1
|
Sanchez C, Ramirez A, Hodgson L. Unravelling molecular dynamics in living cells: Fluorescent protein biosensors for cell biology. J Microsc 2025; 298:123-184. [PMID: 38357769 PMCID: PMC11324865 DOI: 10.1111/jmi.13270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 01/11/2024] [Accepted: 01/22/2024] [Indexed: 02/16/2024]
Abstract
Genetically encoded, fluorescent protein (FP)-based Förster resonance energy transfer (FRET) biosensors are microscopy imaging tools tailored for the precise monitoring and detection of molecular dynamics within subcellular microenvironments. They are characterised by their ability to provide an outstanding combination of spatial and temporal resolutions in live-cell microscopy. In this review, we begin by tracing back on the historical development of genetically encoded FP labelling for detection in live cells, which lead us to the development of early biosensors and finally to the engineering of single-chain FRET-based biosensors that have become the state-of-the-art today. Ultimately, this review delves into the fundamental principles of FRET and the design strategies underpinning FRET-based biosensors, discusses their diverse applications and addresses the distinct challenges associated with their implementation. We place particular emphasis on single-chain FRET biosensors for the Rho family of guanosine triphosphate hydrolases (GTPases), pointing to their historical role in driving our understanding of the molecular dynamics of this important class of signalling proteins and revealing the intricate relationships and regulatory mechanisms that comprise Rho GTPase biology in living cells.
Collapse
Affiliation(s)
- Colline Sanchez
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Andrea Ramirez
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Louis Hodgson
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
2
|
Fuchsberger T, Stockwell I, Woods M, Brzosko Z, Greger IH, Paulsen O. Dopamine increases protein synthesis in hippocampal neurons enabling dopamine-dependent LTP. eLife 2025; 13:RP100822. [PMID: 40063079 PMCID: PMC11893101 DOI: 10.7554/elife.100822] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2025] Open
Abstract
The reward and novelty-related neuromodulator dopamine plays an important role in hippocampal long-term memory, which is thought to involve protein-synthesis-dependent synaptic plasticity. However, the direct effects of dopamine on protein synthesis, and the functional implications of newly synthesised proteins for synaptic plasticity, have not yet been investigated. We have previously reported that timing-dependent synaptic depression (t-LTD) can be converted into potentiation by dopamine application during synaptic stimulation (Brzosko et al., 2015) or postsynaptic burst activation (Fuchsberger et al., 2022). Here, we show that dopamine increases protein synthesis in mouse hippocampal CA1 neurons, enabling dopamine-dependent long-term potentiation (DA-LTP), which is mediated via the Ca2+-sensitive adenylate cyclase (AC) subtypes 1/8, cAMP, and cAMP-dependent protein kinase (PKA). We found that neuronal activity is required for the dopamine-induced increase in protein synthesis. Furthermore, dopamine induced a protein-synthesis-dependent increase in the AMPA receptor subunit GluA1, but not GluA2. We found that DA-LTP is absent in GluA1 knock-out mice and that it requires calcium-permeable AMPA receptors. Taken together, our results suggest that dopamine together with neuronal activity controls synthesis of plasticity-related proteins, including GluA1, which enable DA-LTP via a signalling pathway distinct from that of conventional LTP.
Collapse
Affiliation(s)
- Tanja Fuchsberger
- Department of Physiology, Development and Neuroscience, Physiological Laboratory, University of CambridgeCambridgeUnited Kingdom
| | - Imogen Stockwell
- Neurobiology Division, MRC Laboratory of Molecular BiologyCambridgeUnited Kingdom
| | - Matty Woods
- Department of Physiology, Development and Neuroscience, Physiological Laboratory, University of CambridgeCambridgeUnited Kingdom
| | - Zuzanna Brzosko
- Department of Physiology, Development and Neuroscience, Physiological Laboratory, University of CambridgeCambridgeUnited Kingdom
| | - Ingo H Greger
- Neurobiology Division, MRC Laboratory of Molecular BiologyCambridgeUnited Kingdom
| | - Ole Paulsen
- Department of Physiology, Development and Neuroscience, Physiological Laboratory, University of CambridgeCambridgeUnited Kingdom
| |
Collapse
|
3
|
Shan-Ni L, Liang H, Yasui Y, Ninomiya K, Uehara T, Nishimura T, Kobayashi K. Leptin on the apical surface inhibits casein production and STAT5 phosphorylation in mammary epithelial cells. Exp Cell Res 2024; 443:114330. [PMID: 39536931 DOI: 10.1016/j.yexcr.2024.114330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 11/16/2024]
Abstract
Leptin is a peptide hormone present in both the blood and milk. A close relationship between leptin and milk production in lactating mammary glands has been previously reported. However, how leptin influences milk production in lactating mammary glands remains unclear. Also, whether leptin in milk or blood influences mammary epithelial cells (MECs) during lactation needs further investigation. This study investigated the effects of leptin on mouse MECs using a culture model in which MECs produced milk components and formed less permeable tight junctions. Our results showed that β-casein production in MEC was inhibited by leptin in a concentration-dependent manner. Leptin also inactivated the signal transducer and activator of transcription 5 (STAT5), a transcription factor that facilitates milk production in MECs. Leptin treatment induced the activation of p38 and c-Jun N-terminal kinase (JNK) in MEC before STAT5 inactivation, and anisomycin, an activator of p38 and JNK, induced the inactivation of STAT5. Furthermore, leptin exposure on the apical surface of MECs inhibited β-casein production and inactivated STAT5. However, leptin exposure on the basolateral surface hardly caused these effects. These findings suggested that milk leptin, but not plasma leptin, inhibited milk production in MECs.
