scope of this review is limited to focus on molecular mechanisms underlying HO-1 expression and the significance of targeted induction of HO-1 as a strategy to achieve chemoprevention and chemoprotection.
Collapse
Affiliation(s)
- Auemduan Prawan
- National Research Laboratory of Molecular Carcinogenesis and Chemoprevention, College of Pharmacy, Seoul National University, Seoul 151-742, South Korea
| | | | | |
Collapse
|
47
|
Srisook K, Kim C, Cha YN. Molecular mechanisms involved in enhancing HO-1 expression: de-repression by heme and activation by Nrf2, the "one-two" punch. Antioxid Redox Signal 2005; 7:1674-87. [PMID: 16356129 DOI: 10.1089/ars.2005.7.1674] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Heme oxygenase (HO)-1 is a stress response protein, which confers cytoprotection against oxidative injury and provides a vital function in maintaining tissue homeostasis. Molecular mechanisms involved in the inducible transcription of ho-1 occurring in response to numerous and diverse stressful conditions have remained elusive. Since the discovery of E1 and E2, the two upstream enhancers regulating induction of ho-1 transcription in 1989, there have been many studies dealing with molecular mechanisms involved in enhancing HO-1 expression. In this commentary, recent advances in our understanding of the mechanisms involved in the induction of HO-1 expression in mammalian cells are summarized with some supportive results reported by others. Currently available data indicate that activation of ho-1 transcription involves both the heme (native substrate)-dependent selective alleviation of repressor and the oxidative stress-dependent activation of transcriptional activator. The stress-released free-heme (HO-1 substrate) from hemoproteins involved in causing oxidative stress itself appears to act as a molecular switch controlling the repressor- activator antagonism on the enhancer sequences of ho-1. Thus, induction of HO-1 appears to operate in a manner like a simple feedback loop. dox Signal. 7, 1674-1687.
Collapse
Affiliation(s)
- Klaokwan Srisook
- Department of Pharmacology and Toxicology, College of Medicine, Inha University, Incheon, South Korea
| | | | | |
Collapse
|
48
|
Ho AK, McNeil L, Terriff D, Price DM, Chik CL. Role of protein turnover in the activation of p38 mitogen-activated protein kinase in rat pinealocytes. Biochem Pharmacol 2005; 70:1840-50. [PMID: 16236272 DOI: 10.1016/j.bcp.2005.09.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2005] [Revised: 09/15/2005] [Accepted: 09/16/2005] [Indexed: 11/17/2022]
Abstract
Differences in the time profiles of activation between p38MAPK and p42/44MAPK by norepinephrine (NE) in rat pinealocytes suggest involvement of mechanisms other than the phosphorylation cascades in their activation. In the present study we investigated whether protein turnover played a role in regulating p38MAPK activation in the rat pineal gland. NE stimulation caused an increase in MAPK kinase3/6 (MKK 3/6) and p38MAPK phosphorylation that occurred in the absence of changes in the mRNA or protein levels of p38MAPK or MKK3/6. The stimulatory effect of NE on phosphorylated MKK3/6 and p38MAPK, but not phosphorylated p42/44MAPK, was blocked by treatment with actinomycin or cycloheximide, indicating a requirement of transcription and translation in activation of the p38MAPK but not the p42/44MAPK pathway. Moreover, inhibition of proteasomes by clasto-lactacystin beta-lactone or Z-Leu-Leu-Leu-CHO (MG132) selectively increased basal and NE-stimulated phosphorylated MKK3/6 and p38MAPK levels without affecting the mRNA or protein levels of MKK3 or p38MAPK. In contrast, the effect of proteasomal inhibition on NE-stimulated p42/44MAPK phosphorylation was inhibitory. Treatment with MG132 also reduced the decline in the phosphorylated levels of NE-stimulated MKK3/6 and p38MAPK that normally follows beta-adrenergic blockade. Together, our results indicate that p38MAPK but not p42/44MAPK activation in the rat pineal gland is tightly coupled to protein synthesis and degradation. The synthesis of an activator upstream of MKK3/6 is required for the NE-activation of p38MAPK.
