151
|
Yin Y, Ma P, Wang S, Zhang Y, Han R, Huo C, Wu M, Deng H. The CRTC-CREB axis functions as a transcriptional sensor to protect against proteotoxic stress in Drosophila. Cell Death Dis 2022; 13:688. [PMID: 35933423 PMCID: PMC9357022 DOI: 10.1038/s41419-022-05122-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 01/21/2023]
Abstract
cAMP Responsible Element Binding Protein (CREB) is an evolutionarily conserved transcriptional factor that regulates cell growth, synaptic plasticity and so on. In this study, we unexpectedly found proteasome inhibitors, such as MLN2238, robustly increase CREB activity in adult flies through a large-scale compound screening. Mechanistically, reactive oxidative species (ROS) generated by proteasome inhibition are required and sufficient to promote CREB activity through c-Jun N-terminal kinase (JNK). In 293 T cells, JNK activation by MLN2238 is also required for increase of CREB phosphorylation at Ser133. Meanwhile, transcriptome analysis in fly intestine identified a group of genes involved in redox and proteostatic regulation are augmented by overexpressing CRTC (CREB-regulated transcriptional coactivator). Intriguingly, CRTC overexpression in muscles robustly restores protein folding and proteasomal activity in a fly Huntington's disease (HD) model, and ameliorates HD related pathogenesis, such as protein aggregates, motility, and lifespan. Moreover, CREB activity increases during aging, and further enhances its activity can suppress protein aggregates in aged muscles. Together, our results identified CRTC/CREB downstream ROS/JNK signaling as a conserved sensor to tackle oxidative and proteotoxic stresses. Boosting CRTC/CREB activity is a potential therapeutic strategy to treat aging related protein aggregation diseases.
Collapse
Affiliation(s)
- Youjie Yin
- grid.24516.340000000123704535 Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, School of Life Sciences and Technology, Tongji University, Shanghai, 20092 China
| | - Peng Ma
- grid.24516.340000000123704535 Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, School of Life Sciences and Technology, Tongji University, Shanghai, 20092 China
| | - Saifei Wang
- grid.24516.340000000123704535 Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, School of Life Sciences and Technology, Tongji University, Shanghai, 20092 China
| | - Yao Zhang
- grid.24516.340000000123704535 Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, School of Life Sciences and Technology, Tongji University, Shanghai, 20092 China
| | - Ruolei Han
- grid.24516.340000000123704535 Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, School of Life Sciences and Technology, Tongji University, Shanghai, 20092 China
| | - Chunyu Huo
- grid.24516.340000000123704535 Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, School of Life Sciences and Technology, Tongji University, Shanghai, 20092 China
| | - Meixian Wu
- grid.24516.340000000123704535 Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, School of Life Sciences and Technology, Tongji University, Shanghai, 20092 China
| | - Hansong Deng
- grid.24516.340000000123704535 Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, School of Life Sciences and Technology, Tongji University, Shanghai, 20092 China
| |
Collapse
|
152
|
Yoshikawa T, Fukuda A, Omatsu M, Namikawa M, Sono M, Yuichi F, Masuda T, Araki O, Nagao M, Ogawa S, Masuo K, Goto N, Hiramatsu Y, Muta Y, Tsuda M, Maruno T, Nakanishi Y, Kawada K, Takaishi S, Seno H. JNK pathway plays a critical role for expansion of human colorectal cancer in the context of BRG1 suppression. Cancer Sci 2022; 113:3417-3427. [PMID: 35924439 PMCID: PMC9530857 DOI: 10.1111/cas.15520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 07/14/2022] [Accepted: 07/26/2022] [Indexed: 12/09/2022] Open
Abstract
Tumor stem cells (TSCs), capable of self‐renewal and continuous production of progeny cells, could be potential therapeutic targets. We have recently reported that chromatin remodeling regulator Brg1 is required for maintenance of murine intestinal TSCs and stemness feature of human colorectal cancer (CRC) cells by inhibiting apoptosis. However, it is still unclear how BRG1 suppression changes the underlying intracellular mechanisms of human CRC cells. We found that Brg1 suppression resulted in upregulation of the JNK signaling pathway in human CRC cells and murine intestinal TSCs. Simultaneous suppression of BRG1 and the JNK pathway, either by pharmacological inhibition or silencing of c‐JUN, resulted in even stronger inhibition of the expansion of human CRC cells compared to Brg1 suppression alone. Consistently, high c‐JUN expression correlated with worse prognosis for survival in human CRC patients with low BRG1 expression. Therefore, the JNK pathway plays a critical role for expansion and stemness of human CRC cells in the context of BRG1 suppression, and thus a combined blockade of BRG1 and the JNK pathway could be a novel therapeutic approach against human CRC.
Collapse
Affiliation(s)
- Takaaki Yoshikawa
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Gastroenterology and Hepatology, Kitano Hospital, Tazuke Kofukai Medical Research Institute, Osaka, Japan
| | - Akihisa Fukuda
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Mayuki Omatsu
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Mio Namikawa
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Makoto Sono
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Fukunaga Yuichi
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Drug Discovery Medicine, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tomonori Masuda
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Osamu Araki
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Munemasa Nagao
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Satoshi Ogawa
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kenji Masuo
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Laboratory for Malignancy Control Research (DSK project), Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Norihiro Goto
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yukiko Hiramatsu
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yu Muta
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Motoyuki Tsuda
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takahisa Maruno
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuki Nakanishi
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kenji Kawada
- Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shigeo Takaishi
- Laboratory for Malignancy Control Research (DSK project), Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
153
|
Samuvel DJ, Nguyen NT, Jaeschke H, Lemasters JJ, Wang X, Choo YM, Hamann MT, Zhong Z. Platanosides, a Potential Botanical Drug Combination, Decrease Liver Injury Caused by Acetaminophen Overdose in Mice. JOURNAL OF NATURAL PRODUCTS 2022; 85:1779-1788. [PMID: 35815804 PMCID: PMC9788857 DOI: 10.1021/acs.jnatprod.2c00324] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Oxidative stress plays an important role in acetaminophen (APAP)-induced hepatotoxicity. Platanosides (PTSs) isolated from the American sycamore tree (Platanus occidentalis) represent a potential new four-molecule botanical drug class of antibiotics active against drug-resistant infectious disease. Preliminary studies have suggested that PTSs are safe and well tolerated and have antioxidant properties. The potential utility of PTSs in decreasing APAP hepatotoxicity in mice in addition to an assessment of their potential with APAP for the control of infectious diseases along with pain and pyrexia associated with a bacterial infection was investigated. On PTS treatment in mice, serum alanine aminotransferase (ALT) release, hepatic centrilobular necrosis, and 4-hydroxynonenal (4-HNE) were markedly decreased. In addition, inducible nitric oxide synthase (iNOS) expression and c-Jun-N-terminal kinase (JNK) activation decreased when mice overdosed with APAP were treated with PTSs. Computational studies suggested that PTSs may act as JNK-1/2 and Keap1-Nrf2 inhibitors and that the isomeric mixture could provide greater efficacy than the individual molecules. Overall, PTSs represent promising botanical drugs for hepatoprotection and drug-resistant bacterial infections and are effective in protecting against APAP-related hepatotoxicity, which decreases liver necrosis and inflammation, iNOS expression, and oxidative and nitrative stresses, possibly by preventing persistent JNK activation.
Collapse
Affiliation(s)
- Devadoss J. Samuvel
- Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - Nga T. Nguyen
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| | - John J. Lemasters
- Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
- Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, United States
| | - Xiaojuan Wang
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, People Republic of China
| | - Yeun-Mun Choo
- Chemistry Department, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | - Mark T. Hamann
- Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - Zhi Zhong
- Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
154
|
Fu X, Matsui T, Funaba M. Enhancement of vitamin C-induced myogenesis by inhibition of extracellular signal-regulated kinase (ERK) 1/2 pathway. Biochem Biophys Res Commun 2022; 612:57-62. [DOI: 10.1016/j.bbrc.2022.04.103] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 04/15/2022] [Accepted: 04/22/2022] [Indexed: 11/02/2022]
|
155
|
Ethanol Extract of Sargassum siliquastrum Inhibits Lipopolysaccharide-Induced Nitric Oxide Generation by Downregulating the Nuclear Factor-Kappa B Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:6160010. [PMID: 35722164 PMCID: PMC9205721 DOI: 10.1155/2022/6160010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 05/04/2022] [Indexed: 11/17/2022]
Abstract
Sargassum siliquastrum (SS) is an edible brown seaweed widely consumed in Korea and considered a functional food source. Previous studies have reported various biological activities of SS extracts, including antioxidant and hepatoprotective properties. In the present study, we examined the anti-inflammatory effects of the SS extract and assessed the underlying mechanism of action. The SS extract significantly inhibited lipopolysaccharide (LPS)-induced nitric oxide (NO) production in a dose-dependent manner (% of NO production at 500 μg/mL: 60.1 ± 0.9%), with no obvious toxicity. Furthermore, the SS extract inhibited mRNA and protein expression levels of inducible NO synthase, as well as LPS-induced expression and production of proinflammatory cytokines such as IL-1β, IL-6, or TNF-α (IL-6 production (ng/mL) : LPS−: 0.7 ± 0.3; LPS+: 68.1 ± 2.8; LPS + SS extract: 51.9 ± 1.2; TNF-α production (ng/mL) : LPS−: 0.3 ± 0.1; LPS+: 23.0 ± 0.1; LPS + SS extract: 18.2 ± 10.8). Mechanistically, the SS extract attenuated LPS-induced activation of the nuclear factor kappa-light-chain-enhancer of activated B cells (nuclear factor-kappa B, NF-κB) signaling pathway such as phosphorylation of NF-κB p65 and degradation of IκB-α, thereby blocking LPS-induced activation of NF-κB transcriptional activity. The SS extract also enhanced LPS-induced heme oxygenase-1 expression and attenuated LPS-induced cellular reactive oxygen species production (% of ROS production at 500 μg/mL: 52.2 ± 1.3%). Collectively, these findings suggest that the SS extract elicits anti-inflammatory effects in mouse macrophage cells.
Collapse
|
156
|
Sanders J, Castiglione M, Shun T, Vollmer LL, Schurdak ME, Vogt A, Schwacha A. Validation of a high throughput screening assay to identify small molecules that target the eukaryotic replicative helicase. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2022; 27:229-241. [PMID: 35058181 PMCID: PMC9196137 DOI: 10.1016/j.slasd.2021.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Mcm2-7 is the catalytic core of the eukaryotic replicative helicase, which together with CDC45 and the GINS complex unwind parental DNA to generate templates for DNA polymerase. Being a highly regulated and complex enzyme that operates via an incompletely understood multi-step mechanism, molecular probes of Mcm2-7 that interrogate specific mechanistic steps would be useful tools for research and potential future chemotherapy. Based upon a synthetic lethal approach, we previously developed a budding yeast multivariate cell-based high throughput screening (HTS) assay to identify putative Mcm inhibitors by their ability to specifically cause a growth defect in an mcm mutant relative to a wild-type strain[1]. Here, as proof of concept, we used this assay to screen a 1280-member compound library (LOPAC) for potential Mcm2-7 inhibitors. Primary screening and dose-dependent retesting identified twelve compounds from this library that specifically inhibited the growth of the Mcm mutant relative to the corresponding wild-type strain (0.9 % hit rate). Secondary assays were employed to rule out non-specific DNA damaging agents, establish direct protein-ligand interaction via biophysical methods, and verify in vivo DNA replication inhibition via fluorescence activated cell sorter analysis (FACS). We identified one agent (β-carboline-3-carboxylic acid N-methylamide, CMA) that physically bound to the purified Mcm2-7 complex (Kdapp119 µM), and at slightly higher concentrations specifically blocked S-phase cell cycle progression of the wild-type strain. In total, identification of Mcm2-7 as a CMA target validates our synthetic lethal HTS assay paradigm as a tool to identify chemical probes for the Mcm2-7 replicative helicase.
Collapse
Affiliation(s)
- Jordan Sanders
- The Department of Biological Sciences, University of Pittsburgh, Pittsburgh PA 15260
| | - Michael Castiglione
- Drug Discovery Institute, University of Pittsburgh Medical School, Pittsburgh PA 15260
| | - Tongying Shun
- Drug Discovery Institute, University of Pittsburgh Medical School, Pittsburgh PA 15260
| | - Laura L Vollmer
- Drug Discovery Institute, University of Pittsburgh Medical School, Pittsburgh PA 15260
| | - Mark E Schurdak
- Drug Discovery Institute, University of Pittsburgh Medical School, Pittsburgh PA 15260; Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh PA 15260
| | - Andreas Vogt
- Drug Discovery Institute, University of Pittsburgh Medical School, Pittsburgh PA 15260; Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh PA 15260.
| | - Anthony Schwacha
- The Department of Biological Sciences, University of Pittsburgh, Pittsburgh PA 15260.
| |
Collapse
|
157
|
Kuroyanagi G, Tokuda H, Fujita K, Kawabata T, Sakai G, Kim W, Hioki T, Tachi J, Matsushima-Nishiwaki R, Otsuka T, Iida H, Kozawa O. Upregulation of TGF-β-induced HSP27 by HSP90 inhibitors in osteoblasts. BMC Musculoskelet Disord 2022; 23:495. [PMID: 35619094 PMCID: PMC9134601 DOI: 10.1186/s12891-022-05419-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 04/27/2022] [Indexed: 06/02/2024] Open
Abstract
Background Heat shock protein (HSP) 90 functions as a molecular chaperone and is constitutively expressed and induced in response to stress in many cell types. We have previously demonstrated that transforming growth factor-β (TGF-β), the most abundant cytokine in bone cells, induces the expression of HSP27 through Smad2, p44/p42 mitogen-activated protein kinase (MAPK), p38 MAPK, and stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) in mouse osteoblastic MC3T3-E1 cells. This study investigated the effects of HSP90 on the TGF-β-induced HSP27 expression and the underlying mechanism in mouse osteoblastic MC3T3-E1 cells. Methods Clonal osteoblastic MC3T3-E1 cells were treated with the HSP90 inhibitors and then stimulated with TGF-β. HSP27 expression and the phosphorylation of Smad2, p44/p42 MAPK, p38 MAPK, and SAPK/JNK were evaluated by western blot analysis. Result HSP90 inhibitors 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG) and onalespib significantly enhanced the TGF-β-induced HSP27 expression. TGF-β inhibitor SB431542 reduced the enhancement by 17-DMAG or onalespib of the TGF-β-induced HSP27 expression levels. HSP90 inhibitors, geldanamycin, onalespib, and 17-DMAG did not affect the TGF-β-stimulated phosphorylation of Smad2. Geldanamycin did not affect the TGF-β-stimulated phosphorylation of p44/p42 MAPK or p38 MAPK but significantly enhanced the TGF-β-stimulated phosphorylation of SAPK/JNK. Onalespib also increased the TGF-β-stimulated phosphorylation of SAPK/JNK. Furthermore, SP600125, a specific inhibitor for SAPK/JNK, significantly suppressed onalespib or geldanamycin’s enhancing effect of the TGF-β-induced HSP27 expression levels. Conclusion Our results strongly suggest that HSP90 inhibitors upregulated the TGF-β-induced HSP27 expression and that these effects of HSP90 inhibitors were mediated through SAPK/JNK pathway in osteoblasts. Supplementary Information The online version contains supplementary material available at 10.1186/s12891-022-05419-1.
