401
|
Yang Z, Lv J, Lu X, Li X, An X, Wang J, Weng H, Li Y. Emulsified isoflurane induces release of cytochrome C in human neuroblastoma SHSY-5Y cells via JNK (c-Jun N-terminal kinases) signaling pathway. Neurotoxicol Teratol 2018; 65:19-25. [DOI: 10.1016/j.ntt.2017.12.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 11/27/2017] [Accepted: 12/04/2017] [Indexed: 01/01/2023]
|
402
|
Kittaka M, Mayahara K, Mukai T, Yoshimoto T, Yoshitaka T, Gorski JP, Ueki Y. Cherubism Mice Also Deficient in c-Fos Exhibit Inflammatory Bone Destruction Executed by Macrophages That Express MMP14 Despite the Absence of TRAP+ Osteoclasts. J Bone Miner Res 2018; 33:167-181. [PMID: 28914985 PMCID: PMC5771992 DOI: 10.1002/jbmr.3295] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 08/28/2017] [Accepted: 09/07/2017] [Indexed: 11/10/2022]
Abstract
Currently, it is believed that osteoclasts positive for tartrate-resistant acid phosphatase (TRAP+) are the exclusive bone-resorbing cells responsible for focal bone destruction in inflammatory arthritis. Recently, a mouse model of cherubism (Sh3bp2KI/KI ) with a homozygous gain-of-function mutation in the SH3-domain binding protein 2 (SH3BP2) was shown to develop auto-inflammatory joint destruction. Here, we demonstrate that Sh3bp2KI/KI mice also deficient in the FBJ osteosarcoma oncogene (c-Fos) still exhibit noticeable bone erosion at the distal tibia even in the absence of osteoclasts at 12 weeks old. Levels of serum collagen I C-terminal telopeptide (ICTP), a marker of bone resorption generated by matrix metalloproteinases (MMPs), were elevated, whereas levels of serum cross-linked C-telopeptide (CTX), another resorption marker produced by cathepsin K, were not increased. Collagenolytic MMP levels were increased in the inflamed joints of the Sh3bp2KI/KI mice deficient in c-Fos. Resorption pits contained a large number of F4/80+ macrophages and genetic depletion of macrophages rescued these erosive changes. Importantly, administration of NSC405020, an MMP14 inhibitor targeted to the hemopexin (PEX) domain, suppressed bone erosion in c-Fos-deficient Sh3bp2KI/KI mice. After activation of the NF-κB pathway, macrophage colony-stimulating factor (M-CSF)-dependent macrophages from c-Fos-deficient Sh3bp2KI/KI mice expressed increased amounts of MMP14 compared with wild-type macrophages. Interestingly, receptor activator of NF-κB ligand (RANKL)-deficient Sh3bp2KI/KI mice failed to show notable bone erosion, whereas c-Fos deletion did restore bone erosion to the RANKL-deficient Sh3bp2KI/KI mice, suggesting that osteolytic transformation of macrophages requires both loss-of-function of c-Fos and gain-of-function of SH3BP2 in this model. These data provide the first genetic evidence that cells other than osteoclasts can cause focal bone destruction in inflammatory bone disease and suggest that MMP14 is a key mediator conferring pathological bone-resorbing capacity on c-Fos-deficient Sh3bp2KI/KI macrophages. In summary, the paradigm that osteoclasts are the exclusive cells executing inflammatory bone destruction may need to be reevaluated based on our findings with c-Fos-deficient cherubism mice lacking osteoclasts. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Mizuho Kittaka
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Kotoe Mayahara
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO, USA.,Department of Orthodontics, Nihon University, School of Dentistry, Tokyo, Japan
| | - Tomoyuki Mukai
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Tetsuya Yoshimoto
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Teruhito Yoshitaka
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Jeffrey P Gorski
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO, USA.,University of Missouri-Kansas City (UMKC) Center of Excellence in the Study of Dental and Musculoskeletal Tissues (CEMT), Kansas City, MO, USA
| | - Yasuyoshi Ueki
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO, USA.,University of Missouri-Kansas City (UMKC) Center of Excellence in the Study of Dental and Musculoskeletal Tissues (CEMT), Kansas City, MO, USA
| |
Collapse
|
403
|
Valentijn FA, Falke LL, Nguyen TQ, Goldschmeding R. Cellular senescence in the aging and diseased kidney. J Cell Commun Signal 2017; 12:69-82. [PMID: 29260442 PMCID: PMC5842195 DOI: 10.1007/s12079-017-0434-2] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 11/03/2017] [Indexed: 12/19/2022] Open
Abstract
The program of cellular senescence is involved in both the G1 and G2 phase of the cell cycle, limiting G1/S and G2/M progression respectively, and resulting in prolonged cell cycle arrest. Cellular senescence is involved in normal wound healing. However, multiple organs display increased senescent cell numbers both during natural aging and after injury, suggesting that senescent cells can have beneficial as well as detrimental effects in organismal aging and disease. Also in the kidney, senescent cells accumulate in various compartments with advancing age and renal disease. In experimental studies, forced apoptosis induction through the clearance of senescent cells leads to better preservation of kidney function during aging. Recent groundbreaking studies demonstrate that senescent cell depletion through INK-ATTAC transgene-mediated or cell-penetrating FOXO4-DRI peptide induced forced apoptosis, reduced age-associated damage and dysfunction in multiple organs, in particular the kidney, and increased performance and lifespan. Senescence is also involved in oncology and therapeutic depletion of senescent cells by senolytic drugs has been studied in experimental and human cancers. Although studies with senolytic drugs in models of kidney injury are lacking, their dose limiting side effects on other organs suggest that targeted delivery might be needed for successful application of senolytic drugs for treatment of kidney disease. In this review, we discuss (i) current understanding of the mechanisms and associated pathways of senescence, (ii) evidence of senescence occurrence and causality with organ injury, and (iii) therapeutic strategies for senescence depletion (senotherapy) including targeting, all in the context of renal aging and disease.
Collapse
Affiliation(s)
- F A Valentijn
- Department of Pathology, University Medical Center Utrecht, H04.312, Heidelberglaan 110, 3584, CX, Utrecht, The Netherlands
| | - L L Falke
- Department of Pathology, University Medical Center Utrecht, H04.312, Heidelberglaan 110, 3584, CX, Utrecht, The Netherlands
- Department of Internal Medicine, Diakonessenhuis, Utrecht, The Netherlands
| | - T Q Nguyen
- Department of Pathology, University Medical Center Utrecht, H04.312, Heidelberglaan 110, 3584, CX, Utrecht, The Netherlands
| | - Roel Goldschmeding
- Department of Pathology, University Medical Center Utrecht, H04.312, Heidelberglaan 110, 3584, CX, Utrecht, The Netherlands.
| |
Collapse
|
404
|
Fang K, Law IKM, Padua D, Sideri A, Huang V, Kevil CG, Iliopoulos D, Pothoulakis C. MicroRNA-31-3p Is Involved in Substance P (SP)-Associated Inflammation in Human Colonic Epithelial Cells and Experimental Colitis. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 188:586-599. [PMID: 29253460 DOI: 10.1016/j.ajpath.2017.10.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 10/24/2017] [Accepted: 10/30/2017] [Indexed: 02/08/2023]
Abstract
Substance P (SP) mediates colitis. SP signaling regulates the expression of several miRNAs, including miR-31-3p, in human colonocytes. However, the role of miR-31-3p in colitis and the underlying mechanisms has not been elucidated. We performed real-time PCR analysis of miR-31-3p expression in human colonic epithelial cells overexpressing neurokinin-1 receptor (NCM460 NK-1R) in response to SP stimulation and in NCM460 cells after IL-6, IL8, tumor necrosis factor (TNF)-α, and interferon-γ exposure. Functions of miR-31-3p were tested in NCM460-NK-1R cells and the trinitrobenzene sulfonic acid (TNBS) and dextran sodium sulfate (DSS) models of colitis. Targets of miRNA-31-3p were confirmed by Western blot analysis and luciferase reporter assay. Jun N-terminal kinase inhibition decreased SP-induced miR-31-3p expression. miR-31-3p expression was increased in both TNBS- and DSS-induced colitis and human colonic biopsies from ulcerative colitis, compared with controls. Intracolonic administration of a miR-31-3p chemical inhibitor exacerbated TNBS- and DSS-induced colitis and increased colonic TNF-α, CXCL10, and chemokine (C-C motif) ligand 2 (CCL2) mRNA expression. Conversely, overexpression of miR-31-3p ameliorated the severity of DSS-induced colitis. Bioinformatic, luciferase reporter assay, and Western blot analyses identified RhoA as a target of miR-31-3p in NCM460 cells. Constitutive activation of RhoA led to increased expression of CCL2, IL6, TNF-α, and CXCL10 in NCM460-NK-1R cells on SP stimulation. Our results reveal a novel SP-miR-31-3p-RhoA pathway that protects from colitis. The use of miR-31-3p mimics may be a promising approach for colitis treatment.
Collapse
Affiliation(s)
- Kai Fang
- Inflammatory Bowel Disease Center, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, California
| | - Ivy Ka Man Law
- Inflammatory Bowel Disease Center, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, California
| | - David Padua
- Inflammatory Bowel Disease Center, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, California
| | - Aristea Sideri
- Inflammatory Bowel Disease Center, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, California
| | - Vanessa Huang
- Inflammatory Bowel Disease Center, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, California
| | - Christopher G Kevil
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Dimitrios Iliopoulos
- Center for Systems Biomedicine, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, California
| | - Charalabos Pothoulakis
- Inflammatory Bowel Disease Center, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, California.
| |
Collapse
|
405
|
Fung TS, Liu DX. Activation of the c-Jun NH 2-terminal kinase pathway by coronavirus infectious bronchitis virus promotes apoptosis independently of c-Jun. Cell Death Dis 2017; 8:3215. [PMID: 29238080 PMCID: PMC5870581 DOI: 10.1038/s41419-017-0053-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 09/28/2017] [Accepted: 10/11/2017] [Indexed: 12/16/2022]
Abstract
Mitogen-activated protein kinases (MAPKs) are conserved protein kinases that regulate a variety of important cellular signaling pathways. Among them, c-Jun N-terminal kinases (JNK) are known to be activated by various environmental stresses including virus infections. Previously, activation of the JNK pathway has been detected in cells infected with several coronaviruses. However, detailed characterization of the pathway as well as its implication in host-virus interactions has not been fully investigated. Here we report that the JNK pathway was activated in cells infected with the avian coronavirus infectious bronchitis virus (IBV). Of the two known upstream MAPK kinases (MKK), MKK7, but not MKK4, was shown to be responsible for IBV-induced JNK activation. Moreover, knockdown and overexpression experiments demonstrated that JNK served as a pro-apoptotic protein during IBV infection. Interestingly, pro-apoptotic activity of JNK was not mediated via c-Jun, but involved modulation of the anti-apoptotic protein B-cell lymphoma 2 (Bcl2). Taken together, JNK constitutes an important aspect of coronavirus-host interaction, along with other MAPKs.
Collapse
Affiliation(s)
- To Sing Fung
- 0000 0000 9546 5767grid.20561.30South China Agricultural University, Guangdong Province Key Laboratory Microbial Signals & Disease Co, and Integrative Microbiology Research Centre, Guangzhou, 510642 Guangdong, People’s Republic of China
| | - Ding Xiang Liu
- 0000 0000 9546 5767grid.20561.30South China Agricultural University, Guangdong Province Key Laboratory Microbial Signals & Disease Co, and Integrative Microbiology Research Centre, Guangzhou, 510642 Guangdong, People’s Republic of China ,0000 0001 2224 0361grid.59025.3bSchool of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 63755 Singapore
| |
Collapse
|
406
|
Kang HM, Jeong CB, Lee YH, Cui YH, Kim DH, Lee MC, Kim HS, Han J, Hwang DS, Lee SJ, Lee JS. Cross-reactivities of mammalian MAPKs antibodies in rotifer and copepod: Application in mechanistic studies in aquatic ecotoxicology. MARINE POLLUTION BULLETIN 2017; 124:614-623. [PMID: 28012735 DOI: 10.1016/j.marpolbul.2016.11.049] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 11/01/2016] [Accepted: 11/19/2016] [Indexed: 06/06/2023]
Abstract
The mitogen-activated protein kinases (MAPKs) family is known to mediate various biological processes in response to diverse environmental pollutants. Although MAPKs are well characterized and studied in vertebrates, in invertebrates the cross-reactivities of MAPKs antibodies were not clearly known in response to environmental pollutants due to limited information of antibody epitopes with material resources for invertebrates. In this paper, we performed phylogenetic analysis of MAPKs genes in the marine rotifer Brachionus koreanus and the copepods Paracyclopina nana and Tigriopus japonicus. Also in rotifer and copepods, several studies of Western blot of MAPK signaling pathways were shown in response to environmental pollutants, including multi-walled carbon nanotubes (MWCNTs), water-accommodated fractions (WAFs) of crude oil, and microplastics. This paper will provide a better understanding of the underlying mechanistic scenario in terms of cross-reactivities of mammalian antibodies in rotifer and copepod.
