101
|
Sakai H, Kai Y, Oguchi A, Kimura M, Tabata S, Yaegashi M, Saito T, Sato K, Sato F, Yumoto T, Narita M. Curcumin Inhibits 5-Fluorouracil-induced Up-regulation of CXCL1 and CXCL2 of the Colon Associated with Attenuation of Diarrhoea Development. Basic Clin Pharmacol Toxicol 2016; 119:540-547. [PMID: 27194111 DOI: 10.1111/bcpt.12619] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 05/03/2016] [Indexed: 12/27/2022]
Abstract
The compound 5-fluorouracil (5-FU) is used in cancer chemotherapy and is known to cause diarrhoea. We recently reported that chemokine (C-X-C motif) ligand 1 (CXCL1) and neutrophils in the colonic mucosa were markedly increased by the administration of 5-FU in mice. Curcumin has anti-inflammatory, antitumour and antioxidant properties. Therefore, we examined the effect of curcumin on 5-FU-induced diarrhoea development and CXCL1 and CXCL2 up-regulation in the colon. Mice were given 5-FU (50 mg/kg, i.p.) daily for 4 days. Curcumin (100 or 300 mg/kg, p.o.) was administered on the day before the first administration of 5-FU and administered 30 min. before the administration of 5-FU. Gene expression levels of CXCL1 and CXCL2 in the colon were examined by real-time RT-PCR. Curcumin reduced the 5-FU-induced diarrhoea development. Under this condition, the CXCL1 and CXCL2 gene up-regulated by 5-FU administration was inhibited by curcumin. The gene expression of CXCL1 and CXCL2 was also enhanced by 5-FU application in vitro. The 5-FU-induced up-regulated CXCL1 and CXCL2 gene expressions were inhibited by curcumin, Bay-117082 and bortezomib, nuclear factor kappa B (NF-κB) inhibitors, C646, a p300/cyclic adenosine monophosphate response element-binding protein-histone acetyltransferase (HAT) inhibitor. In conclusion, these findings suggested that curcumin prevented the development of diarrhoea by inhibiting NF-κB and HAT activation.
Collapse
Affiliation(s)
- Hiroyasu Sakai
- Department of Analytical Pathophysiology, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Yuki Kai
- Department of Analytical Pathophysiology, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Aya Oguchi
- Department of Pharmacology, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Minami Kimura
- Department of Analytical Pathophysiology, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Shoko Tabata
- Department of Analytical Pathophysiology, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Miyabi Yaegashi
- Department of Analytical Pathophysiology, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Taiki Saito
- Department of Analytical Pathophysiology, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Ken Sato
- Department of Analytical Pathophysiology, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Fumiaki Sato
- Department of Analytical Pathophysiology, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Tetsuro Yumoto
- Department of Analytical Pathophysiology, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Minoru Narita
- Department of Pharmacology, Hoshi University, Shinagawa-ku, Tokyo, Japan
| |
Collapse
|
102
|
Histone deacetylase inhibition enhances antimicrobial peptide but not inflammatory cytokine expression upon bacterial challenge. Proc Natl Acad Sci U S A 2016; 113:E2993-3001. [PMID: 27162363 DOI: 10.1073/pnas.1605997113] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Antimicrobial peptides (AMP) are defense effectors of the innate immunity playing a crucial role in the intestinal homeostasis with commensals and protection against pathogens. Herein we aimed to investigate AMP gene regulation by deciphering specific characteristics allowing their enhanced expression among innate immune genes, particularly those encoding proinflammatory mediators. Our emphasis was on epigenetic regulation of the gene encoding the AMP β-defensin 2 (HBD2), taken as a model of possibly specific induction, upon challenge with a commensal bacterium, compared with the proinflammatory cytokine IL-8. Using an in vitro model of colonic epithelial cells challenged with Escherichia coli K12, we showed that inhibition of histone deacetylases (HDAC) by trichostatin A dramatically enhanced induction of HBD2 expression, without affecting expression of IL-8. This mechanism was supported by an increased phosphorylation of histone H3 on serine S10, preferentially at the HBD2 promoter. This process occurred through activation of the IκB kinase complex, which also led to activation of NF-κB. Moreover, we demonstrated that NF-κB was modified by acetylation upon HDAC inhibition, partly by the histone acetyltransferase p300, and that both NF-κB and p300 supported enhanced induction of HBD2 expression. Furthermore, we identified additional genes belonging to antimicrobial defense and epithelial restitution pathways that showed a similar pattern of epigenetic control. Finally, we confirmed our finding in human colonic primary cells using an ex vivo organoid model. This work opens the way to use epigenetic pharmacology to achieve induction of epithelial antimicrobial defenses, while limiting the deleterious risk of an inflammatory response.
Collapse
|
103
|
Lillico R, Stesco N, Khorshid Amhad T, Cortes C, Namaka MP, Lakowski TM. Inhibitors of enzymes catalyzing modifications to histone lysine residues: structure, function and activity. Future Med Chem 2016; 8:879-97. [PMID: 27173004 DOI: 10.4155/fmc-2016-0021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Gene expression is partly controlled by epigenetic mechanisms including histone-modifying enzymes. Some diseases are caused by changes in gene expression that can be mitigated by inhibiting histone-modifying enzymes. This review covers the enzyme inhibitors targeting histone lysine modifications. We summarize the enzymatic mechanisms of histone lysine acetylation, deacetylation, methylation and demethylation and discuss the biochemical roles of these modifications in gene expression and in disease. We discuss inhibitors of lysine acetylation, deacetylation, methylation and demethylation defining their structure-activity relationships and their potential mechanisms. We show that there are potentially indiscriminant off-target effects on gene expression even with the use of selective epigenetic enzyme inhibitors.
Collapse
Affiliation(s)
- Ryan Lillico
- Faculty of Health Sciences, College of Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada
- Pharmaceutical Analysis Laboratory, College of Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Nicholas Stesco
- Faculty of Health Sciences, College of Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada
- Pharmaceutical Analysis Laboratory, College of Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Tina Khorshid Amhad
- Faculty of Health Sciences, College of Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada
- Joint Laboratory of Biological Psychiatry Between Shantou University Medical College and College of Medicine, University of Manitoba, Winnipeg, MB, Canada
- Department of Human Anatomy and Cell Science, College of Medicine, University of Manitoba, Winnipeg, MB, Canada
- Department of Rehabilitation Medicine, Health Sciences Centre (HSC), Winnipeg, MB, Canada
| | - Claudia Cortes
- Joint Laboratory of Biological Psychiatry Between Shantou University Medical College and College of Medicine, University of Manitoba, Winnipeg, MB, Canada
- Department of Human Anatomy and Cell Science, College of Medicine, University of Manitoba, Winnipeg, MB, Canada
- Department of Rehabilitation Medicine, Health Sciences Centre (HSC), Winnipeg, MB, Canada
| | - Mike P Namaka
- Faculty of Health Sciences, College of Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada
- Joint Laboratory of Biological Psychiatry Between Shantou University Medical College and College of Medicine, University of Manitoba, Winnipeg, MB, Canada
- Department of Human Anatomy and Cell Science, College of Medicine, University of Manitoba, Winnipeg, MB, Canada
- Department of Rehabilitation Medicine, Health Sciences Centre (HSC), Winnipeg, MB, Canada
| | - Ted M Lakowski
- Faculty of Health Sciences, College of Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada
- Pharmaceutical Analysis Laboratory, College of Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
104
|
LPS-induced NFκB enhanceosome requires TonEBP/NFAT5 without DNA binding. Sci Rep 2016; 6:24921. [PMID: 27118681 PMCID: PMC4847014 DOI: 10.1038/srep24921] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 04/07/2016] [Indexed: 12/24/2022] Open
Abstract
NFκB is a central mediator of inflammation. Present inhibitors of NFκB are mostly based on inhibition of essential machinery such as proteasome and protein kinases, or activation of nuclear receptors; as such, they are of limited therapeutic use due to severe toxicity. Here we report an LPS-induced NFκB enhanceosome in which TonEBP is required for the recruitment of p300. Increased expression of TonEBP enhances the NFκB activity and reduced TonEBP expression lowers it. Recombinant TonEBP molecules incapable of recruiting p300 do not stimulate NFκB. Myeloid-specific deletion of TonEBP results in milder inflammation and sepsis. We discover that a natural small molecule cerulenin specifically disrupts the enhanceosome without affecting the activation of NFκB itself. Cerulenin suppresses the pro-inflammatory activation of macrophages and sepsis without detectable toxicity. Thus, the NFκB enhanceosome offers a promising target for useful anti-inflammatory agents.
Collapse
|
105
|
Balasubramaniyan N, Ananthanarayanan M, Suchy FJ. Nuclear factor-κB regulates the expression of multiple genes encoding liver transport proteins. Am J Physiol Gastrointest Liver Physiol 2016; 310:G618-28. [PMID: 26867564 PMCID: PMC4836129 DOI: 10.1152/ajpgi.00363.2015] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 02/04/2016] [Indexed: 02/07/2023]
Abstract
In this study we identified the mechanisms underlying the inhibitory effects of NF-κB on the expression of genes encoding multiple liver transport proteins. Well-conserved NF-κB binding sites were found in the promoters of farnesoid X receptor (FXR)-target genes. An electromobility shift assay (EMSA) demonstrated the specific interaction between the NF-κB p65 protein and a (32)P-labeled BSEP NF-κB response element (NF-κBE). Chromatin immunoprecipitation (ChIP) analysis confirmed binding of NF-κB p65 to the BSEP locus but not the FXRE in vitro. NF-κB p65 overexpression in Huh-7 cells markedly repressed FXR/RXR transactivation of the BSEP, ABCG5/G8, MRP2, and FXR promoters, which was totally reversed by expression of the IκBα super-repressor. NF-κB interacted directly with FXR on coimmunoprecipitation, suggesting another level for the inhibitory effects of NF-κB on FXR-target genes. In vivo ChIP analysis with liver nuclei obtained from mice after 3 days of common bile duct ligation (BDL) or 6 h post-lipopolysaccharide (LPS) injection showed a markedly increased recruitment of NF-κB p65 to the Bsep promoter compared with controls. There was also increased recruitment of the corepressor silencing mediator of retinoic acid and thyroid hormone receptor (SMRT) and histone deacetylase (HDAC)3 and HDAC2 to the NF-κB sites. We also found that NF-κB p65 was recruited to NF-κB binding sites in the promoters of organic solute transporter, OSTα and OSTβ, and unexpectedly activated rather than repressed gene expression. In mouse liver after BDL NF-κB recruitment to Ostα and Ostβ promoters was associated with increased binding of the potent coactivator cAMP response element binding protein (CREB)-binding protein (CBP)/p300 to the NF-κBE and depletion of CBP/p300 at the FXR element. Overall, these studies demonstrate a novel role for NF-κB in adaptation to obstructive and LPS-induced cholestasis acting through recruitment to specific NF-κB binding sites in the promoters of FXR-target genes and possibly through direct interaction with FXR.
Collapse
Affiliation(s)
- Natarajan Balasubramaniyan
- 1Department of Pediatrics, Children's Hospital Colorado Research Institute, University of Colorado School of Medicine, Aurora, Colorado; and
| | | | - Frederick J. Suchy
- 1Department of Pediatrics, Children's Hospital Colorado Research Institute, University of Colorado School of Medicine, Aurora, Colorado; and
| |
Collapse
|
106
|
Huang HY, Chang HF, Tsai MJ, Chen JS, Wang MJ. 6-Mercaptopurine attenuates tumor necrosis factor-α production in microglia through Nur77-mediated transrepression and PI3K/Akt/mTOR signaling-mediated translational regulation. J Neuroinflammation 2016; 13:78. [PMID: 27075886 PMCID: PMC4831152 DOI: 10.1186/s12974-016-0543-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Accepted: 04/07/2016] [Indexed: 02/07/2023] Open
Abstract
Background The pathogenesis of several neurodegenerative diseases often involves the microglial activation and associated inflammatory processes. Activated microglia release pro-inflammatory factors that may be neurotoxic. 6-Mercaptopurine (6-MP) is a well-established immunosuppressive drug. Common understanding of their immunosuppressive properties is largely limited to peripheral immune cells. However, the effect of 6-MP in the central nervous system, especially in microglia in the context of neuroinflammation is, as yet, unclear. Tumor necrosis factor-α (TNF-α) is a key cytokine of the immune system that initiates and promotes neuroinflammation. The present study aimed to investigate the effect of 6-MP on TNF-α production by microglia to discern the molecular mechanisms of this modulation. Methods Lipopolysaccharide (LPS) was used to induce an inflammatory response in cultured primary microglia or murine BV-2 microglial cells. Released TNF-α was measured by enzyme-linked immunosorbent assay (ELISA). Gene expression was determined by real-time reverse transcription polymerase chain reaction (RT-PCR). Signaling molecules were analyzed by western blotting, and activation of NF-κB was measured by ELISA-based DNA binding analysis and luciferase reporter assay. Chromatin immunoprecipitation (ChIP) analysis was performed to examine NF-κB p65 and coactivator p300 enrichments and histone modifications at the endogenous TNF-α promoter. Results Treatment of LPS-activated microglia with 6-MP significantly attenuated TNF-α production. In 6-MP pretreated microglia, LPS-induced MAPK signaling, IκB-α degradation, NF-κB p65 nuclear translocation, and in vitro p65 DNA binding activity were not impaired. However, 6-MP suppressed transactivation activity of NF-κB and TNF-α promoter by inhibiting phosphorylation and acetylation of p65 on Ser276 and Lys310, respectively. ChIP analyses revealed that 6-MP dampened LPS-induced histone H3 acetylation of chromatin surrounding the TNF-α promoter, ultimately leading to a decrease in p65/coactivator-mediated transcription of TNF-α gene. Furthermore, 6-MP enhanced orphan nuclear receptor Nur77 expression. Using RNA interference approach, we further demonstrated that Nur77 upregulation contribute to 6-MP-mediated inhibitory effect on TNF-α production. Additionally, 6-MP also impeded TNF-α mRNA translation through prevention of LPS-activated PI3K/Akt/mTOR signaling cascades. Conclusions These results suggest that 6-MP might have a therapeutic potential in neuroinflammation-related neurodegenerative disorders through downregulation of microglia-mediated inflammatory processes. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0543-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hsin-Yi Huang
- Department of Medical Research, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Hui-Fen Chang
- Department of Medical Research, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Ming-Jen Tsai
- Department of Emergency Medicine, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi, Taiwan
| | - Jhih-Si Chen
- Department of Medical Research, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Mei-Jen Wang
- Department of Medical Research, Buddhist Tzu Chi General Hospital, Hualien, Taiwan.