Collapse
Affiliation(s)
- Gen Kuroyanagi
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, 1, Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan. .,Department of Rehabilitation Medicine, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan. .,Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu, Japan.
| | - Haruhiko Tokuda
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu, Japan.,Department of Clinical Laboratory/Medical Genome Center Biobank, National Center for Geriatrics and Gerontology, Obu, Japan.,Department of Metabolic Research, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Kazuhiko Fujita
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, 1, Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan.,Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Tetsu Kawabata
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, 1, Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan.,Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Go Sakai
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, 1, Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan.,Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Woo Kim
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu, Japan.,Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Tomoyuki Hioki
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu, Japan.,Department of Dermatology, Kizawa Memorial Hospital, Minokamo, Japan
| | - Junko Tachi
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu, Japan.,Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | | | - Takanobu Otsuka
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, 1, Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Hiroki Iida
- Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Osamu Kozawa
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu, Japan
| |
Collapse
|
158
|
Goswami P, Ives AM, Abbott ARN, Bertke AS. Stress Hormones Epinephrine and Corticosterone Selectively Reactivate HSV-1 and HSV-2 in Sympathetic and Sensory Neurons. Viruses 2022; 14:1115. [PMID: 35632856 PMCID: PMC9147053 DOI: 10.3390/v14051115] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 12/16/2022] Open
Abstract
Herpes simplex viruses 1 and 2 (HSV-1 and HSV-2) establish latency in sensory and autonomic neurons, from which they can reactivate to cause recurrent disease throughout the life of the host. Stress is strongly associated with HSV recurrences in humans and animal models. However, the mechanisms through which stress hormones act on the latent virus to cause reactivation are unknown. We show that the stress hormones epinephrine (EPI) and corticosterone (CORT) induce HSV-1 reactivation selectively in sympathetic neurons, but not sensory or parasympathetic neurons. Activation of multiple adrenergic receptors is necessary for EPI-induced HSV-1 reactivation, while CORT requires the glucocorticoid receptor. In contrast, CORT, but not EPI, induces HSV-2 reactivation in both sensory and sympathetic neurons through either glucocorticoid or mineralocorticoid receptors. Reactivation is dependent on different transcription factors for EPI and CORT, and coincides with rapid changes in viral gene expression, although genes differ for HSV-1 and HSV-2, and temporal kinetics differ for EPI and CORT. Thus, stress-induced reactivation mechanisms are neuron-specific, stimulus-specific and virus-specific. These findings have implications for differences in HSV-1 and HSV-2 recurrent disease patterns and frequencies, as well as development of targeted, more effective antivirals that may act on different responses in different types of neurons.
Collapse
Affiliation(s)
- Poorna Goswami
- Translational Biology Medicine and Health, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA;
| | - Angela M. Ives
- Biomedical and Veterinary Science, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA;
| | - Amber R. N. Abbott
- Department of Biology, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA;
| | - Andrea S. Bertke
- Population Health Sciences, Center for Emerging Zoonotic and Arthropod-Borne Pathogens, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
| |
Collapse
|
159
|
Schwärzler J, Mayr L, Vich Vila A, Grabherr F, Niederreiter L, Philipp M, Grander C, Meyer M, Jukic A, Tröger S, Enrich B, Przysiecki N, Tschurtschenthaler M, Sommer F, Kronberger I, Koch J, Hilbe R, Hess MW, Oberhuber G, Sprung S, Ran Q, Koch R, Effenberger M, Kaneider NC, Wieser V, Keller MA, Weersma RK, Aden K, Rosenstiel P, Blumberg RS, Kaser A, Tilg H, Adolph TE. PUFA-Induced Metabolic Enteritis as a Fuel for Crohn's Disease. Gastroenterology 2022; 162:1690-1704. [PMID: 35031299 DOI: 10.1053/j.gastro.2022.01.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 12/16/2021] [Accepted: 01/04/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Crohn's disease (CD) globally emerges with Westernization of lifestyle and nutritional habits. However, a specific dietary constituent that comprehensively evokes gut inflammation in human inflammatory bowel diseases remains elusive. We aimed to delineate how increased intake of polyunsaturated fatty acids (PUFAs) in a Western diet, known to impart risk for developing CD, affects gut inflammation and disease course. We hypothesized that the unfolded protein response and antioxidative activity of glutathione peroxidase 4 (GPX4), which are compromised in human CD epithelium, compensates for metabolic perturbation evoked by dietary PUFAs. METHODS We phenotyped and mechanistically dissected enteritis evoked by a PUFA-enriched Western diet in 2 mouse models exhibiting endoplasmic reticulum (ER) stress consequent to intestinal epithelial cell (IEC)-specific deletion of X-box binding protein 1 (Xbp1) or Gpx4. We translated the findings to human CD epithelial organoids and correlated PUFA intake, as estimated by a dietary questionnaire or stool metabolomics, with clinical disease course in 2 independent CD cohorts. RESULTS PUFA excess in a Western diet potently induced ER stress, driving enteritis in Xbp1-/-IEC and Gpx4+/-IEC mice. ω-3 and ω-6 PUFAs activated the epithelial endoplasmic reticulum sensor inositol-requiring enzyme 1α (IRE1α) by toll-like receptor 2 (TLR2) sensing of oxidation-specific epitopes. TLR2-controlled IRE1α activity governed PUFA-induced chemokine production and enteritis. In active human CD, ω-3 and ω-6 PUFAs instigated epithelial chemokine expression, and patients displayed a compatible inflammatory stress signature in the serum. Estimated PUFA intake correlated with clinical and biochemical disease activity in a cohort of 160 CD patients, which was similarly demonstrable in an independent metabolomic stool analysis from 199 CD patients. CONCLUSIONS We provide evidence for the concept of PUFA-induced metabolic gut inflammation which may worsen the course of human CD. Our findings provide a basis for targeted nutritional therapy.
Collapse
Affiliation(s)
- Julian Schwärzler
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Lisa Mayr
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Arnau Vich Vila
- Department of Gastroenterology and Hepatology, University of Groningen and Groningen University Medical Center, Groningen, the Netherlands
| | - Felix Grabherr
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Lukas Niederreiter
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Maureen Philipp
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Christoph Grander
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Moritz Meyer
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Almina Jukic
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Simone Tröger
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Barbara Enrich
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Nicole Przysiecki
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Markus Tschurtschenthaler
- Institute for Experimental Cancer Therapy, Center for Translational Cancer Research (TranslaTUM), Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; Department of Internal Medicine II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Felix Sommer
- Institute of Clinical Molecular Biology, Christian Albrecht University Kiel and Schleswig-Holstein University Hospital, Kiel, Germany
| | - Irmgard Kronberger
- Department of Visceral, Transplant, and Thoracic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Jakob Koch
- Institute of Human Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Richard Hilbe
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria
| | - Michael W Hess
- Institute of Histology and Embryology, Medical University of Innsbruck, Innsbruck, Austria
| | - Georg Oberhuber
- INNPATH, Innsbruck Medical University Hospital, Innsbruck, Austria
| | - Susanne Sprung
- Department of Pathology, Medical University of Innsbruck, Innsbruck, Austria
| | - Qitao Ran
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, Texas
| | - Robert Koch
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Maria Effenberger
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Nicole C Kaneider
- Division of Gastroenterology and Hepatology, Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Verena Wieser
- Department of Obstetrics and Gynecology, Medical University of Innsbruck, Innsbruck, Austria
| | - Markus A Keller
- Institute of Human Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Rinse K Weersma
- Department of Gastroenterology and Hepatology, University of Groningen and Groningen University Medical Center, Groningen, the Netherlands
| | - Konrad Aden
- Institute of Clinical Molecular Biology, Christian Albrecht University Kiel and Schleswig-Holstein University Hospital, Kiel, Germany
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Christian Albrecht University Kiel and Schleswig-Holstein University Hospital, Kiel, Germany
| | - Richard S Blumberg
- Gastroenterology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Arthur Kaser
- Division of Gastroenterology and Hepatology, Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, and Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
160
|
Liu Q, Pan J, Bao L, Xu C, Qi Y, Jiang B, Wang D, Zhu X, Li X, Zhang H, Bai H, Yang Q, Ma J, Wiemer EAC, Ben J, Chen Q. Major Vault Protein Prevents Atherosclerotic Plaque Destabilization by Suppressing Macrophage ASK1-JNK Signaling. Arterioscler Thromb Vasc Biol 2022; 42:580-596. [PMID: 35387478 DOI: 10.1161/atvbaha.121.316662] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Macrophages are implicated in atherosclerotic plaque instability by inflammation and degradation of extracellular matrix. However, the regulatory mechanisms driving these macrophage-associated processes are not well understood. Here, we aimed to identify the plaque destabilization-associated cytokines and signaling pathways in macrophages. METHODS The atherosclerotic models of myeloid-specific MVP (major vault protein) knockout mice and control mice were generated. Atherosclerotic instability, macrophage inflammatory signaling, and active cytokines released by macrophages were examined in vivo and in vitro by using cellular and molecular biological approaches. RESULTS MVP deficiency in myeloid cells exacerbated murine plaque instability by increasing production of both MMP (matrix metallopeptidase)-9 and proinflammatory cytokines in artery wall. Mechanistically, expression of MMP-9 was mediated via ASK1 (apoptosis signal-regulating kinase 1)-MKK-4 (mitogen-activated protein kinase kinase 4)-JNK (c-Jun N-terminal kinase) signaling in macrophages. MVP and its α-helical domain could bind with ASK1 and inhibit its dimerization and phosphorylation. A 62 amino acid peptide (MVP-[686-747]) in the α-helical domain of MVP showed a crucial role in preventing macrophage MMP-9 production and plaque instability. CONCLUSIONS MVP may act as an inhibitor for ASK1-JNK signaling-mediated MMP-9 production in macrophages and, thereby, attenuate unstable plaque formation. Our findings suggest that suppression of macrophage ASK1-JNK signaling may be a useful strategy antagonizing atherosclerotic diseases.
Collapse
Affiliation(s)
- Qingling Liu
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Junlu Pan
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Linrui Bao
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Chunxiang Xu
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Yu Qi
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Bin Jiang
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Dongdong Wang
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Xudong Zhu
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Xiaoyu Li
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Hanwen Zhang
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Hui Bai
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Qing Yang
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Junqing Ma
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Erik A C Wiemer
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands (E.A.C.W.)
| | - Jingjing Ben
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Qi Chen
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| |
Collapse
|
161
|
Cheng C, Liu M, Gao X, Wu D, Pu M, Ma J, Quinn RJ, Xiao Z, Liu Z. Identifying New Ligands for JNK3 by Fluorescence Thermal Shift Assays and Native Mass Spectrometry. ACS OMEGA 2022; 7:13925-13931. [PMID: 35559183 PMCID: PMC9088906 DOI: 10.1021/acsomega.2c00340] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/05/2022] [Indexed: 06/15/2023]
Abstract
The c-Jun N-terminal kinases (JNKs) are evolutionary highly conserved serine/threonine kinases. Numerous findings suggest that JNK3 is involved in the pathogenesis of neurodegenerative diseases, so the inhibition of JNK3 may be a potential therapeutic intervention. The identification of novel compounds with promising pharmacological properties still represents a challenge. Fluorescence thermal shift screening of a chemically diversified lead-like scaffold library of 2024 pure compounds led to the initial identification of seven JNK3 binding hits, which were classified into four scaffold groups according to their chemical structures. Native mass spectrometry validated the interaction of 4 out of the 7 hits with JNK3. Binding geometries and interactions of the top 2 hits were evaluated by docking into a JNK3 crystal structure. Hit 5 had a K d of 21 μM with JNK3 suggested scaffold 5-(phenylamino)-1H-1,2,3-triazole-4-carboxamide as a novel and selective JNK3 binder.
Collapse
Affiliation(s)
- Chongyun Cheng
- National
Laboratory of Biomacromolecules, Institute
of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- Griffith
Institute for Drug Discovery, Griffith University, Brisbane, Queensland 4111, Australia
- Monash
Biomedicine Discovery Institute, Monash
University, Melbourne, Victoria 3800, Australia
| | - Miaomiao Liu
- Griffith
Institute for Drug Discovery, Griffith University, Brisbane, Queensland 4111, Australia
| | - Xiaoqin Gao
- State
Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical
Sciences, Peking University, Beijing 100191, China
| | - Dong Wu
- iHuman
Institute, ShanghaiTech University, Shanghai 201210, China
| | - Mengchen Pu
- National
Laboratory of Biomacromolecules, Institute
of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jun Ma
- Griffith
Institute for Drug Discovery, Griffith University, Brisbane, Queensland 4111, Australia
| | - Ronald J. Quinn
- Griffith
Institute for Drug Discovery, Griffith University, Brisbane, Queensland 4111, Australia
| | - Zhicheng Xiao
- Monash
Biomedicine Discovery Institute, Monash
University, Melbourne, Victoria 3800, Australia
- Kunming
Medical College, Kunming, Yunnan 650031, China
| | - Zhijie Liu
- National
Laboratory of Biomacromolecules, Institute
of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- iHuman
Institute, ShanghaiTech University, Shanghai 201210, China
- Kunming
Medical College, Kunming, Yunnan 650031, China
| |
Collapse
|
162
|
DOI T, HIOKI T, TACHI J, UEDA K, MATSUSHIMA-NISHIWAKI R, IIDA H, OGURA S, KOZAWA O, TOKUDA H. Oncostatin M reduces the synthesis of macrophage-colony stimulating factor stimulated by TGF-β via suppression of p44/p42 MAP kinase and JNK in osteoblasts. Biomed Res 2022; 43:41-51. [DOI: 10.2220/biomedres.43.41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Tomoaki DOI
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine
| | - Tomoyuki HIOKI
- Department of Pharmacology, Gifu University Graduate School of Medicine
| | - Junko TACHI
- Department of Pharmacology, Gifu University Graduate School of Medicine
| | - Kyohei UEDA
- Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine
| | | | - Hiroki IIDA
- Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine
| | - Shinji OGURA
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine
| | - Osamu KOZAWA
- Department of Pharmacology, Gifu University Graduate School of Medicine
| | - Haruhiko TOKUDA
- Department of Clinical Laboratory/Medical Genome Center, National Center for Geriatrics and Gerontology
| |
Collapse
|
163
|
Li Y, Wei L, Meinsohn MC, Suliman R, Chauvin M, Berstler J, Hartland K, Jensen MM, Sicher NA, Nagykery N, Donahoe PK, Pepin D. A screen of repurposed drugs identifies AMHR2/MISR2 agonists as potential contraceptives. Proc Natl Acad Sci U S A 2022; 119:e2122512119. [PMID: 35380904 PMCID: PMC9169708 DOI: 10.1073/pnas.2122512119] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/22/2022] [Indexed: 11/18/2022] Open
Abstract
We identified the anti-Mullerian hormone (also known as Müllerian inhibiting substance or MIS) as an inhibitory hormone that induces long-term contraception in mammals. The type II receptor to this hormone, AMHR2 (also known as MISR2), represents a promising druggable target for the modulation of female reproduction with a mechanism of action distinct from steroidal contraceptives. We designed an in vitro platform to screen and validate small molecules that can activate MISR2 signaling and suppress ovarian folliculogenesis. Using a bone morphogenesis protein (BMP)–response element luciferase reporter cell–based assay, we screened 5,440 compounds from a repurposed drug library. Positive hits in this screen were tested for specificity and potency in luciferase dose–response assays, and biological activity was tested in ex vivo Mullerian duct regression bioassays. Selected candidates were further evaluated in ex vivo follicle/ovary culture assays and in vivo in mice and rats. Here, we report that SP600125, CYC-116, gandotinib, and ruxolitinib can specifically inhibit primordial follicle activation and repress folliculogenesis by stimulating the MISR2 pathway.