Collapse
Affiliation(s)
- Hye-Min Kang
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Chang-Bum Jeong
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon 16419, South Korea; Department of Chemistry, College of Natural Sciences, Hanyang University, Seoul 04763, South Korea
| | - Young Hwan Lee
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Yan-Hong Cui
- Department of Life Sciences, College of Natural Sciences, Hanyang University, Seoul 04763, South Korea
| | - Duck-Hyun Kim
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Min-Chul Lee
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Hui-Su Kim
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Jeonghoon Han
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Dae-Sik Hwang
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Su-Jae Lee
- Department of Life Sciences, College of Natural Sciences, Hanyang University, Seoul 04763, South Korea
| | - Jae-Seong Lee
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon 16419, South Korea.
| |
Collapse
|
407
|
Massip‐Copiz M, Clauzure M, Valdivieso ÁG, Santa‐Coloma TA. Epiregulin (EREG) is upregulated through an IL‐1β autocrine loop in Caco‐2 epithelial cells with reduced CFTR function. J Cell Biochem 2017; 119:2911-2922. [DOI: 10.1002/jcb.26483] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 10/31/2017] [Indexed: 01/10/2023]
Affiliation(s)
- Macarena Massip‐Copiz
- The Laboratory of Cellular and Molecular BiologyInstitute for Biomedical Research (BIOMED)School of Medical SciencesPontifical Catholic University of Argentina (UCA)The National Scientific and Technical Research Council (CONICET)Buenos AiresArgentina
| | - Mariángeles Clauzure
- The Laboratory of Cellular and Molecular BiologyInstitute for Biomedical Research (BIOMED)School of Medical SciencesPontifical Catholic University of Argentina (UCA)The National Scientific and Technical Research Council (CONICET)Buenos AiresArgentina
| | - Ángel G. Valdivieso
- The Laboratory of Cellular and Molecular BiologyInstitute for Biomedical Research (BIOMED)School of Medical SciencesPontifical Catholic University of Argentina (UCA)The National Scientific and Technical Research Council (CONICET)Buenos AiresArgentina
| | - Tomás A. Santa‐Coloma
- The Laboratory of Cellular and Molecular BiologyInstitute for Biomedical Research (BIOMED)School of Medical SciencesPontifical Catholic University of Argentina (UCA)The National Scientific and Technical Research Council (CONICET)Buenos AiresArgentina
| |
Collapse
|
408
|
Wang J, Guo Z, Fu Y, Wu Z, Huang C, Zheng C, Shar PA, Wang Z, Xiao W, Wang Y. Weak-binding molecules are not drugs?-toward a systematic strategy for finding effective weak-binding drugs. Brief Bioinform 2017; 18:321-332. [PMID: 26962012 DOI: 10.1093/bib/bbw018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Indexed: 12/16/2022] Open
Abstract
Designing maximally selective ligands that act on individual drug targets with high binding affinity has been the central dogma of drug discovery and development for the past two decades. However, many low-affinity drugs that aim for several targets at the same time are found more effective than the high-affinity binders when faced with complex disease conditions, such as cancers, Alzheimer's disease and cardiovascular diseases. The aim of this study was to appreciate the importance and reveal the features of weak-binding drugs and propose an integrated strategy for discovering them. Weak-binding drugs can be characterized by their high dissociation rates and transient interactions with their targets. In addition, network topologies and dynamics parameters involved in the targets of weak-binding drugs also influence the effects of the drugs. Here, we first performed a dynamics analysis for 33 elementary subgraphs to determine the desirable topology and dynamics parameters among targets. Then, by applying the elementary subgraphs to the mitogen-activated protein kinase (MAPK) pathway, several optimal target combinations were obtained. Combining drug-target interaction prediction with molecular dynamics simulation, we got two potential weak-binding drug candidates, luteolin and tanshinone IIA, acting on these targets. Further, the binding affinity of these two compounds to their targets and the anti-inflammatory effects of them were validated through in vitro experiments. In conclusion, weak-binding drugs have real opportunities for maximum efficiency and may show reduced adverse reactions, which can offer a bright and promising future for new drug discovery.
Collapse
Affiliation(s)
- Jinan Wang
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, Shaanxi, China, School of Chemical engineering, Dalian University of Technology, Dalian, Liaoning, China, Beijing University of Chinese Medicine, ChaoYang District, Beijing, China and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Zihu Guo
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, Shaanxi, China, School of Chemical engineering, Dalian University of Technology, Dalian, Liaoning, China, Beijing University of Chinese Medicine, ChaoYang District, Beijing, China and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Yingxue Fu
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, Shaanxi, China, School of Chemical engineering, Dalian University of Technology, Dalian, Liaoning, China, Beijing University of Chinese Medicine, ChaoYang District, Beijing, China and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Ziyin Wu
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, Shaanxi, China, School of Chemical engineering, Dalian University of Technology, Dalian, Liaoning, China, Beijing University of Chinese Medicine, ChaoYang District, Beijing, China and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Chao Huang
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, Shaanxi, China, School of Chemical engineering, Dalian University of Technology, Dalian, Liaoning, China, Beijing University of Chinese Medicine, ChaoYang District, Beijing, China and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Chunli Zheng
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, Shaanxi, China, School of Chemical engineering, Dalian University of Technology, Dalian, Liaoning, China, Beijing University of Chinese Medicine, ChaoYang District, Beijing, China and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Piar Ali Shar
- College of Life Science, Northwest A & F University, Yangling, Shaanxi, 712100, China; Center of Bioinformatics, Northwest A & F University, Yangling, Shaanxi, China
| | - Zhenzhong Wang
- Jiangsu Kanion Pharmaceutical Co. Ltd., Lianyungang, PR China
| | - Wei Xiao
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutical Process, Lianyungang, Jiangsu, China
| | - Yonghua Wang
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, Shaanxi, China, School of Chemical engineering, Dalian University of Technology, Dalian, Liaoning, China, Beijing University of Chinese Medicine, ChaoYang District, Beijing, China and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| |
Collapse
|
409
|
Yang Z, Yang W, Lu M, Li Z, Qiao X, Sun B, Zhang W, Xue D. Role of the c-Jun N-terminal kinase signaling pathway in the activation of trypsinogen in rat pancreatic acinar cells. Int J Mol Med 2017; 41:1119-1126. [PMID: 29207022 DOI: 10.3892/ijmm.2017.3266] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 11/08/2017] [Indexed: 11/06/2022] Open
Abstract
Bile acid causes trypsinogen activation in pancreatic acinar cells through a complex process. Additional research is required to further elucidate which signaling pathways affect trypsinogen activation when activated. the changes in the whole‑genome expression profile of AR42J cells under the effect of taurolithocholic acid 3‑sulfate (TLC‑S) were investigated. Furthermore, gene groups that may play a regulatory role were analyzed using the modular approach of biological networks. The aim of the present study was to improve our understanding of the changes in TLC‑S‑stimulated AR42J cells through a genetic functional modular analysis. whole‑genome expression profile chip arrays were applied to detect genes that were differentially expressed in pancreatic acinar AR42J cells treated with TLC‑S for 20 min. Based on the human protein reference database, a protein‑protein interaction network was obtained, which was then processed by CFinder software to derive 14 modules. Among these 14 modules, the gene ontology biological processes enrichment analysis identified two as modules of interest. Kyoto encyclopedia of genes and genomes map analysis revealed that MAP2K4, MAPK8 and FLNA are part of the c-Jun N-terminal kinase (JNK) pathway. The JNK signaling pathway is involved in regulating trypsinogen activation in rat pancreatic AR42J cells. Next, a regulatory network of seven kinase inhibitors was constructed. SP600125 is an ATP‑competitive, efficient, selective and reversible inhibitor of JNK. the results were verified by four sets of experiments and demonstrated that trypsinogen activation is mediated by the JNK signaling pathway in the pathogenesis of acute pancreatitis (AP). The present study provided a useful reference for better understanding the pathogenesis of AP and identifying new targets to regulate trypsinogen activation, in addition to providing valuable information for the treatment of AP.
Collapse
Affiliation(s)
- Zhengpeng Yang
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Weiguang Yang
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Ming Lu
- Department of Surgery, David Geffen School of Medicine, University of Califonia at Los Angeles, Los Angeles, CA 90095, USA
| | - Zhituo Li
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Xin Qiao
- Department of Surgery, David Geffen School of Medicine, University of Califonia at Los Angeles, Los Angeles, CA 90095, USA
| | - Bei Sun
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Weihui Zhang
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Dongbo Xue
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
410
|
Ábrigo J, Campos F, Simon F, Riedel C, Cabrera D, Vilos C, Cabello-Verrugio C. TGF-β requires the activation of canonical and non-canonical signalling pathways to induce skeletal muscle atrophy. Biol Chem 2017; 399:253-264. [DOI: 10.1515/hsz-2017-0217] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 11/03/2017] [Indexed: 12/20/2022]
Abstract
Abstract
The transforming growth factor type-beta (TGF-β) induces skeletal muscle atrophy characterised by a decrease in the fibre’s diameter and levels of myosin heavy chain (MHC), also as an increase of MuRF-1 expression. In addition, TGF-β induces muscle atrophy by a mechanism dependent on reactive oxygen species (ROS). TGF-β signals by activating both canonical Smad-dependent, and non-canonical signalling pathways such as ERK1/2, JNK1/2, and p38 MAPKs. However, the participation of canonical and non-canonical signalling pathways in the TGF-β atrophic effect on skeletal muscle is unknown. We evaluate the impact of Smad and MAPK signalling pathways on the TGF-β-induced atrophic effect in C2C12 myotubes. The results indicate that TGF-β activates Smad2/3, ERK1/2 and JNK1/2, but not p38 in myotubes. The pharmacological inhibition of Smad3, ERK1/2 and JNK1/2 activation completely abolished the atrophic effect of TGF-β. Finally, the inhibition of these canonical and non-canonical pathways did not decrease the ROS increment, while the inhibition of ROS production entirely abolished the phosphorylation of Smad3, ERK1/2 and JNK1/2. These results suggest that TGF-β requires Smad3, ERK1/2 and JNK1/2 activation to produce skeletal muscle atrophy. Moreover, the induction of ROS by TGF-β is an upstream event to canonical and non-canonical pathways.
Collapse
Affiliation(s)
- Johanna Ábrigo
- Departamento de Ciencias Biológicas , Facultad de Ciencias Biológicas and Facultad de Medicina, Universidad Andres Bello , Avenida República 239 , Santiago 8370146 , Chile
- Millennium Institute on Immunology and Immunotherapy , 8331150 Santiago , Chile
| | - Fabian Campos
- Departamento de Ciencias Biológicas , Facultad de Ciencias Biológicas and Facultad de Medicina, Universidad Andres Bello , Avenida República 239 , Santiago 8370146 , Chile
- Millennium Institute on Immunology and Immunotherapy , 8331150 Santiago , Chile
| | - Felipe Simon
- Departamento de Ciencias Biológicas , Facultad de Ciencias Biológicas and Facultad de Medicina, Universidad Andres Bello , Avenida República 239 , Santiago 8370146 , Chile
- Millennium Institute on Immunology and Immunotherapy , 8331150 Santiago , Chile
| | - Claudia Riedel
- Departamento de Ciencias Biológicas , Facultad de Ciencias Biológicas and Facultad de Medicina, Universidad Andres Bello , Avenida República 239 , Santiago 8370146 , Chile
- Millennium Institute on Immunology and Immunotherapy , 8331150 Santiago , Chile
| | - Daniel Cabrera
- Universidad Bernardo O Higgins, Facultad de Salud , Departamento de Ciencias Químicas y Biológicas , 8370993 Santiago , Chile
- Departamento de Gastroenterología, Facultad de Medicina , Pontificia Universidad Católica de Chile , 8331150 Santiago , Chile
| | - Cristian Vilos
- Laboratory of Nanomedicine and Targeted Delivery, Center for Integrative Medicine and Innovative Science, Faculty of Medicine, and Center for Bioinformatics and Integrative Biology, Faculty of Biological Sciences , Universidad Andres Bello , 8370146 Santiago , Chile
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA) , Universidad de Santiago de Chile , 9170022 Santiago , Chile
| | - Claudio Cabello-Verrugio
- Millennium Institute on Immunology and Immunotherapy , 8331150 Santiago , Chile
- Laboratory of Muscle Pathology, Fragility and Aging , Departmento de Ciencias Biológicas, Facultad de Ciencias Biológicas and Facultad de Medicina , Universidad Andres Bello , Avenida República 239 , Santiago 8370146 , Chile
| |
Collapse
|
411
|
Doytcheva P, Bächler T, Tarasco E, Marzolla V, Engeli M, Pellegrini G, Stivala S, Rohrer L, Tona F, Camici GG, Vanhoutte PM, Matter CM, Lutz TA, Lüscher TF, Osto E. Inhibition of Vascular c-Jun N-Terminal Kinase 2 Improves Obesity-Induced Endothelial Dysfunction After Roux-en-Y Gastric Bypass. J Am Heart Assoc 2017; 6:JAHA.117.006441. [PMID: 29138180 PMCID: PMC5721746 DOI: 10.1161/jaha.117.006441] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background Roux‐en‐Y gastric bypass (RYGB) reduces obesity‐associated comorbidities and cardiovascular mortality. RYGB improves endothelial dysfunction, reducing c‐Jun N‐terminal kinase (JNK) vascular phosphorylation. JNK activation links obesity with insulin resistance and endothelial dysfunction. Herein, we examined whether JNK1 or JNK2 mediates obesity‐induced endothelial dysfunction and if pharmacological JNK inhibition can mimic RYGB vascular benefits. Methods and Results After 7 weeks of a high‐fat high‐cholesterol diet, obese rats underwent RYGB or sham surgery; sham–operated ad libitum–fed rats received, for 8 days, either the control peptide D‐TAT or the JNK peptide inhibitor D‐JNKi‐1 (20 mg/kg per day subcutaneous). JNK peptide inhibitor D‐JNKi‐1 treatment improved endothelial vasorelaxation in response to insulin and glucagon‐like peptide‐1, as observed after RYGB. Obesity increased aortic phosphorylation of JNK2, but not of JNK1. RYGB and JNK peptide inhibitor D‐JNKi‐1 treatment blunted aortic JNK2 phosphorylation via activation of glucagon‐like peptide‐1–mediated signaling. The inhibitory phosphorylation of insulin receptor substrate‐1 was reduced, whereas the protein kinase B/endothelial NO synthase pathway was increased and oxidative stress was decreased, resulting in improved vascular NO bioavailability. Conclusions Decreased aortic JNK2 phosphorylation after RYGB rapidly improves obesity‐induced endothelial dysfunction. Pharmacological JNK inhibition mimics the endothelial protective effects of RYGB. These findings highlight the therapeutic potential of novel strategies targeting vascular JNK2 against the severe cardiovascular disease associated with obesity.