| |
Collapse
|
107
|
Thangjam GS, Birmpas C, Barabutis N, Gregory BW, Clemens MA, Newton JR, Fulton D, Catravas JD. Hsp90 inhibition suppresses NF-κB transcriptional activation via Sirt-2 in human lung microvascular endothelial cells. Am J Physiol Lung Cell Mol Physiol 2016; 310:L964-74. [PMID: 27036868 DOI: 10.1152/ajplung.00054.2016] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 03/13/2016] [Indexed: 11/22/2022] Open
Abstract
The ability of anti-heat shock protein 90 (Hsp90) drugs to attenuate NF-κB-mediated transcription is the major basis for their anti-inflammatory properties. While the molecular mechanisms underlying this effect are not clear, they appear to be distinct in human endothelial cells. We now show for the first time that type 2 sirtuin (Sirt-2) histone deacetylase binds human NF-κB target gene promoter and prevents the recruitment of NF-κB proteins and subsequent assembly of RNA polymerase II complex in human lung microvascular endothelial cells. Hsp90 inhibitors stabilize the Sirt-2/promoter interaction and impose a "transcriptional block," which is reversed by either inhibition or downregulation of Sirt-2 protein expression. Furthermore, this process is independent of NF-κB (p65) Lysine 310 deacetylation, suggesting that it is distinct from known Sirt-2-dependent mechanisms. We demonstrate that Sirt-2 is recruited to NF-κB target gene promoter via interaction with core histones. Upon inflammatory challenge, chromatin remodeling and core histone H3 displacement from the promoter region removes Sirt-2 and allows NF-κB/coactivator recruitment essential for RNA Pol II-dependent mRNA induction. This novel mechanism may have important implications in pulmonary inflammation.
Collapse
Affiliation(s)
- Gagan S Thangjam
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, Virginia
| | - Charalampos Birmpas
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, Virginia
| | - Nektarios Barabutis
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, Virginia
| | - Betsy W Gregory
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, Virginia
| | - Mary Ann Clemens
- Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, Virginia
| | - Joseph R Newton
- Department of Surgery, Eastern Virginia Medical School, Norfolk, Virginia
| | - David Fulton
- Vascular Biology Center, Augusta University, Augusta, Georgia; and
| | - John D Catravas
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, Virginia; School of Medical Diagnostic and Translational Sciences, College of Health Sciences, Old Dominion University, Norfolk, Virginia
| |
Collapse
|
108
|
Tanaka T, Iino M. Nuclear Translocation of p65 is Controlled by Sec6 via the Degradation of IκBα. J Cell Physiol 2016; 231:719-30. [PMID: 26247921 DOI: 10.1002/jcp.25122] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 08/04/2015] [Indexed: 12/18/2022]
Abstract
Nuclear factor-κB (NF-κB) is an inducible transcription factor that mediates immune and inflammatory responses. NF-κB pathways are also involved in cell adhesion, differentiation, proliferation, autophagy, senescence, and protection against apoptosis. The deregulation of NF-κB activity is found in a number of disease states, including cancer, arthritis, chronic inflammation, asthma, neurodegenerative diseases, and heart disease. The 90 kDa ribosomal S6 kinase (p90RSK) family, which is serine/threonine kinases, is phosphorylated by extracellular signal-regulated kinase1/2 (ERK1/2) and is related to NF-κB pathways. Our previous studies revealed that Sec6, a component of the exocyst complex, plays specific roles in cell-cell adhesion and cell cycle arrest. However, the mechanism by which Sec6 regulates the NF-κB signaling pathway is unknown. We demonstrated that Sec6 knockdown inhibited the degradation of IκBα and delayed the nucleus-cytoplasm translocation of p65 in HeLa cells transfected with Sec6 siRNAs after treatment with tumor necrosis factor alpha (TNF-α). Furthermore, the binding of p65 and cAMP response element binding protein (CREB) binding protein (CBP) or p300 decreased and NF-κB related genes which were inhibitors of NF-κB alpha (IκBα), A20, B cell lymphoma protein 2 (Bcl-2), and monocyte chemoattractant protein-1 (MCP-1) were low in cells transfected with Sec6 siRNAs in response to TNF-α stimulation. Sec6 knockdown decreased the expression of p90RSKs and the phosphorylation of ERK or p90RSK1 at Ser380 or IκBα at Ser32. The present study suggests that Sec6 regulates NF-κB transcriptional activity via the control of the phosphorylation of IκBα, p90RSK1, and ERK.
Collapse
Affiliation(s)
- Toshiaki Tanaka
- Department of Anatomy and Cell Biology, School of Medicine, Yamagata University, 2-2-2 Iidanishi, Yamagata, Japan.,Department of Dentistry, Oral and Maxillofacial Surgery, Plastic and Reconstructive Surgery, School of Medicine, Yamagata University, 2-2-2 Iidanishi, Yamagata, Japan
| | - Mitsuyoshi Iino
- Department of Dentistry, Oral and Maxillofacial Surgery, Plastic and Reconstructive Surgery, School of Medicine, Yamagata University, 2-2-2 Iidanishi, Yamagata, Japan
| |
Collapse
|
109
|
The Role of Protein Arginine Methyltransferases in Inflammatory Responses. Mediators Inflamm 2016; 2016:4028353. [PMID: 27041824 PMCID: PMC4793140 DOI: 10.1155/2016/4028353] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 02/14/2016] [Indexed: 12/29/2022] Open
Abstract
Protein arginine methyltransferases (PRMTs) mediate the methylation of a number of protein substrates of arginine residues and serve critical functions in many cellular responses, including cancer development, progression, and aggressiveness, T-lymphocyte activation, and hepatic gluconeogenesis. There are nine members of the PRMT family, which are divided into 4 types (types I–IV). Although most PRMTs do not require posttranslational modification (PTM) to be activated, fine-tuning modifications, such as interactions between cofactor proteins, subcellular compartmentalization, and regulation of RNA, via micro-RNAs, seem to be required. Inflammation is an essential defense reaction of the body to eliminate harmful stimuli, including damaged cells, irritants, or pathogens. However, chronic inflammation can eventually cause several types of diseases, including some cancers, atherosclerosis, rheumatoid arthritis, and periodontitis. Therefore, inflammation responses should be well modulated. In this review, we briefly discuss the role of PRMTs in the control of inflammation. More specifically, we review the roles of four PRMTs (CARM1, PRMT1, PRMT5, and PRMT6) in modulating inflammation responses, particularly in terms of modulating the transcriptional factors or cofactors related to inflammation. Based on the regulatory roles known so far, we propose that PRMTs should be considered one of the target molecule groups that modulate inflammatory responses.
Collapse
|
110
|
van den Bosch T, Boichenko A, Leus NGJ, Ourailidou ME, Wapenaar H, Rotili D, Mai A, Imhof A, Bischoff R, Haisma HJ, Dekker FJ. The histone acetyltransferase p300 inhibitor C646 reduces pro-inflammatory gene expression and inhibits histone deacetylases. Biochem Pharmacol 2015; 102:130-140. [PMID: 26718586 DOI: 10.1016/j.bcp.2015.12.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 12/14/2015] [Indexed: 12/26/2022]
Abstract
Lysine acetylations are reversible posttranslational modifications of histone and non-histone proteins that play important regulatory roles in signal transduction cascades and gene expression. Lysine acetylations are regulated by histone acetyltransferases as writers and histone deacetylases as erasers. Because of their role in signal transduction cascades, these enzymes are important players in inflammation. Therefore, histone acetyltransferase inhibitors could reduce inflammatory responses. Among the few histone acetyltransferase inhibitors described, C646 is one of the most potent (Ki of 0.4μM for histone acetyltransferase p300). C646 was described to affect the NF-κB pathway; an important pathway in inflammatory responses, which is regulated by acetylation. This pathway has been implicated in asthma and COPD. Therefore, we hypothesized that via regulation of the NF-κB signaling pathway, C646 can inhibit pro-inflammatory gene expression, and have potential for the treatment of inflammatory lung diseases. In line with this, we demonstrate here that C646 reduces pro-inflammatory gene expression in RAW264.7 murine macrophages and murine precision-cut lung slices. To unravel its effects on cellular substrates we applied mass spectrometry and found, counterintuitively, a slight increase in acetylation of histone H3. Based on this finding, and structural features of C646, we presumed inhibitory activity of C646 on histone deacetylases, and indeed found inhibition of histone deacetylases from 7μM and higher concentrations. This indicates that C646 has potential for further development towards applications in the treatment of inflammation, however, its newly discovered lack of selectivity at higher concentrations needs to be taken into account.
Collapse
Affiliation(s)
- Thea van den Bosch
- Pharmaceutical Gene Modulation, Groningen Research Institute of Pharmacy, University of Groningen, The Netherlands
| | - Alexander Boichenko
- Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, The Netherlands
| | - Niek G J Leus
- Pharmaceutical Gene Modulation, Groningen Research Institute of Pharmacy, University of Groningen, The Netherlands
| | - Maria E Ourailidou
- Pharmaceutical Gene Modulation, Groningen Research Institute of Pharmacy, University of Groningen, The Netherlands
| | - Hannah Wapenaar
- Pharmaceutical Gene Modulation, Groningen Research Institute of Pharmacy, University of Groningen, The Netherlands
| | - Dante Rotili
- Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, Italy
| | - Antonello Mai
- Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, Italy; Pasteur Institute, Cenci Bolognetti Foundation, Sapienza University of Rome, Italy
| | - Axel Imhof
- Protein Analysis Unit Biomedical Center and Center for Integrated Protein Science Munich, Ludwig-Maximilians University, Planegg-Martinsried, Germany
| | - Rainer Bischoff
- Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, The Netherlands
| | - Hidde J Haisma
- Pharmaceutical Gene Modulation, Groningen Research Institute of Pharmacy, University of Groningen, The Netherlands
| | - Frank J Dekker
- Pharmaceutical Gene Modulation, Groningen Research Institute of Pharmacy, University of Groningen, The Netherlands.
| |
Collapse
|
111
|
Judes G, Rifaï K, Ngollo M, Daures M, Bignon YJ, Penault-Llorca F, Bernard-Gallon D. A bivalent role of TIP60 histone acetyl transferase in human cancer. Epigenomics 2015; 7:1351-63. [PMID: 26638912 DOI: 10.2217/epi.15.76] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Acetylation is a major modification that is required for gene regulation, genome maintenance and metabolism. A dysfunctional acetylation plays an important role in several diseases, including cancer. A group of enzymes-lysine acetyltransferases are responsible for this modification and act in regulation of transcription as cofactors and by acetylation of histones and other proteins. Tip60, a member of MYST family, is expressed ubiquitously and is the acetyltransferase catalytic subunit of human NuA4 complex. This HAT has a well-characterized involvement in many processes, such as cellular signaling, DNA damage repair, transcriptional and cellular cycle. Aberrant lysine acetyltransferase functions promote or suppress tumorigenesis in different cancers such as colon, breast and prostate tumors. Therefore, Tip60 might be a potential and important therapeutic target in the cancer treatment; new histone acetyl transferase inhibitors were identified and are more selective inhibitors of Tip60.