Collapse
Affiliation(s)
- Lu Shan-Ni
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Hokkaido University, North 9, West 9, 060-8589, Sapporo, Japan.
| | - Han Liang
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Hokkaido University, North 9, West 9, 060-8589, Sapporo, Japan.
| | - Yuki Yasui
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Hokkaido University, North 9, West 9, 060-8589, Sapporo, Japan.
| | - Kazuki Ninomiya
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Hokkaido University, North 9, West 9, 060-8589, Sapporo, Japan.
| | - Tamaki Uehara
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Hokkaido University, North 9, West 9, 060-8589, Sapporo, Japan.
| | - Takanori Nishimura
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Hokkaido University, North 9, West 9, 060-8589, Sapporo, Japan.
| | - Ken Kobayashi
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Hokkaido University, North 9, West 9, 060-8589, Sapporo, Japan.
| |
Collapse
|
4
|
Isaac AR, Chauvet MG, Lima-Filho R, Wagner BDA, Caroli BG, Leite REP, Suemoto CK, Nunes PV, De Felice FG, Ferreira ST, Lourenco MV. Defective regulation of the eIF2-eIF2B translational axis underlies depressive-like behavior in mice and correlates with major depressive disorder in humans. Transl Psychiatry 2024; 14:397. [PMID: 39349438 PMCID: PMC11442801 DOI: 10.1038/s41398-024-03128-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 09/18/2024] [Accepted: 09/24/2024] [Indexed: 10/02/2024] Open
Abstract
Major depressive disorder (MDD) is a significant cause of disability in adults worldwide. However, the underlying causes and mechanisms of MDD are not fully understood, and many patients are refractory to available therapeutic options. Impaired control of brain mRNA translation underlies several neurodevelopmental and neurodegenerative conditions, including autism spectrum disorders and Alzheimer's disease (AD). Nonetheless, a potential role for mechanisms associated with impaired translational control in depressive-like behavior remains elusive. A key pathway controlling translation initiation relies on the phosphorylation of the α subunit of eukaryotic initiation factor 2 (eIF2α-P) which, in turn, blocks the guanine exchange factor activity of eIF2B, thereby reducing global translation rates. Here we report that the expression of EIF2B5 (which codes for eIF2Bε, the catalytic subunit of eIF2B) is reduced in postmortem MDD prefrontal cortex from two distinct human cohorts and in the frontal cortex of social isolation-induced depressive-like behavior model mice. Further, pharmacological treatment with anisomycin or with salubrinal, an inhibitor of the eIF2α phosphatase GADD34, induces depressive-like behavior in adult C57BL/6J mice. Salubrinal-induced depressive-like behavior is blocked by ISRIB, a compound that directly activates eIF2B regardless of the phosphorylation status of eIF2α, suggesting that increased eIF2α-P promotes depressive-like states. Taken together, our results suggest that impaired eIF2-associated translational control may participate in the pathophysiology of MDD, and underscore eIF2-eIF2B translational axis as a potential target for the development of novel approaches for MDD and related mood disorders.
Collapse
Affiliation(s)
- Alinny R Isaac
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Multidisciplinary Research Core in Biology (NUMPEX-BIO), Campus Duque de Caxias Professor Geraldo Cidade, Federal University of Rio de Janeiro, Duque de Caxias, RJ, Brazil
| | - Mariana G Chauvet
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Ricardo Lima-Filho
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Beatriz de A Wagner
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Bruno G Caroli
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Renata E P Leite
- Department of Pathology, University of São Paulo Medical School, São Paulo, SP, Brazil
| | - Claudia K Suemoto
- Division of Geriatrics, University of São Paulo Medical School, São Paulo, SP, Brazil
| | - Paula Villela Nunes
- Department of Psychiatry, University of São Paulo Medical School, São Paulo, SP, Brazil
| | - Fernanda G De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences & Department of Psychiatry, Queen's University, Kingston, ON, Canada
- D'Or Institute for Research and Education, Rio de Janeiro, RJ, Brazil
| | - Sergio T Ferreira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- D'Or Institute for Research and Education, Rio de Janeiro, RJ, Brazil
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Mychael V Lourenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
5
|
Sinha NK, McKenney C, Yeow ZY, Li JJ, Nam KH, Yaron-Barir TM, Johnson JL, Huntsman EM, Cantley LC, Ordureau A, Regot S, Green R. The ribotoxic stress response drives UV-mediated cell death. Cell 2024; 187:3652-3670.e40. [PMID: 38843833 PMCID: PMC11246228 DOI: 10.1016/j.cell.2024.05.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 03/03/2024] [Accepted: 05/09/2024] [Indexed: 06/13/2024]
Abstract
While ultraviolet (UV) radiation damages DNA, eliciting the DNA damage response (DDR), it also damages RNA, triggering transcriptome-wide ribosomal collisions and eliciting a ribotoxic stress response (RSR). However, the relative contributions, timing, and regulation of these pathways in determining cell fate is unclear. Here we use time-resolved phosphoproteomic, chemical-genetic, single-cell imaging, and biochemical approaches to create a chronological atlas of signaling events activated in cells responding to UV damage. We discover that UV-induced apoptosis is mediated by the RSR kinase ZAK and not through the DDR. We identify two negative-feedback modules that regulate ZAK-mediated apoptosis: (1) GCN2 activation limits ribosomal collisions and attenuates ZAK-mediated RSR and (2) ZAK activity leads to phosphodegron autophosphorylation and its subsequent degradation. These events tune ZAK's activity to collision levels to establish regimes of homeostasis, tolerance, and death, revealing its key role as the cellular sentinel for nucleic acid damage.