Collapse
Affiliation(s)
- Petia Doytcheva
- Center for Molecular Cardiology, University of Zurich, Switzerland.,University Heart Center, Cardiology, University Hospital Zurich, Switzerland.,Institute of Veterinary Physiology, Vetsuisse Faculty University of Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Switzerland
| | - Thomas Bächler
- Department of Surgery, Cantonal Hospital Fribourg, Fribourg, Switzerland
| | - Erika Tarasco
- Institute of Veterinary Physiology, Vetsuisse Faculty University of Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Switzerland
| | - Vincenzo Marzolla
- Center for Molecular Cardiology, University of Zurich, Switzerland.,University Heart Center, Cardiology, University Hospital Zurich, Switzerland.,Laboratory of Cardiovascular Endocrinology, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Pisana, Rome, Italy
| | - Michael Engeli
- Center for Molecular Cardiology, University of Zurich, Switzerland.,University Heart Center, Cardiology, University Hospital Zurich, Switzerland
| | - Giovanni Pellegrini
- Laboratory for Animal Model Pathology, Institute for Veterinary Pathology, Vetsuisse Faculty University of Zurich, Switzerland
| | - Simona Stivala
- Center for Molecular Cardiology, University of Zurich, Switzerland.,University Heart Center, Cardiology, University Hospital Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Switzerland
| | - Lucia Rohrer
- Institute of Clinical Chemistry, University Hospital Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Switzerland
| | - Francesco Tona
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Italy
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Switzerland.,University Heart Center, Cardiology, University Hospital Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Switzerland
| | - Paul M Vanhoutte
- State Key Laboratory for Pharmaceutical Biotechnologies & Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Schwerzenbach, Switzerland
| | - Christian M Matter
- Center for Molecular Cardiology, University of Zurich, Switzerland.,University Heart Center, Cardiology, University Hospital Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Switzerland
| | - Thomas A Lutz
- Zurich Center for Integrative Human Physiology, University of Zurich, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Switzerland.,University Heart Center, Cardiology, University Hospital Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Switzerland
| | - Elena Osto
- Center for Molecular Cardiology, University of Zurich, Switzerland .,University Heart Center, Cardiology, University Hospital Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Switzerland.,Laboratory of Translational Nutrition Biology Federal Institute of Technology Zurich (ETHZ), Schwerzenbach, Switzerland
| |
Collapse
|
412
|
Levinson S, Cagan RL. Drosophila Cancer Models Identify Functional Differences between Ret Fusions. Cell Rep 2017; 16:3052-3061. [PMID: 27626672 DOI: 10.1016/j.celrep.2016.08.019] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 07/22/2016] [Accepted: 08/05/2016] [Indexed: 12/23/2022] Open
Abstract
We generated and compared Drosophila models of RET fusions CCDC6-RET and NCOA4-RET. Both RET fusions directed cells to migrate, delaminate, and undergo EMT, and both resulted in lethality when broadly expressed. In all phenotypes examined, NCOA4-RET was more severe than CCDC6-RET, mirroring their effects on patients. A functional screen against the Drosophila kinome and a library of cancer drugs found that CCDC6-RET and NCOA4-RET acted through different signaling networks and displayed distinct drug sensitivities. Combining data from the kinome and drug screens identified the WEE1 inhibitor AZD1775 plus the multi-kinase inhibitor sorafenib as a synergistic drug combination that is specific for NCOA4-RET. Our work emphasizes the importance of identifying and tailoring a patient's treatment to their specific RET fusion isoform and identifies a multi-targeted therapy that may prove effective against tumors containing the NCOA4-RET fusion.
Collapse
Affiliation(s)
- Sarah Levinson
- Department of Developmental and Regenerative Biology and Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029-1020, USA
| | - Ross L Cagan
- Department of Developmental and Regenerative Biology and Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029-1020, USA.
| |
Collapse
|
413
|
Differential Regulation of Evoked and Spontaneous Release by Presynaptic NMDA Receptors. Neuron 2017; 96:839-855.e5. [DOI: 10.1016/j.neuron.2017.09.030] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 08/03/2017] [Accepted: 09/19/2017] [Indexed: 01/04/2023]
|
414
|
Wang J, Tai G. Role of C-Jun N-terminal Kinase in Hepatocellular Carcinoma Development. Target Oncol 2017; 11:723-738. [PMID: 27392951 DOI: 10.1007/s11523-016-0446-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hepatocellular carcinoma (HCC) is among the most frequently occurring cancers and the leading causes of cancer mortality worldwide. Identification of the signaling pathways regulating liver carcinogenesis is critical for developing novel chemoprevention and targeted therapies. C-Jun N-terminal kinase (JNK) is a member of a larger group of serine/threonine (Ser/Thr) protein kinases known as the mitogen-activated protein kinase (MAPK) family. JNK is an important signaling component that converts external stimuli into a wide range of cellular responses, including cell proliferation, differentiation, survival, migration, invasion, and apoptosis, as well as the development of inflammation, fibrosis, cancer growth, and metabolic diseases. Because of the essential roles of JNK in these cellular functions, deregulated JNK is often found to contribute to the development of HCC. Recently, the functions and molecular mechanisms of JNK in HCC development have been addressed using mouse models and human HCC cell lines. Furthermore, recent studies demonstrate that the activation of JNK by oncogenes can promote the development of cancers by regulating the transforming growth factor (TGF)-β/Smad pathway, which makes the oncogenes/JNK/Smad signaling pathway an attractive target for cancer therapy. Additionally, JNK-targeted therapy has a broad potential for clinical applications. In summary, we are convinced that promising new avenues for the treatment of HCC by targeting JNK are on the horizon, which will undoubtedly lead to better, more effective, and faster therapies in the years to come.
Collapse
Affiliation(s)
- Juan Wang
- Department of Immunology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Jilin, Changchun, 130021, China
| | - Guixiang Tai
- Department of Immunology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Jilin, Changchun, 130021, China.
| |
Collapse
|
415
|
Busquets O, Ettcheto M, Verdaguer E, Castro-Torres RD, Auladell C, Beas-Zarate C, Folch J, Camins A. JNK1 inhibition by Licochalcone A leads to neuronal protection against excitotoxic insults derived of kainic acid. Neuropharmacology 2017; 131:440-452. [PMID: 29111385 DOI: 10.1016/j.neuropharm.2017.10.030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 09/13/2017] [Accepted: 10/23/2017] [Indexed: 12/16/2022]
Abstract
The mitogen-activated protein kinase family (MAPK) is an important group of enzymes involved in cellular responses to diverse external stimuli. One of the members of this family is the c-Jun-N-terminal kinase (JNK). The activation of the JNK pathway has been largely associated with the pathogenesis that occurs in epilepsy and neurodegeneration. Kainic acid (KA) administration in rodents is an experimental approach that induces status epilepticus (SE) and replicates many of the phenomenological features of human temporal lobe epilepsy (TLE). Recent studies in our group have evidenced that the absence of the JNK1 gene has neuroprotective effects against the damage induced by KA, as it occurs with the absence of JNK3. The aim of the present study was to analyse whether the pharmacological inhibition of JNK1 by Licochalcone A (Lic-A) had similar effects and if it may be considered as a new molecule for the treatment of SE. In order to achieve this objective, animals were pre-treated with Lic-A and posteriorly administered with KA as a model for TLE. In addition, a comparative study with KA was performed between wild type pre-treated with Lic-A and single knock-out transgenic mice for the Jnk1-/- gene. Our results showed that JNK1 inhibition by Lic-A, previous to KA administration, caused a reduction in the convulsive pattern. Furthermore, it reduced phosphorylation levels of the JNK, as well as its activity. In addition, Lic-A prevented hippocampal neuronal degeneration, increased pro-survival anti-apoptotic mechanisms, reduced pro-apoptotic biomarkers, decreased cellular stress and neuroinflammatory processes. Thus, our results suggest that inhibition of the JNK1 by Lic-A has neuroprotective effects and that; it could be a new potential approach for the treatment of SE and neurodegeneration.
Collapse
Affiliation(s)
- Oriol Busquets
- Departament de Bioquímica i Biotecnologia, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Tarragona, Spain; Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain; Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain; Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Miren Ettcheto
- Departament de Bioquímica i Biotecnologia, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Tarragona, Spain; Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain; Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain; Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Ester Verdaguer
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain; Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain; Departament de Biologia Cellular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Ruben D Castro-Torres
- Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain; Departament de Biologia Cellular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain; Departamento de Biología Celular y Molecular, C.U.C.B.A., Universidad de Guadalajara y División de Neurociencias, Sierra Mojada 800, Col. Independencia, Guadalajara, Jalisco 44340, Mexico
| | - Carme Auladell
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain; Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain; Departament de Biologia Cellular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Carlos Beas-Zarate
- Departamento de Biología Celular y Molecular, C.U.C.B.A., Universidad de Guadalajara y División de Neurociencias, Sierra Mojada 800, Col. Independencia, Guadalajara, Jalisco 44340, Mexico
| | - Jaume Folch
- Departament de Bioquímica i Biotecnologia, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Tarragona, Spain; Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Antoni Camins
- Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain; Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain; Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
416
|
Grynberg K, Ma FY, Nikolic-Paterson DJ. The JNK Signaling Pathway in Renal Fibrosis. Front Physiol 2017; 8:829. [PMID: 29114233 PMCID: PMC5660697 DOI: 10.3389/fphys.2017.00829] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 10/06/2017] [Indexed: 01/07/2023] Open
Abstract
Fibrosis of the glomerular and tubulointerstitial compartments is a common feature of chronic kidney disease leading to end-stage renal failure. This fibrotic process involves a number of pathologic mechanisms, including cell death and inflammation. This review focuses on the role of the c-Jun amino terminal kinase (JNK) signaling pathway in the development of renal fibrosis. The JNK pathway is activated in response to various cellular stresses and plays an important role in cell death and inflammation. Activation of JNK signaling is a common feature in most forms of human kidney injury, evident in both intrinsic glomerular and tubular cells as well as in infiltrating leukocytes. Similar patterns of JNK activation are evident in animal models of acute and chronic renal injury. Administration of JNK inhibitors can protect against acute kidney injury and suppress the development of glomerulosclerosis and tubulointerstitial fibrosis. In particular, JNK activation in tubular epithelial cells may be a pivotal mechanism in determining the outcome of both acute kidney injury and progression of chronic kidney disease. JNK signaling promotes tubular epithelial cell production of pro-inflammatory and pro-fibrotic molecules as well as tubular cell de-differentiation toward a mesenchymal phenotype. However, the role of JNK within renal fibroblasts is less well-characterized. The JNK pathway interacts with other pro-fibrotic pathways, most notable with the TGF-β/SMAD pathway. JNK activation can augment TGF-β gene transcription, induce expression of enzymes that activate the latent form of TGF-β, and JNK directly phosphorylates SMAD3 to enhance transcription of pro-fibrotic molecules. In conclusion, JNK signaling plays an integral role in several key mechanisms operating in renal fibrosis. Targeting of JNK enzymes has therapeutic potential for the treatment of fibrotic kidney diseases.
Collapse
Affiliation(s)
- Keren Grynberg
- Department of Nephrology, Monash Medical Centre, Monash University Centre for Inflammatory Diseases, Monash Health, Clayton, VIC, Australia
| | - Frank Y Ma
- Department of Nephrology, Monash Medical Centre, Monash University Centre for Inflammatory Diseases, Monash Health, Clayton, VIC, Australia
| | - David J Nikolic-Paterson
- Department of Nephrology, Monash Medical Centre, Monash University Centre for Inflammatory Diseases, Monash Health, Clayton, VIC, Australia
| |
Collapse
|
417
|
Jiang L, Tang C, Rao J, Xue Q, Wu H, Wu D, Zhang A, Chen L, Shen Z, Lei L. Systematic identification of the druggable interactions between human protein kinases and naturally occurring compounds in endometriosis. Comput Biol Chem 2017; 71:136-143. [PMID: 29096379 DOI: 10.1016/j.compbiolchem.2017.10.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 10/08/2017] [Accepted: 10/14/2017] [Indexed: 01/31/2023]
Abstract
Diverse kinase signaling pathways have been involved in the pathogenesis of endometriosis (EM), which can be modulated either by directly targeting the hub kinases or by indirectly regulating marginal members in the pathways. Here, a systematic kinase-inhibitor interaction profile was created for 8 naturally occurring compounds against 20 human protein kinases. The compounds are all non-sterid that have been reported as pharmacologically active molecular entities potential for EM therapeutics, while the kinases were curated via gene ontology terms enriched from the gene co-citation network with EM. The resulting profile was analyzed at structural, energetic and dynamic levels to identify druggable kinase-compound interactions. The compounds Gossypol, Curcumin and EGCG showed a similar interaction profile across these kinases; they can bind tightly to the top-listed kinases in gene ontology, while the compounds Marrubiin, Apigenin and DIM were predicted to exhibit generally weak affinity for the 20 curated kinases. The JNK kinase, a MAPK family member, was identified as a putative candidate of druggable target for EM therapeutics; the inhibitory activity of eight naturally occurring compounds as well as a sophisticated kinase inhibitor SP600125 against the JNK was tested using enzymatic activity analysis. As might be expected, the Gossypol and EGCG were determined to have high inhibitory activity at namomolar level (IC50=55 and 94nM, respectively), which are comparable with or better than the positive control SP600125 (IC50=76nM), while other tested compounds exhibited weak inhibition (IC50>100nM) or bad potency (IC50=n.d.) against the kinase.
Collapse
Affiliation(s)
- Lai Jiang
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital of Anhui Medical University, Hefei 230001, China
| | - Chaoliang Tang
- Department of Anesthesiology, Anhui Provincial Hospital of Anhui Medical University, Hefei 230001, China; Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jie Rao
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital of Anhui Medical University, Hefei 230001, China
| | - Qing Xue
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital of Anhui Medical University, Hefei 230001, China
| | - Hao Wu
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital of Anhui Medical University, Hefei 230001, China
| | - Dabao Wu
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital of Anhui Medical University, Hefei 230001, China
| | - Aijun Zhang
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital of Anhui Medical University, Hefei 230001, China
| | - Ling Chen
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital of Anhui Medical University, Hefei 230001, China
| | - Zhen Shen
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital of Anhui Medical University, Hefei 230001, China
| | - Lei Lei
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital of Anhui Medical University, Hefei 230001, China.
| |
Collapse
|
418
|
Berntsen HF, Bogen IL, Wigestrand MB, Fonnum F, Walaas SI, Moldes-Anaya A. The fungal neurotoxin penitrem A induces the production of reactive oxygen species in human neutrophils at submicromolar concentrations. Toxicology 2017; 392:64-70. [PMID: 29037868 DOI: 10.1016/j.tox.2017.10.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 10/05/2017] [Accepted: 10/12/2017] [Indexed: 11/24/2022]
Abstract
Penitrem A is a fungal neurotoxin that recurrently causes intoxication in animals, and occasionally also in humans. We have previously reported that penitrem A induced the production of reactive oxygen species (ROS) in rat cerebellar granule cells, opening for a new mechanism of action for the neurotoxin. The aim of this study was to examine the potential of penitrem A to induce ROS production in isolated human neutrophil granulocytes, and to study possible mechanisms involved. Penitrem A significantly increased the production of ROS in human neutrophils at concentrations as low as 0.25μM (40% increase over basal levels), as measured with the DCF fluorescence assay. The EC50 determined for the production of ROS by penitrem A was 3.8μM. The maximal increase in ROS production was approximately 330% over basal levels at a concentration of 12.5μM. ROS formation was significantly inhibited by the antioxidant vitamin E (50μM), the intracellular Ca+2 chelator BAPTA-AM (5μM), the mitogen activated protein kinase kinase (MEK) 1/2 and 5 inhibitor U0126 (1 and 10μM), the p38 mitogen activated protein kinase (MAPK) inhibitor SB203580 (1μM), the c-Jun amino-terminal kinase (JNK) inhibitor SP600125 (10μM), and the calcineurin inhibitors FK-506 and cyclosporine A (1.5 and 0.5μM, respectively). These finding suggest that penitrem A is able to induce an increase in ROS production in neutrophils via the activation of several MAPK-signalling pathways. We suggest that this increase may partly explain the pathophysiology generated by penitrem A neuromycotoxicosis in both humans and animals.