Collapse
Affiliation(s)
- Gaëlle Judes
- Department of Oncogenetics, Centre Jean Perrin, CBRV, 28 place Henri Dunant, 63001 Clermont-Ferrand, France.,EA 4677 'ERTICA', University of Auvergne, 63011 Clermont-Ferrand, France
| | - Khaldoun Rifaï
- Department of Oncogenetics, Centre Jean Perrin, CBRV, 28 place Henri Dunant, 63001 Clermont-Ferrand, France.,EA 4677 'ERTICA', University of Auvergne, 63011 Clermont-Ferrand, France
| | - Marjolaine Ngollo
- Department of Oncogenetics, Centre Jean Perrin, CBRV, 28 place Henri Dunant, 63001 Clermont-Ferrand, France.,EA 4677 'ERTICA', University of Auvergne, 63011 Clermont-Ferrand, France
| | - Marine Daures
- Department of Oncogenetics, Centre Jean Perrin, CBRV, 28 place Henri Dunant, 63001 Clermont-Ferrand, France.,EA 4677 'ERTICA', University of Auvergne, 63011 Clermont-Ferrand, France
| | - Yves-Jean Bignon
- Department of Oncogenetics, Centre Jean Perrin, CBRV, 28 place Henri Dunant, 63001 Clermont-Ferrand, France.,EA 4677 'ERTICA', University of Auvergne, 63011 Clermont-Ferrand, France
| | - Frédérique Penault-Llorca
- EA 4677 'ERTICA', University of Auvergne, 63011 Clermont-Ferrand, France.,Centre Jean Perrin, Laboratory of Biopathology, 63011 Clermont-Ferrand, France
| | - Dominique Bernard-Gallon
- Department of Oncogenetics, Centre Jean Perrin, CBRV, 28 place Henri Dunant, 63001 Clermont-Ferrand, France.,EA 4677 'ERTICA', University of Auvergne, 63011 Clermont-Ferrand, France
| |
Collapse
|
112
|
Kuznetsova T, Wang SY, Rao NA, Mandoli A, Martens JHA, Rother N, Aartse A, Groh L, Janssen-Megens EM, Li G, Ruan Y, Logie C, Stunnenberg HG. Glucocorticoid receptor and nuclear factor kappa-b affect three-dimensional chromatin organization. Genome Biol 2015; 16:264. [PMID: 26619937 PMCID: PMC4665721 DOI: 10.1186/s13059-015-0832-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 11/11/2015] [Indexed: 01/25/2023] Open
Abstract
Background The impact of signal-dependent transcription factors, such as glucocorticoid receptor and nuclear factor kappa-b, on the three-dimensional organization of chromatin remains a topic of discussion. The possible scenarios range from remodeling of higher order chromatin architecture by activated transcription factors to recruitment of activated transcription factors to pre-established long-range interactions. Results Using circular chromosome conformation capture coupled with next generation sequencing and high-resolution chromatin interaction analysis by paired-end tag sequencing of P300, we observed agonist-induced changes in long-range chromatin interactions, and uncovered interconnected enhancer–enhancer hubs spanning up to one megabase. The vast majority of activated glucocorticoid receptor and nuclear factor kappa-b appeared to join pre-existing P300 enhancer hubs without affecting the chromatin conformation. In contrast, binding of the activated transcription factors to loci with their consensus response elements led to the increased formation of an active epigenetic state of enhancers and a significant increase in long-range interactions within pre-existing enhancer networks. De novo enhancers or ligand-responsive enhancer hubs preferentially interacted with ligand-induced genes. Conclusions We demonstrate that, at a subset of genomic loci, ligand-mediated induction leads to active enhancer formation and an increase in long-range interactions, facilitating efficient regulation of target genes. Therefore, our data suggest an active role of signal-dependent transcription factors in chromatin and long-range interaction remodeling. Electronic supplementary material The online version of this article (doi:10.1186/s13059-015-0832-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tatyana Kuznetsova
- Department of Molecular Biology, Faculty of Science Nijmegen, Radboud University, Nijmegen, The Netherlands.
| | - Shuang-Yin Wang
- Department of Molecular Biology, Faculty of Science Nijmegen, Radboud University, Nijmegen, The Netherlands.
| | - Nagesha A Rao
- Department of Molecular Biology, Faculty of Science Nijmegen, Radboud University, Nijmegen, The Netherlands.
| | - Amit Mandoli
- Department of Molecular Biology, Faculty of Science Nijmegen, Radboud University, Nijmegen, The Netherlands.
| | - Joost H A Martens
- Department of Molecular Biology, Faculty of Science Nijmegen, Radboud University, Nijmegen, The Netherlands.
| | - Nils Rother
- Department of Molecular Biology, Faculty of Science Nijmegen, Radboud University, Nijmegen, The Netherlands.
| | - Aafke Aartse
- Department of Molecular Biology, Faculty of Science Nijmegen, Radboud University, Nijmegen, The Netherlands.
| | - Laszlo Groh
- Department of Molecular Biology, Faculty of Science Nijmegen, Radboud University, Nijmegen, The Netherlands.
| | - Eva M Janssen-Megens
- Department of Molecular Biology, Faculty of Science Nijmegen, Radboud University, Nijmegen, The Netherlands.
| | - Guoliang Li
- National Key Laboratory of Crop Genetic Improvement, College of Informatics, Huazhong Agricultural University, Wuhan, China.
| | - Yijun Ruan
- The Jackson Laboratory for Genomic Medicine, and Department of Genetic and Development Biology, University of Connecticut, 400 Farmington Ave., Farmington, CT, 06030, USA.
| | - Colin Logie
- Department of Molecular Biology, Faculty of Science Nijmegen, Radboud University, Nijmegen, The Netherlands.
| | - Hendrik G Stunnenberg
- Department of Molecular Biology, Faculty of Science Nijmegen, Radboud University, Nijmegen, The Netherlands.
| |
Collapse
|
113
|
Tyagi M, Weber J, Bukrinsky M, Simon GL. The effects of cocaine on HIV transcription. J Neurovirol 2015; 22:261-74. [PMID: 26572787 DOI: 10.1007/s13365-015-0398-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 10/01/2015] [Accepted: 10/21/2015] [Indexed: 11/29/2022]
Abstract
Illicit drug users are a high-risk population for infection with the human immunodeficiency virus (HIV). A strong correlation exists between prohibited drug use and an increased rate of HIV transmission. Cocaine stands out as one of the most frequently abused illicit drugs, and its use is correlated with HIV infection and disease progression. The central nervous system (CNS) is a common target for both drugs of abuse and HIV, and cocaine intake further accelerates neuronal injury in HIV patients. Although the high incidence of HIV infection in illicit drug abusers is primarily due to high-risk activities such as needle sharing and unprotected sex, several studies have demonstrated that cocaine enhances the rate of HIV gene expression and replication by activating various signal transduction pathways and downstream transcription factors. In order to generate mature HIV genomic transcript, HIV gene expression has to pass through both the initiation and elongation phases of transcription, which requires discrete transcription factors. In this review, we will provide a detailed analysis of the molecular mechanisms that regulate HIV transcription and discuss how cocaine modulates those mechanisms to upregulate HIV transcription and eventually HIV replication.
Collapse
Affiliation(s)
- Mudit Tyagi
- Division of Infectious Diseases, Department of Medicine, The George Washington University, 2300 Eye Street, N.W., Washington, DC, 20037, USA. .,Department of Microbiology, Immunology and Tropical Medicine, The George Washington University, Washington, DC, 20037, USA.
| | - Jaime Weber
- Division of Infectious Diseases, Department of Medicine, The George Washington University, 2300 Eye Street, N.W., Washington, DC, 20037, USA
| | - Michael Bukrinsky
- Department of Microbiology, Immunology and Tropical Medicine, The George Washington University, Washington, DC, 20037, USA
| | - Gary L Simon
- Division of Infectious Diseases, Department of Medicine, The George Washington University, 2300 Eye Street, N.W., Washington, DC, 20037, USA
| |
Collapse
|
114
|
Chen H, Wang L, Zhou Z, Hou Z, Liu Z, Wang W, Gao D, Gao Q, Wang M, Song L. The comprehensive immunomodulation of NeurimmiRs in haemocytes of oyster Crassostrea gigas after acetylcholine and norepinephrine stimulation. BMC Genomics 2015; 16:942. [PMID: 26576764 PMCID: PMC4650145 DOI: 10.1186/s12864-015-2150-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 10/26/2015] [Indexed: 12/19/2022] Open
Abstract
Background Neural-endocrine-immune (NEI) system is a major modulation network among the nervous, endocrine and immune system and weights greatly in maintaining homeostasis of organisms during stress and infection. Some microRNAs are found interacting with NEI system (designated NeurimmiRs), addressing swift modulations on immune system. The oyster Crassostrea gigas, as an intertidal bivalve, has evolved a primary NEI system. However, the knowledge about NeurimmiRs in oysters remains largely unknown. Results Six small RNA libraries from haemocytes of oysters stimulated with acetylcholine (ACh) and norepinephrine (NE) were sequenced to identify neurotransmitter-responsive miRNAs and survey their immunomodulation roles. A total of 331 miRNAs (132 identified in the present study plus 199 identified previously) were subjected to expression analysis, and twenty-one and sixteen of them were found ACh- or NE-responsive, respectively (FDR < 0.05). Meanwhile, 21 miRNAs exhibited different expression pattern after ACh or NE stimulation. Consequently, 355 genes were predicted as putative targets of these neurotransmitter-responsive miRNAs in oyster. Through gene onthology analysis, multiple genes involved in death, immune system process and response to stimulus were annotated to be modulated by NeurimmiRs. Besides, a significant decrease in haemocyte phagocytosis and late-apoptosis or necrosis rate was observed after ACh and NE stimulation (p < 0.05) while early-apoptosis rate remained unchanged. Conclusions A comprehensive immune-related network involving PRRs, intracellular receptors, signaling transducers and immune effectors was proposed to be modulated by ACh- and NE-responsive NeurimmiRs, which would be indispensable for oyster haemocytes to respond against stress and infection. Characterization of the NeurimmiRs would be an essential step to understand the NEI system of invertebrate and the adaptation mechanism of oyster. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-2150-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hao Chen
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Lingling Wang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.
| | - Zhi Zhou
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
| | - Zhanhui Hou
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
| | - Zhaoqun Liu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Weilin Wang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Dahai Gao
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
| | - Qiang Gao
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
| | - Mengqiang Wang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
| | - Linsheng Song
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China. .,Key Laboratory of Mariculture & Stock enhancement in North China's Sea, Ministry of Agriculture, Dalian Ocean University, Dalian, 116023, China.
| |
Collapse
|
115
|
NFκB- and AP-1-mediated DNA looping regulates matrix metalloproteinase-9 transcription in TNF-α-treated human leukemia U937 cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2015; 1849:1248-59. [DOI: 10.1016/j.bbagrm.2015.07.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 07/29/2015] [Accepted: 07/31/2015] [Indexed: 01/08/2023]
|
116
|
Gobejishvili L, Ghare S, Khan R, Cambon A, Barker DF, Barve S, McClain C, Hill D. Misoprostol modulates cytokine expression through a cAMP pathway: Potential therapeutic implication for liver disease. Clin Immunol 2015; 161:291-9. [PMID: 26408955 DOI: 10.1016/j.clim.2015.09.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 07/09/2015] [Accepted: 09/15/2015] [Indexed: 12/20/2022]
Abstract
Dysregulated cytokine metabolism plays a critical role in the pathogenesis of many forms of liver disease, including alcoholic and non-alcoholic liver disease. In this study we examined the efficacy of Misoprostol in modulating LPS-inducible TNFα and IL-10 expression in healthy human subjects and evaluated molecular mechanisms for Misoprostol modulation of cytokines in vitro. Healthy subjects were given 14day courses of Misoprostol at doses of 100, 200, and 300μg four times a day, in random order. Baseline and LPS-inducible cytokine levels were examined ex vivo in whole blood at the beginning and the end of the study. Additionally, in vitro studies were performed using primary human PBMCs and the murine macrophage cell line, RAW 264.7, to investigate underlying mechanisms of misoprostol on cytokine production. Administration of Misoprostol reduced LPS inducible TNF production by 29%, while increasing IL-10 production by 79% in human subjects with no significant dose effect on ex vivo cytokine activity; In vitro, the effect of Misoprostol was largely mediated by increased cAMP levels and consequent changes in CRE and NFκB activity, which are critical for regulating IL-10 and TNF expression. Additionally, chromatin immunoprecipitation (ChIP) studies demonstrated that Misoprostol treatment led to changes in transcription factor and RNA Polymerase II binding, resulting in changes in mRNA levels. In summary, Misoprostol was effective at beneficially modulating TNF and IL-10 levels both in vivo and in vitro; these studies suggest a potential rationale for Misoprostol use in ALD, NASH and other liver diseases where inflammation plays an etiologic role.
Collapse
Affiliation(s)
- Leila Gobejishvili
- Department of Internal Medicine, University of Louisville, United States; Department of Pharmacology & Toxicology, University of Louisville, United States.
| | - Smita Ghare
- Department of Internal Medicine, University of Louisville, United States
| | - Rehan Khan
- RK Gastroenterology & Hepatology, Coppell, TX, United States
| | - Alexander Cambon
- Department of Bioinformatics & Biostatistics, University of Louisville, United States
| | - David F Barker
- Department of Internal Medicine, University of Louisville, United States
| | - Shirish Barve
- Department of Internal Medicine, University of Louisville, United States; Department of Pharmacology & Toxicology, University of Louisville, United States
| | - Craig McClain
- Department of Internal Medicine, University of Louisville, United States; Department of Pharmacology & Toxicology, University of Louisville, United States; VA Medical Center, Louisville, KY, United States
| | - Daniell Hill
- Department of Internal Medicine, University of Louisville, United States; VA Medical Center, Louisville, KY, United States
| |
Collapse
|
117
|
SUMOylation-disrupting WAS mutation converts WASp from a transcriptional activator to a repressor of NF-κB response genes in T cells. Blood 2015; 126:1670-82. [PMID: 26261240 DOI: 10.1182/blood-2015-05-646182] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 08/08/2015] [Indexed: 11/20/2022] Open
Abstract
In Wiskott-Aldrich syndrome (WAS), immunodeficiency and autoimmunity often comanifest, yet how WAS mutations misregulate chromatin-signaling in Thelper (TH) cells favoring development of auto-inflammation over protective immunity is unclear. Previously, we identified an essential promoter-specific, coactivator role of nuclear-WASp in TH1 gene transcription. Here we identify small ubiquitin-related modifier (SUMO)ylation as a novel posttranslational modification of WASp, impairment of which converts nuclear-WASp from a transcriptional coactivator to a corepressor of nuclear factor (NF)-κB response genes in human (TH)1-differentiating cells. V75M, one of many disease-causing mutations occurring in SUMO*motif (72-ψψψψKDxxxxSY-83) of WASp, compromises WASp-SUMOylation, associates with COMMD1 to attenuate NF-κB signaling, and recruits histone deacetylases-6 (HDAC6) to p300-marked promoters of NF-κB response genes that pattern immunity but not inflammation. Consequently, proteins mediating adaptive immunity (IFNG, STAT1, TLR1) are deficient, whereas those mediating auto-inflammation (GM-CSF, TNFAIP2, IL-1β) are paradoxically increased in TH1 cells expressing SUMOylation-deficient WASp. Moreover, SUMOylation-deficient WASp favors ectopic development of the TH17-like phenotype (↑IL17A, IL21, IL22, IL23R, RORC, and CSF2) under TH1-skewing conditions, suggesting a role for WASp in modulating TH1/TH17 plasticity. Notably, pan-histone deacetylase inhibitors lift promoter-specific repression imposed by SUMOylation-deficient WASp and restore misregulated gene expression. Our findings uncovering a SUMOylation-based mechanism controlling WASp's dichotomous roles in transcription may have implications for personalized therapy for patients carrying mutations that perturb WASp-SUMOylation.