Collapse
Affiliation(s)
- Niladri K Sinha
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Connor McKenney
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Biochemistry, Cellular and Molecular Biology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Zhong Y Yeow
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jeffrey J Li
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ki Hong Nam
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Tomer M Yaron-Barir
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA; Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA; Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Jared L Johnson
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Emily M Huntsman
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
| | - Lewis C Cantley
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Alban Ordureau
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - Sergi Regot
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Rachel Green
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
6
|
Ke D, Xu H, Han J, Dai H, Wang X, Luo J, Yu Y, Xu J. Curcumin suppresses RANKL-induced osteoclast precursor autophagy in osteoclastogenesis by inhibiting RANK signaling and downstream JNK-BCL2-Beclin1 pathway. Biomed J 2024; 47:100605. [PMID: 37179010 PMCID: PMC10839592 DOI: 10.1016/j.bj.2023.100605] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 01/30/2023] [Accepted: 05/08/2023] [Indexed: 05/15/2023] Open
Abstract
BACKGROUND Curcumin ameliorates bone loss by inhibiting osteoclastogenesis. Curcumin inhibits RANKL-promoted autophagy in osteoclast precursors (OCPs), which mediates its anti-osteoclastogenic effect. But the role of RANKL signaling in curcumin-regulated OCP autophagy is unknown. This study aimed to explore the relationship between curcumin, RANKL signaling, and OCP autophagy during osteoclastogenesis. METHODS We investigated the role of curcumin in RANKL-related molecular signaling in OCPs, and identified the significance of RANK-TRAF6 signaling in curcumin-treated osteoclastogenesis and OCP autophagy using flow sorting and lentiviral transduction. Tg-hRANKL mice were used to observe the in vivo effects of curcumin on RANKL-regulated bone loss, osteoclastogenesis, and OCP autophagy. The significance of JNK-BCL2-Beclin1 pathway in curcumin-regulated OCP autophagy with RANKL was explored via rescue assays and BCL2 phosphorylation detection. RESULTS Curcumin inhibited RANKL-related molecular signaling in OCPs, and repressed osteoclast differentiation and autophagy in sorted RANK+ OCPs but did not affect those of RANK- OCPs. Curcumin-inhibited osteoclast differentiation and OCP autophagy were recovered by TRAF6 overexpression. But curcumin lost these effects under TRAF6 knockdown. Furthermore, curcumin prevented the decrease in bone mass and the increase in trabecular osteoclast formation and autophagy in RANK+ OCPs in Tg-hRANKL mice. Additionally, curcumin-inhibited OCP autophagy with RANKL was reversed by JNK activator anisomycin and TAT-Beclin1 overexpressing Beclin1. Curcumin inhibited BCL2 phosphorylation at Ser70 and enhanced protein interaction between BCL2 and Beclin1 in OCPs. CONCLUSIONS Curcumin suppresses RANKL-promoted OCP autophagy by inhibiting signaling pathway downstream of RANKL, contributing to its anti-osteoclastogenic effect. Moreover, JNK-BCL2-Beclin1 pathway plays an important role in curcumin-regulated OCP autophagy.
Collapse
Affiliation(s)
- Dianshan Ke
- Department of Orthopedics, Fujian Provincial Hospital, Fuzhou, Fujian, China; Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Haoying Xu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Junyong Han
- Institute for Immunology, Fujian Academy of Medical Sciences, Fuzhou, Fujian, China
| | - Hanhao Dai
- Department of Orthopedics, Fujian Provincial Hospital, Fuzhou, Fujian, China; Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Xinwen Wang
- Department of Orthopedics, Dongguan People's Hospital, Southern Medical University, Dongguan, Guangdong, China
| | - Jun Luo
- Department of Orthopedics, Fujian Provincial Hospital, Fuzhou, Fujian, China; Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Yunlong Yu
- Department of Orthopedics, Fujian Provincial Hospital, Fuzhou, Fujian, China; Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China.
| | - Jie Xu
- Department of Orthopedics, Fujian Provincial Hospital, Fuzhou, Fujian, China; Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China.