Collapse
Affiliation(s)
- H F Berntsen
- Department of Administration, Lab Animal Unit, National Institute of Occupational Health, P.O. Box 8149 Dep, 0033 Oslo, Norway
| | - I L Bogen
- Oslo University Hospital, Department of Forensic Sciences, Section of Drug Abuse Research, P.O. Box 4950 Nydalen, N-0424 Oslo, Norway
| | - M B Wigestrand
- Institute of Basic Medical Sciences, Department of Biochemistry, University of Oslo, P.O. Box 1112 Blindern, N-0317 Oslo, Norway
| | - F Fonnum
- Institute of Basic Medical Sciences, Department of Biochemistry, University of Oslo, P.O. Box 1112 Blindern, N-0317 Oslo, Norway
| | - S I Walaas
- Institute of Basic Medical Sciences, Department of Biochemistry, University of Oslo, P.O. Box 1112 Blindern, N-0317 Oslo, Norway
| | - A Moldes-Anaya
- Section of Chemistry and Toxicology, Norwegian Veterinary Institute, P.O. Box 750 Sentrum, N-0106 Oslo, Norway; R&D Section, PET-center, University Hospital of North Norway (UNN), P.O. Box 100 Langnes, N-9038 Tromsø, Norway.
| |
Collapse
|
419
|
Moussa RS, Kovacevic Z, Bae DH, Lane DJR, Richardson DR. Transcriptional regulation of the cyclin-dependent kinase inhibitor, p21 CIP1/WAF1, by the chelator, Dp44mT. Biochim Biophys Acta Gen Subj 2017; 1862:761-774. [PMID: 29032246 DOI: 10.1016/j.bbagen.2017.10.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 10/10/2017] [Accepted: 10/11/2017] [Indexed: 01/04/2023]
Abstract
BACKGROUND The cyclin-dependent kinase inhibitor, p21, is well known for its role in cell cycle arrest. Novel anti-cancer agents that deplete iron pools demonstrate marked anti-tumor activity and are also active in regulating p21 expression. These agents induce p21 mRNA levels independently of the tumor suppressor, p53, and differentially regulate p21 protein expression depending on the cell-type. Several chelators, including an analogue of the potent anti-tumor agent, di-2-pyridylketone 4,4-dimethyl-3-thiosemicarbazone (Dp44mT), have entered clinical trials, and thus, their molecular mechanism of action is crucial to assess. Hence, this investigation examined how several iron chelators transcriptionally regulate p21. METHODS Promoter-deletion constructs; luciferase assays; RT-PCR; western analysis; gene silencing; co-immunoprecipitation. RESULTS The transcriptional regulation of the p21 promoter by iron chelators was demonstrated to be dependent on the chelator and cell-type examined. The potent anti-cancer chelator, Dp44mT, induced p21 promoter activity in SK-MEL-28 melanoma cells, but not in MCF-7 breast cancer cells. Further analysis of the p21 promoter identified a 50-bp region between -104 and -56-bp that was required for Dp44mT-induced activation in SK-MEL-28 cells. This region contained several Sp1-binding sites and mutational analysis of this region revealed the Sp1-3-binding site played a significant role in Dp44mT-induced activation of p21. Further, co-immunoprecipitation demonstrated that Dp44mT induced a marked increase in the interactions between Sp1 and the transcription factors, estrogen receptor-α and c-Jun. CONCLUSIONS AND GENERAL SIGNIFICANCE Dp44mT-induced p21 promoter activation via the Sp1-3-binding site and increased Sp1/ER-α and Sp1/c-Jun complex formation in SK-MEL-28 cells, suggesting these complexes were involved in p21 promoter activation.
Collapse
Affiliation(s)
- Rayan S Moussa
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Zaklina Kovacevic
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Dong-Hun Bae
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Darius J R Lane
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Des R Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia.
| |
Collapse
|
420
|
Nanni M, Ranieri D, Raffa S, Torrisi MR, Belleudi F. Interplay between FGFR2b-induced autophagy and phagocytosis: role of PLCγ-mediated signalling. J Cell Mol Med 2017; 22:668-683. [PMID: 28994193 PMCID: PMC6193413 DOI: 10.1111/jcmm.13352] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 07/19/2017] [Indexed: 12/25/2022] Open
Abstract
Signalling of the epithelial splicing variant of the fibroblast growth factor receptor 2 (FGFR2b) induces both autophagy and phagocytosis in human keratinocytes. Here, we investigated, in the cell model of HaCaT keratinocytes, whether the two processes might be related and the possible involvement of PLCγ signalling. Using fluorescence and electron microscopy, we demonstrated that the FGFR2b-induced phagocytosis and autophagy involve converging autophagosomal and phagosomal compartments. Moreover, the forced expression of FGFR2b signalling mutants and the use of specific inhibitors of FGFR2b substrates showed that the receptor-triggered autophagy requires PLCγ signalling, which in turn activates JNK1 via PKCδ. Finally, we found that in primary human keratinocytes derived from light or dark pigmented skin and expressing different levels of FGFR2b, the rate of phagocytosis and autophagy and the convergence of the two intracellular pathways are dependent on the level of receptor expression, suggesting that FGFR2b signalling would control in vivo the number of melanosomes in keratinocytes, determining skin pigmentation.
Collapse
Affiliation(s)
- Monica Nanni
- Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Danilo Ranieri
- Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Salvatore Raffa
- Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy.,S. Andrea University Hospital, Rome, Italy
| | - Maria Rosaria Torrisi
- Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy.,S. Andrea University Hospital, Rome, Italy
| | - Francesca Belleudi
- Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
421
|
t-BuOOH induces ferroptosis in human and murine cell lines. Arch Toxicol 2017; 92:759-775. [PMID: 28975372 DOI: 10.1007/s00204-017-2066-y] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 09/14/2017] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS)-induced apoptosis has been extensively studied. Increasing evidence suggests that ROS, for instance, induced by hydrogen peroxide (H2O2), might also trigger regulated necrotic cell death pathways. Almost nothing is known about the cell death pathways triggered by tertiary-butyl hydroperoxide (t-BuOOH), a widely used inducer of oxidative stress. The lipid peroxidation products induced by t-BuOOH are involved in the pathophysiology of many diseases, such as cancer, cardiovascular diseases, or diabetes. In this study, we exposed murine fibroblasts (NIH3T3) or human keratinocytes (HaCaT) to t-BuOOH (50 or 200 μM, respectively) which induced a rapid necrotic cell death. Well-established regulators of cell death, i.e., p53, poly(ADP)ribose polymerase-1 (PARP-1), the stress kinases p38 and c-Jun N-terminal-kinases 1/2 (JNK1/2), or receptor-interacting serine/threonine protein kinase 1 (RIPK1) and 3 (RIPK3), were not required for t-BuOOH-mediated cell death. Using the selective inhibitors ferrostatin-1 (1 μM) and liproxstatin-1 (1 μM), we identified ferroptosis, a recently discovered cell death mechanism dependent on iron and lipid peroxidation, as the main cell death pathway. Accordingly, t-BuOOH exposure resulted in a ferrostatin-1- and liproxstatin-1-sensitive increase in lipid peroxidation and cytosolic ROS. Ferroptosis was executed independently from other t-BuOOH-mediated cellular damages, i.e., loss of mitochondrial membrane potential, DNA double-strand breaks, or replication block. H2O2 did not cause ferroptosis at equitoxic concentrations (300 μM) and induced a (1) lower and (2) ferrostatin-1- or liproxstatin-1-insensitive increase in lipid peroxidation. We identify that t-BuOOH and H2O2 produce a different pattern of lipid peroxidation, thereby leading to different cell death pathways and present t-BuOOH as a novel inducer of ferroptosis.
Collapse
|
422
|
NLRP3 Phosphorylation Is an Essential Priming Event for Inflammasome Activation. Mol Cell 2017; 68:185-197.e6. [PMID: 28943315 DOI: 10.1016/j.molcel.2017.08.017] [Citation(s) in RCA: 362] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 07/11/2017] [Accepted: 08/21/2017] [Indexed: 12/12/2022]
Abstract
Many infections and stress signals can rapidly activate the NLRP3 inflammasome to elicit robust inflammatory responses. This activation requires a priming step, which is thought to be mainly for upregulating NLRP3 transcription. However, recent studies report that the NLRP3 inflammasome can be activated independently of transcription, suggesting that the priming process has unknown essential regulatory steps. Here, we report that JNK1-mediated NLRP3 phosphorylation at S194 is a critical priming event and is essential for NLRP3 inflammasome activation. We show that NLRP3 inflammasome activation is disrupted in NLRP3-S194A knockin mice. JNK1-mediated NLRP3 S194 phosphorylation is critical for NLRP3 deubiquitination and facilitates its self-association and the subsequent inflammasome assembly. Importantly, we demonstrate that blocking S194 phosphorylation prevents NLRP3 inflammasome activation in cryopyrin-associated periodic syndromes (CAPS). Thus, our study reveals a key priming molecular event that is a prerequisite for NLRP3 inflammasome activation. Inhibiting NLRP3 phosphorylation could be an effective treatment for NLRP3-related diseases.
Collapse
|
423
|
Blocking autophagy enhances the apoptotic effect of 18β-glycyrrhetinic acid on human sarcoma cells via endoplasmic reticulum stress and JNK activation. Cell Death Dis 2017; 8:e3055. [PMID: 28933787 PMCID: PMC5636985 DOI: 10.1038/cddis.2017.441] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 07/30/2017] [Accepted: 08/01/2017] [Indexed: 12/13/2022]
Abstract
Sarcoma, a rare form of cancer, is unlike the much more common carcinomas as it occurs in a distinct type of tissue. The potent antitumor effects of 18β-glycyrrhetinic acid (GA), a novel naturally derived agent, have been demonstrated in various cancers. However, the effect of GA on human sarcoma, and the underlying mechanisms, remain to be elucidated. In the current study, we show that GA inhibits sarcoma cell proliferation by inducing G0/G1-phase arrest. Exposure to GA resulted in the activation of caspase-3, -8, and -9, indicating that GA induced apoptosis through both extrinsic and intrinsic pathways. In addition, the autophagy pathway, characterized by the conversion of LC3-I to LC3- II, was activated, resulting in increased Beclin-1 protein levels, decreased p62 expression, and stimulation of autophagic flux. The present findings showed that GA stimulated autophagy by inducing endoplasmic reticulum (ER) stress via the IRE1–JNK pathway. Our data supported the prosurvival role of GA-induced autophagy when the autophagy pathway was blocked with specific chemical inhibitors. Finally, GA markedly reduced sarcoma growth, with little organ-related toxicity, in vivo. The present results suggest that the combination of GA with a specific autophagy inhibitor represents a promising therapeutic approach for the treatment of sarcoma.
Collapse
|
424
|
Tuure L, Hämäläinen M, Whittle BJ, Moilanen E. Microsomal Prostaglandin E Synthase-1 Expression in Inflammatory Conditions Is Downregulated by Dexamethasone: Seminal Role of the Regulatory Phosphatase MKP-1. Front Pharmacol 2017; 8:646. [PMID: 28983247 PMCID: PMC5613146 DOI: 10.3389/fphar.2017.00646] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 08/31/2017] [Indexed: 11/13/2022] Open
Abstract
Microsomal prostaglandin E synthase-1 (mPGES-1) is an inducible enzyme situated downstream of cyclo-oxygenase-2, promoting the excessive PGE2 production in inflammation. Dexamethasone is known to suppress mPGES-1 but the mechanisms regulating mPGES-1 expression remain poorly known. MKP-1 is a phosphatase controlling the proinflammatory MAP kinase pathways p38 and JNK, thus limiting the inflammatory responses. We have now investigated the role of MKP-1 and MAP kinases p38 and JNK in the regulation of mPGES-1 expression by dexamethasone. Dexamethasone increased MKP-1 and decreased mPGES-1 expression in J774 macrophages and in peritoneal macrophages from wild-type but not from MKP-1 deficient mice. Dexamethasone also reduced p38 and JNK phosphorylation along with enhancement of MKP-1, while inhibition of JNK reduced mPGES-1 expression. These findings were also translated to in vivo conditions as dexamethasone downregulated mPGES-1 expression in paw inflammation in wild-type but not in MKP-1 deficient mice. In conclusion, dexamethasone was found to downregulate mPGES-1 expression through enhanced MKP-1 expression and reduced JNK phosphorylation in inflammatory conditions. The results extend the understanding on the regulation of mPGES-1 expression and highlight the potential of MKP-1 as an anti-inflammatory drug target.