Collapse
|
118
|
Abstract
Antiretroviral therapy (ART) inhibits HIV-1 replication, but the virus persists in latently infected resting memory CD4+ T cells susceptible to viral reactivation. The virus-encoded early gene product Tat activates transcription of the viral genome and promotes exponential viral production. Here we show that the Tat inhibitor didehydro-cortistatin A (dCA), unlike other antiretrovirals, reduces residual levels of viral transcription in several models of HIV latency, breaks the Tat-mediated transcriptional feedback loop, and establishes a nearly permanent state of latency, which greatly diminishes the capacity for virus reactivation. Importantly, treatment with dCA induces inactivation of viral transcription even after its removal, suggesting that the HIV promoter is epigenetically repressed. Critically, dCA inhibits viral reactivation upon CD3/CD28 or prostratin stimulation of latently infected CD4+ T cells from HIV-infected subjects receiving suppressive ART. Our results suggest that inclusion of a Tat inhibitor in current ART regimens may contribute to a functional HIV-1 cure by reducing low-level viremia and preventing viral reactivation from latent reservoirs. Antiretroviral therapy (ART) reduces HIV-1 replication to very low levels, but the virus persists in latently infected memory CD4+ T cells, representing a long-lasting source of resurgent virus upon ART interruption. Based on the mode of action of didehydro-cortistatin A (dCA), a Tat-dependent transcription inhibitor, our work highlights an alternative approach to current HIV-1 eradication strategies to decrease the latent reservoir. In our model, dCA blocks the Tat feedback loop initiated after low-level basal reactivation, blocking transcriptional elongation and hence viral production from latently infected cells. Therefore, dCA combined with ART would be aimed at delaying or halting ongoing viral replication, reactivation, and replenishment of the latent viral reservoir. Thus, the latent pool of cells in an infected individual would be stabilized, and death of the long-lived infected memory T cells would result in a continuous decay of this pool over time, possibly culminating in the long-awaited sterilizing cure.
Collapse
|
119
|
Ma B, Fey M, Hottiger MO. WNT/β-catenin signaling inhibits CBP-mediated RelA acetylation and expression of proinflammatory NF-κB target genes. J Cell Sci 2015; 128:2430-6. [PMID: 26021349 DOI: 10.1242/jcs.168542] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 05/21/2015] [Indexed: 12/24/2022] Open
Abstract
The discovery of functional crosstalk between WNT and nuclear factor κB (NF-κB) signaling has established a more complex role for these two pathways in inflammation and cancer. However, the molecular mechanisms of the crosstalk and its biological consequences are largely unknown. Here, we show that WNT/β-catenin signaling selectively inhibits the expression of a proinflammatory subset of IL-1β-induced NF-κB target genes. WNT/β-catenin signaling does not affect nuclear translocation of the RelA subunit of NF-κB or its association with CBP (also known as CREBBP), but reduces CBP-mediated acetylation and chromatin recruitment of RelA. Thus, β-catenin selectively regulates NF-κB gene expression through its negative effects on RelA acetylation. This anti-inflammatory effect may be relevant for cancer treatment.
Collapse
Affiliation(s)
- Bin Ma
- Institute of Veterinary Biochemistry and Molecular Biology, University of Zurich, Zurich 8057, Switzerland Division of Immunology, University Children's Hospital Zurich, Zurich 8032, Switzerland
| | - Monika Fey
- Institute of Veterinary Biochemistry and Molecular Biology, University of Zurich, Zurich 8057, Switzerland
| | - Michael O Hottiger
- Institute of Veterinary Biochemistry and Molecular Biology, University of Zurich, Zurich 8057, Switzerland
| |
Collapse
|
120
|
Abstract
Hyaluronan (HA) is a critical component of cancer microenvironment that is known to increase tumor progression and aggressiveness. The synthesis of HA starts from the cytosolic precursors UDP-N-acetylglucosamine and UDP-glucuronic acid. These two sugar nucleotides have several functions in addition to glycoconjugate synthesis and glucuronidation reactions, each of which can have a critical role in cancer. HA is synthesized by a family of three HA synthase (HAS) enzymes and, in this review, we described the main posttranslational modifications that are known to regulate HA metabolism. In particular, as the main HAS in adult tissues is HAS2, we focused on the role of AMPK-mediated phosphorylation and glycosylation by O-linked N-acetylglucosamine (O-GlcNAcylation) of HAS2 which mediate HAS2 inactivation and activation, respectively. HA catabolism, furnishing glucuronic acid and N-acetylglucosamine, can represent for a cancer cell a valid source of substrates to sustain complex tumor metabolism, and we highlight a presumable metabolic fate of such sugars in tumor cells.
Collapse
Affiliation(s)
- Davide Vigetti
- Dipartimento di Scienze Chirurgiche e Morfologiche, Università degli Studi dell'Insubria, Varese, Italy
| | - Alberto Passi
- Dipartimento di Scienze Chirurgiche e Morfologiche, Università degli Studi dell'Insubria, Varese, Italy.
| |
Collapse
|
121
|
Impact on antibody responses of B-cell-restricted transgenic expression of a viral gene inhibiting activation of NF-κB and NFAT. Arch Virol 2015; 160:1477-88. [PMID: 25864175 DOI: 10.1007/s00705-015-2419-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Accepted: 03/31/2015] [Indexed: 10/23/2022]
Abstract
In this work, we have assessed the impact in vivo of the evasion gene A238L of African swine fever virus, an inhibitor of both NF-κB- and NFAT-mediated transcription. The A238L gene was selectively expressed in mouse B lymphocytes using the promoter and enhancer sequences of the mouse Ig μ heavy chain. The IgM primary and IgG2b secondary serological responses and the number of splenic germinal centres in response to the TD antigens DNP-keyhole limpet hemocyanin and sheep red blood cells, respectively, were both lower in the transgenic mice, whereas the response to the TI type-1 and type-2 antigens DNP-Ficoll and DNP-LPS, respectively, were normal, except for the increased levels of IgG3 at day 14 in the DNP-LPS-immunized mice. Thus, it appears that neither p65 (NF-κB) nor NFAT is essential for B-cell development but, in a manner that is still unclear, may be relevant for their function.
Collapse
|
122
|
Wollebo HS, Bellizzi A, Cossari DH, Safak M, Khalili K, White MK. Epigenetic regulation of polyomavirus JC involves acetylation of specific lysine residues in NF-κB p65. J Neurovirol 2015; 21:679-87. [PMID: 25791343 DOI: 10.1007/s13365-015-0326-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 01/09/2015] [Accepted: 01/28/2015] [Indexed: 12/22/2022]
Abstract
Progressive multifocal leukoencephalopathy (PML) is a fatal demyelinating disease caused by neurotropic polyomavirus, JC virus (JCV), a virus that causes lytic infection of CNS glial cells. After primary infection, JCV is controlled by the immune system but virus persists asymptomatically. Rarely, when immune function is impaired, it can reemerge to cause PML. The mechanisms of JCV persistence and reactivation are not well understood but our earlier work implicated epigenetic control by protein acetylation since histone deacetylase inhibitors such as trichostatin A (TSA) strongly stimulate JCV transcription. Since both TNF-α and TSA activate JCV transcription via the same unique NF-κB site in the JCV control region, we investigated a role for acetylation of NF-κB in JCV regulation. A site-directed mutagenesis strategy was employed targeting the known lysine acetylation sites of NF-κB p65: K218, K221, and K310. We individually mutated each lysine to arginine, which cannot be acetylated and retains a positive charge like lysine. K218R and K221R impaired transactivation of JCV early promoter transcription either alone or combined with TSA treatment or coexpression of acetyltransferase transcriptional coactivator p300 but K310R was largely without effect. Mutation of lysine to glutamine gives mutants with a negative charge like acetyllysine. However, K218Q and K221Q showed impaired activity and only K310Q showed enhanced transactivation. NF-κB acetylation can regulate several aspects of the process of activation including complex formation with IκB, translocation to the nucleus, and DNA binding and transcriptional activation. Cell fractionation studies revealed that the mutants had no defect in translocation to the nucleus whereas gel shift studies revealed reduced binding to the JCV NF-κB site. Thus, acetylation regulates NF-κB p65 activity toward JCV at the level of p65 binding to the JCV control region and activation of JCV transcription.
Collapse
Affiliation(s)
- Hassen S Wollebo
- Center for Neurovirology, Department of Neuroscience, Temple University School of Medicine, Room 756 MERB, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Anna Bellizzi
- Center for Neurovirology, Department of Neuroscience, Temple University School of Medicine, Room 756 MERB, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Dominique H Cossari
- Center for Neurovirology, Department of Neuroscience, Temple University School of Medicine, Room 756 MERB, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Mahmut Safak
- Center for Neurovirology, Department of Neuroscience, Temple University School of Medicine, Room 756 MERB, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Kamel Khalili
- Center for Neurovirology, Department of Neuroscience, Temple University School of Medicine, Room 756 MERB, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Martyn K White
- Center for Neurovirology, Department of Neuroscience, Temple University School of Medicine, Room 756 MERB, 3500 N. Broad Street, Philadelphia, PA, 19140, USA.
| |
Collapse
|
123
|
Vahid F, Zand H, Nosrat-Mirshekarlou E, Najafi R, Hekmatdoost A. The role dietary of bioactive compounds on the regulation of histone acetylases and deacetylases: a review. Gene 2015; 562:8-15. [PMID: 25701602 DOI: 10.1016/j.gene.2015.02.045] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Revised: 01/25/2015] [Accepted: 02/15/2015] [Indexed: 12/18/2022]
Abstract
Nutrigenomics is an area of epigenomics that explores and defines the rapidly evolving field of diet-genome interactions. Lifestyle and diet can significantly influence epigenetic mechanisms, which cause heritable changes in gene expression without changes in DNA sequence. Nutrient-dependent epigenetic variations can significantly affect genome stability, mRNA and protein expression, and metabolic changes, which in turn influence food absorption and the activity of its constituents. Dietary bioactive compounds can affect epigenetic alterations, which are accumulated over time and are shown to be involved in the pathogenesis of age-related diseases such as diabetes, cancer, and cardiovascular disease. Histone acetylation is an epigenetic modification mediated by histone acetyl transferases (HATs) and histone deacetylases (HDACs) critically involved in regulating affinity binding between the histones and DNA backbone. The HDAC-mediated increase in histone affinity to DNA causes DNA condensation, preventing transcription, whereas HAT-acetylated chromatin is transcriptionally active. HDAC and HAT activities are reported to be associated with signal transduction, cell growth and death, as well as with the pathogenesis of various diseases. The aim of this review was to evaluate the role of diet and dietary bioactive compounds on the regulation of HATs and HDACs in epigenetic diseases. Dietary bioactive compounds such as genistein, phenylisothiocyanate, curcumin, resveratrol, indole-3-carbinol, and epigallocatechin-3-gallate can regulate HDAC and HAT activities and acetylation of histones and non-histone chromatin proteins, and their health benefits are thought to be attributed to these epigenetic mechanisms. The intake of dietary compounds that regulate epigenetic modifications can provide significant health effects and may prevent various pathological processes involved in the development of cancer and other life-threatening diseases.
Collapse
Affiliation(s)
- F Vahid
- Department of Nutritional Sciences, Nutrition and Food Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Nutrition and Food Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - H Zand
- Department of Nutritional Sciences, Nutrition and Food Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Nutrition and Food Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Cell and Molecular Science and Nutrition, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - E Nosrat-Mirshekarlou
- Department of Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada R3T 2N2
| | - R Najafi
- Department of Nutritional Sciences, Nutrition and Food Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Nutrition and Food Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - A Hekmatdoost
- Department of Nutritional Sciences, Nutrition and Food Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Nutrition and Food Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
124
|
Abstract
Acute pancreatitis is an inflammatory process of the pancreatic gland that eventually may lead to a severe systemic inflammatory response. A key event in pancreatic damage is the intracellular activation of NF-κB and zymogens, involving also calcium, cathepsins, pH disorders, autophagy, and cell death, particularly necrosis. This review focuses on the new role of redox signaling in acute pancreatitis. Oxidative stress and redox status are involved in the onset of acute pancreatitis and also in the development of the systemic inflammatory response, being glutathione depletion, xanthine oxidase activation, and thiol oxidation in proteins critical features of the disease in the pancreas. On the other hand, the release of extracellular hemoglobin into the circulation from the ascitic fluid in severe necrotizing pancreatitis enhances lipid peroxidation in plasma and the inflammatory infiltrate into the lung and up-regulates the HIF-VEGF pathway, contributing to the systemic inflammatory response. Therefore, redox signaling and oxidative stress contribute to the local and systemic inflammatory response during acute pancreatitis.