| |
Collapse
|
7
|
Menchinskaya ES, Dyshlovoy SA, Venz S, Jacobsen C, Hauschild J, Rohlfing T, Silchenko AS, Avilov SA, Balabanov S, Bokemeyer C, Aminin DL, von Amsberg G, Honecker F. Anticancer Activity of the Marine Triterpene Glycoside Cucumarioside A 2-2 in Human Prostate Cancer Cells. Mar Drugs 2023; 22:20. [PMID: 38248645 PMCID: PMC10817243 DOI: 10.3390/md22010020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/24/2023] [Accepted: 12/24/2023] [Indexed: 01/23/2024] Open
Abstract
Despite recent advances in the treatment of metastatic castration-resistant prostate cancer (CRPC), treatment is inevitably hampered by the development of drug resistance. Thus, new drugs are urgently needed. We investigated the efficacy, toxicity, and mechanism of action of the marine triterpene glycoside cucumarioside A2-2 (CA2-2) using an in vitro CRPC model. CA2-2 induced a G2/M-phase cell cycle arrest in human prostate cancer PC-3 cells and caspase-dependent apoptosis executed via an intrinsic pathway. Additionally, the drug inhibited the formation and growth of CRPC cell colonies at low micromolar concentrations. A global proteome analysis performed using the 2D-PAGE technique, followed by MALDI-MS and bioinformatical evaluation, revealed alterations in the proteins involved in cellular processes such as metastatic potential, invasion, and apoptosis. Among others, the regulation of keratin 81, CrkII, IL-1β, and cathepsin B could be identified by our proteomics approach. The effects were validated on the protein level by a 2D Western blotting analysis. Our results demonstrate the promising anticancer activity of CA2-2 in a prostate cancer model and provide insights on the underlying mode of action.
Collapse
Affiliation(s)
- Ekaterina S. Menchinskaya
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald Tumorzentrum—University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (C.J.); (J.H.); (T.R.); (C.B.); (G.v.A.); (F.H.)
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 159 Prospect 100-letiya Vladivostoka, Vladivostok 690022, Russia; (A.S.S.); (S.A.A.); (D.L.A.)
| | - Sergey A. Dyshlovoy
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald Tumorzentrum—University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (C.J.); (J.H.); (T.R.); (C.B.); (G.v.A.); (F.H.)
| | - Simone Venz
- Department of Medical Biochemistry and Molecular Biology, University of Greifswald, 17475 Greifswald, Germany;
| | - Christine Jacobsen
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald Tumorzentrum—University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (C.J.); (J.H.); (T.R.); (C.B.); (G.v.A.); (F.H.)
| | - Jessica Hauschild
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald Tumorzentrum—University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (C.J.); (J.H.); (T.R.); (C.B.); (G.v.A.); (F.H.)
| | - Tina Rohlfing
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald Tumorzentrum—University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (C.J.); (J.H.); (T.R.); (C.B.); (G.v.A.); (F.H.)
| | - Aleksandra S. Silchenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 159 Prospect 100-letiya Vladivostoka, Vladivostok 690022, Russia; (A.S.S.); (S.A.A.); (D.L.A.)
| | - Sergey A. Avilov
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 159 Prospect 100-letiya Vladivostoka, Vladivostok 690022, Russia; (A.S.S.); (S.A.A.); (D.L.A.)
| | - Stefan Balabanov
- Division of Hematology, University Hospital Zurich, 8091 Zurich, Switzerland;
| | - Carsten Bokemeyer
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald Tumorzentrum—University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (C.J.); (J.H.); (T.R.); (C.B.); (G.v.A.); (F.H.)
| | - Dmitry L. Aminin
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 159 Prospect 100-letiya Vladivostoka, Vladivostok 690022, Russia; (A.S.S.); (S.A.A.); (D.L.A.)
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, No. 100, Shin-Chuan 1st Road, Sanmin District, Kaohsiung City 80708, Taiwan
| | - Gunhild von Amsberg
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald Tumorzentrum—University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (C.J.); (J.H.); (T.R.); (C.B.); (G.v.A.); (F.H.)
- Martini-Klinik, Prostate Cancer Center, University Hospital Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Friedemann Honecker
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald Tumorzentrum—University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (C.J.); (J.H.); (T.R.); (C.B.); (G.v.A.); (F.H.)
- Tumor and Breast Center Eastern Switzerland, 9016 St. Gallen, Switzerland
| |
Collapse
|
8
|
Koirala R, Fongsaran C, Poston T, Rogge M, Rogers B, Thune R, Dubytska L. Edwardsiella ictaluri T3SS effector EseN is a phosphothreonine lyase that inactivates ERK1/2, p38, JNK, and PDK1 and modulates cell death in infected macrophages. Microbiol Spectr 2023; 11:e0300323. [PMID: 37796003 PMCID: PMC10714789 DOI: 10.1128/spectrum.03003-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 08/22/2023] [Indexed: 10/06/2023] Open
Abstract
IMPORTANCE This work has global significance in the catfish industry, which provides food for increasing global populations. E. ictaluri is a leading cause of disease loss, and EseN is an important player in E. ictaluri virulence. The E. ictaluri T3SS effector EseN plays an essential role in establishing infection, but the specific role EseN plays is not well characterized. EseN belongs to a family of phosphothreonine lyase effectors that specifically target host mitogen activated protein kinase (MAPK) pathways important in regulating host responses to infection. No phosphothreonine lyase equivalents are known in eukaryotes, making this family of effectors an attractive target for indirect narrow-spectrum antibiotics. Targeting of major vault protein and PDK1 kinase by EseN has not been reported in EseN homologs in other pathogens and may indicate unique functions of E. ictaluri EseN. EseN targeting of PDK1 is particularly interesting in that it is linked to an extraordinarily diverse group of cellular functions.