Collapse
Affiliation(s)
- Yi Li
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - Lina Wei
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - Marie-Charlotte Meinsohn
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - Rana Suliman
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - Maeva Chauvin
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - Jim Berstler
- Center for the Development of Therapeutics (CDoT), Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142
| | - Kate Hartland
- Center for the Development of Therapeutics (CDoT), Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142
| | - Mark M Jensen
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - Natalie A Sicher
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - Nicholas Nagykery
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - Patricia K Donahoe
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - David Pepin
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
164
|
Niu J, Holland SM, Ketschek A, Collura KM, Hesketh NL, Hayashi T, Gallo G, Thomas GM. Palmitoylation couples the kinases DLK and JNK3 to facilitate prodegenerative axon-to-soma signaling. Sci Signal 2022; 15:eabh2674. [PMID: 35349303 PMCID: PMC12049159 DOI: 10.1126/scisignal.abh2674] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Dual leucine-zipper kinase (DLK; a MAP3K) mediates neuronal responses to diverse injuries and insults through the c-Jun N-terminal kinase (JNK) family of mitogen-activated protein kinases (MAPKs). Here, we identified two ways through which DLK is coupled to the neural-specific isoform JNK3 to control prodegenerative signaling. JNK3 catalyzed positive feedback phosphorylation of DLK that further activated DLK and locked the DLK-JNK3 module in a highly active state. Neither homologous MAP3Ks nor a homologous MAPK could support this positive feedback loop. Unlike the related JNK1 isoform JNK2 and JNK3 promote prodegenerative axon-to-soma signaling and were endogenously palmitoylated. Moreover, palmitoylation targeted both DLK and JNK3 to the same axonal vesicles, and JNK3 palmitoylation was essential for axonal retrograde signaling in response to optic nerve crush injury in vivo. These findings provide previously unappreciated insights into DLK-JNK signaling relevant to neuropathological conditions and answer long-standing questions regarding the selective prodegenerative roles of JNK2 and JNK3.
Collapse
Affiliation(s)
- Jingwen Niu
- Shriners Hospitals Pediatric Research Center (Center for Neurorehabilitation and Neural Repair), Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Sabrina M. Holland
- Shriners Hospitals Pediatric Research Center (Center for Neurorehabilitation and Neural Repair), Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Andrea Ketschek
- Shriners Hospitals Pediatric Research Center (Center for Neurorehabilitation and Neural Repair), Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Kaitlin M. Collura
- Shriners Hospitals Pediatric Research Center (Center for Neurorehabilitation and Neural Repair), Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Natasha L. Hesketh
- Shriners Hospitals Pediatric Research Center (Center for Neurorehabilitation and Neural Repair), Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Takashi Hayashi
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central6 (6-10), 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Gianluca Gallo
- Shriners Hospitals Pediatric Research Center (Center for Neurorehabilitation and Neural Repair), Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA
- Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, 3500 N. Broad St., Philadelphia, PA 19140, USA
| | - Gareth M. Thomas
- Shriners Hospitals Pediatric Research Center (Center for Neurorehabilitation and Neural Repair), Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA
- Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, 3500 N. Broad St., Philadelphia, PA 19140, USA
| |
Collapse
|
165
|
Monosodium urate crystals regulate a unique JNK-dependent macrophage metabolic and inflammatory response. Cell Rep 2022; 38:110489. [PMID: 35263587 DOI: 10.1016/j.celrep.2022.110489] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 12/20/2021] [Accepted: 02/14/2022] [Indexed: 12/21/2022] Open
Abstract
Monosodium urate crystals (MSUc) induce inflammation in vivo without prior priming, raising the possibility of an initial cell-autonomous phase. Here, using genome-wide transcriptomic analysis and biochemical assays, we demonstrate that MSUc alone induce a metabolic-inflammatory transcriptional program in non-primed human and murine macrophages that is markedly distinct to that induced by LPS. Genes uniquely upregulated in response to MSUc belong to lipid and amino acid metabolism, glycolysis, and SLC transporters. This upregulation leads to a metabolic rewiring in sera from individuals and mice with acute gouty arthritis. Mechanistically, the initiating inflammatory-metabolic changes in acute gout flares are regulated through a persistent expression and increased binding of JUN to the promoter of target genes through JNK signaling-but not P38-in a process that is different than after LPS stimulation and independent of inflammasome activation. Finally, pharmacological JNK inhibition limits MSUc-induced inflammation in animal models of acute gouty inflammation.
Collapse
|
166
|
Zhu Y, Shuai W, Zhao M, Pan X, Pei J, Wu Y, Bu F, Wang A, Ouyang L, Wang G. Unraveling the Design and Discovery of c-Jun N-Terminal Kinase Inhibitors and Their Therapeutic Potential in Human Diseases. J Med Chem 2022; 65:3758-3775. [PMID: 35200035 DOI: 10.1021/acs.jmedchem.1c01947] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
c-Jun N-terminal kinases (JNKs), members of the mitogen-activated protein kinase (MAPK) family, are encoded by three genes: jnk1, jnk2, and jnk3. JNKs are involved in the pathogenesis and development of many diseases, such as neurodegenerative diseases, inflammation, and cancers. Therefore, JNKs have become important therapeutic targets. Many JNK inhibitors have been discovered, and some have been introduced into clinical trials. However, the study of isoform-selective JNK inhibitors is still a challenging task. To further develop novel JNK inhibitors with clinical value, a comprehensive understanding of JNKs and their corresponding inhibitors is required. In this Perspective, we introduced the JNK signaling pathways and reviewed different chemical types of JNK inhibitors, focusing on their structure-activity relationships and biological activities. The challenges and strategies for the development of JNK inhibitors are also discussed. It is hoped that this Perspective will provide valuable references for the development of novel selective JNK inhibitors.
Collapse
Affiliation(s)
- Yumeng Zhu
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Wen Shuai
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Min Zhao
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Xiaoli Pan
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Junping Pei
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Yongya Wu
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Faqian Bu
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Aoxue Wang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Liang Ouyang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Guan Wang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
167
|
Suzuki T, Hoshina M, Nishijima S, Hoshina N, Kikuguchi C, Tomohiro T, Fukao A, Fujiwara T, Yamamoto T. Regulation of CCR4-NOT complex deadenylase activity and cellular responses by MK2-dependent phosphorylation of CNOT2. RNA Biol 2022; 19:234-246. [PMID: 35129087 PMCID: PMC8820811 DOI: 10.1080/15476286.2021.2021676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
CCR4-NOT complex-mediated mRNA deadenylation serves critical functions in multiple biological processes, yet how this activity is regulated is not fully understood. Here, we show that osmotic stress induces MAPKAPK-2 (MK2)-mediated phosphorylation of CNOT2. Programmed cell death is greatly enhanced by osmotic stress in CNOT2-depleted cells, indicating that CNOT2 is responsible for stress resistance of cells. Although wild-type (WT) and non-phosphorylatable CNOT2 mutants reverse this sensitivity, a phosphomimetic form of CNOT2, in which serine at the phosphorylation site is replaced with glutamate, does not have this function. We also show that mRNAs have elongated poly(A) tails in CNOT2-depleted cells and that introduction of CNOT2 WT or a non-phosphorylatable mutant, but not phosphomimetic CNOT2, renders their poly(A) tail lengths comparable to those in control HeLa cells. Consistent with this, the CCR4-NOT complex containing phosphomimetic CNOT2 exhibits less deadenylase activity than that containing CNOT2 WT. These data suggest that CCR4-NOT complex deadenylase activity is regulated by post-translational modification, yielding dynamic control of mRNA deadenylation.
Collapse
Affiliation(s)
- Toru Suzuki
- Laboratory for Immunogenetics, Center for Integrative Medical Sciences, Riken, Yokohama, Japan
| | - Miyuki Hoshina
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Japan
| | - Saori Nishijima
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Japan
| | - Naosuke Hoshina
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Japan
| | - Chisato Kikuguchi
- Laboratory for Immunogenetics, Center for Integrative Medical Sciences, Riken, Yokohama, Japan
| | - Takumi Tomohiro
- Laboratory of Biochemistry, Kindai University, Higashi-Osaka, Japan
| | - Akira Fukao
- Laboratory of Biochemistry, Kindai University, Higashi-Osaka, Japan
| | | | - Tadashi Yamamoto
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Japan
| |
Collapse
|
168
|
Yu G, Yu H, Yang Q, Wang J, Fan H, Liu G, Wang L, Bello BK, Zhao P, Zhang H, Dong J. Vibrio harveyi infections induce production of proinflammatory cytokines in murine peritoneal macrophages via activation of p38 MAPK and NF-κB pathways, but reversed by PI3K/AKT pathways. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 127:104292. [PMID: 34656643 DOI: 10.1016/j.dci.2021.104292] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 10/09/2021] [Accepted: 10/09/2021] [Indexed: 06/13/2023]
Abstract
Vibrio harveyi is a zoonotic pathogen that can infect humans through wounds and cause severe inflammatory responses. Previous studies have reported that the Toll like receptors (TLR) mediated MAPK, AKT and NF-κB signaling pathways are involved in innate immune system resistance to pathogen invasion. However, the molecular mechanism of these pathways, as well as their involvement in V. harveyi infection remains elusive. This study established a V. harveyi infection model using murine peritoneal macrophages (PMs). Various techniques, including western blotting, ELISA, RT-qPCR, immunofluorescence, inhibition assays, were used to explore the roles of TLRs, MAPK, AKT and NF-κB signaling pathways in V. harveyi-induced inflammatory responses. ELISA assays showed that V. harveyi infection triggered proinflammatory cytokines secretion in PMs. RT-qPCR and inhibition assays showed that TLR2 participated in V. harveyi infection and up-regulated the proinflammatory cytokines secretion in murine PMs. Western blotting data showed that the phosphorylation of p38, JNK, AKT, and NF-κB p65 were significantly increased partly mediated by TLR2. In addition, immunofluorescence assays revealed that the NF-κB p65 translocated into nucleus in response to V. harveyi infection. The secretion of IL-1β, IL-6, IL-12, and TNF-α were considerably reduced when the p38 MAPK and NF-κB signaling pathways were blocked, whereas blocking of AKT significantly increased the expression of IL-1β, IL-6, IL-12, and TNF-α. These findings indicate that V. harveyi infection induces inflammatory responses in murine PMs via activation of p38 MAPK and NF-κB pathways, which are partly mediated by TLR2, but are inhibited by PI3K/AKT pathways.
Collapse
Affiliation(s)
- Guili Yu
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Hong Yu
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Qiankun Yang
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Jinxin Wang
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Hui Fan
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Gang Liu
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Lei Wang
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, 222005, China; Department of Vascular Surgery, The First People's Hospital of Lianyungang, Lianyungang, 222002, China
| | | | - Panpan Zhao
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, 222005, China; Key Laboratory of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, PR China.
| | - Honggang Zhang
- Department of Vascular Surgery, The First People's Hospital of Lianyungang, Lianyungang, 222002, China.
| | - Jingquan Dong
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, 222005, China.
| |
Collapse
|
169
|
Aldosari Z, Abbasian N, Robinson K, Bevington A, Watson E. Low pH up-regulates interleukin-6 mRNA in L6-G8C5 rat skeletal muscle cells independent of pH sensing by SNAT2(SLC38A2) transporters. FASEB Bioadv 2022; 4:138-152. [PMID: 35141477 PMCID: PMC8814557 DOI: 10.1096/fba.2021-00088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 10/14/2021] [Accepted: 10/22/2021] [Indexed: 11/14/2022] Open
Abstract
Exercise is known to create a transient, but potent increase in skeletal muscle expression of potentially anti-inflammatory myokine interleukin-6 (IL-6). This effect may be clinically important in managing chronic inflammatory states. It has previously been proposed that lactic acidosis following exercise promotes this IL-6 up-regulation, but the mechanism of this acidosis effect is unknown. Rat skeletal muscle cell line L6-G8C5 has been used previously to model metabolic effects of acidosis, sensing low pH through the resulting inhibition of amino acid transporter SNAT2(SLC38A2). Use of ionophore ionomycin to model the rise in intracellular Ca2+ concentration occurring in contracting muscle strongly up-regulates IL-6 mRNA in L6-G8C5 myotubes. This study used this model to test the hypothesis that low extracellular pH (7.1) enhances ionomycin-induced IL-6 mRNA up-regulation by inhibiting SNAT2. Incubation of L6-G8C5 myotubes for 6 h with 0.5 µM ionomycin at control pH (7.4) resulted in a 15-fold increase in IL-6 mRNA which was further enhanced (1.74-fold) at pH 7.1. In contrast low pH had no significant effect on IL-6 mRNA without ionomycin, nor on the IL-6 mRNA increase that was induced by cyclic stretch. Even though pH 7.1 halved the transport activity of SNAT2, alternative methods of SNAT2 inhibition (JNK inhibitor SP600125; SNAT2 antagonist MeAIB; or SNAT2 silencing with siRNA) did not mimic the enhancing effect of low pH on IL-6 mRNA. On the contrary, JNK inhibition blunted the effect of pH 7.1 with ionomycin, but had no effect at pH 7.4. It is concluded that low pH promotes Ca2+/ionomycin-induced up-regulation of IL-6 mRNA through a novel SNAT2-independent JNK-dependent pH-sensing pathway not previously described in this skeletal muscle model.
Collapse
Affiliation(s)
- Ziyad Aldosari
- Department of Respiratory SciencesUniversity of LeicesterLeicesterUK
- Department of Medical Laboratories SciencesCollege of Applied Medical Sciences in AlquwayiyahShaqra UniversityRiyadhSaudi Arabia
| | - Nima Abbasian
- School of Life and Medical SciencesUniversity of HertfordshireHatfieldUK
| | | | - Alan Bevington
- Department of Respiratory SciencesUniversity of LeicesterLeicesterUK
| | - Emma Watson
- Department of Cardiovascular SciencesUniversity of LeicesterLeicesterUK
| |
Collapse
|
170
|
Guo M, Zhang M, Cao X, Fang X, Li K, Qin L, He Y, Zhao J, Xu Y, Liu X, Li X. Notch4 mediates vascular remodeling via ERK/JNK/P38 MAPK signaling pathways in hypoxic pulmonary hypertension. Respir Res 2022; 23:6. [PMID: 35016680 PMCID: PMC8753901 DOI: 10.1186/s12931-022-01927-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/03/2022] [Indexed: 12/17/2022] Open
Abstract
Background Hypoxic pulmonary hypertension (HPH) is a chronic progressive advanced disorder pathologically characterized by pulmonary vascular remodeling. Notch4 as a cell surface receptor is critical for vascular development. However, little is known about the role and mechanism of Notch4 in the development of hypoxic vascular remodeling. Methods Lung tissue samples were collected to detect the expression of Notch4 from patients with HPH and matched controls. Human pulmonary artery smooth muscle cells (HPASMCs) were cultured in hypoxic and normoxic conditions. Real-time quantitative PCR and western blotting were used to examine the mRNA and protein levels of Notch4. HPASMCs were transfected with small interference RNA (siRNA) against Notch4 or Notch4 overexpression plasmid, respectively. Cell viability, cell proliferation, apoptosis, and migration were assessed using Cell Counting Kit-8, Edu, Annexin-V/PI, and Transwell assay. The interaction between Notch4 and ERK, JNK, P38 MAPK were analyzed by co-immunoprecipitation. Adeno-associated virus 1-mediated siRNA against Notch4 (AAV1-si-Notch4) was injected into the airways of hypoxic rats. Right ventricular systolic pressure (RVSP), right ventricular hypertrophy and pulmonary vascular remodeling were evaluated. Results In this study, we demonstrate that Notch4 is highly expressed in the media of pulmonary vascular and is upregulated in lung tissues from patients with HPH and HPH rats compared with control groups. In vitro, hypoxia induces the high expression of Delta-4 and Notch4 in HPASMCs. The increased expression of Notch4 promotes HPASMCs proliferation and migration and inhibits cells apoptosis via ERK, JNK, P38 signaling pathways. Furthermore, co-immunoprecipitation result elucidates the interaction between Notch4 and ERK/JNK/P38. In vivo, silencing Notch4 partly abolished the increase in RVSP and pulmonary vascular remodeling caused by hypoxia in HPH rats. Conclusions These findings reveal an important role of the Notch4-ERK/JNK/P38 MAPK axis in hypoxic pulmonary remodeling and provide a potential therapeutic target for patients with HPH. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-01927-9.