Collapse
|
125
|
Impact of viral activators and epigenetic regulators on HIV-1 LTRs containing naturally occurring single nucleotide polymorphisms. BIOMED RESEARCH INTERNATIONAL 2015; 2015:320642. [PMID: 25629043 PMCID: PMC4299542 DOI: 10.1155/2015/320642] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 09/29/2014] [Accepted: 10/14/2014] [Indexed: 12/17/2022]
Abstract
Following human immunodeficiency virus type 1 (HIV-1) integration into host cell DNA, the viral promoter can become transcriptionally silent in the absence of appropriate signals and factors. HIV-1 gene expression is dependent on regulatory elements contained within the long terminal repeat (LTR) that drive the synthesis of viral RNAs and proteins through interaction with multiple host and viral factors. Previous studies identified single nucleotide polymorphisms (SNPs) within CCAAT/enhancer binding protein (C/EBP) site I and Sp site III (3T, C-to-T change at position 3, and 5T, C-to-T change at position 5 of the binding site, respectively, when compared to the consensus B sequence) that are low affinity binding sites and correlate with more advanced stages of HIV-1 disease. Stably transfected cell lines containing the wild type, 3T, 5T, and 3T5T LTRs were developed utilizing bone marrow progenitor, T, and monocytic cell lines to explore the LTR phenotypes associated with these genotypic changes from an integrated chromatin-based microenvironment. Results suggest that in nonexpressing cell clones LTR-driven gene expression occurs in a SNP-specific manner in response to LTR activation or treatment with trichostatin A treatment, indicating a possible cell type and SNP-specific mechanism behind the epigenetic control of LTR activation.
Collapse
|
126
|
Jia Z, Nallasamy P, Liu D, Shah H, Li JZ, Chitrakar R, Si H, McCormick J, Zhu H, Zhen W, Li Y. Luteolin protects against vascular inflammation in mice and TNF-alpha-induced monocyte adhesion to endothelial cells via suppressing IΚBα/NF-κB signaling pathway. J Nutr Biochem 2014; 26:293-302. [PMID: 25577468 DOI: 10.1016/j.jnutbio.2014.11.008] [Citation(s) in RCA: 144] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 10/06/2014] [Accepted: 11/13/2014] [Indexed: 01/09/2023]
Abstract
Vascular inflammation plays a significant role in the pathogenesis of atherosclerosis. Luteolin, a naturally occurring flavonoid present in many medicinal plants and some commonly consumed fruits and vegetables, has received wide attention for its potential to improve vascular function in vitro. However, its effect in vivo and the molecular mechanism of luteolin at physiological concentrations remain unclear. Here, we report that luteolin as low as 0.5 μM significantly inhibited tumor necrosis factor (TNF)-α-induced adhesion of monocytes to human EA.hy 926 endothelial cells, a key event in triggering vascular inflammation. Luteolin potently suppressed TNF-α-induced expression of the chemokine monocyte chemotactic protein-1 (MCP-1) and adhesion molecules intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1), key mediators involved in enhancing endothelial cell-monocyte interaction. Furthermore, luteolin inhibited TNF-α-induced nuclear factor (NF)-κB transcriptional activity, IκBα degradation, expression of IκB kinase β and subsequent NF-κB p65 nuclear translocation in endothelial cells, suggesting that luteolin can inhibit inflammation by suppressing NF-κB signaling. In an animal study, C57BL/6 mice were fed a diet containing 0% or 0.6% luteolin for 3 weeks, and luteolin supplementation greatly suppressed TNF-α-induced increase in circulating levels of MCP-1/JE, CXCL1/KC and sICAM-1 in C57BL/6 mice. Consistently, dietary intake of luteolin significantly reduced TNF-α-stimulated adhesion of monocytes to aortic endothelial cells ex vivo. Histology shows that luteolin treatment prevented the eruption of endothelial lining in the intima layer of the aorta and preserved elastin fibers' delicate organization as shown by Verhoeff-Van Gieson staining. Immunohistochemistry studies further show that luteolin treatment also reduced VCAM-1 and monocyte-derived F4/80-positive macrophages in the aorta of TNF-α-treated mice. In conclusion, luteolin protects against TNF-α-induced vascular inflammation in both in vitro and in vivo models. This anti-inflammatory effect of luteolin may be mediated via inhibition of the NF-κB-mediated pathway.
Collapse
Affiliation(s)
- Zhenquan Jia
- Department of Biology, The University of North Carolina at Greensboro, Greensboro, NC 27412.
| | - Palanisamy Nallasamy
- Department of Biology, The University of North Carolina at Greensboro, Greensboro, NC 27412
| | - Dongmin Liu
- Departments of Human Nutrition, Foods and Exercise, College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, VA 24062, USA.
| | - Halley Shah
- Department of Biology, The University of North Carolina at Greensboro, Greensboro, NC 27412
| | - Jason Z Li
- College and Graduate School of Arts & Sciences, University of Virginia, Charlottesville, VA 22904
| | - Rojin Chitrakar
- Department of Biology, The University of North Carolina at Greensboro, Greensboro, NC 27412
| | - Hongwei Si
- Department of Family Consumer Sciences, College of Agriculture, Human and Natural Sciences, Tennessee State University, Nashville, TN 37209, USA
| | - John McCormick
- Department of Biology, The University of North Carolina at Greensboro, Greensboro, NC 27412
| | - Hong Zhu
- Department of Pharmacology, Campbell University, School of Osteopathic Medicine, Buies Creek, NC 27506, USA
| | - Wei Zhen
- Departments of Human Nutrition, Foods and Exercise, College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, VA 24062, USA
| | - Yunbo Li
- Department of Biology, The University of North Carolina at Greensboro, Greensboro, NC 27412; Department of Pharmacology, Campbell University, School of Osteopathic Medicine, Buies Creek, NC 27506, USA.
| |
Collapse
|
127
|
Tang ZH, Wang L, Zeng F, Zhang K. Human genetics of diabetic retinopathy. J Endocrinol Invest 2014; 37:1165-74. [PMID: 25201002 DOI: 10.1007/s40618-014-0172-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 08/25/2014] [Indexed: 01/03/2023]
Abstract
There is evidence demonstrating that genetic factors contribute to the risk of diabetic retinopathy (DR). Genetics variants, structural variants (copy number variation, CNV) and epigenetic changes play important roles in the development of DR. Genetic linkage and association studies have uncovered a number of genetic loci and common genetic variants susceptibility to DR. CNV and interactions of gene by environment have also been detected by association analysis. Apart from nucleus genome, mitochondrial DNA plays critical roles in regulation of development of DR. Epigenetic studies have indicated epigenetic changes in chromatin affecting gene transcription in response to environmental stimuli, which provided a large body of evidence of regulating development of diabetes mellitus. Identification of genetic variants and epigenetic changes contributed to risk or protection of DR will benefit uncovering the complex mechanism underlying DR. This review focused on the current knowledge of the genetic and epigenetic basis of DR.
Collapse
Affiliation(s)
- Z-H Tang
- Department of Endocrinology and Metabolism, Shanghai Tongji Hospital, Tongji University School of Medicine, Room 517 Building 2nd, NO. 389 Xincun Road, Shanghai, 200063, China,
| | | | | | | |
Collapse
|
128
|
Wright RC, Khakhar A, Eshleman JR, Ostermeier M. Advancements in the development of HIF-1α-activated protein switches for use in enzyme prodrug therapy. PLoS One 2014; 9:e114032. [PMID: 25426963 PMCID: PMC4245239 DOI: 10.1371/journal.pone.0114032] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 11/03/2014] [Indexed: 12/25/2022] Open
Abstract
While gene-directed enzyme prodrug therapy has shown potential as a cancer therapeutic in animal and clinical trials, concerns over the efficacy, selectivity, and safety of gene delivery vehicles have restricted its advance. In an attempt to relieve some of the demands on targeted gene delivery vehicles and achieve the full potential of enzyme prodrug therapy, cancer-targeted activity can be engineered into the enzyme itself. We previously engineered a switchable prodrug-activating enzyme that selectively kills human cancer cells accumulating the cancer marker hypoxia-inducible factor-1α (HIF-1α). This HIF-1α-activated protein switch (Haps59) is designed to increase its ability to convert the prodrug 5-fluorocytosine into the chemotherapeutic 5-fluorouracil in a HIF-1α-dependent manner. However, in cancer cell lines expressing Haps59 the 5FC sensitivity difference between the presence and absence of HIF-1α was not as large as desired. In this work, we aimed to improve the cancer specificity of this switch via a directed evolution approach utilizing random mutagenesis, linker mutagenesis, and random insertion and circular permutation. We identified improved HIF-1α-activated protein switches that confer E. coli with modest increases in HIF-1α-dependent 5FC toxicity. Additionally, the current bottleneck in the development of improved HIF-1α-activated protein switches is screening switch candidates in mammalian cells. To accommodate higher throughput and reduce experimental variability, we explored the use of Flp recombinase-mediated isogenic integration in 293 cells. These experiments raised the possibility that Haps59 can be activated by other interactors of the CH1 domain, and experiments in E. coli indicated that CITED2 can also activate Haps59. Although many CH1 binding partners are also oncogenes, CH1's promiscuous binding and subsequent off-target activation of Haps59 needs to be examined under normal physiological conditions to identify off-target activators. With aberrant activating molecules identified, further directed evolution can be performed to improve the cancer specificity of HIF-1α-activated protein switches.
Collapse
Affiliation(s)
- R. Clay Wright
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Arjun Khakhar
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - James R. Eshleman
- Departments of Pathology and Oncology, Sol Goldman Pancreatic Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Marc Ostermeier
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
129
|
Locus-specific epigenetic remodeling controls addiction- and depression-related behaviors. Nat Neurosci 2014; 17:1720-7. [PMID: 25347353 DOI: 10.1038/nn.3871] [Citation(s) in RCA: 170] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 10/16/2014] [Indexed: 12/11/2022]
Abstract
Chronic exposure to drugs of abuse or stress regulates transcription factors, chromatin-modifying enzymes and histone post-translational modifications in discrete brain regions. Given the promiscuity of the enzymes involved, it has not yet been possible to obtain direct causal evidence to implicate the regulation of transcription and consequent behavioral plasticity by chromatin remodeling that occurs at a single gene. We investigated the mechanism linking chromatin dynamics to neurobiological phenomena by applying engineered transcription factors to selectively modify chromatin at a specific mouse gene in vivo. We found that histone methylation or acetylation at the Fosb locus in nucleus accumbens, a brain reward region, was sufficient to control drug- and stress-evoked transcriptional and behavioral responses via interactions with the endogenous transcriptional machinery. This approach allowed us to relate the epigenetic landscape at a given gene directly to regulation of its expression and to its subsequent effects on reward behavior.
Collapse
|
130
|
Chung MH, Kim DH, Na HK, Kim JH, Kim HN, Haegeman G, Surh YJ. Genistein inhibits phorbol ester-induced NF-κB transcriptional activity and COX-2 expression by blocking the phosphorylation of p65/RelA in human mammary epithelial cells. Mutat Res 2014; 768:74-83. [PMID: 24742714 DOI: 10.1016/j.mrfmmm.2014.04.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2014] [Revised: 03/30/2014] [Accepted: 04/03/2014] [Indexed: 06/03/2023]
Abstract
Genistein, an isoflavone present in soy products, has chemopreventive effects on mammary carcinogenesis. In the present study, we have investigated the effects of genistein on phorbol ester-induced expression of cyclooxygenase-2 (COX-2) that plays an important role in the pathophysiology of inflammation-associated carcinogenesis. Pretreatment of cultured human breast epithelial (MCF10A) cells with genistein reduced COX-2 expression induced by 12-O-tetradecanoylphorbol-13-acetate (TPA). There are multiple lines of evidence supporting that the induction of COX-2 is regulated by the eukaryotic transcription factor NF-κB. Genistein failed to inhibit TPA-induced nuclear translocation and DNA binding of NF-κB as well as degradation of IκB. However, genistein abrogated the TPA-induced transcriptional activity of NF-κB as determined by the luciferase reporter gene assay. Genistein inhibited phosphorylation of the p65 subunit of NF-κB and its interaction with cAMP regulatory element-binding protein-binding protein (CBP)/p300 and TATA-binding protein (TBP). TPA-induced NF-κB phosphorylation was abolished by pharmacological inhibition of extracellular signal-regulated kinase (ERK). Likewise, pharmacologic inhibition or dominant negative mutation of ERK suppressed phosphorylation of p65. The above findings, taken together, suggest that genistein inhibits TPA-induced COX-2 expression in MCF10A cells by blocking ERK-mediated phosphorylation of p65 and its subsequent interaction with CBP and TBP.