Collapse
Affiliation(s)
- Ranjan Koirala
- Department of Biological Sciences and Chemistry, Southern University and A & M College, Baton Rouge, Louisiana, USA
| | - Chanida Fongsaran
- Department of Biological Sciences and Chemistry, Southern University and A & M College, Baton Rouge, Louisiana, USA
| | - Tanisha Poston
- Department of Biological Sciences and Chemistry, Southern University and A & M College, Baton Rouge, Louisiana, USA
| | - Matthew Rogge
- Department of Biology, University of Wisconsin-Stevens Point, Stevens Point, Wisconsin, USA
| | - Bryan Rogers
- Department of Biological Sciences and Chemistry, Southern University and A & M College, Baton Rouge, Louisiana, USA
| | - Ronald Thune
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, USA
| | - Lidiya Dubytska
- Department of Biological Sciences and Chemistry, Southern University and A & M College, Baton Rouge, Louisiana, USA
| |
Collapse
|
9
|
Yu J, Li X, Cao J, Zhu T, Liang S, Du L, Cao M, Wang H, Zhang Y, Zhou Y, Shen B, Feng J, Zhang J, Wang J, Jin J. Components of the JNK-MAPK pathway play distinct roles in hepatocellular carcinoma. J Cancer Res Clin Oncol 2023; 149:17495-17509. [PMID: 37902853 DOI: 10.1007/s00432-023-05473-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/10/2023] [Indexed: 11/01/2023]
Abstract
PURPOSE Mitogen-activated protein kinases (MAPK), specifically the c-Jun N-terminal kinase (JNK)-MAPK subfamily, play a crucial role in the development of various cancers, including hepatocellular carcinoma (HCC). However, the specific roles of JNK1/2 and their upstream regulators, MKK4/7, in HCC carcinogenesis remain unclear. METHODS In this study, we performed differential expression analysis of JNK-MAPK components at both the transcriptome and protein levels using TCGA and HPA databases. We utilized Kaplan-Meier survival plots and receiver operating characteristic (ROC) curve analysis to evaluate the prognostic performance of a risk scoring model based on these components in the TCGA-HCC cohort. Additionally, we conducted immunoblotting, apoptosis analysis with FACS and soft agar assays to investigate the response of JNK-MAPK pathway components to various death stimuli (TRAIL, TNF-α, anisomycin, and etoposide) in HCC cell lines. RESULTS JNK1/2 and MKK7 levels were significantly upregulated in HCC samples compared to paracarcinoma tissues, whereas MKK4 was downregulated. ROC analyses suggested that JNK2 and MKK7 may serve as suitable diagnostic genes for HCC, and high JNK2 expression correlated with significantly poorer overall survival. Knockdown of JNK1 enhanced TRAIL-induced apoptosis in hepatoma cells, while JNK2 knockdown reduced TNF-α/cycloheximide (CHX)-and anisomycin-induced apoptosis. Neither JNK1 nor JNK2 knockdown affected etoposide-induced apoptosis. Furthermore, MKK7 knockdown augmented TNF-α/CHX- and TRAIL-induced apoptosis and inhibited colony formation in hepatoma cells. CONCLUSION Targeting MKK7, rather than JNK1/2 or MKK4, may be a promising therapeutic strategy to inhibit the JNK-MAPK pathway in HCC therapy.
Collapse
Affiliation(s)
- Jijun Yu
- School of Basic Medicine, Hainan Medical University, Haikou, 571199, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Xinying Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Junxia Cao
- Department of Molecular Immunology, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Ting Zhu
- Beijing No. 80 High School, Beijing, 100102, China
| | - Shuifeng Liang
- School of Basic Medicine, Hainan Medical University, Haikou, 571199, China
| | - Le Du
- School of Basic Medicine, Hainan Medical University, Haikou, 571199, China
| | - Meng Cao
- School of Basic Medicine, Hainan Medical University, Haikou, 571199, China
| | - Haitao Wang
- Department of Hematology, The Fifth Medical Center of Chinese, PLA General Hospital, Beijing, 100071, China
| | - Yaolin Zhang
- Department of Molecular Immunology, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Yinxi Zhou
- School of Basic Medicine, Hainan Medical University, Haikou, 571199, China
| | - Beifen Shen
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
- Department of Molecular Immunology, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Jiannan Feng
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Jiyan Zhang
- Department of Molecular Immunology, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China.
| | - Jing Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.
| | - Jianfeng Jin
- School of Basic Medicine, Hainan Medical University, Haikou, 571199, China.