Collapse
Affiliation(s)
- A K Ho
- Department of Physiology, University of Alberta, Edmonton, Alta., Canada T6G 2H7.
| | | | | | | | | |
Collapse
|
49
|
Zwerina J, Tzima S, Hayer S, Redlich K, Hoffmann O, Hanslik-Schnabel B, Smolen JS, Kollias G, Schett G. Heme oxygenase 1 (HO‐1) regulates osteoclastogenesis and bone resorption. FASEB J 2005; 19:2011-3. [PMID: 16234431 DOI: 10.1096/fj.05-4278fje] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Heme oxygenase 1 (HO-1) plays an important role in vascular disease, transplantation, and inflammation. In animal models of acute and chronic inflammation, induction of HO-1 has anti-inflammatory and cytoprotective properties. Since inflammation is an important trigger of osteoclastogenesis, we hypothesized that HO-1 might influence osteoclastogenesis. We investigated the effects of induction of HO-1 on osteoclast formation in vitro and in vivo. Furthermore, we addressed the role of HO-1 in inflammatory bone loss in humans. When HO-1 was induced by hemin in vitro, a significant dose-dependent inhibition of osteoclastogenesis was observed. Up-regulation of HO-1 was mediated by activation of MAPK and primarily prevented differentiation of osteoclast precursors to osteoclasts, whereas it did not affect mature osteoclasts. Anti-osteoclastogenic properties of hemin were based on a down-regulation of c-fms, RANK, TRAF-6, and c-fos. In addition, induction of HO-1 inhibited TNF-triggered osteoclast differentiation in vitro as well as LPS-driven inflammatory bone loss in vivo. Furthermore, HO-1 induction suppressed osteoclastogenesis and bone destruction in a TNF-mediated arthritis. In line, assessment of synovial tissue from rheumatoid arthritis patients revealed that osteoclasts are usually HO-1 negative. Moreover, serum levels of bilirubin, a metabolite of HO-1, were elevated in rheumatoid arthritis patients without bone damage, suggesting HO-1 affects bone loss in humans. In summary, these data indicate that HO-1 negatively regulates osteoclastogenesis, leading to a positive net balance of bone.
Collapse
Affiliation(s)
- Jochen Zwerina
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Yu Y, Yong J, Li X, Qing T, Qin H, Xiong X, You J, Ding M, Deng H. The proteasomal inhibitor MG132 increases the efficiency of mouse embryo production after cloning by electrofusion. Reproduction 2005; 130:553-8. [PMID: 16183873 DOI: 10.1530/rep.1.00758] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In this study, we cloned mice from ES cells by a post-electrofusion MG132 treatment and improved development of cloned embryos with a sequential cultivation protocol. When 5 μM MG132, a proteasome inhibitor, were used to treat the reconstructed embryos, the capacity of in vitro development, implantation and full-term development were significantly improved. Blastocyst formation rates of the reconstructed embryos from X4 ES cells (F1 strain derived from C57BL/6 × 129sv) and J1 ES cells obtained with or without MG132 treatment were 66.9% and 26.6%, and 66.1% and 34.5% respectively (P < 0.05). A total of 146 two-cell embryos cloned from X4 ES cells with MG132 treatment were transferred to recipients, and five cloned pups (3.4%) were born, of which four survived. When the same numbers of two-cell embryos cloned from X4 ES cells without MG132 treatment were transferred, however, no live-born mice were obtained. When embryos cloned from J1 ES cells without MG132 treatment were cultured in KSOM medium for 54 h followed by culture in CZB medium containing 5.6 mM glucose for 42 h, the blastocyst rate was significantly higher than when they were cultured in KSOM continuously for 96 h (34.5% vs 17.1%). However, sequential cultivation did not improve the development of embryos cloned with MG132 treatment and that of parthenotes. In conclusion, MG132 treatment increased the developmental potential of reconstructed mouse embryos, and sequential cultivation improved development of the embryos cloned by electrofusion without MG132 treatment.
Collapse
Affiliation(s)
- Yuansong Yu
- Department of Cell Biology, College of Life Sciences, Beijing University, Beijing 100871, China, Beijing Laboratory Animal Research Center, Beijing 100012, China
| | | | | | | | | | | | | | | | | |
Collapse
|