Collapse
Affiliation(s)
- Myung-Hoon Chung
- Research Institute for Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Do-Hee Kim
- Research Institute for Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Hye-Kyung Na
- Department of Food and Nutrition, Sungshin Women's University, Seoul, South Korea
| | - Jung-Hwan Kim
- Research Institute for Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Ha-Na Kim
- Research Institute for Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, South Korea
| | | | - Young-Joon Surh
- Research Institute for Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, South Korea; Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul, South Korea; Cancer Research Institute, Seoul National University, Seoul, South Korea.
| |
Collapse
|
131
|
Marwarha G, Raza S, Meiers C, Ghribi O. Leptin attenuates BACE1 expression and amyloid-β genesis via the activation of SIRT1 signaling pathway. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1842:1587-95. [PMID: 24874077 PMCID: PMC4125440 DOI: 10.1016/j.bbadis.2014.05.015] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 05/13/2014] [Accepted: 05/16/2014] [Indexed: 02/07/2023]
Abstract
The aspartyl protease β-site AβPP-cleaving enzyme 1 (BACE1) catalyzes the rate-limiting step in Aβ production, a peptide at the nexus of neurodegenerative cascades in Alzheimer Disease (AD). The adipocytokine leptin has been demonstrated to reduce Aβ production and decrease BACE1 activity and expression levels. However, the signaling cascades involved in the leptin-induced mitigation in Aβ levels and BACE1 expression levels have not been elucidated. We have demonstrated that the transcription factor nuclear factor - kappa B (NF-κB) positively regulates BACE1 transcription. NF-κB activity is tightly regulated by the mammalian sirtuin SIRT1. Multiple studies have cogently evinced that leptin activates the metabolic master regulator SIRT1. In this study, we determined the extent to which SIRT1 expression and activity regulate the leptin-induced attenuation in BACE1 expression and Aβ levels in cultured human neuroblastoma SH-SY5Y cells. This study also elucidated and delineated the signal transduction pathways involved in the leptin induced mitigation in BACE1 expression. Our results demonstrate for the first time that leptin attenuates the activation and transcriptional activity of NF-κB by reducing the acetylation of the p65 subunit in a SIRT1-dependent manner. Furthermore, our data shows that leptin reduces the NF-κB-mediated transcription of BACE1 and consequently reduces Amyloid-β genesis. Our study provides a valuable insight and a novel mechanism by which leptin reduces BACE1 expression and Amyloid-β production and may help design potential therapeutic interventions.
Collapse
Affiliation(s)
- Gurdeep Marwarha
- Department of Pharmacology, Physiology and Therapeutics, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | - Shaneabbas Raza
- Department of Pharmacology, Physiology and Therapeutics, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | - Craig Meiers
- Department of Pharmacology, Physiology and Therapeutics, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | - Othman Ghribi
- Department of Pharmacology, Physiology and Therapeutics, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND 58202, USA.
| |
Collapse
|
132
|
Sun S, Guo M, Zhang JB, Ha A, Yokoyama KK, Chiu RH. Cyclophilin A (CypA) interacts with NF-κB subunit, p65/RelA, and contributes to NF-κB activation signaling. PLoS One 2014; 9:e96211. [PMID: 25119989 PMCID: PMC4130471 DOI: 10.1371/journal.pone.0096211] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 04/04/2014] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Peptidyl-prolyl isomerase cyclophilin A (CypA) plays important roles in signaling, protein translocation, inflammation, and cancer formation. However, little is known about the mechanisms by which CypA exerts its effects. C57BL/6 Ppia (encoding CypA)-deficient embryonic fibroblasts show reduced activation of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), the p65/RelA subunit, suggesting that CypA may mediate modulation of NF-κB activity to exert its biological effects. METHODOLOGY Western blotting and qRT-PCR analyses were used to evaluate the association of CypA deficiency with reduced activation of NF-κB/p65 at the protein level. GST pull-down and co-immunoprecipitation were used to examine interactions between CypA and p65/RelA. Truncation mutants and site-directed mutagenesis were used to determine the sequences of p65/RelA required for interactions with CypA. Enhancement of p65/RelA nuclear translocation by CypA was assessed by co-transfection and immunofluorescent imaging. Treatment of cells with cycloheximide that were harvested at various time points for Western blot analyses was carried out to evaluate p65/RelA protein stability. The functional activity of NF-κB was assessed by electrophoretic mobility-shift assays (EMSA), luciferase assays, and changes in expression levels of target genes. RESULTS GST pull-down assays in vitro and co-immunoprecipitation analyses in vivo provided evidence for protein-protein interactions. These interactions were further supported by identification of a CypA-binding consensus-like sequence within NF-κB subunit p65 at the N-terminal 170-176 amino acid residues. Significantly, CypA provided stability for NF-κB p65 and promoted NF-κB p65 nuclear translocation, resulting in increased nuclear accumulation and enhanced NF-κB activity. CONCLUSIONS Our findings revealed important mechanisms that regulate NF-κB activation, and offer new insights into the role of CypA in aberrant activation of NF-κB-mediated signaling for altered expression of its target genes, resulting in pathological effects in various diseases.
Collapse
Affiliation(s)
- Shan Sun
- Dental and Craniofacial Research Institute and School of Dentistry, University of California, Los Angeles, CA, United States of America
| | - Mian Guo
- Dental and Craniofacial Research Institute and School of Dentistry, University of California, Los Angeles, CA, United States of America
- Department of Neurosurgery, the 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilonjiang, China
| | - James Beiji Zhang
- Dental and Craniofacial Research Institute and School of Dentistry, University of California, Los Angeles, CA, United States of America
| | - Albert Ha
- Dental and Craniofacial Research Institute and School of Dentistry, University of California, Los Angeles, CA, United States of America
| | | | - Robert H. Chiu
- Dental and Craniofacial Research Institute and School of Dentistry, University of California, Los Angeles, CA, United States of America
- Surgical Oncology & Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, United States of America
| |
Collapse
|
133
|
Pal S, Bhattacharjee A, Ali A, Mandal NC, Mandal SC, Pal M. Chronic inflammation and cancer: potential chemoprevention through nuclear factor kappa B and p53 mutual antagonism. JOURNAL OF INFLAMMATION-LONDON 2014; 11:23. [PMID: 25152696 PMCID: PMC4142057 DOI: 10.1186/1476-9255-11-23] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 06/28/2014] [Indexed: 12/13/2022]
Abstract
Activation of nuclear factor-kappa B (NF- κB) as a mechanism of host defense against infection and stress is the central mediator of inflammatory responses. A normal (acute) inflammatory response is activated on urgent basis and is auto-regulated. Chronic inflammation that results due to failure in the regulatory mechanism, however, is largely considered as a critical determinant in the initiation and progression of various forms of cancer. Mechanistically, NF- κB favors this process by inducing various genes responsible for cell survival, proliferation, migration, invasion while at the same time antagonizing growth regulators including tumor suppressor p53. It has been shown by various independent investigations that a down regulation of NF- κB activity directly, or indirectly through the activation of the p53 pathway reduces tumor growth substantially. Therefore, there is a huge effort driven by many laboratories to understand the NF- κB signaling pathways to intervene the function of this crucial player in inflammation and tumorigenesis in order to find an effective inhibitor directly, or through the p53 tumor suppressor. We discuss here on the role of NF- κB in chronic inflammation and cancer, highlighting mutual antagonism between NF- κB and p53 pathways in the process. We also discuss prospective pharmacological modulators of these two pathways, including those that were already tested to affect this mutual antagonism.
Collapse
Affiliation(s)
- Srabani Pal
- Pharmacognosy and Phytotherapy laboratory, Division of Pharmacognosy, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Ashish Bhattacharjee
- Department of Biotechnology, National Institute of Technology, Durgapur-713209, India
| | - Asif Ali
- Division of Molecular Medicine, Bose Institute, Kolkata 700054, India
| | | | - Subhash C Mandal
- Pharmacognosy and Phytotherapy laboratory, Division of Pharmacognosy, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Mahadeb Pal
- Division of Molecular Medicine, Bose Institute, Kolkata 700054, India
| |
Collapse
|
134
|
Thangjam GS, Dimitropoulou C, Joshi AD, Barabutis N, Shaw MC, Kovalenkov Y, Wallace CM, Fulton DJ, Patel V, Catravas JD. Novel mechanism of attenuation of LPS-induced NF-κB activation by the heat shock protein 90 inhibitor, 17-N-allylamino-17-demethoxygeldanamycin, in human lung microvascular endothelial cells. Am J Respir Cell Mol Biol 2014; 50:942-52. [PMID: 24303801 DOI: 10.1165/rcmb.2013-0214oc] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Heat shock protein (hsp) 90 inhibition attenuates NF-κB activation and blocks inflammation. However, the precise mechanism of NF-κB regulation by hsp90 in the endothelium is not clear. We investigated the mechanisms of hsp90 inhibition by 17-N-allylamino-17-demethoxygeldanamycin (17-AAG) on NF-κB activation by LPS in primary human lung microvascular endothelial cells. Transcriptional activation of NF-κB was measured by luciferase reporter assay, gene expression by real-time RT-PCR, DNA binding of transcription factors by chromatin immunoprecipitation assay, protein-protein interaction by coimmunoprecipitation/immunoblotting, histone deacetylase (HDAC)/histone acetyltransferase enzyme activity by fluorometry, and nucleosome eviction by partial microccocal DNase digestion. In human lung microvascular endothelial cells, 17-AAG-induced degradation of IKBα was accomplished regardless of the phosphorylation/ubiquitination state of the protein. Hence, 17-AAG did not block LPS-induced NF-κB nuclear translocation and DNA binding activity. Instead, 17-AAG blocked the recruitment of the coactivator, cAMP response element binding protein binding protein, and prevented the assembly of a transcriptionally competent RNA polymerase II complex at the κB elements of the IKBα (an NF-κB-responsive gene) promoter. The effect of LPS on IKBα mRNA expression was associated with rapid deacetylation of histone-H3(Lys9) and a dramatic down-regulation of core histone H3 binding. Even though treatment with an HDAC inhibitor produced the same effect as hsp90 inhibition, the effect of 17-AAG was independent of HDAC. We conclude that hsp90 inhibition attenuates NF-κB transcriptional activation by preventing coactivator recruitment and nucleosome eviction from the target promoter in human lung endothelial cells.
Collapse
|
135
|
Park CK, Lee Y, Kim KH, Lee ZH, Joo M, Kim HH. Nrf2 is a novel regulator of bone acquisition. Bone 2014; 63:36-46. [PMID: 24521946 DOI: 10.1016/j.bone.2014.01.025] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 01/15/2014] [Accepted: 01/31/2014] [Indexed: 12/31/2022]
Abstract
Nuclear factor E2 p45-related factor 2 (Nrf2) is a transcription factor involved in the expression of cytoprotective genes induced by external stresses. We investigated the role of Nrf2 in osteoclast and osteoblast differentiation. Nrf2 knockdown or deletion increased osteoclastic differentiation from bone marrow-derived macrophages (BMMs) through the upregulation of NF-κB, c-Fos, and NFATc1 transcription factors. Nrf2 also inhibited osteoblast differentiation and mineralization via suppression of key regulatory proteins, such as Runx2, osteocalcin, and osterix. Micro-computed tomography and histomorphometric analyses showed an increase in bone mass of Nrf2 knockout compared to that of wild type mice. In addition, the mineral apposition rate and the number of osteoblasts in bone were higher in Nrf2 knockout mice. However, bone resorption parameters, namely DPD and CTX levels, were not affected by Nrf2 deletion. In a coculture condition where calvarial osteoblasts and BMMs from wild type and Nrf2 knockout mice were grown, deletion of Nrf2 in osteoblasts markedly reduced osteoclast formation. This effect was due to an increase in OPG expression in Nrf2 knockout osteoblasts. Taken as a whole, these results indicate that Nrf2 is intrinsically inhibitory to both osteoblast and osteoclast differentiation but its effect on osteoblasts is dominant to its effect on osteoclasts in vivo.
Collapse
Affiliation(s)
- Cheol Kyu Park
- Department of Cell and Developmental Biology, BK21 Program, Dental Research Institute, Seoul National University, Seoul 110-749, Republic of Korea
| | - Youngkyun Lee
- Department of Biochemistry, School of Dentistry, Kyungpook National University, Daegu 700-412, Republic of Korea
| | - Kyun Ha Kim
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan 626-870, Republic of Korea
| | - Zang Hee Lee
- Department of Cell and Developmental Biology, BK21 Program, Dental Research Institute, Seoul National University, Seoul 110-749, Republic of Korea
| | - Myungsoo Joo
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan 626-870, Republic of Korea.
| | - Hong-Hee Kim
- Department of Cell and Developmental Biology, BK21 Program, Dental Research Institute, Seoul National University, Seoul 110-749, Republic of Korea.
| |
Collapse
|
136
|
HPV16 E2-mediated potentiation of NF-κB activation induced by TNF-α involves parallel activation of STAT3 with a reduction in E2-induced apoptosis. Mol Cell Biochem 2014; 394:77-90. [PMID: 24833467 DOI: 10.1007/s11010-014-2083-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 05/03/2014] [Indexed: 02/08/2023]
Abstract
Human papilloma virus is associated with cervical and other tumors, and several cellular conditions also play an important role in carcinogenesis. Human papilloma virus (HPV)-infected cells exhibit activation of NF-κB and STAT3 (mediators of inflammation), but little is known about their regulation by HPV. This study attempts to understand the role of HPV16 E2, an important early protein of HPV16, in the regulation of NF-κB and STAT3 by reporter assays, quantitative reverse transcriptase-polymerase chain reaction, and immunoblotting. We demonstrate that E2 enhances NF-κB activation induced by TNF-α, a proinflammatory cytokine, in both non-tumor- and tumor-derived epithelial cell lines besides potentiating STAT3 transcriptional activity induced by TNF-α in HEK293 cells. E2 increases the expression of RelA and its transcriptional activation, and retention of E2 was observed in the nucleus with significant interaction with RelA (immunoprecipitation) upon TNF-α treatment. Transfection with shRNA-RelA or pretreatment with a STAT3 inhibitor had a negative effect on the ability of E2 to enhance TNF-α-induced NF-κB activation. Experiments with co-expression of a mutant of STAT3 with E2 also suggested that the activation of STAT3 is indispensible for TNF-α-induced NF-κB activation. Inhibition of STAT3 activation enhanced E2-induced apoptosis, whereas parallel activation of NF-κB and STAT3 by the combined action of E2 and TNF-α increased the expression of their common targets, cyclin-D1, c-Myc, survivin, and Bcl-2, leading to a decrease in E2-induced apoptosis (viability and cell cycle). Our results reveal novel mechanisms by which E2 may regulate NF-κB and STAT3 activation in the presence of TNF-α with implications on the survival of HPV-infected cells.