| |
Collapse
|
10
|
Rajabalee N, Siushansian H, Weerapura M, Berton S, Berbatovci F, Hooks B, Geoffrion M, Yang D, Harper ME, Rayner K, Blais A, Sun J. ATF2 orchestrates macrophage differentiation and activation to promote antibacterial responses. J Leukoc Biol 2023; 114:280-298. [PMID: 37403209 DOI: 10.1093/jleuko/qiad076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 04/22/2023] [Accepted: 06/15/2023] [Indexed: 07/06/2023] Open
Abstract
The differentiation and activation of macrophages are critical regulatory programs that are central to host inflammation and pathogen defense. However, the transcriptional regulatory pathways involved in these programs are not well understood. Herein, we demonstrate that the activity and expression of the transcription factor ATF2 is precisely regulated during primary human monocyte-to-macrophage differentiation and that its activation is linked to M1 polarization and antibacterial responses. Genetic perturbation experiments demonstrated that deletion of ATF2 (THP-ΔATF2) resulted in irregular and abnormal macrophage morphology, whereas macrophages overexpressing ATF2 (THP-ATF2) developed round and pancake-like morphology, resembling classically activated (M1) macrophages. Mechanistically, we show that ATF2 binds to the core promoter of PPM1A, a phosphatase that regulates monocyte-to-macrophage differentiation, to regulate its expression. Functionally, overexpression of ATF2 sensitized macrophages to M1 polarization, resulting in increased production of major histocompatibility complex class II, IL-1β, and IP-10; improved phagocytic capacity; and enhanced control of the intracellular pathogen Mycobacterium tuberculosis. Gene expression profiling revealed that overexpression of ATF2 reprogramed macrophages to promote antibacterial pathways enriched in chemokine signaling, metabolism, and antigen presentation. Consistent with pathways analysis, metabolic profiling revealed that genetic overexpression or stimuli-induced activation of ATF2 alters the metabolic capacity of macrophages and primes these cells for glycolytic metabolism during M1 polarization or bacterial infection. Our findings reveal that ATF2 plays a central role during macrophage differentiation and M1 polarization to enhance the functional capacities of macrophages.
Collapse
Affiliation(s)
- Nusrah Rajabalee
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| | - Hannah Siushansian
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| | - Milani Weerapura
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| | - Stefania Berton
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| | - Fjolla Berbatovci
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| | - Breana Hooks
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| | - Michele Geoffrion
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa Heart Institute, 40 Ruskin Road, Ottawa, Ontario K1Y 4W7, Canada
| | - Dabo Yang
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| | - Mary-Ellen Harper
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| | - Katey Rayner
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa Heart Institute, 40 Ruskin Road, Ottawa, Ontario K1Y 4W7, Canada
| | - Alexandre Blais
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
- Éric Poulin Centre for Neuromuscular Disease, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| | - Jim Sun
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| |
Collapse
|
11
|
Melamed Kadosh D, Beenstock J, Engelberg D, Admon A. Differential Modulation of the Phosphoproteome by the MAP Kinases Isoforms p38α and p38β. Int J Mol Sci 2023; 24:12442. [PMID: 37569817 PMCID: PMC10419006 DOI: 10.3390/ijms241512442] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/29/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
The p38 members of the mitogen-activated protein kinases (MAPKs) family mediate various cellular responses to stress conditions, inflammatory signals, and differentiation factors. They are constitutively active in chronic inflammatory diseases and some cancers. The differences between their transient effects in response to signals and the chronic effect in diseases are not known. The family is composed of four isoforms, of which p38α seems to be abnormally activated in diseases. p38α and p38β are almost identical in sequence, structure, and biochemical and pharmacological properties, and the specific unique effects of each of them, if any, have not yet been revealed. This study aimed to reveal the specific effects induced by p38α and p38β, both when transiently activated in response to stress and when chronically active. This was achieved via large-scale proteomics and phosphoproteomics analyses using stable isotope labeling of two experimental systems: one, mouse embryonic fibroblasts (MEFs) deficient in each of these p38 kinases and harboring either an empty vector or vectors expressing p38αWT, p38βWT, or intrinsically active variants of these MAPKs; second, induction of transient stress by exposure of MEFs, p38α-/-, and p38β-/- MEFs to anisomycin. Significant differences in the repertoire of the proteome and phosphoproteome between cells expressing active p38α and p38β suggest distinct roles for each kinase. Interestingly, in both cases, the constitutive activation induced adaptations of the cells to the chronic activity so that known substrates of p38 were downregulated. Within the dramatic effect of p38s on the proteome and phosphoproteome, some interesting affected phosphorylation sites were those found in cancer-associated p53 and Hspb1 (HSP27) proteins and in cytoskeleton-associated proteins. Among these, was the stronger direct phosphorylation by p38α of p53-Ser309, which was validated on the Ser315 in human p53. In summary, this study sheds new light on the differences between chronic and transient p38α and p38β signaling and on the specific targets of these two kinases.