Collapse
Affiliation(s)
- Mingzhou Guo
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China.,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China
| | - Mengzhe Zhang
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China.,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China
| | - Xiaopei Cao
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyu Fang
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China.,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China
| | - Ke Li
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China.,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China
| | - Lu Qin
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China.,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China
| | - Yuanzhou He
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China.,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China
| | - Jianping Zhao
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China.,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China
| | - Yongjian Xu
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China.,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China
| | - Xiansheng Liu
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China.,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China
| | - Xiaochen Li
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China. .,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China.
| |
Collapse
|
171
|
Liu Y, Li Z, Wang Y, Cai Q, Liu H, Xu C, Zhang F. IL-15 Participates in the Pathogenesis of Polycystic Ovary Syndrome by Affecting the Activity of Granulosa Cells. Front Endocrinol (Lausanne) 2022; 13:787876. [PMID: 35250857 PMCID: PMC8894602 DOI: 10.3389/fendo.2022.787876] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 01/18/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Low-grade chronic inflammation may contribute to the pathogenesis of polycystic ovary syndrome (PCOS). Interleukin-15 (IL-15) is a proinflammatory cytokine involved in the development of chronic inflammation leading to obesity-associated metabolic syndrome. However, the concentration of IL-15 in follicular fluid of patients with PCOS has yet been evaluated. OBJECTIVES The aim of this study is to evaluate the expression level of IL-15 in both patients with PCOS and PCOS mice model and investigate the functional effect of IL-15 on ovarian granulosa cells. METHODS The level of IL-15 in follicular fluid (FF) was measured using cytokine array and enzyme linked immunosorbent assay (ELISA) in two cohorts from 23 PCOS patients and 18 normo-ovulatory controls. PCOS mice model was induced by subcutaneously implanted with letrozole pellet for 21 days. The expression level of IL-15 in serum, ovarian, and subcutaneous adipose tissue in PCOS mice model was measured by ELISA, real-time polymerase chain reaction (RT-PCR), immunohistochemistry (IHC), and immunofluorescence. The effect of IL-15 on the proliferation and apoptosis of the KGN cells and mouse ovarian granulosa cells (GCs) were detected by CCK-8 assay and flow cytometry, respectively. Transcript expression of 17α-hydroxylase17,20-lyase (CYP17A1), cytochrome P450 family 19 subfamily A member 1(CYP19A1), FSH receptor (FSHR), steroidogenic acute regulatory protein (StAR), and proinflammatory cytokine were quantified using RT-PCR. The protein level and phosphorylation level of p38 MAPK and JNK are detected by Western blot. Concentration of dehydroepiandrosterone sulfate (DHEAS) and progesterone (P)were measured by ELISA. RESULTS IL-15 expression in follicular fluid of patients with PCOS was significantly elevated compared with the control group, and similar results were observed in the ovarian and subcutaneous adipose tissue of PCOS mice models. Furthermore, the elevated FF IL-15 levels have a positive correlation with the serum testosterone levels. FSHR co-localized with IL-15 indicating that IL-15 production originate from ovarian granulose cells. IL-15 treatment inhibited proliferation and promoted apoptosis of KGN cells and mouse GCs. Moreover, IL-15 upregulated the transcription levels of CYP17A1, IL-1b and Ifng KGN cells. Similar results were observed in mouse GCs except concentration of DHEAS was higher in IL-15 treatment. IL-15 promoted p38 MAPK and JNK phosphorylation in KGN cells, treating KGN cells with p38 MAPK inhibitor SP600125 and JNK inhibitor SB203580 could reverse the effect of IL-15 on the proliferation and function of KGN cells. CONCLUSION The results indicate that IL-15 is involved in the pathogenesis of PCOS potentially by affecting survival, the inflammation state and steroidogenesis of granulosa cells. The practical significance of this association between IL-15 and the pathogenesis of PCOS needs further investigation.
Collapse
Affiliation(s)
- Yan Liu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Zhi Li
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Yang Wang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Qingqing Cai
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Haiou Liu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Congjian Xu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
- Department of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, China
- *Correspondence: Feifei Zhang, ; Congjian Xu,
| | - Feifei Zhang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Department of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, China
- *Correspondence: Feifei Zhang, ; Congjian Xu,
| |
Collapse
|
172
|
de Klerk DJ, de Keijzer MJ, Dias LM, Heemskerk J, de Haan LR, Kleijn TG, Franchi LP, Heger M. Strategies for Improving Photodynamic Therapy Through Pharmacological Modulation of the Immediate Early Stress Response. Methods Mol Biol 2022; 2451:405-480. [PMID: 35505025 DOI: 10.1007/978-1-0716-2099-1_20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Photodynamic therapy (PDT) is a minimally to noninvasive treatment modality that has emerged as a promising alternative to conventional cancer treatments. PDT induces hyperoxidative stress and disrupts cellular homeostasis in photosensitized cancer cells, resulting in cell death and ultimately removal of the tumor. However, various survival pathways can be activated in sublethally afflicted cancer cells following PDT. The acute stress response is one of the known survival pathways in PDT, which is activated by reactive oxygen species and signals via ASK-1 (directly) or via TNFR (indirectly). The acute stress response can activate various other survival pathways that may entail antioxidant, pro-inflammatory, angiogenic, and proteotoxic stress responses that culminate in the cancer cell's ability to cope with redox stress and oxidative damage. This review provides an overview of the immediate early stress response in the context of PDT, mechanisms of activation by PDT, and molecular intervention strategies aimed at inhibiting survival signaling and improving PDT outcome.
Collapse
Affiliation(s)
- Daniel J de Klerk
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Mark J de Keijzer
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Lionel M Dias
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Faculdade de Ciências da Saúde (FCS-UBI), Universidade da Beira Interior, Covilhã, Portugal
| | - Jordi Heemskerk
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
| | - Lianne R de Haan
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Tony G Kleijn
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Leonardo P Franchi
- Departamento de Bioquímica e Biologia Molecular, Instituto de Ciências Biológicas (ICB) 2, Universidade Federal de Goiás (UFG), Goiânia, GO, Brazil
- Faculty of Philosophy, Department of Chemistry, Center of Nanotechnology and Tissue Engineering-Photobiology and Photomedicine Research Group, Sciences, and Letters of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Michal Heger
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China.
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands.
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
173
|
Fan Y, Zhang G, Zhao K, Fu W, Chen S, Liu J, Liu W, Peng L, Ren L, Liu S, Xiao Y. Characteristics of SP600125 Induced Tetraploid Cells in Comparison With Diploid and Tetraploid Cells of Fish. Front Genet 2021; 12:781007. [PMID: 34938322 PMCID: PMC8685524 DOI: 10.3389/fgene.2021.781007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/08/2021] [Indexed: 11/13/2022] Open
Abstract
In our previous research, SP600125 (Anthrapyrazolone) was used to induce autotetraploid of crucian carp cells (SP4N cells), and tetraploid fry was generated from the SP4N cells by somatic cell nuclear transfer technique. However, it is still unclear about biological characteristics of the SP4N cells. In this article, the cytological characteristic and gene expression profiles of the SP4N cells are investigated in comparison with the crucian carp cells (2N cells) and the tetraploid crucian carp cells (CC4N cells). The SP4N cells have tetraploid characteristics in terms of morphology and DNA ploidy levels, and their chromosome behavior is stable during the cell proliferation. The migration ability and the mtDNA copy number of SP4N cells are both lower than those in the CC4N cells and the 2N cells, but there exist giant mitochondria in the SP4N cells. The similar expression trends in the cell cycle regulation genes of the SP4N cells and 2N cells, while the corresponding expression profiles are clearly different between the SP4N cells and the CC4N cells. Moreover, the significant difference genes are associated with energy metabolism pathways among the SP4N cells, 2N cells and CC4N cells. These results can provide deeper understanding of SP600125 induction, as well as finding applications in polyploidization breeding of fish species.
Collapse
Affiliation(s)
- Yunpeng Fan
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, China.,College of Life Sciences, Hunan Normal University, Changsha, China
| | - Guangjing Zhang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, China.,College of Life Sciences, Hunan Normal University, Changsha, China
| | - Kaiyue Zhao
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, China.,College of Life Sciences, Hunan Normal University, Changsha, China
| | - Wen Fu
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, China.,College of Life Sciences, Hunan Normal University, Changsha, China
| | - Shujuan Chen
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, China.,College of Life Sciences, Hunan Normal University, Changsha, China
| | - Jinhui Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, China.,College of Life Sciences, Hunan Normal University, Changsha, China
| | - Wenbin Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, China.,College of Life Sciences, Hunan Normal University, Changsha, China
| | - Liangyue Peng
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, China.,College of Life Sciences, Hunan Normal University, Changsha, China
| | - Li Ren
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, China.,College of Life Sciences, Hunan Normal University, Changsha, China
| | - Shaojun Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, China.,College of Life Sciences, Hunan Normal University, Changsha, China
| | - Yamei Xiao
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, China.,College of Life Sciences, Hunan Normal University, Changsha, China
| |
Collapse
|
174
|
Lactobacillus casei and Epidermal Growth Factor Prevent Osmotic Stress-Induced Tight Junction Disruption in Caco-2 Cell Monolayers. Cells 2021; 10:cells10123578. [PMID: 34944085 PMCID: PMC8700399 DOI: 10.3390/cells10123578] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 01/08/2023] Open
Abstract
Osmotic stress plays a crucial role in the pathogenesis of many gastrointestinal diseases. Lactobacillus casei and epidermal growth factor (EGF) effects on the osmotic stress-induced epithelial junctional disruption and barrier dysfunction were investigated. Caco-2 cell monolayers were exposed to osmotic stress in the presence or absence of L. casei or EGF, and the barrier function was evaluated by measuring inulin permeability. Tight junction (TJ) and adherens junction integrity were assessed by immunofluorescence confocal microscopy. The role of signaling molecules in the L. casei and EGF effects was determined by using selective inhibitors. Data show that pretreatment of cell monolayers with L. casei or EGF attenuates osmotic stress-induced TJ and adherens junction disruption and barrier dysfunction. EGF also blocked osmotic stress-induced actin cytoskeleton remodeling. U0126 (MEK1/2 inhibitor), the MAP kinase inhibitor, blocked EGF-mediated epithelial protection from osmotic stress. In contrast, the L. casei-mediated epithelial protection from osmotic stress was unaffected by U0126, AG1478 (EGFR tyrosine kinase inhibitor), SP600125 (JNK1/2 inhibitor), or SB202190 (P38 MAP kinase inhibitor). On the other hand, Ro-32-0432 (PKC inhibitor) blocked the L. casei-mediated prevention of osmotic stress-induced TJ disruption and barrier dysfunction. The combination of EGF and L. casei is more potent in protecting the barrier function from osmotic stress. These findings suggest that L. casei and EGF ameliorate osmotic stress-induced disruption of apical junctional complexes and barrier dysfunction in the intestinal epithelium by distinct signaling mechanisms.
Collapse
|
175
|
Park SH, Hwang I, McNaughton DA, Kinross AJ, Howe EN, He Q, Xiong S, Kilde MD, Lynch VM, Gale PA, Sessler JL, Shin I. Synthetic Na +/K + exchangers promote apoptosis by disturbing cellular cation homeostasis. Chem 2021; 7:3325-3339. [PMID: 38239771 PMCID: PMC10795848 DOI: 10.1016/j.chempr.2021.08.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A number of artificial cation ionophores (or transporters) have been developed for basic research and biomedical applications. However, their mechanisms of action and the putative correlations between changes in intracellular cation concentrations and induced cell death remain poorly understood. Here, we show that three hemispherand-strapped calix[4]pyrrole-based ion-pair receptors act as efficient Na+/K+ exchangers in the presence of Cl- in liposomal models and promote Na+ influx and K+ efflux (Na+/K+ exchange) in cancer cells to induce apoptosis. Mechanistic studies reveal that these cation exchangers induce endoplasmic reticulum (ER) stress in cancer cells by perturbing intracellular cation homeostasis, promote generation of reactive oxygen species, and eventually enhance mitochondria-mediated apoptosis. However, they neither induce osmotic stress nor affect autophagy. This study provides support for the notion that synthetic receptors, which perturb cellular cation homeostasis, may provide new small molecules with potentially useful apoptotic activity.