Collapse
Affiliation(s)
- Lauri Tuure
- The Immunopharmacology Research Group, Faculty of Medicine and Life Sciences, University of Tampere, Tampere University HospitalTampere, Finland
| | - Mari Hämäläinen
- The Immunopharmacology Research Group, Faculty of Medicine and Life Sciences, University of Tampere, Tampere University HospitalTampere, Finland
| | - Brendan J Whittle
- The Immunopharmacology Research Group, Faculty of Medicine and Life Sciences, University of Tampere, Tampere University HospitalTampere, Finland.,William Harvey Research Institute, Barts and the London School of MedicineLondon, United Kingdom
| | - Eeva Moilanen
- The Immunopharmacology Research Group, Faculty of Medicine and Life Sciences, University of Tampere, Tampere University HospitalTampere, Finland
| |
Collapse
|
425
|
Nguyen AH, Elliott IA, Wu N, Matsumura C, Vogelauer M, Attar N, Dann A, Ghukasyan R, Toste PA, Patel SG, Williams JL, Li L, Dawson DW, Radu C, Kurdistani SK, Donahue TR. Histone deacetylase inhibitors provoke a tumor supportive phenotype in pancreatic cancer associated fibroblasts. Oncotarget 2017; 8:19074-19088. [PMID: 27894105 PMCID: PMC5386671 DOI: 10.18632/oncotarget.13572] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 11/07/2016] [Indexed: 12/13/2022] Open
Abstract
Although histone deacetylase inhibitors (HDACi) are a promising class of anti-cancer drugs, thus far, they have been unsuccessful in early phase clinical trials for pancreatic ductal adenocarcinoma (PDAC). One potential reason for their poor efficacy is the tumor stroma, where cancer-associated fibroblasts (CAFs) are a prominent cell type and a source of resistance to cancer therapies. Here, we demonstrate that stromal fibroblasts contribute to the poor efficacy of HDACi's in PDAC. HDACi-treated fibroblasts show increased biological aggressiveness and are characterized by increased secretion of pro-inflammatory tumor-supportive cytokines and chemokines. We find that HDAC2 binds to the enhancer and promoter regions of pro-inflammatory genes specifically in CAFs and in silico analysis identified AP-1 to be the most frequently associated transcription factor bound in these regions. Pharmacologic inhibition of pathways upstream of AP-1 suppresses the HDACi-induced inflammatory gene expression and tumor-supportive responses in fibroblasts. Our findings demonstrate that the combination of HDACi's with chemical inhibitors of the AP-1 signaling pathway attenuate the inflammatory phenotype of fibroblasts and may improve the efficacy of HDACi in PDAC and, potentially, in other solid tumors rich in stroma.
Collapse
Affiliation(s)
- Andrew H Nguyen
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Irmina A Elliott
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Nanping Wu
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Cynthia Matsumura
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Maria Vogelauer
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Narsis Attar
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Amanda Dann
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Razmik Ghukasyan
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Paul A Toste
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Sanjeet G Patel
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Jennifer L Williams
- Department of Surgery, Harbor-UCLA Medical Center, Torrance, California, USA
| | - Luyi Li
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - David W Dawson
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Caius Radu
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Siavash K Kurdistani
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Timothy R Donahue
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
426
|
Díaz M, González R, Plano D, Palop JA, Sanmartín C, Encío I. A diphenyldiselenide derivative induces autophagy via JNK in HTB-54 lung cancer cells. J Cell Mol Med 2017; 22:289-301. [PMID: 28922542 PMCID: PMC5742718 DOI: 10.1111/jcmm.13318] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 06/19/2017] [Indexed: 12/21/2022] Open
Abstract
Symmetric aromatic diselenides are potential anticancer agents with strong cytotoxic activity. In this study, the in vitro anticancer activities of a novel series of diarylseleno derivatives from the diphenyldiselenide (DPDS) scaffold were evaluated. Most of the compounds exhibited high efficacy for inducing cytotoxicity against different human cancer cell lines. DPDS 2, the compound with the lowest mean GI50 value, induced both caspase-dependent apoptosis and arrest at the G0 /G1 phase in acute lymphoblastic leucemia CCRF-CEM cells. Consistent with this, PARP cleavage; enhanced caspase-2, -3, -8 and -9 activity; reduced CDK4 expression and increased levels of p53 were detected in these cells upon DPDS 2 treatment. Mutated p53 expressed in CCRF-CEM cells retains its transactivating activity. Therefore, increased levels of p21CIP1 and BAX proteins were also detected. On the other hand, DPDS 6, the compound with the highest selectivity index for cancer cells, resulted in G2 /M cell cycle arrest and caspase-independent cell death in p53 deficient HTB-54 lung cancer cells. Autophagy inhibitors 3-methyladenine, wortmannin and chloroquine inhibited DPDS 6-induced cell death. Consistent with autophagy, increased LC3-II and decreased SQSTM1/p62 levels were detected in HTB-54 cells in response to DPDS 6. Induction of JNK phosphorylation and a reduction in phospho-p38 MAPK were also detected. Moreover, the JNK inhibitor SP600125-protected HTB-54 cells from DPDS 6-induced cell death indicating that JNK activation is involved in DPDS 6-induced autophagy. These results highlight the anticancer effects of these derivatives and warrant future studies examining their clinical potential.
Collapse
Affiliation(s)
- Marta Díaz
- Department of Organic and Pharmaceutical Chemistry, Faculty of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain.,Instituto de Investigaciones Sanitarias de Navarra (IDISNA), Pamplona, Spain
| | - Roncesvalles González
- Departamento de Ciencias de la Salud, Universidad Pública de Navarra, Pamplona, Spain
| | - Daniel Plano
- Department of Organic and Pharmaceutical Chemistry, Faculty of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain.,Instituto de Investigaciones Sanitarias de Navarra (IDISNA), Pamplona, Spain
| | - Juan Antonio Palop
- Department of Organic and Pharmaceutical Chemistry, Faculty of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain.,Instituto de Investigaciones Sanitarias de Navarra (IDISNA), Pamplona, Spain
| | - Carmen Sanmartín
- Department of Organic and Pharmaceutical Chemistry, Faculty of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain.,Instituto de Investigaciones Sanitarias de Navarra (IDISNA), Pamplona, Spain
| | - Ignacio Encío
- Departamento de Ciencias de la Salud, Universidad Pública de Navarra, Pamplona, Spain
| |
Collapse
|
427
|
Sato-Matsubara M, Matsubara T, Daikoku A, Okina Y, Longato L, Rombouts K, Thuy LTT, Adachi J, Tomonaga T, Ikeda K, Yoshizato K, Pinzani M, Kawada N. Fibroblast growth factor 2 (FGF2) regulates cytoglobin expression and activation of human hepatic stellate cells via JNK signaling. J Biol Chem 2017; 292:18961-18972. [PMID: 28916723 PMCID: PMC5706471 DOI: 10.1074/jbc.m117.793794] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 09/11/2017] [Indexed: 12/19/2022] Open
Abstract
Cytoglobin (CYGB) belongs to the mammalian globin family and is exclusively expressed in hepatic stellate cells (HSCs) in the liver. In addition to its gas-binding ability, CYGB is relevant to hepatic inflammation, fibrosis, and cancer because of its anti-oxidative properties; however, the regulation of CYGB gene expression remains unknown. Here, we sought to identify factors that induce CYGB expression in HSCs and to clarify the molecular mechanism involved. We used the human HSC cell line HHSteC and primary human HSCs isolated from intact human liver tissues. In HHSteC cells, treatment with a culture supplement solution that included fibroblast growth factor 2 (FGF2) increased CYGB expression with concomitant and time-dependent α-smooth muscle actin (αSMA) down-regulation. We found that FGF2 is a key factor in inducing the alteration in both CYGB and αSMA expression in HHSteCs and primary HSCs and that FGF2 triggered the rapid phosphorylation of both c-Jun N-terminal kinase (JNK) and c-JUN. Both the JNK inhibitor PS600125 and transfection of c-JUN-targeting siRNA abrogated FGF2-mediated CYGB induction, and conversely, c-JUN overexpression induced CYGB and reduced αSMA expression. Chromatin immunoprecipitation analyses revealed that upon FGF2 stimulation, phospho-c-JUN bound to its consensus motif (5'-TGA(C/G)TCA), located -218 to -222 bases from the transcription initiation site in the CYGB promoter. Of note, in bile duct-ligated mice, FGF2 administration ameliorated liver fibrosis and significantly reduced HSC activation. In conclusion, FGF2 triggers CYGB gene expression and deactivation of myofibroblastic human HSCs, indicating that FGF2 has therapeutic potential for managing liver fibrosis.
Collapse
Affiliation(s)
| | - Tsutomu Matsubara
- Department of Anatomy and Regenerative Biology, Graduate School of Medicine, Osaka City University, Osaka 545-8585, Japan
| | | | | | - Lisa Longato
- the Regenerative Medicine and Fibrosis Group, Institute for Liver and Digestive Health, University College London, Royal Free, London NW3 2PF, United Kingdom, and
| | - Krista Rombouts
- the Regenerative Medicine and Fibrosis Group, Institute for Liver and Digestive Health, University College London, Royal Free, London NW3 2PF, United Kingdom, and
| | | | - Jun Adachi
- the Laboratory of Proteome Research, Proteome Research Center, National Institute of Biomedical Innovation, Osaka 567-0085, Japan
| | - Takeshi Tomonaga
- the Laboratory of Proteome Research, Proteome Research Center, National Institute of Biomedical Innovation, Osaka 567-0085, Japan
| | - Kazuo Ikeda
- Department of Anatomy and Regenerative Biology, Graduate School of Medicine, Osaka City University, Osaka 545-8585, Japan
| | | | - Massimo Pinzani
- the Regenerative Medicine and Fibrosis Group, Institute for Liver and Digestive Health, University College London, Royal Free, London NW3 2PF, United Kingdom, and
| | | |
Collapse
|
428
|
Disrupting CCT-β : β-tubulin selectively kills CCT-β overexpressed cancer cells through MAPKs activation. Cell Death Dis 2017; 8:e3052. [PMID: 28906489 PMCID: PMC5636972 DOI: 10.1038/cddis.2017.425] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 07/12/2017] [Accepted: 07/24/2017] [Indexed: 12/21/2022]
Abstract
We have previously demonstrated the ability of I-Trp to disrupt the protein–protein interaction of β-tubulin with chaperonin-containing TCP-1β (CCT-β). This caused more severe apoptosis in multidrug-resistant MES-SA/Dx5, compared to MES-SA, due to its higher CCT-β overexpression. In this study, we screened a panel of cancer cell lines, finding CCT-β overexpression in the triple-negative breast cancer cell line MDA-MB-231, colorectal cancer cell lines Colo205 and HCT116, and a gastric cancer cell line MKN-45. Thus, I-Trp killed these cancers with sub- to low-μM EC50, whereas it was non-toxic to MCF-10A. We then synthesized analogs of I-Trp and evaluated their cytotoxicity. Furthermore, apoptotic mechanism investigations revealed the activation of both protein ubiquitination/degradation and ER-associated protein degradation pathways. These pathways proceeded through activation of MAPKs at the onset of CCT-β : β-tubulin complex disruption. We thus establish an effective strategy to treat CCT-β overexpressed cancers by disrupting the CCT-β : β-tubulin complex.
Collapse
|
429
|
Lessel W, Silver A, Jechorek D, Guenther T, Roehl FW, Kalinski T, Roessner A, Poehlmann-Nitsche A. Inactivation of JNK2 as carcinogenic factor in colitis-associated and sporadic colorectal carcinogenesis. Carcinogenesis 2017; 38:559-569. [PMID: 28383667 DOI: 10.1093/carcin/bgx032] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 03/28/2017] [Indexed: 12/12/2022] Open
Abstract
We recently reported that dysregulated c-Jun N-terminal kinases (JNK) activity causes defective cell cycle checkpoint control, inducing neoplastic transformation in a cellular ulcerative colitis (UC) model. In the quiescent chronic phase of UC, p-p54 JNK was down-regulated and p-p46 JNK was up-regulated. Both were up-regulated in the acute phase. Consequently, increased p21WAF1 and γ-H2AX, two JNK-regulated proteins, induced cell cycle arrest. Their down-regulation led to checkpoint override, causing increased proliferation and undetected DNA damage in quiescent chronic phase, all characteristics of tumorigenesis. We investigated expression of p-JNK2, p-JNK1-3, p21WAF1, γ-H2AX and Ki67 by immunohistochemistry in cases of quiescent UC (QUC), active UC (AUC), UC-dysplasia and UC-related colorectal carcinoma (UC-CRC). Comparison was made to normal healthy colorectal mucosa, sporadic adenoma and colorectal carcinoma (CRC), diverticulitis and Crohns disease (CD). We found p-JNK2 up-regulation in AUC and its early down-regulation in UC-CRC and CRC carcinogenesis. With down-regulated p-JNK2, p21WAF1 was also decreased. Ki67 was inversely expressed, showing increased proliferation early in UC-CRC and CRC carcinogenesis. p-JNK1-3 was increased in AUC and QUC. Less increased γ-H2AX in UC-CRC compared to CRC gave evidence that colitis-triggered inflammation masks DNA damage, thus contributing to neoplastic transformation. We hypothesize that JNK-dependent cell cycle arrest is important in AUC, while chronic inflammation causes dysregulated JNK activity in quiescent phase that may contribute to checkpoint override, promoting UC carcinogenesis. We suggest restoring p-JNK2 expression as a novel therapeutic strategy to early prevent the development of UC-related cancer.
Collapse
Affiliation(s)
- Wiebke Lessel
- Department of Pathology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Andrew Silver
- Colorectal Cancer Genetics, Centre for Genomic and Child Health, Blizard Institute, Barts and The London School of Medicine and Dentistry, E1 2A London, UK
| | - Doerthe Jechorek
- Department of Pathology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Thomas Guenther
- Department of Pathology, 22339 Hamburg, Germany.,Academic Department of Histopathology, St. Mark's Hospital, HA1 3UJ Harrow, Middlesex, UK
| | - Friedrich-Wilhelm Roehl
- Department of Biometrics and Medical Informatics, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | | | - Albert Roessner
- Department of Pathology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | | |
Collapse
|
430
|
Astaxanthin acts via LRP-1 to inhibit inflammation and reverse lipopolysaccharide-induced M1/M2 polarization of microglial cells. Oncotarget 2017; 8:69370-69385. [PMID: 29050210 PMCID: PMC5642485 DOI: 10.18632/oncotarget.20628] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 08/17/2017] [Indexed: 12/17/2022] Open
Abstract
Microglia become activated during neuroinflammation and produce neurotoxic and neurotrophic factors, depending on whether they acquire M1 proinflammatory or M2 anti-inflammatory phenotypes. Astaxanthin (ATX), a natural carotenoid, has anti-inflammatory and neuroprotective effects. We investigated whether ATX could reverse M1/M2 polarization and suppress neuroinflammation via low-density lipoprotein receptor-related protein-1 (LRP-1). We observed increased expression of M1 (TNF-α, IL-1β, and CD86) and decreased expression of M2 (Arg-1, IL-10, and CD206) markers in BV2 microglial cells stimulated with lipopolysaccharide (LPS). These alterations were reversed by pretreating the cells with ATX. Activation of the NF-κB and JNK pathways was observed upon LPS stimulation, which was reversed by ATX. ATX-induced M2 polarization was attenuated by inhibition of NF-κB and JNK. Pretreatment of LPS-stimulated BV2 cells with ATX resulted in increased LRP-1 expression. The addition of receptor-associated protein, an LRP-1 antagonist, ameliorated ATX-induced inactivation of NF-κB and JNK signaling, and M2 polarization. ATX promotes M2 polarization to suppress neuroinflammation via LRP-1 by inhibiting NF-κB and JNK signaling. This novel mechanism may suppress neuroinflammation in diseases such as Alzheimer’s disease.