Collapse
Affiliation(s)
| | - Jonah Beenstock
- Department of Biological Chemistry, Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel;
| | - David Engelberg
- Department of Biological Chemistry, Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel;
- Singapore-HUJ Alliance for Research and Enterprise, Mechanisms of Liver Inflammatory Diseases Program, National University of Singapore, Singapore 138602, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
| | - Arie Admon
- Faculty of Biology, Technion—Israel Institute of Technology, Haifa 3200003, Israel;
| |
Collapse
|
12
|
Yin K, Tong M, Suttapitugsakul S, Xu S, Wu R. Global quantification of newly synthesized proteins reveals cell type- and inhibitor-specific effects on protein synthesis inhibition. PNAS NEXUS 2023; 2:pgad168. [PMID: 37275259 PMCID: PMC10235912 DOI: 10.1093/pnasnexus/pgad168] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/05/2023] [Accepted: 05/15/2023] [Indexed: 06/07/2023]
Abstract
Manipulation of protein synthesis is commonly applied to uncover protein functions and cellular activities. Multiple inhibitors with distinct mechanisms have been widely investigated and employed in bio-related research, but it is extraordinarily challenging to measure and evaluate the synthesis inhibition efficiencies of individual proteins by different inhibitors at the proteome level. Newly synthesized proteins are the immediate and direct products of protein synthesis, and thus their comprehensive quantification provides a unique opportunity to study protein inhibition. Here, we systematically investigate protein inhibition and evaluate different popular inhibitors, i.e. cycloheximide, puromycin, and anisomycin, through global quantification of newly synthesized proteins in several types of human cells (A549, MCF-7, Jurkat, and THP-1 cells). The inhibition efficiencies of protein synthesis are comprehensively measured by integrating azidohomoalanine-based protein labeling, selective enrichment, a boosting approach, and multiplexed proteomics. The same inhibitor results in dramatic variation of the synthesis inhibition efficiencies for different proteins in the same cells, and each inhibitor exhibits unique preferences. Besides cell type- and inhibitor-specific effects, some universal rules are unraveled. For instance, nucleolar and ribosomal proteins have relatively higher inhibition efficiencies in every type of cells treated with each inhibitor. Moreover, proteins intrinsically resistant or sensitive to the inhibition are identified and found to have distinct functions. Systematic investigation of protein synthesis inhibition in several types of human cells by different inhibitors provides valuable information about the inhibition of protein synthesis, advancing our understanding of inhibiting protein synthesis.
Collapse
Affiliation(s)
| | | | - Suttipong Suttapitugsakul
- School of Chemistry and Biochemistry and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Senhan Xu
- School of Chemistry and Biochemistry and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Ronghu Wu
- To whom correspondence should be addressed:
| |
Collapse
|
13
|
Ke X, Yu S, Situ S, Lin Z, Yuan Y. Morroniside inhibits Beclin1-dependent autophagic death and Bax-dependent apoptosis in cardiomyocytes through repressing BCL2 phosphorylation. In Vitro Cell Dev Biol Anim 2023:10.1007/s11626-023-00768-0. [PMID: 37155079 DOI: 10.1007/s11626-023-00768-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 04/13/2023] [Indexed: 05/10/2023]
Abstract
Morroniside can prevent myocardial injury caused by ischemia and hypoxia, which can be used to treat acute myocardial infarction (AMI). Hypoxia can cause apoptosis and autophagic death of cardiomyocytes. Morroniside has the ability to inhibit apoptosis and autophagy. However, the relationship between Morroniside-protected cardiomyocytes and two forms of death is unclear. The effects of Morroniside on the proliferation, apoptosis level, and autophagic activity of rat cardiomyocyte line H9c2 under hypoxia were first observed. Next, the roles of Morroniside in the phosphorylation of JNK and BCL2, BCL2-Beclin1, and BCL2-Bax complexes as well as mitochondrial membrane potential in H9c2 cells were evaluated upon hypoxia. Finally, the significance of BCL2 or JNK in Morroniside-regulated autophagy, apoptosis, and proliferation in H9c2 cells was assessed by combining Morroniside and BCL2 competitive inhibitor (ABT-737) or JNK activator (Anisomycin). Our results showed that hypoxia promoted autophagy and apoptosis of H9c2 cells, and inhibited their proliferation. However, Morroniside could block the effect of hypoxia on H9c2 cells. In addition, Morroniside could inhibit JNK phosphorylation, BCL2 phosphorylation at the Ser70 and Ser87 sites, and the dissociation of BCL2-Beclin1 and BCL2-Bax complexes in H9c2 cells upon hypoxia. Moreover, the reduction of mitochondrial membrane potential in H9c2 cells caused by hypoxia was improved by Morroniside administration. Importantly, the inhibited autophagy, apoptosis, and promoted proliferation in H9c2 cells by Morroniside were reversed by the application of ABT-737 or Anisomycin. Overall, Morroniside inhibits Beclin1-dependent autophagic death and Bax-dependent apoptosis via JNK-mediated BCL2 phosphorylation, thereby improving the survival of cardiomyocytes under hypoxia.
Collapse
Affiliation(s)
- Xueping Ke
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
- The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, 528200, China
| | - Shicheng Yu
- Department of Medicine, Liwan Central Hospital of Guangzhou, Guangzhou, 510145, China
| | - Shubiao Situ
- Department of Medicine, Liwan Central Hospital of Guangzhou, Guangzhou, 510145, China
| | - Zhenqian Lin
- Department of Cardiology, Henan Provincial Chest Hospital, Zhengzhou, 450008, Henan, China
| | - Yiqiang Yuan
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China.