Collapse
Affiliation(s)
- Sang-Hyun Park
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
- These authors contributed equally
| | - Inhong Hwang
- Department of Chemistry, The University of Texas at Austin, 105 East 24th Street, Stop A5300, Austin, TX 78712, USA
- These authors contributed equally
| | - Daniel A. McNaughton
- School of Chemistry (F11), The University of Sydney, Sydney, NSW 2006, Australia
- These authors contributed equally
| | - Airlie J. Kinross
- School of Chemistry (F11), The University of Sydney, Sydney, NSW 2006, Australia
| | - Ethan N.W. Howe
- School of Chemistry (F11), The University of Sydney, Sydney, NSW 2006, Australia
- Present address: GlaxoSmithKline, GSK Jurong, 1 Pioneer Sector 1, Singapore 628413
| | - Qing He
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P.R. China
| | - Shenglun Xiong
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P.R. China
| | - Martin Drøhse Kilde
- Department of Chemistry, The University of Texas at Austin, 105 East 24th Street, Stop A5300, Austin, TX 78712, USA
- Present address: Department of Chemistry, University of Copenhagen, Universitetsparken 5, DK-2100 Copenhagen Ø, Denmark
| | - Vincent M. Lynch
- Department of Chemistry, The University of Texas at Austin, 105 East 24th Street, Stop A5300, Austin, TX 78712, USA
| | - Philip A. Gale
- School of Chemistry (F11), The University of Sydney, Sydney, NSW 2006, Australia
- The University of Sydney Nano Institute (SydneyNano), The University of Sydney, Sydney, NSW 2006, Australia
| | - Jonathan L. Sessler
- Department of Chemistry, The University of Texas at Austin, 105 East 24th Street, Stop A5300, Austin, TX 78712, USA
- Lead contact
| | - Injae Shin
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
176
|
Stoll K, Bergmann M, Spiliotis M, Brehm K. A MEKK1 - JNK mitogen activated kinase (MAPK) cascade module is active in Echinococcus multilocularis stem cells. PLoS Negl Trop Dis 2021; 15:e0010027. [PMID: 34879059 PMCID: PMC8687709 DOI: 10.1371/journal.pntd.0010027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 12/20/2021] [Accepted: 11/25/2021] [Indexed: 11/18/2022] Open
Abstract
Background The metacestode larval stage of the fox-tapeworm Echinococcus multilocularis causes alveolar echinococcosis by tumour-like growth within the liver of the intermediate host. Metacestode growth and development is stimulated by host-derived cytokines such as insulin, fibroblast growth factor, and epidermal growth factor via activation of cognate receptor tyrosine kinases expressed by the parasite. Little is known, however, concerning signal transmission to the parasite nucleus and cross-reaction with other parasite signalling systems. Methodology/Principal findings Using bioinformatic approaches, cloning, and yeast two-hybrid analyses we identified a novel mitogen-activated kinase (MAPK) cascade module that consists of E. multilocularis orthologs of the tyrosine kinase receptor interactor Growth factor receptor-bound 2, EmGrb2, the MAPK kinase kinase EmMEKK1, a novel MAPK kinase, EmMKK3, and a close homolog to c-Jun N-terminal kinase (JNK), EmMPK3. Whole mount in situ hybridization analyses indicated that EmMEKK1 and EmMPK3 are both expressed in E. multilocularis germinative (stem) cells but also in differentiated or differentiating cells. Treatment with the known JNK inhibitor SP600125 led to a significantly reduced formation of metacestode vesicles from stem cells and to a specific reduction of proliferating stem cells in mature metacestode vesicles. Conclusions/Significance We provide evidence for the expression of a MEKK1-JNK MAPK cascade module which, in mammals, is crucially involved in stress responses, cytoskeletal rearrangements, and apoptosis, in E. multilocularis stem cells. Inhibitor studies indicate an important role of JNK signalling in E. multilocularis stem cell survival and/or maintenance. Our data are relevant for molecular and cellular studies into crosstalk signalling mechanisms that govern Echinococcus stem cell function and introduce the JNK signalling cascade as a possible target of chemotherapeutics against echinococcosis. The metacestode larva of the tapeworm E. multilocularis grows infiltrative, like a malignant tumour, within the liver of the host thus causing the lethal disease alveolar echinococcosis. Previous work established that the metacestode senses signals of host hormones and cytokines by expressing surface receptors that share high homology with respective host receptors. However, little is known how these signals are transmitted from the parasite cell surface to the nucleus to alter gene expression. In this work, the authors present a module of several protein kinases that typically transmit cytokine signals from surface receptors to central regulators called mitogen-activated protein kinases (MAPK). The authors demonstrate that this module is active in parasite stem cells, which drive the development of metacestode larva. They also show that inhibitors directed against one component of the module, EmMPK3, affect maintenance and/or survival of stem cells in the metacestode and prevent the formation of metacestode larva from parasite cell cultures. This information facilitates molecular and cellular studies to unravel the complex signalling network that regulate Echinococcus stem cell proliferation in response to host signals. Furthermore, these data could open new ways of anti-parasitic chemotherapy by introducing EmMPK3 as a possible drug target.
Collapse
Affiliation(s)
- Kristin Stoll
- University of Würzburg, Institute of Hygiene and Microbiology, Würzburg, Germany
| | - Monika Bergmann
- University of Würzburg, Institute of Hygiene and Microbiology, Würzburg, Germany
| | - Markus Spiliotis
- University of Würzburg, Institute of Hygiene and Microbiology, Würzburg, Germany
| | - Klaus Brehm
- University of Würzburg, Institute of Hygiene and Microbiology, Würzburg, Germany
- * E-mail:
| |
Collapse
|
177
|
Tanimura K, Yamada T, Horinaka M, Katayama Y, Fukui S, Morimoto K, Nakano T, Tokuda S, Morimoto Y, Iwasaku M, Kaneko Y, Uchino J, Yoneda K, Yano S, Sakai T, Takayama K. Inhibition of c-Jun N-terminal kinase signaling increased apoptosis and prevented the emergence of ALK-TKI-tolerant cells in ALK-rearranged non-small cell lung cancer. Cancer Lett 2021; 522:119-128. [PMID: 34534615 DOI: 10.1016/j.canlet.2021.09.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 08/25/2021] [Accepted: 09/10/2021] [Indexed: 10/20/2022]
Abstract
Anaplastic lymphoma kinase-tyrosine kinase inhibitors (ALK-TKIs) have improved clinical outcomes in non-small cell lung cancer (NSCLC) harboring ALK- rearrangements. However, a small population of tumor cells survives due to adaptive resistance under drug pressure and ultimately acquires drug resistance. Thus, it is necessary to elucidate the mechanisms underlying the prevention of drug resistance to improve the prognosis of patients with ALK-rearranged NSCLC. We identified novel adaptive resistance, generated through c-Jun N-terminal kinase (JNK)/c-Jun signaling, to initial ALK-TKIs-alectinib and brigatinib-in ALK-rearranged NSCLC. Inhibition of JNK/c-Jun axis showed suppression of growth and promotion of apoptosis induced by ALK-TKIs in drug-tolerant cells. JNK inhibition, in combination with the use of ALK-TKIs, increased cell apoptosis through repression of the Bcl-xL proteins, compared with ALK-TKI monotherapy. Importantly, combination therapy targeting JNK and ALK significantly delayed the regrowth following cessation of these treatments. Together, our results demonstrated that JNK pathway activation plays a pivotal role in the intrinsic resistance to ALK-TKIs and the emergence of ALK-TKI-tolerant cells in ALK-rearranged NSCLC, thus indicating that optimal inhibition of tolerant signals combined with ALK-TKIs may potentially improve the outcome of ALK-rearranged NSCLC.
Collapse
Affiliation(s)
- Keiko Tanimura
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Tadaaki Yamada
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan.
| | - Mano Horinaka
- Department of Drug Discovery Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Yuki Katayama
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Sarina Fukui
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Kenji Morimoto
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Takayuki Nakano
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Shinsaku Tokuda
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Yoshie Morimoto
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Masahiro Iwasaku
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Yoshiko Kaneko
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Junji Uchino
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Kazue Yoneda
- Second Department of Surgery, University of Occupational and Environmental Health, Kitakyushu-shi, Fukuoka, 807-8555, Japan
| | - Seiji Yano
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, Kakuma, Kanazawa, 920-1192, Japan
| | - Toshiyuki Sakai
- Department of Drug Discovery Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Koichi Takayama
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| |
Collapse
|
178
|
Fetuin-A regulates adipose tissue macrophage content and activation in insulin resistant mice through MCP-1 and iNOS: involvement of IFNγ-JAK2-STAT1 pathway. Biochem J 2021; 478:4027-4043. [PMID: 34724561 DOI: 10.1042/bcj20210442] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/29/2021] [Accepted: 11/01/2021] [Indexed: 12/12/2022]
Abstract
In the context of obesity-induced adipose tissue (AT) inflammation, migration of macrophages and their polarization from predominantly anti-inflammatory to proinflammatory subtype is considered a pivotal event in the loss of adipose insulin sensitivity. Two major chemoattractants, monocyte chemoattractant protein-1 (MCP-1) and Fetuin-A (FetA), have been reported to stimulate macrophage migration into inflamed AT instigating inflammation. Moreover, FetA could notably modulate macrophage polarization, yet the mechanism(s) is unknown. The present study was undertaken to elucidate the mechanistic pathway involved in the actions of FetA and MCP-1 in obese AT. We found that FetA knockdown in high fat diet (HFD) fed mice could significantly subdue the augmented MCP-1 expression and reduce adipose tissue macrophage (ATM) content thereby indicating that MCP-1 is being regulated by FetA. Additionally, knockdown of FetA in HFD mice impeded the expression of inducible nitric oxide synthase (iNOS) reverting macrophage activation from mostly proinflammatory to anti-inflammatory state. It was observed that the stimulating effect of FetA on MCP-1 and iNOS was mediated through interferon γ (IFNγ) induced activation of JAK2-STAT1-NOX4 pathway. Furthermore, we detected that the enhanced IFNγ expression was accounted by the stimulatory effect of FetA upon the activities of both cJun and JNK. Taken together, our findings revealed that obesity-induced FetA acts as a master upstream regulator of AT inflammation by regulating MCP-1 and iNOS expression through JNK-cJun-IFNγ-JAK2-STAT1 signaling pathway. This study opened a new horizon in understanding the regulation of ATM content and activation in conditions of obesity-induced insulin resistance.
Collapse
|
179
|
11H-Indeno[1,2-b]quinoxalin-11-one 2-(4-ethylbenzylidene)hydrazone. MOLBANK 2021. [DOI: 10.3390/m1299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
11H-Indeno[1,2-b]quinoxaline derivatives present an important type of nitrogen-containing heterocyclic compound that are useful intermediate products in organic synthesis and have potential pharmaceutical applications. A new 11H-indeno[1,2-b]quinoxalin-11-one-2-(4-ethylbenzylidene)hydrazone (compound 3) was synthesized. Compound 3 is the first example of an azine derivative based on the 11H-indeno[1,2-b]quinoxaline system. The Z,E-isomerism of compound 3 was investigated by DFT calculations. Bioavailability was evaluated in silico using ADME predictions. According to the ADME results, compound 3 is potentially highly bioavailable and has potential to be used for the treatment of neuroinflammation and ischemia–reperfusion injury.
Collapse
|
180
|
Vesicular formation regulated by ERK/MAPK pathway mediates human erythroblast enucleation. Blood Adv 2021; 5:4648-4661. [PMID: 34551066 PMCID: PMC8759143 DOI: 10.1182/bloodadvances.2021004859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 07/17/2021] [Indexed: 11/20/2022] Open
Abstract
ERK pathway plays a key role in enucleation of human orthochromatic erythroblasts. ERK regulates human erythroblast enucleation by affecting vesicular formation.
Enucleation is a key event in mammalian erythropoiesis responsible for the generation of enucleated reticulocytes. Although progress is being made in developing mechanistic understanding of enucleation, our understanding of mechanisms for enucleation is still incomplete. The MAPK pathway plays diverse roles in biological processes, but its role in erythropoiesis has yet to be fully defined. Analysis of RNA-sequencing data revealed that the MAPK pathway is significantly upregulated during human terminal erythroid differentiation. The MAPK pathway consists of 3 major signaling cassettes: MEK/ERK, p38, and JNK. In the present study, we show that among these 3 cassettes, only ERK was significantly upregulated in late-stage human erythroblasts. The increased expression of ERK along with its increased phosphorylation suggests a potential role for ERK activation in enucleation. To explore this hypothesis, we treated sorted populations of human orthochromatic erythroblasts with the MEK/ERK inhibitor U0126 and found that U0126 inhibited enucleation. In contrast, inhibitors of either p38 or JNK had no effect on enucleation. Mechanistically, U0126 selectively inhibited formation/accumulation of cytoplasmic vesicles and endocytosis of the transferrin receptor without affecting chromatin condensation, nuclear polarization, or enucleosome formation. Treatment with vacuolin-1 that induces vacuole formation partially rescued the blockage of enucleation by U0126. Moreover, phosphoproteomic analysis revealed that inactivation of the ERK pathway led to downregulation of the endocytic recycling pathway. Collectively, our findings uncovered a novel role of ERK activation in human erythroblast enucleation by modulating vesicle formation and have implications for understanding anemia associated with defective enucleation.
Collapse
|
181
|
Somorjai IML, Ehebauer MT, Escrivà H, Garcia-Fernàndez J. JNK Mediates Differentiation, Cell Polarity and Apoptosis During Amphioxus Development by Regulating Actin Cytoskeleton Dynamics and ERK Signalling. Front Cell Dev Biol 2021; 9:749806. [PMID: 34778260 PMCID: PMC8586503 DOI: 10.3389/fcell.2021.749806] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/17/2021] [Indexed: 11/13/2022] Open
Abstract
c-Jun N-terminal kinase (JNK) is a multi-functional protein involved in a diverse array of context-dependent processes, including apoptosis, cell cycle regulation, adhesion, and differentiation. It is integral to several signalling cascades, notably downstream of non-canonical Wnt and mitogen activated protein kinase (MAPK) signalling pathways. As such, it is a key regulator of cellular behaviour and patterning during embryonic development across the animal kingdom. The cephalochordate amphioxus is an invertebrate chordate model system straddling the invertebrate to vertebrate transition and is thus ideally suited for comparative studies of morphogenesis. However, next to nothing is known about JNK signalling or cellular processes in this lineage. Pharmacological inhibition of JNK signalling using SP600125 during embryonic development arrests gastrula invagination and causes convergence extension-like defects in axial elongation, particularly of the notochord. Pharynx formation and anterior oral mesoderm derivatives like the preoral pit are also affected. This is accompanied by tissue-specific transcriptional changes, including reduced expression of six3/6 and wnt2 in the notochord, and ectopic wnt11 in neurulating embryos treated at late gastrula stages. Cellular delamination results in accumulation of cells in the gut cavity and a dorsal fin-like protrusion, followed by secondary Caspase-3-mediated apoptosis of polarity-deficient cells, a phenotype only partly rescued by co-culture with the pan-Caspase inhibitor Z-VAD-fmk. Ectopic activation of extracellular signal regulated kinase (ERK) signalling in the neighbours of extruded notochord and neural cells, possibly due to altered adhesive and tensile properties, as well as defects in cellular migration, may explain some phenotypes caused by JNK inhibition. Overall, this study supports conserved functions of JNK signalling in mediating the complex balance between cell survival, apoptosis, differentiation, and cell fate specification during cephalochordate morphogenesis.
Collapse
Affiliation(s)
- Ildiko M L Somorjai
- School of Biology, University of St Andrews, St Andrews, United Kingdom.,Sorbonne Université, CNRS, Biologie Intégrative des Organismes Marins, Observatoire Océanologique, Banyuls-sur-Mer, France.,Departament de Genètica, Microbiologia i Estadística, University of Barcelona, Barcelona, Spain
| | | | - Hector Escrivà
- Sorbonne Université, CNRS, Biologie Intégrative des Organismes Marins, Observatoire Océanologique, Banyuls-sur-Mer, France
| | - Jordi Garcia-Fernàndez
- Departament de Genètica, Microbiologia i Estadística, University of Barcelona, Barcelona, Spain.,Institut de Biomedicina, University of Barcelona, Barcelona, Spain
| |
Collapse
|
182
|
Lepore A, Choy PM, Lee NCW, Carella MA, Favicchio R, Briones-Orta MA, Glaser SS, Alpini G, D'Santos C, Tooze RM, Lorger M, Syn WK, Papakyriakou A, Giamas G, Bubici C, Papa S. Phosphorylation and Stabilization of PIN1 by JNK Promote Intrahepatic Cholangiocarcinoma Growth. Hepatology 2021; 74:2561-2579. [PMID: 34048060 DOI: 10.1002/hep.31983] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 04/30/2021] [Accepted: 05/16/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Intrahepatic cholangiocarcinoma (ICC) is a highly aggressive type of liver cancer in urgent need of treatment options. Aberrant activation of the c-Jun N-terminal kinase (JNK) pathway is a key feature in ICC and an attractive candidate target for its treatment. However, the mechanisms by which constitutive JNK activation promotes ICC growth, and therefore the key downstream effectors of this pathway, remain unknown for their applicability as therapeutic targets. Our aim was to obtain a better mechanistic understanding of the role of JNK signaling in ICC that could open up therapeutic opportunities. APPROACH AND RESULTS Using loss-of-function and gain-of-function studies in vitro and in vivo, we show that activation of the JNK pathway promotes ICC cell proliferation by affecting the protein stability of peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (PIN1), a key driver of tumorigenesis. PIN1 is highly expressed in ICC primary tumors, and its expression positively correlates with active JNK. Mechanistically, the JNK kinases directly bind to and phosphorylate PIN1 at Ser115, and this phosphorylation prevents PIN1 mono-ubiquitination at Lys117 and its proteasomal degradation. Moreover, pharmacological inhibition of PIN1 through all-trans retinoic acid, a Food and Drug Administration-approved drug, impairs the growth of both cultured and xenografted ICC cells. CONCLUSIONS Our findings implicate the JNK-PIN1 regulatory axis as a functionally important determinant for ICC growth, and provide a rationale for therapeutic targeting of JNK activation through PIN1 inhibition.