Collapse
|
431
|
Kainuma S, Tokuda H, Yamamoto N, Kuroyanagi G, Fujita K, Kawabata T, Sakai G, Matsushima-Nishiwaki R, Kozawa O, Otsuka T. Heat shock protein 27 (HSPB1) suppresses the PDGF-BB-induced migration of osteoblasts. Int J Mol Med 2017; 40:1057-1066. [PMID: 28902366 PMCID: PMC5593454 DOI: 10.3892/ijmm.2017.3119] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 08/28/2017] [Indexed: 12/13/2022] Open
Abstract
Heat shock protein 27 (HSP27/HSPB1), one of the small heat shock proteins, is constitutively expressed in various tissues. HSP27 and its phosphorylation state participate in the regulation of multiple physiological and pathophysiological cell functions. However, the exact roles of HSP27 in osteoblasts remain unclear. In the present study, we investigated the role of HSP27 in the platelet-derived growth factor‑BB (PDGF‑BB)‑stimulated migration of osteoblast-like MC3T3-E1 cells. PDGF-BB by itself barely upregulated the expression of HSP27 protein, but stimulated the phosphorylation of HSP27 in these cells. The PDGF-BB‑induced cell migration was significantly downregulated by HSP27 overexpression. The PDGF-BB-induced migrated cell numbers of the wild‑type HSP27-overexpressing cells and the phospho‑mimic HSP27-overexpressing (3D) cells were less than those of the unphosphorylatable HSP27-overexpressing (3A) cells. PD98059, an inhibitor of MEK1/2, SB203580, an inhibitor of p38 mitogen-activated protein kinase, and SP600125, an inhibitor of stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) reduced the PDGF-BB-induced migration of these cells, whereas Akt inhibitor or rapamycin, an inhibitor of upstream kinase of p70 S6 kinase (mTOR), barely affected the migration. However, the PDGF-BB-induced phosphorylation of p44/p42 MAPΚ, p38 MAPK and SAPK/JNK was not affected by HSP27 overexpression. There were no significant differences in the phosphorylation of p44/p42 MAPΚ, p38 MAP kinase or SAPK/JNK between the 3D cells and the 3A cells. These results strongly suggest that HSP27 functions as a negative regulator in the PDGF-BB-stimulated migration of osteoblasts, and the suppressive effect is amplified by the phosphorylation state of HSP27.
Collapse
Affiliation(s)
- Shingo Kainuma
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Haruhiko Tokuda
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Naohiro Yamamoto
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Gen Kuroyanagi
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Kazuhiko Fujita
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Tetsu Kawabata
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Go Sakai
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | | | - Osamu Kozawa
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Takanobu Otsuka
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| |
Collapse
|
432
|
Mandić AD, Bennek E, Verdier J, Zhang K, Roubrocks S, Davis RJ, Denecke B, Gassler N, Streetz K, Kel A, Hornef M, Cubero FJ, Trautwein C, Sellge G. c-Jun N-terminal kinase 2 promotes enterocyte survival and goblet cell differentiation in the inflamed intestine. Mucosal Immunol 2017; 10:1211-1223. [PMID: 28098247 DOI: 10.1038/mi.2016.125] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 11/15/2016] [Indexed: 02/06/2023]
Abstract
c-Jun N-terminal kinases (JNKs) contribute to immune signaling but their functional role during intestinal mucosal inflammation has remained ill defined. Using genetic mouse models, we characterized the role of JNK1 and JNK2 during homeostasis and acute colitis. Epithelial apoptosis, regeneration, differentiation, and barrier function were analyzed in intestinal epithelium-specific (ΔIEC) or complete JNK1 and bone marrow chimeric or complete JNK2 deficient mice as well as double-knockout animals (JNK1ΔIECJNK2-/-) during homeostasis and acute dextran sulfate sodium (DSS)-induced colitis. Results were confirmed using human HT-29 cells and wild-type or JNK2-deficient mouse intestinal organoid cultures. We show that nonhematopoietic JNK2 but not JNK1 expression confers protection from DSS-induced intestinal inflammation reducing epithelial barrier dysfunction and enterocyte apoptosis. JNK2 additionally enhanced Atonal homolog 1 expression, goblet cell and enteroendocrine cell differentiation, and mucus production under inflammatory conditions. Our results identify a protective role of epithelial JNK2 signaling to maintain mucosal barrier function, epithelial cell integrity, and mucus layer production in the event of inflammatory tissue damage.
Collapse
Affiliation(s)
- A D Mandić
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - E Bennek
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - J Verdier
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - K Zhang
- Department of Microbiology, University Hospital RWTH Aachen, Aachen, Germany
| | - S Roubrocks
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - R J Davis
- Howard Hughes Medical Institute and University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - B Denecke
- Interdisciplinary Center for Clinical Research (IZKF), University Hospital RWTH Aachen, Aachen, Germany
| | - N Gassler
- Institute of Pathology, Klinikum Braunschweig, Braunschweig, Germany
| | - K Streetz
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - A Kel
- GeneXplain GmbH, Wolfenbüttel, Germany
| | - M Hornef
- Department of Microbiology, University Hospital RWTH Aachen, Aachen, Germany
| | - F J Cubero
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - C Trautwein
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - G Sellge
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
433
|
Molecular screening and analysis of novel therapeutic inhibitors against c-Jun N-terminal kinase. Med Chem Res 2017. [DOI: 10.1007/s00044-017-1919-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
434
|
Che KF, Kaarteenaho R, Lappi-Blanco E, Levänen B, Sun J, Wheelock Å, Palmberg L, Sköld CM, Lindén A. Interleukin-26 Production in Human Primary Bronchial Epithelial Cells in Response to Viral Stimulation: Modulation by Th17 cytokines. Mol Med 2017; 23:247-257. [PMID: 28853490 DOI: 10.2119/molmed.2016.00064] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 08/21/2017] [Indexed: 12/22/2022] Open
Abstract
Interleukin (IL)-26 is abundant in human airways and this cytokine is involved in the local immune response to a bacterial stimulus in vivo. Specifically, local exposure to the toll-like receptor (TLR) 4 agonist endotoxin does increase IL-26 in human airways and this cytokine potentiates chemotactic responses in human neutrophils. In addition to T-helper (Th) 17 cells, alveolar macrophages can produce IL-26, but it remains unknown whether this cytokine can also be produced in the airway mucosa per se in response to a viral stimulus. Here, we evaluated whether this is the case using primary bronchial epithelial cells from the airway epithelium in vitro, and exploring the signaling mechanisms involved, including the modulatory effects of additional Th17 cytokines. Finally, we assessed IL-26 and its archetype signaling responses in healthy human airways in vivo. We found increased transcription and release of IL-26 protein after stimulation with the viral-related double stranded (ds) RNA polyinosinic-polycytidylic acid (poly-IC) and showed that this IL-26 release involved mitogen-activated protein (MAP) kinases and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). The release of IL-26 in response to a viral stimulus was modulated by additional Th17 cytokines. Moreover, there was transcription of IL26 mRNA and expression of the protein in epithelial cells of bronchial brush and tissue biopsies respectively after harvest in vivo. In addition, the extracellular IL-26 protein concentrations in bronchoalveolar lavage (BAL) samples did correlate with increased epithelial cell transcription of an archetype intracellular signaling molecule downstream of the IL-26-receptor complex, STAT1, in the bronchial brush biopsies. Thus, our study suggests that viral stimulation causes the production of IL-26 in lining epithelial cells of human airway structural cells that constitute a critical immune barrier and that this production is modulated by Th17 cytokines.
Collapse
Affiliation(s)
- Karlhans Fru Che
- Unit for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, SE-171 77 Stockholm, Sweden
| | - Riitta Kaarteenaho
- Unit of Medicine and Clinical Research, Pulmonary Division, University of Eastern Finland and Center of Medicine and Clinical Research, Division of Respiratory Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Elisa Lappi-Blanco
- Department of Pathology, Center for Cancer Research and Translational Medicine, University of Oulu, Oulu, Finland
| | - Bettina Levänen
- Unit for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, SE-171 77 Stockholm, Sweden
| | - Jitong Sun
- Unit for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, SE-171 77 Stockholm, Sweden
| | - Åsa Wheelock
- Respiratory Medicine Unit. Center for Molecular Medicine, Department of Medicine Solna, Karolinska Institutet, SE-171 76 Stockholm
| | - Lena Palmberg
- Unit for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, SE-171 77 Stockholm, Sweden
| | - C Magnus Sköld
- Respiratory Medicine Unit. Center for Molecular Medicine, Department of Medicine Solna, Karolinska Institutet, SE-171 76 Stockholm.,Lung Allergy Clinic, Karolinska University Hospital Solna, Stockholm, SE-171 76 Stockholm, Sweden
| | - Anders Lindén
- Unit for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, SE-171 77 Stockholm, Sweden.,Lung Allergy Clinic, Karolinska University Hospital Solna, Stockholm, SE-171 76 Stockholm, Sweden
| |
Collapse
|
435
|
Oben KZ, Alhakeem SS, McKenna MK, Brandon JA, Mani R, Noothi SK, Jinpeng L, Akunuru S, Dhar SK, Singh IP, Liang Y, Wang C, Abdel-Latif A, Stills HF, St Clair DK, Geiger H, Muthusamy N, Tohyama K, Gupta RC, Bondada S. Oxidative stress-induced JNK/AP-1 signaling is a major pathway involved in selective apoptosis of myelodysplastic syndrome cells by Withaferin-A. Oncotarget 2017; 8:77436-77452. [PMID: 29100399 PMCID: PMC5652791 DOI: 10.18632/oncotarget.20497] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 07/16/2017] [Indexed: 02/07/2023] Open
Abstract
Myelodysplastic syndromes (MDS) are a diverse group of malignant clonal hematopoietic stem cell disorders characterized by ineffective hematopoiesis, dysplastic cell morphology in one or more hematopoietic lineages, and a risk of progression to acute myeloid leukemia (AML). Approximately 50% of MDS patients respond to current FDA-approved drug therapies but a majority of responders relapse within 2-3 years. There is therefore a compelling need to identify potential new therapies for MDS treatment. We utilized the MDS-L cell line to investigate the anticancer potential and mechanisms of action of a plant-derived compound, Withaferin A (WFA), in MDS. WFA was potently cytotoxic to MDS-L cells but had no significant effect on the viability of normal human primary bone marrow cells. WFA also significantly reduced engraftment of MDS-L cells in a xenotransplantation model. Through transcriptome analysis, we identified reactive oxygen species (ROS)-activated JNK/AP-1 signaling as a major pathway mediating apoptosis of MDS-L cells by WFA. We conclude that the molecular mechanism mediating selective cytotoxicity of WFA on MDS-L cells is strongly associated with induction of ROS. Therefore, pharmacologic manipulation of redox biology could be exploited as a selective therapeutic target in MDS.
Collapse
Affiliation(s)
- Karine Z Oben
- Markey Cancer Center and Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA
| | - Sara S Alhakeem
- Markey Cancer Center and Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA
| | - Mary K McKenna
- Markey Cancer Center and Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA
| | - Jason A Brandon
- Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Rajeswaran Mani
- Comprehensive Cancer Center and Department of Internal Medicine, Ohio State University, Columbus, OH 43210, USA
| | - Sunil K Noothi
- Markey Cancer Center and Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA
| | - Liu Jinpeng
- Biostatistics Core, Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Shailaja Akunuru
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229, USA
| | - Sanjit K Dhar
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Inder P Singh
- Department of Natural Products, National Institute of Pharmaceutical Research, S.A.S Nagar, Punjab 160062, India
| | - Ying Liang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Chi Wang
- Biostatistics Core, Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Ahmed Abdel-Latif
- Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Harold F Stills
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA
| | - Daret K St Clair
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Hartmut Geiger
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229, USA
| | - Natarajan Muthusamy
- Comprehensive Cancer Center and Department of Internal Medicine, Ohio State University, Columbus, OH 43210, USA
| | - Kaoru Tohyama
- Department of Laboratory Medicine, Kawasaki Medical School, Kurashiki, Okayama 701-0192, Japan
| | - Ramesh C Gupta
- Department of Pharmacology and Toxicology, and James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Subbarao Bondada
- Markey Cancer Center and Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
436
|
Role of the DNA repair glycosylase OGG1 in the activation of murine splenocytes. DNA Repair (Amst) 2017; 58:13-20. [PMID: 28843610 DOI: 10.1016/j.dnarep.2017.08.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 08/07/2017] [Accepted: 08/08/2017] [Indexed: 01/06/2023]
Abstract
OGG1 (8-oxoguanine-DNA glycosylase) is the major DNA repair glycosylase removing the premutagenic DNA base modification 8-oxo-7,8-dihydroguanine (8-oxoG) from the genome of mammalian cells. In addition, there is accumulating evidence that OGG1 and its substrate 8-oxoG might function in the regulation of certain genes, which could account for an attenuated immune response observed in Ogg1-/- mice in several settings. Indications for at least two different mechanisms have been obtained. Thus, OGG1 could either act as an ancillary transcription factor cooperating with the lysine-specific demethylase LSD1 or as an activator of small GTPases. Here, we analysed the activation by lipopolysaccaride (LPS) of primary splenocytes obtained from two different Ogg1-/- mouse strains. We found that the induction of TNF-α expression was reduced in splenocytes (in particular macrophages) of both Ogg1-/- strains. Notably, an inhibitor of LSD1, OG-L002, reduced the induction of TNF-α mRNA in splenocytes from wild-type mice to the level observed in splenocytes from Ogg1-/- mice and had no influence in the latter cells. In contrast, inhibitors of the MAP kinases p38 and JNK as well as the antioxidant N-acetylcysteine attenuated the LPS-stimulated TNF-α expression both in the absence and presence of OGG1. The free base 8-oxo-7,8-dihydroguanine had no influence on the TNF-α expression in the splenocytes. The data demonstrate that OGG1 plays a role in an LSD1-dependent pathway of LPS-induced macrophage activation in mice.