Collapse
|
137
|
Nallasamy P, Si H, Babu PVA, Pan D, Fu Y, Brooke EAS, Shah H, Zhen W, Zhu H, Liu D, Li Y, Jia Z. Sulforaphane reduces vascular inflammation in mice and prevents TNF-α-induced monocyte adhesion to primary endothelial cells through interfering with the NF-κB pathway. J Nutr Biochem 2014; 25:824-33. [PMID: 24880493 DOI: 10.1016/j.jnutbio.2014.03.011] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 02/24/2014] [Accepted: 03/12/2014] [Indexed: 01/10/2023]
Abstract
Sulforaphane, a naturally occurring isothiocyanate present in cruciferous vegetables, has received wide attention for its potential to improve vascular function in vitro. However, its effect in vivo and the molecular mechanism of sulforaphane at physiological concentrations remain unclear. Here, we report that a sulforaphane concentration as low as 0.5 μM significantly inhibited tumor necrosis factor-α (TNF-α)-induced adhesion of monocytes to human umbilical vein endothelial cells, a key event in the pathogenesis of atherosclerosis both in static and under flow conditions. Such physiological concentrations of sulforaphane also significantly suppressed TNF-α-induced production of monocyte chemotactic protein-1 and adhesion molecules including soluble vascular adhesion molecule-1 and soluble E-selectin, key mediators in the regulation of enhanced endothelial cell-monocyte interaction. Furthermore, sulforaphane inhibited TNF-α-induced nuclear factor (NF)-κB transcriptional activity, Inhibitor of NF-κB alpha (IκBα) degradation and subsequent NF-κB p65 nuclear translocation in endothelial cells, suggesting that sulforaphane can inhibit inflammation by suppressing NF-κB signaling. In an animal study, sulforaphane (300 ppm) in a mouse diet significantly abolished TNF-α-increased ex vivo monocyte adhesion and circulating adhesion molecules and chemokines in C57BL/6 mice. Histology showed that sulforaphane treatment significantly prevented the eruption of endothelial lining in the intima layer of the aorta and preserved elastin fibers' delicate organization, as shown by Verhoeff-van Gieson staining. Immunohistochemistry studies showed that sulforaphane treatment also reduced vascular adhesion molecule-1 and monocyte-derived F4/80-positive macrophages in the aorta of TNF-α-treated mice. In conclusion, sulforaphane at physiological concentrations protects against TNF-α-induced vascular endothelial inflammation, in both in vitro and in vivo models. This anti-inflammatory effect of sulforaphane may be, at least in part, associated with interfering with the NF-κB pathway.
Collapse
Affiliation(s)
- Palanisamy Nallasamy
- Department of Biology, The University of North Carolina at Greensboro, Greensboro, NC 27412, USA
| | - Hongwei Si
- Department of Family and Consumer Sciences, College of Agriculture, Human and Natural Sciences, Tennessee State University, Nashville, TN 37209, USA
| | | | - Dengke Pan
- Department of Gene and Cell Engineering, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Yu Fu
- Departments of Human Nutrition, Foods and Exercise, College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, VA 24062, USA
| | - Elizabeth A S Brooke
- Department of Biology, The University of North Carolina at Greensboro, Greensboro, NC 27412, USA
| | - Halley Shah
- Department of Biology, The University of North Carolina at Greensboro, Greensboro, NC 27412, USA
| | - Wei Zhen
- Departments of Human Nutrition, Foods and Exercise, College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, VA 24062, USA
| | - Hong Zhu
- Department of Pharmacology, School of Osteopathic Medicine, Campbell University, Buies Creek, NC 27506, USA
| | - Dongmin Liu
- Departments of Human Nutrition, Foods and Exercise, College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, VA 24062, USA.
| | - Yunbo Li
- Department of Pharmacology, School of Osteopathic Medicine, Campbell University, Buies Creek, NC 27506, USA.
| | - Zhenquan Jia
- Department of Biology, The University of North Carolina at Greensboro, Greensboro, NC 27412, USA.
| |
Collapse
|
138
|
Interaction of transactive response DNA binding protein 43 with nuclear factor κB in mild cognitive impairment with episodic memory deficits. Acta Neuropathol Commun 2014; 2:37. [PMID: 24690380 PMCID: PMC4230634 DOI: 10.1186/2051-5960-2-37] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 03/20/2014] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Transactive response DNA binding protein 43 (TDP-43) is detected in pathological inclusions in many cases of Alzheimer's disease (AD) and mild cognitive impairment (MCI), but its pathological role in AD and MCI remains unknown. Recently, TDP-43 was reported to contribute to pathogenesis in amyotrophic lateral sclerosis through its interaction with p65 nuclear factor κB (NF-κB) resulting in abnormal hyperactivation of this signaling pathway in motor neurons. Hence, we investigated the interaction of TDP-43 with p65 in the temporal cortex of subjects with a clinical diagnosis of MCI (n = 12) or AD (n = 12) as well as of age-matched controls with no cognitive impairment (NCI, n = 12). RESULTS Immunoprecipitation and immunofluorescence approaches revealed a robust interaction of TDP-43 with p65 in the nucleus of temporal lobe neurons in four individuals with MCI (named MCI-p). These MCI-p cases exhibited high expression levels of soluble TDP-43, p65, phosphorylated p65 and low expression levels of β-amyloid 40 when compared to AD or NCI cases. The analysis of cognitive performance tests showed that MCI-p individuals presented intermediate deficits of global cognition and episodic memory between those of AD cases and of NCI cases and MCI cases with no interaction of TDP-43 with p65. CONCLUSIONS From these results, we propose that enhanced NF-κB activation due to TDP-43 and p65 interaction may contribute to neuronal dysfunction in MCI individuals with episodic memory deficits. Accordingly, treatment with inhibitors of NF-κB activation may be considered for MCI individuals with episodic memory deficits.
Collapse
|
139
|
Kim HJ, Lee W, Yun JM. Luteolin inhibits hyperglycemia-induced proinflammatory cytokine production and its epigenetic mechanism in human monocytes. Phytother Res 2014; 28:1383-91. [PMID: 24623679 DOI: 10.1002/ptr.5141] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 01/26/2014] [Accepted: 02/16/2014] [Indexed: 12/26/2022]
Abstract
Hyperglycemia is a key feature in diabetes. Hyperglycemia has been implicated as a major contributor to several complications of diabetes. High glucose levels induce the release of proinflammatory cytokines. Luteolin is a flavone isolated from celery, green pepper, perilla leaf, and chamomile tea. Luteolin has been reported to possess antimutagenic, antitumorigenic, antioxidant, and anti-inflammatory properties. In this study, we investigated the effects of luteolin on proinflammatory cytokine secretion and its underlying epigenetic regulation in high-glucose-induced human monocytes. Human monocytic (THP-1) cells were cultured under controlled (14.5 mM mannitol), normoglycemic (NG, 5.5 mM glucose), or hyperglycemic (HG, 20 mM glucose) conditions, in the absence or presence of luteolin. Luteolin (3-10 μM) was added for 48 h. While hyperglycemic conditions significantly induced histone acetylation, NF-κB activation, and proinflammatory cytokine (IL-6 and TNF-α) release from THP-1 cells, luteolin suppressed NF-κB activity and cytokine release. Luteolin also significantly reduced CREB-binding protein/p300 (CBP/p300) gene expression, as well as the levels of acetylation and histone acetyltransferase (HAT) activity of the CBP/p300 protein, which is a known NF-κB coactivator. These results suggest that luteolin inhibits HG-induced cytokine production in monocytes, through epigenetic changes involving NF-κB. We therefore suggest that luteolin may be a potential candidate for the treatment and prevention of diabetes and its complications.
Collapse
Affiliation(s)
- Hye Joo Kim
- Pharmacology Research Center, Korea Research Institute of Chemical Technology, Daejeon, 305-600, South Korea
| | | | | |
Collapse
|
140
|
Yang YI, Ahn JH, Lee KT, Shih IM, Choi JH. RSF1 is a positive regulator of NF-κB-induced gene expression required for ovarian cancer chemoresistance. Cancer Res 2014; 74:2258-69. [PMID: 24566868 DOI: 10.1158/0008-5472.can-13-2459] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Overexpression or amplification of the RSF1 gene has been associated with poor prognosis in various human cancers, including ovarian cancer. In previous work, RSF1 was identified as an amplified gene that facilitated the development of paclitaxel-resistant ovarian cancer. In the present study, we further demonstrated that RSF1 expression inversely correlated with paclitaxel response in patients with ovarian cancer and the mouse xenograft model. In addition, RSF1-overexpressing paclitaxel-resistant ovarian cancer cell lines were found to express elevated levels of genes regulated by NF-κB, including some involved with the evasion of apoptosis (CFLAR, XIAP, BCL2, and BCL2L1) and inflammation (PTGS2). In addition, ectopic expression of RSF1 using Tet-off inducible SKOV3 cells significantly enhanced NF-κB-dependent gene expression and transcriptional activation of NF-κB. An RSF1 knockdown using short hairpin RNAs suppressed these same pathways. Moreover, pretreatment with NF-κB inhibitors or downregulation of NF-κB-regulated gene expression considerably enhanced paclitaxel sensitivity in RSF1-overexpressing OVCAR3 and/or RSF1-induced SKOV3 cells. A coimmunoprecipitation assay revealed that RSF1 interacts with NF-κB and CREB-binding protein, a ubiquitous coactivator for NF-κB. Recruitment of RSF1 to the NF-κB binding element in the PTGS2 and XIAP promoters was demonstrated by the chromatin immunoprecipitation assay. Furthermore, hSNF2H, a well-known binding partner of RSF1, was partially involved in the interaction between RSF1 and NF-κB. Taken together, these data suggest that RSF1 may function as a coactivator for NF-κB, consequently augmenting expression of genes necessary for the development of chemoresistance in ovarian cancer cells.
Collapse
Affiliation(s)
- Yeong-In Yang
- Authors' Affiliations: Department of Life and Nanopharmaceutical Science; Division of Molecular Biology, College of Pharmacy, Kyung Hee University, Seoul, South Korea; and Department of Pathology and Oncology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | | | | | | | | |
Collapse
|
141
|
Mbonye U, Karn J. Transcriptional control of HIV latency: cellular signaling pathways, epigenetics, happenstance and the hope for a cure. Virology 2014; 454-455:328-39. [PMID: 24565118 DOI: 10.1016/j.virol.2014.02.008] [Citation(s) in RCA: 179] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 01/23/2014] [Accepted: 02/07/2014] [Indexed: 02/06/2023]
Abstract
Replication-competent latent HIV-1 proviruses that persist in the genomes of a very small subset of resting memory T cells in infected individuals under life-long antiretroviral therapy present a major barrier towards viral eradication. Multiple molecular mechanisms are required to repress the viral trans-activating factor Tat and disrupt the regulatory Tat feedback circuit leading to the establishment of the latent viral reservoir. In particular, latency is due to a combination of transcriptional silencing of proviruses via host epigenetic mechanisms and restrictions on the expression of P-TEFb, an essential co-factor for Tat. Induction of latent proviruses in the presence of antiretroviral therapy is expected to enable clearance of latently infected cells by viral cytopathic effects and host antiviral immune responses. An in-depth comprehensive understanding of the molecular control of HIV-1 transcription should inform the development of optimal combinatorial reactivation strategies that are intended to purge the latent viral reservoir.
Collapse
Affiliation(s)
- Uri Mbonye
- Department of Molecular Biology and Microbiology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, United States
| | - Jonathan Karn
- Department of Molecular Biology and Microbiology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, United States.
| |
Collapse
|
142
|
Haery L, Lugo-Picó JG, Henry RA, Andrews AJ, Gilmore TD. Histone acetyltransferase-deficient p300 mutants in diffuse large B cell lymphoma have altered transcriptional regulatory activities and are required for optimal cell growth. Mol Cancer 2014; 13:29. [PMID: 24529102 PMCID: PMC3930761 DOI: 10.1186/1476-4598-13-29] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 02/05/2014] [Indexed: 11/10/2022] Open
Abstract
Background Recent genome-wide studies have shown that approximately 30% of diffuse large B-cell lymphoma (DLBCL) cases harbor mutations in the histone acetyltransferase (HAT) coactivators p300 or CBP. The majority of these mutations reduce or eliminate the catalytic HAT activity. We previously demonstrated that the human DLBCL cell line RC-K8 expresses a C-terminally truncated, HAT-defective p300 protein (p300ΔC-1087), whose expression is essential for cell proliferation. Methods Using results from large-scale DLBCL studies, we have identified and characterized a second C-terminally truncated, HAT-defective p300 mutant, p300ΔC-820, expressed in the SUDHL2 DLBCL cell line. Properties of p300ΔC-820 were characterized in the SUDHL2 DLBCL cell line by Western blotting, co-immunoprecipitation, and shRNA gene knockdown, as well by using cDNA expression vectors for p300ΔC-820 in pull-down assays, transcriptional reporter assays, and immunofluorescence experiments. A mass spectrometry-based method was used to compare the histone acetylation profile of DLBCL cell lines expressing various levels of wild-type p300. Results We show that the SUDHL2 cell line expresses a C-terminally truncated, HAT-defective form of p300 (p300ΔC-820), but no wild-type p300. The p300ΔC-820 protein has a wild-type ability to localize to subnuclear “speckles,” but has a reduced ability to enhance transactivation by transcription factor REL. Knockdown of p300ΔC-820 in SUDHL2 cells reduced their proliferation and soft agar colony-forming ability. In RC-K8 cells, knockdown of p300ΔC-1087 resulted in increased expression of mRNA and protein for REL target genes A20 and IκBα, two genes that have been shown to limit the growth of RC-K8 cells when overexpressed. Among a panel of B-lymphoma cell lines, low-level expression of full-length p300 protein, which is characteristic of the SUDHL2 and RC-K8 cells, was associated with decreased acetylation of histone H3 at lysines 14 and 18. Conclusions The high prevalence of p300 mutations in DLBCL suggests that HAT-deficient p300 activity defines a subtype of DLBCL, which we have investigated using human DLBCL cell lines RC-K8 and SUDHL2. Our results suggest that truncated p300 proteins contribute to DLBCL cell growth by affecting the expression of specific genes, perhaps through a mechanism that involves alterations in global histone acetylation.