Collapse
Affiliation(s)
- Alessio Lepore
- Leeds Institute of Medical Research at St. James', Faculty of Medicine and Health, University of Leeds, St. James' University Hospital, Leeds, United Kingdom
| | - Pui Man Choy
- Institute of Hepatology, Foundation for Liver Research and Birkbeck University of London, London, United Kingdom
| | - Nathan C W Lee
- Leeds Institute of Medical Research at St. James', Faculty of Medicine and Health, University of Leeds, St. James' University Hospital, Leeds, United Kingdom
| | - Maria Annunziata Carella
- Center for Genome Engineering and Maintenance, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Rosy Favicchio
- Department of Surgery and Cancer, Imperial College, London, United Kingdom
| | - Marco A Briones-Orta
- Institute of Hepatology, Foundation for Liver Research and Birkbeck University of London, London, United Kingdom
- Department of Infectious Disease, Imperial College, London, United Kingdom
| | - Shannon S Glaser
- Department of Medical Physiology, Texas A&M University, Bryan, TX
| | - Gianfranco Alpini
- Division of Gastroenterology, Department of Medicine, Richard L. Roudebush VA Medical Center, Indiana University, Indianapolis, IN
| | - Clive D'Santos
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Reuben M Tooze
- Leeds Institute of Medical Research at St. James', Faculty of Medicine and Health, University of Leeds, St. James' University Hospital, Leeds, United Kingdom
| | - Mihaela Lorger
- Leeds Institute of Medical Research at St. James', Faculty of Medicine and Health, University of Leeds, St. James' University Hospital, Leeds, United Kingdom
| | - Wing-Kin Syn
- Institute of Hepatology, Foundation for Liver Research and Birkbeck University of London, London, United Kingdom
- Section of Gastroenterology, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC
- Division of Gastroenterology and Hepatology, Department of Medicine, Medical University of South Carolina, Charleston, SC
- Department of Physiology, Faculty of Medicine and Nursing, University of Basque Country UPV/EHU, Leioa, Spain
| | - Athanasios Papakyriakou
- Institute of Biosciences and Applications, National Center for Scientific Research, Athens, Greece
| | - Georgios Giamas
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, United Kingdom
| | - Concetta Bubici
- Center for Genome Engineering and Maintenance, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Salvatore Papa
- Leeds Institute of Medical Research at St. James', Faculty of Medicine and Health, University of Leeds, St. James' University Hospital, Leeds, United Kingdom
- Institute of Hepatology, Foundation for Liver Research and Birkbeck University of London, London, United Kingdom
| |
Collapse
|
183
|
Świerczek-Lasek B, Dudka D, Bauer D, Czajkowski T, Ilach K, Streminska W, Kominek A, Piwocka K, Ciemerych MA, Archacka K. Comparison of Differentiation Pattern and WNT/SHH Signaling in Pluripotent Stem Cells Cultured under Different Conditions. Cells 2021; 10:cells10102743. [PMID: 34685722 PMCID: PMC8534321 DOI: 10.3390/cells10102743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 10/01/2021] [Accepted: 10/08/2021] [Indexed: 11/16/2022] Open
Abstract
Pluripotent stem cells (PSCs) are characterized by the ability to self-renew as well as undergo multidirectional differentiation. Culture conditions have a pivotal influence on differentiation pattern. In the current study, we compared the fate of mouse PSCs using two culture media: (1) chemically defined, free of animal reagents, and (2) standard one relying on the serum supplementation. Moreover, we assessed the influence of selected regulators (WNTs, SHH) on PSC differentiation. We showed that the differentiation pattern of PSCs cultured in both systems differed significantly: cells cultured in chemically defined medium preferentially underwent ectodermal conversion while their endo- and mesodermal differentiation was limited, contrary to cells cultured in serum-supplemented medium. More efficient ectodermal differentiation of PSCs cultured in chemically defined medium correlated with higher activity of SHH pathway while endodermal and mesodermal conversion of cells cultured in serum-supplemented medium with higher activity of WNT/JNK pathway. However, inhibition of either canonical or noncanonical WNT pathway resulted in the limitation of endo- and mesodermal conversion of PSCs. In addition, blocking WNT secretion led to the inhibition of PSC mesodermal differentiation, confirming the pivotal role of WNT signaling in this process. In contrast, SHH turned out to be an inducer of PSC ectodermal, not mesodermal differentiation.
Collapse
Affiliation(s)
- Barbara Świerczek-Lasek
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland; (B.Ś.-L.); (D.D.); (D.B.); (T.C.); (K.I.); (W.S.); (M.A.C.)
| | - Damian Dudka
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland; (B.Ś.-L.); (D.D.); (D.B.); (T.C.); (K.I.); (W.S.); (M.A.C.)
| | - Damian Bauer
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland; (B.Ś.-L.); (D.D.); (D.B.); (T.C.); (K.I.); (W.S.); (M.A.C.)
| | - Tomasz Czajkowski
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland; (B.Ś.-L.); (D.D.); (D.B.); (T.C.); (K.I.); (W.S.); (M.A.C.)
| | - Katarzyna Ilach
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland; (B.Ś.-L.); (D.D.); (D.B.); (T.C.); (K.I.); (W.S.); (M.A.C.)
| | - Władysława Streminska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland; (B.Ś.-L.); (D.D.); (D.B.); (T.C.); (K.I.); (W.S.); (M.A.C.)
| | - Agata Kominek
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland; (A.K.); (K.P.)
| | - Katarzyna Piwocka
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland; (A.K.); (K.P.)
| | - Maria A. Ciemerych
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland; (B.Ś.-L.); (D.D.); (D.B.); (T.C.); (K.I.); (W.S.); (M.A.C.)
| | - Karolina Archacka
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland; (B.Ś.-L.); (D.D.); (D.B.); (T.C.); (K.I.); (W.S.); (M.A.C.)
- Correspondence: ; Tel.: +48-22-55-42-203
| |
Collapse
|
184
|
Cho H, Hah JM. A Perspective on the Development of c-Jun N-terminal Kinase Inhibitors as Therapeutics for Alzheimer's Disease: Investigating Structure through Docking Studies. Biomedicines 2021; 9:biomedicines9101431. [PMID: 34680547 PMCID: PMC8533360 DOI: 10.3390/biomedicines9101431] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 09/28/2021] [Accepted: 10/08/2021] [Indexed: 11/26/2022] Open
Abstract
c-Jun N-terminal kinase (JNK) plays an important role in cell death caused by various stimuli. Because the isoform JNK3 is mainly expressed in the brain, it is believed to play a pivotal role in various neurodegenerative diseases, including Alzheimer’s disease (AD) and Parkinson’s disease (PD), which still lack plausible therapeutics. To develop a novel and selective JNK3 inhibitor, we conducted a decadal review (2011 to 2021) of published articles on JNK inhibitors, particularly those focusing on a structural perspective and docking insights. We observed the structures of three isoforms of JNK, namely holo-proteins and co-crystal structures, with JNK3 inhibitors and summarized the significant structural aspects of selective JNK3 inhibitors as AD therapeutics.
Collapse
Affiliation(s)
- Hyunwook Cho
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan 15588, Korea;
- Center for Proteinopathy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan 15588, Korea
| | - Jung-Mi Hah
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan 15588, Korea;
- Center for Proteinopathy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan 15588, Korea
- Correspondence: ; Tel.: +82-31-400-5803
| |
Collapse
|
185
|
Chen YL, Ren Y, Rosa RH, Kuo L, Hein TW. Contributions of Sodium-Hydrogen Exchanger 1 and Mitogen-Activated Protein Kinases to Enhanced Retinal Venular Constriction to Endothelin-1 in Diabetes. Diabetes 2021; 70:2353-2363. [PMID: 34353852 PMCID: PMC8576499 DOI: 10.2337/db20-0889] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 07/28/2021] [Indexed: 11/13/2022]
Abstract
Diabetes elevates endothelin-1 (ET-1) in the vitreous and enhances constriction of retinal venules to this peptide. However, mechanisms contributing to ET-1-induced constriction of retinal venules are incompletely understood. We examined roles of sodium-hydrogen exchanger 1 (NHE1), protein kinase C (PKC), mitogen-activated protein kinases (MAPKs), and extracellular calcium (Ca2+) in retinal venular constriction to ET-1 and the impact of diabetes on these signaling molecules. Retinal venules were isolated from control pigs and pigs with streptozocin-induced diabetes for in vitro studies. ET-1-induced vasoconstriction was abolished in the absence of extracellular Ca2+ and sensitive to c-Jun N-terminal kinase (JNK) inhibitor SP600125 but unaffected by extracellular signal-regulated kinase (ERK) inhibitor PD98059, p38 kinase inhibitor SB203580, or broad-spectrum PKC inhibitor Gö 6983. Diabetes (after 2 weeks) enhanced venular constriction to ET-1, which was insensitive to PD98059 and Gö 6983 but was prevented by NHE1 inhibitor cariporide, SB203580, and SP600125. In conclusion, extracellular Ca2+ entry and activation of JNK, independent of ERK and PKC, mediate constriction of retinal venules to ET-1. Diabetes activates p38 MAPK and NHE1, which cause enhanced venular constriction to ET-1. Treatments targeting these vascular molecules may lessen retinal complications in early diabetes.
Collapse
Affiliation(s)
- Yen-Lin Chen
- Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Bryan, TX
| | - Yi Ren
- Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Bryan, TX
| | - Robert H Rosa
- Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Bryan, TX
- Department of Ophthalmology, Baylor Scott & White Eye Institute, Temple, TX
| | - Lih Kuo
- Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Bryan, TX
| | - Travis W Hein
- Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Bryan, TX
| |
Collapse
|
186
|
Ishijima T, Nakajima K. Inflammatory cytokines TNFα, IL-1β, and IL-6 are induced in endotoxin- stimulated microglia through different signaling cascades. Sci Prog 2021; 104:368504211054985. [PMID: 34821182 PMCID: PMC10450609 DOI: 10.1177/00368504211054985] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
By using an animal model in which inflammatory cytokines are induced in lipopolysaccharide (LPS)-injected rat brain, we investigated the induction of tumor necrosis factor alpha (TNFα), interleukin-1beta (IL-1β), and IL-6. Immunoblotting and immunohistochemistry revealed that all three cytokines were transiently induced in the cerebral cortex at about 12 h after LPS injection. To clarify which glial cell type induced the cytokines, we examined the respective abilities of astrocytes and microglia in vitro. Primary microglia largely induced TNFα, IL-1β and IL-6 in response to LPS, but primary astrocytes induced only limited levels of TNFα. Thus, we used specific inhibitors to focus on microglia in surveying signaling molecules involved in the induction of TNFα, IL-1β, and IL-6. The experiments using mitogen-activated protein kinases (MAPK) inhibitors revealed that c-Jun N-terminal kinase (JNK)/p38, external signal regulated kinase (ERK)/JNK, and ERK/JNK/p38 are necessary for the induction of TNFα, IL-1β, and IL-6, respectively. The experiments using protein kinase C (PKC) inhibitor clarified that PKCα is required for the induction of all these cytokines in LPS-stimulated microglia. Furthermore, LPS-dependent IL-1β/IL-6 induction was suppressed by pretreatment with a nitric oxide (NO) scavenger, suggesting that NO is involved in the signaling cascade of IL-1β/IL-6 induction. Thus, an inducible NO synthase induced in the LPS-injected cerebral cortex might be related to the induction of IL-1β/IL-6 through the production of NO in vivo. Taken together, these results demonstrated that microglia induce different kinds of inflammatory cytokine through specific combinations of MAPKs and by the presence or absence of NO.
Collapse
Affiliation(s)
- Takashi Ishijima
- Department of Science and Engineering for Sustainable Innovation, Faculty of Science and Engineering, Glycan & Life Systems Integration Center, Soka University, Tokyo, Japan
| | - Kazuyuki Nakajima
- Department of Science and Engineering for Sustainable Innovation, Faculty of Science and Engineering, Glycan & Life Systems Integration Center, Soka University, Tokyo, Japan
| |
Collapse
|
187
|
Fermaintt CS, Takahashi-Ruiz L, Liang H, Mooberry SL, Risinger AL. Eribulin Activates the cGAS-STING Pathway via the Cytoplasmic Accumulation of Mitochondrial DNA. Mol Pharmacol 2021; 100:309-318. [PMID: 34312217 PMCID: PMC8626644 DOI: 10.1124/molpharm.121.000297] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 07/14/2021] [Indexed: 11/23/2022] Open
Abstract
Microtubule-targeting agents (MTAs), including both microtubule stabilizers and destabilizers are highly effective chemotherapeutic drugs used in the treatment of solid tumors and hematologic malignancies. In addition to the shared ability of all MTAs to block cell cycle progression, growing evidence shows that different agents of this class can also have mechanistically distinct effects on nonmitotic microtubule-dependent cellular processes, including cellular signaling and transport. Herein, we test the biologic hypothesis that MTAs used in the treatment of triple-negative breast cancer (TNBC) can differentially affect innate immune signaling pathways independent of their antimitotic effects. Our data demonstrate that the microtubule destabilizer eribulin, but not the microtubule stabilizer paclitaxel, induces cGAS-STING-dependent expression of interferon-β in both myeloid and TNBC cells. Activation of the cGAS-STING pathway by eribulin was further found to be mediated by the accumulation of cytoplasmic mitochondrial DNA. Together, these findings provide mechanistic insight into how eribulin can induce innate immune signaling independent of its antimitotic or cytotoxic effects. SIGNIFICANCE STATEMENT: Microtubule-targeting agents (MTAs) are often used in the treatment of breast cancer and have been used in combination with immune checkpoint inhibitors to improve efficacy. Although all clinically approved MTAs share an antimitotic mechanism of action, their distinct effects on interphase microtubules can promote differential downstream signaling consequences. This work shows that the microtubule destabilizer eribulin, but not the microtubule stabilizer paclitaxel, activates the cGAS-STING innate immune signaling pathway through the accumulation of mitochondrial DNA in the cytoplasm.