Collapse
|
437
|
Janczar S, Nautiyal J, Xiao Y, Curry E, Sun M, Zanini E, Paige AJ, Gabra H. WWOX sensitises ovarian cancer cells to paclitaxel via modulation of the ER stress response. Cell Death Dis 2017; 8:e2955. [PMID: 28749468 PMCID: PMC5550887 DOI: 10.1038/cddis.2017.346] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 06/09/2017] [Accepted: 06/14/2017] [Indexed: 12/22/2022]
Abstract
There are clear gaps in our understanding of genes and pathways through which cancer cells facilitate survival strategies as they become chemoresistant. Paclitaxel is used in the treatment of many cancers, but development of drug resistance is common. Along with being an antimitotic agent paclitaxel also activates endoplasmic reticulum (ER) stress. Here, we examine the role of WWOX (WW domain containing oxidoreductase), a gene frequently lost in several cancers, in mediating paclitaxel response. We examine the ER stress-mediated apoptotic response to paclitaxel in WWOX-transfected epithelial ovarian cancer (EOC) cells and following siRNA knockdown of WWOX. We show that WWOX-induced apoptosis following exposure of EOC cells to paclitaxel is related to ER stress and independent of the antimitotic action of taxanes. The apoptotic response to ER stress induced by WWOX re-expression could be reversed by WWOX siRNA in EOC cells. We report that paclitaxel treatment activates both the IRE-1 and PERK kinases and that the increase in paclitaxel-mediated cell death through WWOX is dependent on active ER stress pathway. Log-rank analysis of overall survival (OS) and progression-free survival (PFS) in two prominent EOC microarray data sets (Tothill and The Cancer Genome Atlas), encompassing ~800 patients in total, confirmed clinical relevance to our findings. High WWOX mRNA expression predicted longer OS and PFS in patients treated with paclitaxel, but not in patients who were treated with only cisplatin. The association of WWOX and survival was dependent on the expression level of glucose-related protein 78 (GRP78), a key ER stress marker in paclitaxel-treated patients. We conclude that WWOX sensitises EOC to paclitaxel via ER stress-induced apoptosis, and predicts clinical outcome in patients. Thus, ER stress response mechanisms could be targeted to overcome chemoresistance in cancer.
Collapse
Affiliation(s)
- Szymon Janczar
- Department of Surgery and Cancer, Molecular Therapeutics Unit and Ovarian Cancer Action Research Centre, Imperial College London, Hammersmith Hospital, London, UK.,Department of Pediatrics, Oncology, Hematology and Diabetology, Medical University of Lodz, Lodz, Poland
| | - Jaya Nautiyal
- Department of Surgery and Cancer, Molecular Therapeutics Unit and Ovarian Cancer Action Research Centre, Imperial College London, Hammersmith Hospital, London, UK
| | - Yi Xiao
- Department of Surgery and Cancer, Molecular Therapeutics Unit and Ovarian Cancer Action Research Centre, Imperial College London, Hammersmith Hospital, London, UK
| | - Edward Curry
- Department of Surgery and Cancer, Molecular Therapeutics Unit and Ovarian Cancer Action Research Centre, Imperial College London, Hammersmith Hospital, London, UK
| | - Mingjun Sun
- Department of Surgery and Cancer, Molecular Therapeutics Unit and Ovarian Cancer Action Research Centre, Imperial College London, Hammersmith Hospital, London, UK
| | - Elisa Zanini
- Department of Surgery and Cancer, Molecular Therapeutics Unit and Ovarian Cancer Action Research Centre, Imperial College London, Hammersmith Hospital, London, UK
| | - Adam Jw Paige
- Department of Surgery and Cancer, Molecular Therapeutics Unit and Ovarian Cancer Action Research Centre, Imperial College London, Hammersmith Hospital, London, UK.,Department of Life Sciences, University of Bedfordshire, Luton, UK
| | - Hani Gabra
- Department of Surgery and Cancer, Molecular Therapeutics Unit and Ovarian Cancer Action Research Centre, Imperial College London, Hammersmith Hospital, London, UK.,Clinical Discovery Unit, Early Clinical Development, AstraZeneca, Cambridge, UK
| |
Collapse
|
438
|
Phosphatase wild-type p53-induced phosphatase 1 controls the development of T H9 cells and allergic airway inflammation. J Allergy Clin Immunol 2017; 141:2168-2181. [PMID: 28732646 DOI: 10.1016/j.jaci.2017.06.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 06/02/2017] [Accepted: 06/20/2017] [Indexed: 12/24/2022]
Abstract
BACKGROUND Allergic asthma is one of the most common diseases worldwide, resulting in a burden of diseases. No available therapeutic regimens can cure asthma thus far. OBJECTIVE We sought to identify new molecular targets for TH9 cell-mediated allergic airway inflammation. METHODS Wild-type p53-induced phosphatase 1 (Wip1) gene knockout mice, Wip1 inhibitor-treated mice, and ovalbumin-induced allergic airway inflammation mouse models were used to characterize the roles of Wip1 in allergic airway inflammation. The induction of TH cell subsets in vitro, real-time PCR, immunoblots, luciferase assays, and chromatin immunoprecipitation assays were used to determine the regulatory pathways of Wip1 in TH9 differentiation. RESULTS Here we demonstrate that Wip1-deficient mice are less prone to allergic airway inflammation, as indicated by the decreased pathologic alterations in lungs. Short-term treatment with a Wip1-specific inhibitor significantly ameliorates allergic inflammation progression. Intriguingly, Wip1 selectively impaired TH9 but not TH1, TH2, and TH17 cell differentiation. Biochemical assays show that Wip1 deficiency increases c-Jun/c-Fos activity in a c-Jun N-terminal kinase-dependent manner and that c-Jun/c-Fos directly binds to Il9 promoter and inhibits Il9 transcription. CONCLUSION Wip1 controls TH9 cell development through regulating c-Jun/c-Fos activity on the Il9 promoter and is important for the pathogenesis of allergic airway inflammation. These findings shed light on the previously unrecognized roles of Wip1 in TH9 cell differentiation. The inhibitory effects of a Wip1 inhibitor on the pathogenesis of allergic airway inflammation can have important implications for clinical application of Wip1 inhibitors in allergy therapies.
Collapse
|
439
|
Wang T, Liao Y, Sun Q, Tang H, Wang G, Zhao F, Jin Y. Upregulation of Matrix Metalloproteinase-9 in Primary Cultured Rat Astrocytes Induced by 2-Chloroethanol Via MAPK Signal Pathways. Front Cell Neurosci 2017; 11:218. [PMID: 28769771 PMCID: PMC5516094 DOI: 10.3389/fncel.2017.00218] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 07/06/2017] [Indexed: 01/25/2023] Open
Abstract
2-Chloroethanol (2-CE) is one of the reactive metabolites of 1,2-DCE in vivo, which might contribute to brain edema formation induced by 1,2-dichloroethane (1,2-DCE) poisoning. Thus, the purpose of this study was to explore the roles of mitogen-activated protein kinase (MAPK) signal pathways in upregulation of matrix metalloproteinase-9 (MMP-9) in 2-CE exposed rat astrocytes. Expression of p38 MAPK (p38), extracellular signal regulated protein kinase (ERK), c-Jun N-terminal kinase (JNK) and MMP-9 at both protein and gene levels in rat astrocytes were determined using western blot and real-time RT-PCR methods. The results showed that both protein and mRNA levels of MMP-9 in 2-CE exposed astrocytes significantly increased. Meanwhile, protein levels of phosphorylated p38 (p-p38), ERK1/2 (p-ERK1/2) and JNK1/2 (p-JNK1/2) in 2-CE exposed astrocytes also significantly increased. In addition, both protein and mRNA levels of MMP-9 significantly decreased in response to reduced protein levels of p-p38, p-ERK1/2 and p-JNK1/2 achieved by supplement with their specific inhibitors, indicating that activation of MAPK signal pathways might play an important role in upregulation of MMP-9 expression at the transcriptional level in 2-CE exposed astrocytes. Furthermore, since pretreatment of n-acetyl-l-cysteine (NAC), a powerful antioxidant amino acid, could attenuate the elevated levels of MMP-9, p-p38, p-ERK2 and p-JNK1/2 in 2-CE exposed astrocytes, activation of MAPK signal pathways in 2-CE exposed astrocytes could be mediated partially by reactive oxygen species (ROS), which was most likely generated in the metabolism of 2-CE.
Collapse
Affiliation(s)
- Tong Wang
- Department of Occupational and Environmental Health, School of Public Health, China Medical UniversityShenyang, China
| | - Yingjun Liao
- Department of Physiology, China Medical UniversityShenyang, China
| | - Qi Sun
- Department of Occupational and Environmental Health, School of Public Health, China Medical UniversityShenyang, China
| | - Hongge Tang
- Department of Occupational and Environmental Health, School of Public Health, China Medical UniversityShenyang, China
| | - Gaoyang Wang
- Department of Occupational and Environmental Health, School of Public Health, China Medical UniversityShenyang, China
| | - Fenghong Zhao
- Department of Occupational and Environmental Health, School of Public Health, China Medical UniversityShenyang, China
| | - Yaping Jin
- Department of Occupational and Environmental Health, School of Public Health, China Medical UniversityShenyang, China
| |
Collapse
|
440
|
Subedi L, Venkatesan R, Kim SY. Neuroprotective and Anti-Inflammatory Activities of Allyl Isothiocyanate through Attenuation of JNK/NF-κB/TNF-α Signaling. Int J Mol Sci 2017; 18:ijms18071423. [PMID: 28671636 PMCID: PMC5535914 DOI: 10.3390/ijms18071423] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 06/28/2017] [Accepted: 06/29/2017] [Indexed: 12/23/2022] Open
Abstract
Allyl isothiocyanate (AITC), present in Wasabia japonica (wasabi), is an aliphatic isothiocyanate derived from the precursor sinigrin, which is a glucosinolate present in vegetables of the Brassica family. Traditionally, it has been used to treat rheumatic arthralgia, blood circulation, and pain. This study focuses on its anti-apoptotic activity through the regulation of lipopolysaccharide (LPS)-induced neuroinflammation. Furthermore, we assessed its neuroprotective efficacy, which it achieves through the upregulation of nerve growth factor (NGF) production. Pretreatment with AITC significantly inhibited inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) expression, decreased tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), prostaglandin E2 (PGE2), and nitric oxide (NO) production in activated microglia, and increased the nerve growth factor (NGF) and neurite outgrowth in neuroblastoma cells. AITC inhibited the nuclear factor (NF-κB-mediated transcription by modulating mitogen activated protein kinase (MAPK) signaling, particularly downregulating c-Jun N-terminal kinase (JNK) phosphorylation, which was followed by a reduction in the TNF-α expression in activated microglia. This promising effect of AITC in controlling JNK/NF-κB/TNF-α cross-linking maintains the Bcl-2 gene family and protects neuroblastoma cells from activated microglia-induced toxicity. These findings provide novel insights into the anti-neuroinflammatory effects of AITC on microglial cells, which may have clinical significance in neurodegeneration.
Collapse
Affiliation(s)
- Lalita Subedi
- Laboratory of Pharmacognosy, College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, #191, Hambakmoero, Yeonsu-gu, Incheon 21936, Korea.
| | - Ramu Venkatesan
- Laboratory of Pharmacognosy, College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, #191, Hambakmoero, Yeonsu-gu, Incheon 21936, Korea.
| | - Sun Yeou Kim
- Laboratory of Pharmacognosy, College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, #191, Hambakmoero, Yeonsu-gu, Incheon 21936, Korea.
| |
Collapse
|
441
|
Waetzig V, Belzer M, Haeusgen W, Boehm R, Cascorbi I, Herdegen T. Crosstalk control and limits of physiological c-Jun N-terminal kinase activity for cell viability and neurite stability in differentiated PC12 cells. Mol Cell Neurosci 2017; 82:12-22. [DOI: 10.1016/j.mcn.2017.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 02/28/2017] [Accepted: 04/13/2017] [Indexed: 10/19/2022] Open
|
442
|
Huang W, Solouki S, Koylass N, Zheng SG, August A. ITK signalling via the Ras/IRF4 pathway regulates the development and function of Tr1 cells. Nat Commun 2017; 8:15871. [PMID: 28635957 PMCID: PMC5482062 DOI: 10.1038/ncomms15871] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 05/05/2017] [Indexed: 12/16/2022] Open
Abstract
Type 1 regulatory T (Tr1) cells differentiate in response to signals engaging the T cell receptor (TCR), express high levels of the immunosuppressive cytokine IL-10, but not Foxp3, and can suppress inflammation and promote immune tolerance. Here we show that ITK, an important modulator of TCR signalling, is required for the TCR-induced development of Tr1 cells in various organs, and in the mucosal system during parasitic and viral infections. ITK kinase activity is required for mouse and human Tr1 cell differentiation. Tr1 cell development and suppressive function of Itk deficient cells can be restored by the expression of the transcription factor interferon regulatory factor 4 (IRF4). Downstream of ITK, Ras activity is responsible for Tr1 cell induction, as expression of constitutively active HRas rescues IRF4 expression and Tr1 cell differentiation in Itk-/- cells. We conclude that TCR/ITK signalling through the Ras/IRF4 pathway is required for functional development of Tr1 cells.