Collapse
Affiliation(s)
| | | | | | | | - Thomas D Gilmore
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA.
| |
Collapse
|
143
|
Nakajima T, Aratani S, Nakazawa M, Hirose T, Fujita H, Nishioka K. Implications of transcriptional coactivator CREB binding protein complexes in rheumatoid arthritis. Mod Rheumatol 2014. [DOI: 10.3109/s10165-003-0258-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
144
|
|
145
|
Kim SW, Lee HK, Shin JH, Lee JK. Up-down regulation of HO-1 and iNOS gene expressions by ethyl pyruvate via recruiting p300 to Nrf2 and depriving It from p65. Free Radic Biol Med 2013; 65:468-476. [PMID: 23891677 DOI: 10.1016/j.freeradbiomed.2013.07.028] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 06/27/2013] [Accepted: 07/19/2013] [Indexed: 11/22/2022]
Abstract
Ethyl pyruvate (EP), a simple ester of pyruvic acid, has been shown to exert robust neuroprotection in various neuropathological conditions, such as, cerebral ischemia and KA-induced seizure animal models. The neuroprotective effect of EP is attributable to the anti-inflammatory, anti-oxidative, and anti-apoptotic effects. In the present study, we investigated convergence of anti-inflammatory and anti-oxidative functions of EP and present a novel molecular mechanism underlying anti-inflammatory effects of EP, which is conveyed by p300, a transcriptional co-activator for both Nuclear factor E2-related factor 2 (Nrf2) and p65. In BV2 cells, a microglia cell line, EP induced translocation of Nrf2 from the cytosol to the nucleus and enhanced the expression of hemeoxygenase 1 (HO-1) in a dose-dependent manner and 1h incubation with 10mM EP increased HO-1 to 4.9-fold. Nrf2 was found to translocate from the cytosol to the nucleus beginning 30 min after EP-treatment and binds to the antioxidant response element (ARE) located on HO-1 promoter. Interestingly, LPS-induced inducible NO synthase (iNOS) induction was substantially suppressed in EP-pre-treated BV2 cells and it was reverted by Nrf2 knockdown. We found that EP-induced Nrf2 accumulation in the nucleus recruits p300, a transcriptional co-activator of both Nrf2 and p65, inhibiting p65-p300 interaction. Competition between Nrf2 and p65 for p300 binding was confirmed by glutathione S-transferase (GST) pull down assay and reporter gene analysis. These results demonstrate that EP induced nuclear translocation of Nrf2 which binds to ARE along with p300 and hampers iNOS expression via depleting p300 from p65. This is a novel anti-inflammatory mechanism conveyed by EP, which enhances protective effect by converging anti-inflammatory and anti-oxidative effects and might be applicable to various Nrf2-activating agents, such as phytochemicals.
Collapse
Affiliation(s)
- Seung-Woo Kim
- Department of Anatomy, Center for Advanced Medical Education (BK21 project), Inha University School of Medicine, Incheon, Korea
| | - Hye-Kyung Lee
- Department of Anatomy, Center for Advanced Medical Education (BK21 project), Inha University School of Medicine, Incheon, Korea
| | - Joo-Hyun Shin
- Department of Anatomy, Center for Advanced Medical Education (BK21 project), Inha University School of Medicine, Incheon, Korea
| | - Ja-Kyeong Lee
- Department of Anatomy, Center for Advanced Medical Education (BK21 project), Inha University School of Medicine, Incheon, Korea.
| |
Collapse
|
146
|
Mukherjee SP, Behar M, Birnbaum HA, Hoffmann A, Wright PE, Ghosh G. Analysis of the RelA:CBP/p300 interaction reveals its involvement in NF-κB-driven transcription. PLoS Biol 2013; 11:e1001647. [PMID: 24019758 PMCID: PMC3760798 DOI: 10.1371/journal.pbio.1001647] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 07/23/2013] [Indexed: 11/18/2022] Open
Abstract
NF-κB plays a vital role in cellular immune and inflammatory response, survival, and proliferation by regulating the transcription of various genes involved in these processes. To activate transcription, RelA (a prominent NF-κB family member) interacts with transcriptional co-activators like CREB-binding protein (CBP) and its paralog p300 in addition to its cognate κB sites on the promoter/enhancer regions of DNA. The RelA:CBP/p300 complex is comprised of two components--first, DNA binding domain of RelA interacts with the KIX domain of CBP/p300, and second, the transcriptional activation domain (TAD) of RelA binds to the TAZ1 domain of CBP/p300. A phosphorylation event of a well-conserved RelA(Ser276) is prerequisite for the former interaction to occur and is considered a decisive factor for the overall RelA:CBP/p300 interaction. The role of the latter interaction in the transcription of RelA-activated genes remains unclear. Here we provide the solution structure of the latter component of the RelA:CBP complex by NMR spectroscopy. The structure reveals the folding of RelA-TA2 (a section of TAD) upon binding to TAZ1 through its well-conserved hydrophobic sites in a series of grooves on the TAZ1 surface. The structural analysis coupled with the mechanistic studies by mutational and isothermal calorimetric analyses allowed the design of RelA-mutants that selectively abrogated the two distinct components of the RelA:CBP/p300 interaction. Detailed studies of these RelA mutants using cell-based techniques, mathematical modeling, and genome-wide gene expression analysis showed that a major set of the RelA-activated genes, larger than previously believed, is affected by this interaction. We further show how the RelA:CBP/p300 interaction controls the nuclear response of NF-κB through the negative feedback loop of NF-κB pathway. Additionally, chromatin analyses of RelA target gene promoters showed constitutive recruitment of CBP/p300, thus indicating a possible role of CBP/p300 in recruitment of RelA to its target promoter sites.
Collapse
Affiliation(s)
- Sulakshana P. Mukherjee
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California, United States of America
- Department of Molecular Biology and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Marcelo Behar
- Signaling Systems Laboratory, University of California, San Diego, La Jolla, California, United States of America
| | - Harry A. Birnbaum
- Signaling Systems Laboratory, University of California, San Diego, La Jolla, California, United States of America
| | - Alexander Hoffmann
- Signaling Systems Laboratory, University of California, San Diego, La Jolla, California, United States of America
| | - Peter E. Wright
- Department of Molecular Biology and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California, United States of America
- * E-mail: (GG); (PEW)
| | - Gourisankar Ghosh
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California, United States of America
- * E-mail: (GG); (PEW)
| |
Collapse
|
147
|
Haselow K, Bode JG, Wammers M, Ehlting C, Keitel V, Kleinebrecht L, Schupp AK, Häussinger D, Graf D. Bile acids PKA-dependently induce a switch of the IL-10/IL-12 ratio and reduce proinflammatory capability of human macrophages. J Leukoc Biol 2013; 94:1253-64. [PMID: 23990628 DOI: 10.1189/jlb.0812396] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
That cholestatic conditions are accompanied by an enhanced susceptibility to bacterial infection in human and animal models is a known phenomenon. This correlates with the observation that bile acids have suppressive effects on cells of innate and adaptive immunity. The present study provides evidence that in human macrophages, bile acids inhibit the LPS-induced expression of proinflammatory cytokines without affecting the expression of the anti-inflammatory cytokine IL-10. This results in a macrophage phenotype that is characterized by an increased IL-10/IL-12 ratio. Correspondingly, bile acids suppress basal phagocytic activity of human macrophages. These effects of bile acids can be mimicked by cAMP, which is presumably induced TGR5-dependently. The data provided further suggest that in primary human macrophages, modulation of the macrophage response toward LPS by bile acids involves activation of CREB, disturbed nuclear translocation of NF-κB, and PKA-dependent enhancement of LPS-induced cFos expression. The increase in cFos expression is paralleled by an enhanced formation of a protein complex comprising cFos and the p65 subunit of NF-κB. In summary, the data provided suggest that in human macrophages, bile acids induce an anti-inflammatory phenotype characterized by an increased IL-10/IL-12 ratio via activation of PKA and thereby, prevent their activation as classically activated macrophages. This bile acid-induced modulation of macrophage function may also be responsible for the experimentally and clinically observed anti-inflammatory and immunosuppressive effects of bile acids.
Collapse
Affiliation(s)
- Katrin Haselow
- 1.Hepatology und Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, D-40225 Düsseldorf, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
148
|
Hoesel B, Schmid JA. The complexity of NF-κB signaling in inflammation and cancer. Mol Cancer 2013; 12:86. [PMID: 23915189 PMCID: PMC3750319 DOI: 10.1186/1476-4598-12-86] [Citation(s) in RCA: 2504] [Impact Index Per Article: 208.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 07/30/2013] [Indexed: 02/07/2023] Open
Abstract
The NF-κB family of transcription factors has an essential role in inflammation and innate immunity. Furthermore, NF-κB is increasingly recognized as a crucial player in many steps of cancer initiation and progression. During these latter processes NF-κB cooperates with multiple other signaling molecules and pathways. Prominent nodes of crosstalk are mediated by other transcription factors such as STAT3 and p53 or the ETS related gene ERG. These transcription factors either directly interact with NF-κB subunits or affect NF-κB target genes. Crosstalk can also occur through different kinases, such as GSK3-β, p38, or PI3K, which modulate NF-κB transcriptional activity or affect upstream signaling pathways. Other classes of molecules that act as nodes of crosstalk are reactive oxygen species and miRNAs. In this review, we provide an overview of the most relevant modes of crosstalk and cooperativity between NF-κB and other signaling molecules during inflammation and cancer.
Collapse
Affiliation(s)
- Bastian Hoesel
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| | - Johannes A Schmid
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| |
Collapse
|
149
|
Shin SY, Lee JM, Lim Y, Lee YH. Transcriptional regulation of the growth-regulated oncogene α gene by early growth response protein-1 in response to tumor necrosis factor α stimulation. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2013; 1829:1066-74. [PMID: 23872552 DOI: 10.1016/j.bbagrm.2013.07.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 07/11/2013] [Accepted: 07/12/2013] [Indexed: 12/15/2022]
Abstract
Growth-regulated oncogene α (GROα) plays an important role in a wide range of normal and pathological conditions, including inflammation, angiogenesis, wound healing, tumor invasion, and metastasis. Egr-1 is a member of the zinc-finger transcription factor family induced by diverse stimuli, including TNFα. However, the role of Egr-1 in GROα expression was previously unknown. This study shows that Egr-1 directly binds to the GROα promoter and transactivates the GROα gene. Silencing of Egr-1 by expression of Egr-1 siRNA abrogated TNFα-induced GROα transcription. We also found that Egr-1 mediates ERK and JNK MAPK-dependent GROα transcription upon TNFα stimulation. Our findings suggest that Egr-1 may play an important role in tumor development through transactivation of the GROα gene in response to TNFα within the tumor microenvironment.
Collapse
Affiliation(s)
- Soon Young Shin
- Department of Biological Sciences, College of Biological Science and Biotechnology, Konkuk University, Seoul 143-701, Republic of Korea; Cancer and Metabolism Institute, Konkuk University, Seoul 143-701, Republic of Korea.
| | | | | | | |
Collapse
|
150
|
Heyne K, Winter C, Gerten F, Schmidt C, Roemer K. A novel mechanism of crosstalk between the p53 and NFκB pathways: MDM2 binds and inhibits p65RelA. Cell Cycle 2013; 12:2479-92. [PMID: 23839035 DOI: 10.4161/cc.25495] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The inflammation regulating transcription factor NFκB and the tumor-suppressing transcription factor p53 can act as functional antagonists. Chronic inflammation (NFκB activity) may contribute to the development of cancer through the inhibition of p53 function, while, conversely, p53 activity may dampen inflammation. Here we report that the E3 ubiquitin ligase MDM2, whose gene is transcriptionally activated by both NFκB and p53, can bind and inhibit the p65RelA subunit of NFκB. The interaction is mediated through the N-terminal and the acidic/zinc finger domains of MDM2 on the one hand and through the N-terminal Rel homology domain of p65RelA on the other hand. Co-expression of MDM2 and p65RelA caused ubiquitination of the latter in the nucleus, and this modification was dependent of a functional MDM2 RING domain. Conversely, inhibition of endogenous MDM2 by small-molecule inhibitors or siRNA significantly reduced the ubiquitination of ectopic and endogenous p65RelA. MDM2 was able to equip p65RelA with mutated ubiquitin moieties capable of multiple monoubiquitination but incapable of polyubiquitination; moreover, MDM2 failed to destabilize p65RelA detectably, suggesting that the ubiquitin modification of p65RelA by MDM2 was mostly regulatory rather than stability-determining. MDM2 inhibited the NFκB-mediated transactivation of a reporter gene and the binding of NFκB to its DNA binding motif in vitro. Finally, knockdown of endogenous MDM2 increased the activity of endogenous NFκB as a transactivator. Thus, MDM2 can act as a direct negative regulator of NFκB by binding and inhibiting p65RelA.
Collapse
Affiliation(s)
- Kristina Heyne
- Internal Medicine I and José Carreras Center; University of Saarland Medical Center; Homburg, Saarland, Germany
| | | | | | | | | |
Collapse
|