Collapse
Affiliation(s)
- Charles S Fermaintt
- Department of Pharmacology (C.S.F., L.T.-R., H.L., S.L.M., A.L.R.), and Mays Cancer Center (C.S.F., S.L.M., A.L.R.), University of Texas Health Science Center San Antonio, San Antonio, Texas
| | - Leila Takahashi-Ruiz
- Department of Pharmacology (C.S.F., L.T.-R., H.L., S.L.M., A.L.R.), and Mays Cancer Center (C.S.F., S.L.M., A.L.R.), University of Texas Health Science Center San Antonio, San Antonio, Texas
| | - Huiyun Liang
- Department of Pharmacology (C.S.F., L.T.-R., H.L., S.L.M., A.L.R.), and Mays Cancer Center (C.S.F., S.L.M., A.L.R.), University of Texas Health Science Center San Antonio, San Antonio, Texas
| | - Susan L Mooberry
- Department of Pharmacology (C.S.F., L.T.-R., H.L., S.L.M., A.L.R.), and Mays Cancer Center (C.S.F., S.L.M., A.L.R.), University of Texas Health Science Center San Antonio, San Antonio, Texas
| | - April L Risinger
- Department of Pharmacology (C.S.F., L.T.-R., H.L., S.L.M., A.L.R.), and Mays Cancer Center (C.S.F., S.L.M., A.L.R.), University of Texas Health Science Center San Antonio, San Antonio, Texas
| |
Collapse
|
188
|
Cao Y, Wang Q, Liu C, Wang W, Lai S, Zou H, Tao E, Wang F, Wan L. Capn4 aggravates angiotensin II-induced cardiac hypertrophy by activating the IGF-AKT signaling pathway. J Biochem 2021; 171:53-61. [PMID: 34580724 DOI: 10.1093/jb/mvab100] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 09/22/2021] [Indexed: 11/12/2022] Open
Abstract
Capn4 belongs to a family of calpains that participate in a wide variety of biological functions, but little is known about the role of Capn4 in cardiac disease. Here, we show that the expression of Capn4 was significantly increased in Angiotensin II (Ang II)-treated cardiomyocytes and Ang II-induced cardiac hypertrophic mouse hearts. Importantly, in agreement with the Capn4 expression patterns, the maximal calpain activity measured in heart homogenates was elevated in Ang II-treated mice, and oral coadministration of SNJ-1945 (calpain inhibitor) attenuated the total calpain activity measured in vitro. Functional assays indicated that overexpression of Capn4 obviously aggravated Ang II-induced cardiac hypertrophy, whereas Capn4 knockdown resulted in the opposite phenotypes. Further investigation demonstrated that Capn4 maintained the activation of the insulin-like growth factor (IGF)-AKT signaling pathway in cardiomyocytes by increasing c-Jun expression. Mechanistic investigations revealed that Capn4 directly bound and stabilized c-Jun, and knockdown of Capn4 increased the ubiquitination level of c-Jun in cardiomyocytes. Additionally, our results demonstrated that the antihypertrophic effect of Capn4 silencing was partially dependent on the inhibition of c-Jun. Overall, these data suggested that Capn4 contributes to cardiac hypertrophy by enhancing the c-Jun-mediated IGF-AKT signaling pathway and could be a potential therapeutic target for hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
- Yuanping Cao
- Department of Cardiac Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Qun Wang
- Department of Cardiac Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Caiyun Liu
- Operating Room, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Wenjun Wang
- Department of Cardiac Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Songqing Lai
- Department of Cardiac Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Huaxi Zou
- Department of Cardiac Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Ende Tao
- Department of Cardiac Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Fudong Wang
- Department of Cardiac Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Li Wan
- Department of Cardiac Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| |
Collapse
|
189
|
Liakhov SA, Schepetkin IA, Karpenko OS, Duma HI, Haidarzhy NM, Kirpotina LN, Kovrizhina AR, Khlebnikov AI, Bagryanskaya IY, Quinn MT. Novel c-Jun N-Terminal Kinase (JNK) Inhibitors with an 11 H-Indeno[1,2- b]quinoxalin-11-one Scaffold. Molecules 2021; 26:molecules26185688. [PMID: 34577159 PMCID: PMC8464905 DOI: 10.3390/molecules26185688] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/10/2021] [Accepted: 09/13/2021] [Indexed: 11/16/2022] Open
Abstract
c-Jun N-terminal kinase (JNK) plays a central role in stress signaling pathways implicated in important pathological processes, including rheumatoid arthritis and ischemia-reperfusion injury. Therefore, inhibition of JNK is of interest for molecular targeted therapy to treat various diseases. We synthesized 13 derivatives of our reported JNK inhibitor 11H-indeno[1,2-b]quinoxalin-11-one oxime and evaluated their binding to the three JNK isoforms and their biological effects. Eight compounds exhibited submicromolar binding affinity for at least one JNK isoform. Most of these compounds also inhibited lipopolysaccharide (LPS)-induced nuclear factor-κB/activating protein 1 (NF-κB/AP-1) activation and interleukin-6 (IL-6) production in human monocytic THP1-Blue cells and human MonoMac-6 cells, respectively. Selected compounds (4f and 4m) also inhibited LPS-induced c-Jun phosphorylation in MonoMac-6 cells, directly confirming JNK inhibition. We conclude that indenoquinoxaline-based oximes can serve as specific small-molecule modulators for mechanistic studies of JNKs, as well as potential leads for the development of anti-inflammatory drugs.
Collapse
Affiliation(s)
- Serhii A. Liakhov
- A.V. Bogatsky Physico-Chemical Institute, National Academy of Sciences of Ukraine, 65080 Odessa, Ukraine; (S.A.L.); (O.S.K.); (H.I.D.)
| | - Igor A. Schepetkin
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA; (I.A.S.); (L.N.K.)
| | - Olexander S. Karpenko
- A.V. Bogatsky Physico-Chemical Institute, National Academy of Sciences of Ukraine, 65080 Odessa, Ukraine; (S.A.L.); (O.S.K.); (H.I.D.)
| | - Hanna I. Duma
- A.V. Bogatsky Physico-Chemical Institute, National Academy of Sciences of Ukraine, 65080 Odessa, Ukraine; (S.A.L.); (O.S.K.); (H.I.D.)
| | | | - Liliya N. Kirpotina
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA; (I.A.S.); (L.N.K.)
| | - Anastasia R. Kovrizhina
- Kizhner Research Center, Tomsk Polytechnic University, 634050 Tomsk, Russia; (A.R.K.); (A.I.K.)
| | - Andrei I. Khlebnikov
- Kizhner Research Center, Tomsk Polytechnic University, 634050 Tomsk, Russia; (A.R.K.); (A.I.K.)
| | - Irina Y. Bagryanskaya
- Vorozhtsov Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia;
| | - Mark T. Quinn
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA; (I.A.S.); (L.N.K.)
- Correspondence: ; Tel.: +406-994-4707; Fax: +406-994-4303
| |
Collapse
|
190
|
Mbonye U, Leskov K, Shukla M, Valadkhan S, Karn J. Biogenesis of P-TEFb in CD4+ T cells to reverse HIV latency is mediated by protein kinase C (PKC)-independent signaling pathways. PLoS Pathog 2021; 17:e1009581. [PMID: 34529720 PMCID: PMC8478230 DOI: 10.1371/journal.ppat.1009581] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 09/28/2021] [Accepted: 09/04/2021] [Indexed: 01/09/2023] Open
Abstract
The switch between HIV latency and productive transcription is regulated by an auto-feedback mechanism initiated by the viral trans-activator Tat, which functions to recruit the host transcription elongation factor P-TEFb to proviral HIV. A heterodimeric complex of CDK9 and one of three cyclin T subunits, P-TEFb is expressed at vanishingly low levels in resting memory CD4+ T cells and cellular mechanisms controlling its availability are central to regulation of the emergence of HIV from latency. Using a well-characterized primary T-cell model of HIV latency alongside healthy donor memory CD4+ T cells, we characterized specific T-cell receptor (TCR) signaling pathways that regulate the generation of transcriptionally active P-TEFb, defined as the coordinate expression of cyclin T1 and phospho-Ser175 CDK9. Protein kinase C (PKC) agonists, such as ingenol and prostratin, stimulated active P-TEFb expression and reactivated latent HIV with minimal cytotoxicity, even in the absence of intracellular calcium mobilization with an ionophore. Unexpectedly, inhibition-based experiments demonstrated that PKC agonists and TCR-mobilized diacylglycerol signal through MAP kinases ERK1/2 rather than through PKC to effect the reactivation of both P-TEFb and latent HIV. Single-cell and bulk RNA-seq analyses revealed that of the four known isoforms of the Ras guanine nucleotide exchange factor RasGRP, RasGRP1 is by far the predominantly expressed diacylglycerol-dependent isoform in CD4+ T cells. RasGRP1 should therefore mediate the activation of ERK1/2 via Ras-Raf signaling upon TCR co-stimulation or PKC agonist challenge. Combined inhibition of the PI3K-mTORC2-AKT-mTORC1 pathway and the ERK1/2 activator MEK prior to TCR co-stimulation abrogated active P-TEFb expression and substantially suppressed latent HIV reactivation. Therefore, contrary to prevailing models, the coordinate reactivation of P-TEFb and latent HIV in primary T cells following either TCR co-stimulation or PKC agonist challenge is independent of PKC but rather involves two complementary signaling arms of the TCR cascade, namely, RasGRP1-Ras-Raf-MEK-ERK1/2 and PI3K-mTORC2-AKT-mTORC1.
Collapse
Affiliation(s)
- Uri Mbonye
- Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
- * E-mail: (UM); (JK)
| | - Konstantin Leskov
- Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Meenakshi Shukla
- Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Saba Valadkhan
- Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Jonathan Karn
- Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
- * E-mail: (UM); (JK)
| |
Collapse
|
191
|
Kobayashi Y, Tomoshige S, Imakado K, Sekino Y, Koganezawa N, Shirao T, Diniz GB, Miyamoto T, Saito Y. Ciliary GPCR-based transcriptome as a key regulator of cilia length control. FASEB Bioadv 2021; 3:744-767. [PMID: 34485842 PMCID: PMC8409570 DOI: 10.1096/fba.2021-00029] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/08/2021] [Accepted: 06/14/2021] [Indexed: 12/28/2022] Open
Abstract
The primary cilium is a plasma membrane-protruding sensory organelle that efficiently conveys signaling cascades in a highly ordered microenvironment. Its signaling is mediated, in part, by a limited set of GPCRs preferentially enriched in the cilium membrane. This includes melanin-concentrating hormone (MCH) receptor 1 (MCHR1), which plays a role in feeding and mood. In addition to its receptor composition, the length of the cilium is a characteristic parameter that is implicated in its function. We previously found that MCH can dynamically shorten cilia length via the Gi/o and Akt pathways in both MCHR1-expressing hTERT-RPE1 cells (hRPE1 cells) and rat hippocampal neurons. However, the detailed mechanisms by which MCH regulates cilia length through ciliary MCHR1 remains unclear. In this study, we aimed to determine the transcriptome changes in MCHR1-expressing hRPE1 cells in response to MCH to identify the target molecules involved in cilia length regulation via MCHR1 activation. RNA sequencing analysis of ciliated cells subjected to MCH treatment showed upregulation of 424 genes and downregulation of 112 genes compared with static control cells. Validation by quantitative real-time PCR, knocking down, and CRISPR/Cas9-mediated knockout technology identified a molecule, PDZ and LIM domain-containing protein 5 (PDLIM5). Thus, it was considered as the most significant key factor for MCHR1-mediated shortening of cilia length. Additional analyses revealed that the actin-binding protein alpha-actinin 1/4 is a crucial downstream target of the PDLIM5 signaling pathway that exerts an effect on MCHR1-induced cilia shortening. In the endogenous MCHR1-expressing hippocampus, transcriptional upregulation of PDLIM5 and actinin 1/4, following the application of MCH, was detected when the MCHR1-positive cilia were shortened. Together, our transcriptome study based on ciliary MCHR1 function uncovered a novel and important regulatory step underlying cilia length control. These results will potentially serve as a basis for understanding the mechanism underlying the development of obesity and mood disorders.
Collapse
Affiliation(s)
- Yuki Kobayashi
- Graduate School of Integrated Sciences for LifeHiroshima UniversityHiroshimaJapan
| | - Sakura Tomoshige
- Graduate School of Integrated Sciences for LifeHiroshima UniversityHiroshimaJapan
| | - Kosuke Imakado
- Graduate School of Integrated Sciences for LifeHiroshima UniversityHiroshimaJapan
| | - Yuko Sekino
- Endowed Laboratory of Human Cell‐Based Drug DiscoveryGraduate School of Pharmaceutical SciencesThe University of TokyoTokyoJapan
| | - Noriko Koganezawa
- Department of Neurobiology and BehaviorGraduate School of MedicineGunma UniversityMaebashiJapan
| | - Tomoaki Shirao
- Department of Neurobiology and BehaviorGraduate School of MedicineGunma UniversityMaebashiJapan
- AlzMed, Inc.TokyoJapan
| | - Giovanne B. Diniz
- California National Primate Research CenterUniversity of CaliforniaDavisCAUSA
| | - Tatsuo Miyamoto
- Department of Genetics and Cell BiologyResearch Institute for Radiation Biology and MedicineHiroshima UniversityHiroshimaJapan
| | - Yumiko Saito
- Graduate School of Integrated Sciences for LifeHiroshima UniversityHiroshimaJapan
| |
Collapse
|
192
|
Ren L, Zhang X, Li J, Yan X, Gao X, Cui J, Tang C, Liu S. Diverse transcriptional patterns of homoeologous recombinant transcripts in triploid fish (Cyprinidae). SCIENCE CHINA. LIFE SCIENCES 2021; 64:1491-1501. [PMID: 33420922 DOI: 10.1007/s11427-020-1749-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 08/13/2020] [Indexed: 11/29/2022]
Abstract
Homoeologous recombination (HR), the exchange of homoeologous chromosomes, contributes to subgenome adaptation to diverse environments by producing various phenotypes. However, the potential relevance of HR and innate immunity is rarely described in triploid cyprinid fish species. In our study, two allotriploid genotypes (R2C and RC2), whose innate immunity was stronger than their inbred parents (Carassius auratus red var. and Cyprinus carpio L.), were obtained from backcrossing between male allotetraploids of C. auratus red var.×C. carpio L. and females of their two inbred parents, respectively. The work detected 140 HRs shared between the two triploids at the genomic level. Further, transcriptions of 54 homoeologous recombinant genes (HRGs) in R2C and 65 HRGs in RC2 were detected using both Illumina and PacBio data. Finally, by comparing expressed recombinant reads to total expressed reads in each of the genes, a range of 0.1%-10% was observed in most of the 99-193 HRGs, of which six recombinant genes were classified as "response to stimulus". These results not only provide a novel way to predict HRs in allopolyploids based on cross prediction at both genomic and transcriptional levels, but also insight into the potential relationship between HRs related to innate immunity and adaptation of the triploids and allotetraploids.
Collapse
Affiliation(s)
- Li Ren
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, 410081, China.,College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Xueyin Zhang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, 410081, China.,College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Jiaming Li
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, 410081, China.,College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Xiaojing Yan
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, 410081, China.,College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Xin Gao
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, 410081, China.,College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Jialin Cui
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, 410081, China.,College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Chenchen Tang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, 410081, China.,College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Shaojun Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, 410081, China. .,College of Life Sciences, Hunan Normal University, Changsha, 410081, China.
| |
Collapse
|
193
|
Hioki T, Tokuda H, Tanabe K, Kim W, Tachi J, Yamaguchi S, Matsushima-Nishiwaki R, Kozawa O, Iida H. Amplification by tramadol of PGD 2-induced osteoprotegerin synthesis in osteoblasts: Involvement of μ-opioid receptor and 5-HT transporter. Prostaglandins Leukot Essent Fatty Acids 2021; 172:102323. [PMID: 34392133 DOI: 10.1016/j.plefa.2021.102323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/21/2021] [Accepted: 08/03/2021] [Indexed: 12/01/2022]
Abstract
Tramadol, a weak μ-opioid receptor (MOR) agonist with inhibitory effects on the reuptake of serotonin (5-hydroxytryptamine; 5-HT) and norepinephrine, is an effective analgesic to chronic pains. Osteoprotegerin produced by osteoblasts is essential for bone remodeling to suppress osteoclastic bone resorption. We previously reported that prostaglandin D2 (PGD2) induces osteoprotegerin synthesis whereby p44/p42 mitogen-activated protein (MAP) kinase, p38 MAP kinase and stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) are involved in osteoblast-like MC3T3-E1 cells. Herein, we investigated the mechanism underlying the effect of tramadol on the PGD2-induced osteoprotegerin synthesis in these cells. Tramadol enhanced the PGD2-induced release and mRNA expression of osteoprotegerin. Naloxone, a MOR antagonist, reduced the amplification by tramadol of the PGD2-stimulated osteoprotegerin release. Not the selective norepinephrine reuptake inhibitor reboxetine but the selective serotonin reuptake inhibitors fluvoxamine and sertraline upregulated the PGD2-induced osteoprotegerin release, which was further amplified by morphine. Tramadol enhanced PGD2-stimulated phosphorylation of p38 MAP kinase and SAPK/JNK, but not p44/p42 MAP kinase. Both SB203580 and SP600125 suppressed the tramadol effect to enhance the PGD2-stimulated osteoprotegerin release. Tramadol enhanced the PGE2-induced osteoprotegerin release as well as PGD2. These results suggest that tramadol amplifies the PGD2-induced osteoprotegerin synthesis at the upstream of p38 MAP kinase and SAPK/JNK in the involvement of both MOR and 5-HT transporter in osteoblasts.