Collapse
Affiliation(s)
- Weishan Huang
- Center for Clinical Immunology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, China
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York 14853, USA
| | - Sabrina Solouki
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York 14853, USA
| | - Nicholas Koylass
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York 14853, USA
| | - Song-Guo Zheng
- Center for Clinical Immunology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, China
- Department of Medicine, Milton S. Hershey Medical Center, Pennsylvania State University, Hershey, Pennsylvania 17033, USA
| | - Avery August
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York 14853, USA
| |
Collapse
|
443
|
Ashenden M, van Weverwijk A, Murugaesu N, Fearns A, Campbell J, Gao Q, Iravani M, Isacke CM. An In Vivo Functional Screen Identifies JNK Signaling As a Modulator of Chemotherapeutic Response in Breast Cancer. Mol Cancer Ther 2017; 16:1967-1978. [PMID: 28611109 DOI: 10.1158/1535-7163.mct-16-0731] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 04/15/2017] [Accepted: 05/18/2017] [Indexed: 11/16/2022]
Abstract
Chemotherapy remains the mainstay of treatment for advanced breast cancer; however, resistance is an inevitable event for the majority of patients with metastatic disease. Moreover, there is little information available to guide stratification of first-line chemotherapy, crucial given the common development of multidrug resistance. Here, we describe an in vivo screen to interrogate the response to anthracycline-based chemotherapy in a syngeneic metastatic breast cancer model and identify JNK signaling as a key modulator of chemotherapy response. Combining in vitro and in vivo functional analyses, we demonstrate that JNK inhibition both promotes tumor cell cytostasis and blocks activation of the proapoptotic protein Bax, thereby antagonizing chemotherapy-mediated cytotoxicity. To investigate the clinical relevance of this dual role of JNK signaling, we developed a proliferation-independent JNK activity signature and demonstrate high JNK activity to be enriched in triple-negative and basal-like breast cancer subtypes. Consistent with the dual role of JNK signaling in vitro, high-level JNK pathway activation in triple-negative breast cancers is associated both with poor patient outcome in the absence of chemotherapy treatment and, in neoadjuvant clinical studies, is predictive of enhanced chemotherapy response. These data highlight the potential of monitoring JNK activity as early biomarker of response to chemotherapy and emphasize the importance of rational treatment regimes, particularly when combining cytostatic and chemotherapeutic agents. Mol Cancer Ther; 16(9); 1967-78. ©2017 AACR.
Collapse
Affiliation(s)
- Matthew Ashenden
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Antoinette van Weverwijk
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Nirupa Murugaesu
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Antony Fearns
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - James Campbell
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Qiong Gao
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Marjan Iravani
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Clare M Isacke
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom.
| |
Collapse
|
444
|
Khorasanizadeh M, Eskian M, Gelfand EW, Rezaei N. Mitogen-activated protein kinases as therapeutic targets for asthma. Pharmacol Ther 2017; 174:112-126. [DOI: 10.1016/j.pharmthera.2017.02.024] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
445
|
Börchers S, Babaei R, Klimpel C, Duque Escobar J, Schröder S, Blume R, Malik MNH, Oetjen E. TNFα-induced DLK activation contributes to apoptosis in the beta-cell line HIT. Naunyn Schmiedebergs Arch Pharmacol 2017; 390:813-825. [DOI: 10.1007/s00210-017-1385-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 05/17/2017] [Indexed: 12/20/2022]
|
446
|
Boubriak II, Malhas AN, Drozdz MM, Pytowski L, Vaux DJ. Stress-induced release of Oct-1 from the nuclear envelope is mediated by JNK phosphorylation of lamin B1. PLoS One 2017; 12:e0177990. [PMID: 28542436 PMCID: PMC5443517 DOI: 10.1371/journal.pone.0177990] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 05/06/2017] [Indexed: 02/08/2023] Open
Abstract
The nuclear lamina can bind and sequester transcription factors (TFs), a function lost if the lamina is abnormal, with missing or mutant lamin proteins. We now show that TF sequestration is not all-or-nothing, but a dynamic physiological response to external signals. We show that the binding of the ubiquitous TF, Oct-1, to lamin B1 was reversed under conditions of cellular stress caused, inter alia, by the chemical methylating agent methylmethanesulfonate (MMS). A search for lamin B1 post-translational modifications that might mediate changes in Oct-1 binding using kinase inhibitors uncovered a role for c-Jun N-terminal kinase (JNK). Phosphoproteomic and site-directed mutagenesis analyses of lamin B1 isolated from control and MMS-treated nuclei identified T575 as a JNK site phosphorylated after stress. A new phospho-T575 specific anti-peptide antibody confirmed increased interphase cellular T575 phosphorylation after cell exposure to certain stress conditions, enabling us to conclude that lamin B1 acts as an interphase kinase target, releasing Oct-1 to execute a protective response to stress.
Collapse
Affiliation(s)
- Ivan I. Boubriak
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Ashraf N. Malhas
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Marek M. Drozdz
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Lior Pytowski
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - David J. Vaux
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
447
|
Gautam J, Ku JM, Regmi SC, Jeong H, Wang Y, Banskota S, Park MH, Nam TG, Jeong BS, Kim JA. Dual Inhibition of NOX2 and Receptor Tyrosine Kinase by BJ-1301 Enhances Anticancer Therapy Efficacy via Suppression of Autocrine-Stimulatory Factors in Lung Cancer. Mol Cancer Ther 2017; 16:2144-2156. [PMID: 28536313 DOI: 10.1158/1535-7163.mct-16-0915] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 04/18/2017] [Accepted: 05/17/2017] [Indexed: 11/16/2022]
Abstract
NADPH oxidase-derived reactive oxygen species (ROS) potentiate receptor tyrosine kinase (RTK) signaling, resulting in enhanced angiogenesis and tumor growth. In this study, we report that BJ-1301, a hybrid of pyridinol and alpha-tocopherol, exerts anticancer effects by dual inhibition of NADPH oxidase and RTK activities in endothelial and lung cancer cells. BJ-1301 suppresses ROS production by blocking translocation of NADPH oxidase cytosolic subunits to the cell membrane, thereby inhibiting activation. The potency of RTK inhibition by BJ-1301 was lower than that of sunitinib (a multi-RTK inhibitor), but the inhibition of downstream signaling pathways (e.g., ROS generation) and subsequent biological changes (e.g., NOX2 induction) by BJ-1301 was superior. Consistently, BJ-1301 inhibited cisplatin-resistant lung cancer cell proliferation more than sunitinib did. In xenograft chick or mouse tumor models, BJ-1301 inhibited lung tumor growth, to an extent greater than that of sunitinib or cisplatin. Treatments with BJ-1301 induced regression of tumor growth, potentially due to downregulation of autocrine-stimulatory ligands for RTKs, such as TGFα and stem cell factor, in tumor tissues. Taken together, the current study demonstrates that BJ-1301 is a promising anticancer drug for the treatment of lung cancer. Mol Cancer Ther; 16(10); 2144-56. ©2017 AACR.
Collapse
Affiliation(s)
- Jaya Gautam
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Jin-Mo Ku
- Bio-Center, Gyeonggi Institute of Science and Technology Promotion, Suwon, Republic of Korea
| | | | - Hyunyoung Jeong
- Departments of Pharmacy Practice and Biopharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| | - Ying Wang
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Suhrid Banskota
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Myo-Hyeon Park
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Tae-Gyu Nam
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Republic of Korea
| | - Byeong-Seon Jeong
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea.
| | - Jung-Ae Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea.
| |
Collapse
|
448
|
Shen J, Chen X, Jia H, Meyers CA, Shrestha S, Asatrian G, Ding C, Tsuei R, Zhang X, Peault B, Ting K, Soo C, James AW. Effects of WNT3A and WNT16 on the Osteogenic and Adipogenic Differentiation of Perivascular Stem/Stromal Cells. Tissue Eng Part A 2017; 24:68-80. [PMID: 28463594 DOI: 10.1089/ten.tea.2016.0387] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Human perivascular stem/stromal cells (hPSC) are a multipotent mesenchymogenic stromal cell population defined by their perivascular locale. Recent studies have demonstrated the high potential for clinical translation of this fluorescence-activated cell sorting (FACS)-derived cell population for autologous bone tissue engineering. However, the mechanisms underlying the osteogenic differentiation of PSC are incompletely understood. The current study investigates the roles of canonical and noncanonical Wnt signaling in the osteogenic and adipogenic differentiation of PSC. Results showed that both canonical and noncanonical Wnt signaling activity transiently increased during PSC osteogenic differentiation in vitro. Sustained WNT3A treatment significantly decreased PSC osteogenic differentiation. Conversely, sustained treatment with Wnt family member 16 (WNT16), a mixed canonical and noncanonical ligand, increased osteogenic differentiation in a c-Jun N-terminal kinase (JNK) pathway-dependent manner. Conversely, WNT16 knockdown significantly diminished PSC osteogenic differentiation. Finally, WNT16 but not WNT3A increased the adipogenic differentiation of PSC. These results indicate the importance of regulation of canonical and noncanonical Wnt signaling for PSC fate and differentiation. Moreover, these data suggest that WNT16 plays a functional and necessary role in PSC osteogenesis.
Collapse
Affiliation(s)
- Jia Shen
- 1 Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA , Los Angeles, California
| | - Xuepeng Chen
- 2 Department of Orthodontics, Stomatological Hospital, Zhejiang University , Hangzhou, China
| | - Haichao Jia
- 3 Department of Orthodontics, School of Stomatology, Capital Medical University , Beijing, China
| | - Carolyn A Meyers
- 4 Department of Pathology, Johns Hopkins University , Baltimore, Maryland
| | - Swati Shrestha
- 1 Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA , Los Angeles, California
| | - Greg Asatrian
- 1 Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA , Los Angeles, California
| | - Catherine Ding
- 1 Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA , Los Angeles, California
| | - Rebecca Tsuei
- 1 Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA , Los Angeles, California
| | - Xinli Zhang
- 1 Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA , Los Angeles, California
| | - Bruno Peault
- 5 Department of Orthopaedic Surgery, UCLA and Orthopaedic Hospital, Orthopaedic Hospital Research Center , Los Angeles, California.,6 Center for Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh , Edinburgh, United Kingdom
| | - Kang Ting
- 1 Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA , Los Angeles, California
| | - Chia Soo
- 5 Department of Orthopaedic Surgery, UCLA and Orthopaedic Hospital, Orthopaedic Hospital Research Center , Los Angeles, California.,7 Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California , Los Angeles, Los Angeles, California
| | - Aaron W James
- 4 Department of Pathology, Johns Hopkins University , Baltimore, Maryland.,5 Department of Orthopaedic Surgery, UCLA and Orthopaedic Hospital, Orthopaedic Hospital Research Center , Los Angeles, California
| |
Collapse
|
449
|
Hein TW, Xu W, Xu X, Kuo L. Acute and Chronic Hyperglycemia Elicit JIP1/JNK-Mediated Endothelial Vasodilator Dysfunction of Retinal Arterioles. Invest Ophthalmol Vis Sci 2017; 57:4333-40. [PMID: 27556216 PMCID: PMC5015966 DOI: 10.1167/iovs.16-19990] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Hyperglycemia, a hallmark of diabetes mellitus, is associated with retinal inflammation and impairment of endothelium-dependent nitric oxide (NO)–mediated dilation of retinal arterioles. However, molecular mechanisms involved in this diminished endothelial vasodilator function remain unclear. We examined whether inflammatory stress-activated kinases, c-Jun N-terminal kinase (JNK) and p38, contribute to retinal arteriolar dysfunction during exposure to acute and chronic hyperglycemia. Methods Retinal arterioles were isolated from streptozocin-induced diabetic pigs (2 weeks; chronic hyperglycemia, 471 ± 23 mg/dL) or age-matched control pigs (euglycemia, 79 ± 5 mg/dL), and then cannulated and pressurized for vasoreactivity study. For acute hyperglycemia study, vessels from nondiabetic pigs were exposed intraluminally to high glucose (25 mM ≈ 450 mg/dL) for 2 hours, and normal glucose (5 mM ≈ 90 mg/dL) served as the control. Results Endothelium-dependent vasodilation to bradykinin was reduced in a similar manner after exposure to acute or chronic hyperglycemia. Administration of NO synthase inhibitor NG-nitro-L-arginine methyl ester (L-NAME) nearly abolished vasodilations either in control (euglycemia and normal glucose) or hyperglycemic (acute and chronic) vessels. Treatment of either acute or chronic hyperglycemic vessels with JNK inhibitor SP600125 or JNK-interacting protein-1 (JIP1) inhibitor BI-78D3, but not p38 inhibitor SB203580, preserved bradykinin-induced dilation in an L-NAME–sensitive manner. By contrast, endothelium-independent vasodilation to sodium nitroprusside was unaffected by acute or chronic hyperglycemia. Conclusions Activation of JIP1/JNK signaling in retinal arterioles during exposure to acute or chronic hyperglycemia leads to selective impairment of endothelium-dependent NO-mediated dilation. Therapeutic targeting of the vascular JNK pathway may improve retinal endothelial vasodilator function during early diabetes.
Collapse
Affiliation(s)
- Travis W Hein
- Department of Surgery, Baylor Scott & White Eye Institute, College of Medicine, Texas A&M Health Science Center, Temple, Texas, United States
| | - Wenjuan Xu
- Department of Surgery, Baylor Scott & White Eye Institute, College of Medicine, Texas A&M Health Science Center, Temple, Texas, United States
| | - Xin Xu
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Temple, Texas, United States
| | - Lih Kuo
- Department of Surgery, Baylor Scott & White Eye Institute, College of Medicine, Texas A&M Health Science Center, Temple, Texas, United States 2Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Temple, Texas, United States
| |
Collapse
|
450
|
Novel tumor-suppressor function of KLF4 in pediatric T-cell acute lymphoblastic leukemia. Exp Hematol 2017; 53:16-25. [PMID: 28479419 DOI: 10.1016/j.exphem.2017.04.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 04/21/2017] [Accepted: 04/22/2017] [Indexed: 02/07/2023]
Abstract
Acute lymphoblastic leukemia (ALL) is the most common hematological malignancy in pediatric patients. Despite advances in the treatment of this disease, many children with T-cell ALL (T-ALL) die from disease relapse due to low responses to standard chemotherapy and the lack of a targeted therapy that selectively eradicates the chemoresistant leukemia-initiating cells (LICs) responsible for disease recurrence. We reported recently that the reprogramming factor Krüppel-like factor 4 (KLF4) has a tumor-suppressive function in children with T-ALL. KLF4 silencing by promoter deoxyribonucleic acid (DNA) methylation in patients with T-ALL leads to aberrant activation of the mitogen-activated protein kinase kinase MAP2K7 and the downstream c-Jun NH2-terminal kinase (JNK) pathway that controls the expansion of leukemia cells via c-Jun and activating transcription factor 2. This pathway can be inhibited with small molecules and therefore has the potential to eliminate LICs and eradicate disease in combination with standard therapy for patients with refractory and relapsed disease. The present review summarizes the role of the KLF4-MAP2K7 pathway in T-ALL pathogenesis and the function of JNK and MAP2K7 in carcinogenesis and therapy.
Collapse
|