Collapse
Affiliation(s)
- Tomoyuki Hioki
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of Dermatology, Kizawa Memorial Hospital, Minokamo, Gifu 505-8503, Japan; Department of Metabolic Research, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan
| | - Haruhiko Tokuda
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of Metabolic Research, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan; Department of Clinical Laboratory/Medical Genome Center, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan
| | - Kumiko Tanabe
- Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Woo Kim
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Junko Tachi
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Shinobu Yamaguchi
- Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | | | - Osamu Kozawa
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of Metabolic Research, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan.
| | - Hiroki Iida
- Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| |
Collapse
|
194
|
Aguado J, Chaggar HK, Gómez‐Inclán C, Shaker MR, Leeson HC, Mackay‐Sim A, Wolvetang EJ. Inhibition of the cGAS-STING pathway ameliorates the premature senescence hallmarks of Ataxia-Telangiectasia brain organoids. Aging Cell 2021; 20:e13468. [PMID: 34459078 PMCID: PMC8441292 DOI: 10.1111/acel.13468] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 07/04/2021] [Accepted: 08/14/2021] [Indexed: 12/13/2022] Open
Abstract
Ataxia-telangiectasia (A-T) is a genetic disorder caused by the lack of functional ATM kinase. A-T is characterized by chronic inflammation, neurodegeneration and premature ageing features that are associated with increased genome instability, nuclear shape alterations, micronuclei accumulation, neuronal defects and premature entry into cellular senescence. The causal relationship between the detrimental inflammatory signature and the neurological deficiencies of A-T remains elusive. Here, we utilize human pluripotent stem cell-derived cortical brain organoids to study A-T neuropathology. Mechanistically, we show that the cGAS-STING pathway is required for the recognition of micronuclei and induction of a senescence-associated secretory phenotype (SASP) in A-T olfactory neurosphere-derived cells and brain organoids. We further demonstrate that cGAS and STING inhibition effectively suppresses self-DNA-triggered SASP expression in A-T brain organoids, inhibits astrocyte senescence and neurodegeneration, and ameliorates A-T brain organoid neuropathology. Our study thus reveals that increased cGAS and STING activity is an important contributor to chronic inflammation and premature senescence in the central nervous system of A-T and constitutes a novel therapeutic target for treating neuropathology in A-T patients.
Collapse
Affiliation(s)
- Julio Aguado
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandSaint LuciaQueenslandAustralia
| | - Harman K. Chaggar
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandSaint LuciaQueenslandAustralia
- Present address:
Cellesce Ltd, Cardiff MedicentreHeath ParkCardiffUK
| | - Cecilia Gómez‐Inclán
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandSaint LuciaQueenslandAustralia
| | - Mohammed R. Shaker
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandSaint LuciaQueenslandAustralia
| | - Hannah C. Leeson
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandSaint LuciaQueenslandAustralia
| | - Alan Mackay‐Sim
- Department of NeurogeneticsKolling InstituteSydney Medical SchoolUniversity of SydneySydneyNew South WalesAustralia
- Griffith Institute for Drug DiscoveryGriffith UniversityNathanQueenslandAustralia
| | - Ernst J. Wolvetang
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandSaint LuciaQueenslandAustralia
| |
Collapse
|
195
|
Inhibition of the MAP2K7-JNK pathway with 5Z-7-oxozeaenol induces apoptosis in T-cell acute lymphoblastic leukemia. Oncotarget 2021; 12:1787-1801. [PMID: 34504651 PMCID: PMC8416565 DOI: 10.18632/oncotarget.28040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 07/28/2021] [Indexed: 01/10/2023] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive pediatric leukemia with a worse prognosis than most frequent B-cell ALL due to a high incidence of treatment failures and relapse. Our previous work showed that loss of the pioneer factor KLF4 in a NOTCH1-induced T-ALL mouse model accelerated the development of leukemia through expansion of leukemia-initiating cells and activation of the MAP2K7 pathway. Similarly, epigenetic silencing of the KLF4 gene in children with T-ALL was associated with MAP2K7 activation. Here, we showed the small molecule 5Z-7-oxozeaenol (5Z7O) induces dose-dependent cytotoxicity in a panel of T-ALL cell lines mainly through inhibition of the MAP2K7-JNK pathway, which further validates MAP2K7 as a therapeutic target. Mechanistically, 5Z7O-mediated apoptosis was caused by the downregulation of regulators of the G2/M checkpoint and the inhibition of survival pathways. The anti-leukemic capacity of 5Z7O was evaluated using leukemic cells from two mouse models of T-ALL and patient-derived xenograft cells generated using lymphoblasts from pediatric T-ALL patients. Finally, a combination of 5Z7O with dexamethasone, a drug used in frontline therapy, showed synergistic induction of cytotoxicity. In sum, we report here that MAP2K7 inhibition thwarts survival mechanisms in T-ALL cells and warrants future pre-clinical studies for high-risk and relapsed patients.
Collapse
|
196
|
The Novel Antitumor Compound HCA Promotes Glioma Cell Death by Inducing Endoplasmic Reticulum Stress and Autophagy. Cancers (Basel) 2021; 13:cancers13174290. [PMID: 34503102 PMCID: PMC8428344 DOI: 10.3390/cancers13174290] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/11/2021] [Accepted: 08/23/2021] [Indexed: 12/24/2022] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive type of primary brain tumor in adults, and the median survival of patients with GBM is 14.5 months. Melitherapy is an innovative therapeutic approach to treat different diseases, including cancer, and it is based on the regulation of cell membrane composition and structure, which modulates relevant signal pathways. Here, we have tested the effects of 2-hydroxycervonic acid (HCA) on GBM cells and xenograft tumors. HCA was taken up by cells and it compromised the survival of several human GBM cell lines in vitro, as well as the in vivo growth of xenograft tumors (mice) derived from these cells. HCA appeared to enhance ER stress/UPR signaling, which consequently induced autophagic cell death of the GBM tumor cells. This negative effect of HCA on GBM cells may be mediated by the JNK/c-Jun/CHOP/BiP axis, and it also seems to be provoked by the cellular metabolite of HCA, C21:5n-3 (heneicosapentaenoic acid). These results demonstrate the efficacy of the melitherapeutic treatment used and the potential of using C21:5n-3 as an efficacy biomarker for this treatment. Given the safety profile in animal models, the data presented here provide evidence that HCA warrants further clinical study as a potential therapy for GBM, currently an important unmet medical need.
Collapse
|
197
|
Understanding Abnormal c-JNK/p38MAPK Signaling Overactivation Involved in the Progression of Multiple Sclerosis: Possible Therapeutic Targets and Impact on Neurodegenerative Diseases. Neurotox Res 2021; 39:1630-1650. [PMID: 34432262 DOI: 10.1007/s12640-021-00401-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 08/01/2021] [Accepted: 08/09/2021] [Indexed: 12/16/2022]
Abstract
Demyelination, immune dysregulation, and neuroinflammation are the most common triggers of motor neuron disorders such as multiple sclerosis (MS). MS is a chronic demyelinating neurodegenerative disease of the central nervous system caused by abnormal immune activation, which causes myelin sheath damage. Cell signal transduction pathways are required for a variety of physiological and pathological processes in the brain. When these signaling systems become overactive, they can lead to disease progression. In various physiological conditions, abnormal mitogen-activated protein kinase (MAPK) activation is associated with several physiological dysfunctions that cause neurodegeneration. Previous research indicates that c-JNK and p38MAPK signaling play critical roles in neuronal growth and differentiation. c-JNK/p38MAPK is a member of the MAPK family, which regulates metabolic pathways, cell proliferation, differentiation, and apoptosis that control certain neurological activities. During brain injuries, c-JNK/p38MAPK also affects neuronal elastic properties, nerve growth, and cognitive processing. This review systematically linked abnormal c-JNK/p38MAPK signaling activation to multiple neuropathological pathways in MS and related neurological dysfunctions. MS progression is linked to genetic defects, oligodendrocyte destruction, glial overactivation, and immune dysregulation. We concluded that inhibiting both the c-JNK/p38MAPK signaling pathways can promote neuroprotection and neurotrophic effects against the clinical-pathological presentation of MS and influence other neurological disorders. As a result, the potential benefits of c-JNK/p38MAPK downregulation for the development of disease-modifying treatment interventions in the future could include MS prevention and related neurocomplications.
Collapse
|
198
|
Combined intermittent fasting and ERK inhibition enhance the anti-tumor effects of chemotherapy via the GSK3β-SIRT7 axis. Nat Commun 2021; 12:5058. [PMID: 34433808 PMCID: PMC8387475 DOI: 10.1038/s41467-021-25274-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 07/29/2021] [Indexed: 12/28/2022] Open
Abstract
Dietary interventions such as intermittent fasting (IF) have emerged as an attractive strategy for cancer therapies; therefore, understanding the underlying molecular mechanisms is pivotal. Here, we find SIRT7 decline markedly attenuates the anti-tumor effect of IF. Mechanistically, AMP-activated protein kinase (AMPK) phosphorylating SIRT7 at T263 triggers further phosphorylation at T255/S259 by glycogen synthase kinase 3β (GSK3β), which stabilizes SIRT7 by decoupling E3 ligase UBR5. SIRT7 hyperphosphorylation achieves anti-tumor activity by disrupting the SKP2-SCF E3 ligase, thus preventing SKP2-mediated K63-linked AKT polyubiquitination and subsequent activation. In contrast, GSK3β-SIRT7 axis is inhibited by EGF/ERK2 signaling, with ERK2 inactivating GSK3β, thus accelerating SIRT7 degradation. Unfavorably, glucose deprivation or chemotherapy hijacks the GSK3β-SIRT7 axis via ERK2, thus activating AKT and ensuring survival. Notably, Trametinib, an FDA-approved MEK inhibitor, enhances the efficacy of combination therapy with doxorubicin and IF. Overall, we have revealed the GSK3β-SIRT7 axis that must be fine-tuned in the face of the energetic and oncogenic stresses in malignancy. The combination of intermittent fasting and chemotherapy can improve the response to treatment. Here, the authors show that SIRT7 activation is required to inactivate Akt during intermittent fasting and that the combination of intermittent fasting and inhibitors that block the Erk pathway can improve efficacy of treatment.
Collapse
|
199
|
Mollinedo F, Gajate C. Direct Endoplasmic Reticulum Targeting by the Selective Alkylphospholipid Analog and Antitumor Ether Lipid Edelfosine as a Therapeutic Approach in Pancreatic Cancer. Cancers (Basel) 2021; 13:4173. [PMID: 34439330 PMCID: PMC8394177 DOI: 10.3390/cancers13164173] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/11/2021] [Accepted: 08/15/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), the most common malignancy of the pancreas, shows a dismal and grim overall prognosis and survival rate, which have remained virtually unchanged for over half a century. PDAC is the most lethal of all cancers, with the highest mortality-to-incidence ratio. PDAC responds poorly to current therapies and remains an incurable malignancy. Therefore, novel therapeutic targets and drugs are urgently needed for pancreatic cancer treatment. Selective induction of apoptosis in cancer cells is an appealing approach in cancer therapy. Apoptotic cell death is highly regulated by different signaling routes that involve a variety of subcellular organelles. Endoplasmic reticulum (ER) stress acts as a double-edged sword at the interface of cell survival and death. Pancreatic cells exhibit high hormone and enzyme secretory functions, and thereby show a highly developed ER. Thus, pancreatic cancer cells display a prominent ER. Solid tumors have to cope with adverse situations in which hypoxia, lack of certain nutrients, and the action of certain antitumor agents lead to a complex interplay and crosstalk between ER stress and autophagy-the latter acting as an adaptive survival response. ER stress also mediates cell death induced by a number of anticancer drugs and experimental conditions, highlighting the pivotal role of ER stress in modulating cell fate. The alkylphospholipid analog prototype edelfosine is selectively taken up by tumor cells, accumulates in the ER of a number of human solid tumor cells-including pancreatic cancer cells-and promotes apoptosis through a persistent ER-stress-mediated mechanism both in vitro and in vivo. Here, we discuss and propose that direct ER targeting may be a promising approach in the therapy of pancreatic cancer, opening up a new avenue for the treatment of this currently incurable and deadly cancer. Furthermore, because autophagy acts as a cytoprotective response to ER stress, potentiation of the triggering of a persistent ER response by combination therapy, together with the use of autophagy blockers, could improve the current gloomy expectations for finding a cure for this type of cancer.
Collapse
Affiliation(s)
- Faustino Mollinedo
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine, C/Ramiro de Maeztu 9, E-28040 Madrid, Spain;
| | | |
Collapse
|
200
|
Lin HH, Robertson KL, Bisbee HA, Farkas ME. Oncogenic and Circadian Effects of Small Molecules Directly and Indirectly Targeting the Core Circadian Clock. Integr Cancer Ther 2021; 19:1534735420924094. [PMID: 32493076 PMCID: PMC7273620 DOI: 10.1177/1534735420924094] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Circadian rhythms are essential for controlling the cell cycle, cellular
proliferation, and apoptosis, and hence are tightly linked to cell fate. Several
recent studies have used small molecules to affect circadian oscillations;
however, their concomitant cellular effects were not assessed, and they have not
been compared under similar experimental conditions. In this work, we use five
molecules, grouped into direct versus indirect effectors of the circadian clock,
to modulate periods in a human osteosarcoma cell line (U2OS) and determine their
influences on cellular behaviors, including motility and colony formation.
Luciferase reporters, whose expression was driven via Bmal1- or
Per2-promoters, were used to facilitate the visualization
and quantitative analysis of circadian oscillations. We show that all molecules
increase or decrease the circadian periods of Bmal1 and
Per2 in a dose-dependent manner, but period length does not
correlate with the extent of cell migration or proliferation. Nonetheless,
molecules that affected circadian oscillations to a greater degree resulted in
substantial influence on cellular behaviors (ie, motility and colony formation),
which may also be attributable to noncircadian targets. Furthermore, we find
that the ability and extent to which the molecules are able to affect
oscillations is independent of whether they are direct or indirect modulators.
Because of the numerous connections and feedback between the circadian clock and
other pathways, it is important to consider the effects of both in assessing
these and other compounds.
Collapse
Affiliation(s)
- Hui-Hsien Lin
- University of Massachusetts Amherst, Amherst, MA, USA
| | | | | | | |
Collapse
|