- Department of Cardiology, Zheng Zhou NO.7 People's Hospital, No.17, Jingnan 5th Road, Zhengzhou Economic and Technological Development Zone, Zhengzhou, 450016, China.
| |
Collapse
|
14
|
Martina JA, Jeong E, Puertollano R. p38 MAPK-dependent phosphorylation of TFEB promotes monocyte-to-macrophage differentiation. EMBO Rep 2023; 24:e55472. [PMID: 36507874 PMCID: PMC9900348 DOI: 10.15252/embr.202255472] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 11/14/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022] Open
Abstract
The transcription factor EB (TFEB) regulates energy homeostasis and cellular response to a wide variety of stress conditions, including nutrient deprivation, oxidative stress, organelle damage, and pathogens. Here we identify S401 as a novel phosphorylation site within the TFEB proline-rich domain. Phosphorylation of S401 increases significantly in response to oxidative stress, UVC light, growth factors, and LPS, whereas this increase is prevented by p38 MAPK inhibition or depletion, revealing a new role for p38 MAPK in TFEB regulation. Mutation of S401 in THP1 cells demonstrates that the p38 MAPK/TFEB pathway plays a particularly relevant role during monocyte differentiation into macrophages. TFEB-S401A monocytes fail to upregulate the expression of multiple immune genes in response to PMA-induced differentiation, including critical cytokines, chemokines, and growth factors. Polarization of M0 macrophages into M1 inflammatory macrophages is also aberrant in TFEB-S401A cells. These results indicate that TFEB-S401 phosphorylation links differentiation signals to the transcriptional control of monocyte differentiation.
Collapse
Affiliation(s)
- José A Martina
- Cell and Developmental Biology CenterNational Heart, Lung, and Blood Institute, National Institutes of HealthBethesdaMDUSA
| | - Eutteum Jeong
- Cell and Developmental Biology CenterNational Heart, Lung, and Blood Institute, National Institutes of HealthBethesdaMDUSA
| | - Rosa Puertollano
- Cell and Developmental Biology CenterNational Heart, Lung, and Blood Institute, National Institutes of HealthBethesdaMDUSA
| |
Collapse
|
15
|
Cohen LD, Ziv T, Ziv NE. Synapse integrity and function: Dependence on protein synthesis and identification of potential failure points. Front Mol Neurosci 2022; 15:1038614. [PMID: 36583084 PMCID: PMC9792512 DOI: 10.3389/fnmol.2022.1038614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/07/2022] [Indexed: 12/14/2022] Open
Abstract
Synaptic integrity and function depend on myriad proteins - labile molecules with finite lifetimes that need to be continually replaced with freshly synthesized copies. Here we describe experiments designed to expose synaptic (and neuronal) properties and functions that are particularly sensitive to disruptions in protein supply, identify proteins lost early upon such disruptions, and uncover potential, yet currently underappreciated failure points. We report here that acute suppressions of protein synthesis are followed within hours by reductions in spontaneous network activity levels, impaired oxidative phosphorylation and mitochondrial function, and, importantly, destabilization and loss of both excitatory and inhibitory postsynaptic specializations. Conversely, gross impairments in presynaptic vesicle recycling occur over longer time scales (days), as does overt cell death. Proteomic analysis identified groups of potentially essential 'early-lost' proteins including regulators of synapse stability, proteins related to bioenergetics, fatty acid and lipid metabolism, and, unexpectedly, numerous proteins involved in Alzheimer's disease pathology and amyloid beta processing. Collectively, these findings point to neuronal excitability, energy supply and synaptic stability as early-occurring failure points under conditions of compromised supply of newly synthesized protein copies.
Collapse
Affiliation(s)
- Laurie D. Cohen
- Technion Faculty of Medicine, Rappaport Institute and Network Biology Research Laboratories, Haifa, Israel
| | - Tamar Ziv
- Smoler Proteomics Center, Lokey Interdisciplinary Center for Life Sciences & Engineering, Technion, Haifa, Israel
| | - Noam E. Ziv
- Technion Faculty of Medicine, Rappaport Institute and Network Biology Research Laboratories, Haifa, Israel,*Correspondence: Noam E. Ziv,
| |
Collapse
|
16
|
Kobayashi K, Wakasa H, Han L, Koyama T, Tsugami Y, Nishimura T. Lactose on the basolateral side of mammary epithelial cells inhibits milk production concomitantly with signal transducer and activator of transcription 5 inactivation. Cell Tissue Res 2022; 389:501-515. [PMID: 35748981 DOI: 10.1007/s00441-022-03651-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 06/09/2022] [Indexed: 11/29/2022]
Abstract
Mammary epithelial cells (MECs) are the only cells capable of synthesizing lactose. During lactation, alveolar MECs secrete lactose through the apical membrane into the alveolar lumen, whereas alveolar tight junctions (TJs) block the leakage of lactose into the basolateral sides of the MECs. However, lactose leaks from the alveolar lumen into the blood plasma in the mastitis and after weaning. This exposes the basolateral membrane of MECs to lactose. The relationship between lactose in blood plasma and milk production has been suggested. The present study determined whether lactose exposure on the basolateral membrane of mouse MECs adversely affects milk production in vitro. Restricted exposure to lactose on the basolateral side of the MECs was performed using a culture model, in which MECs on the cell culture insert exhibit milk production and less-permeable TJs. The results indicated that lactose exposure on the basolateral side inhibited casein and lipid production in the MECs. Interestingly, lactose exposure on the apical side did not show detectable effects on milk production in the MECs. Basolateral lactose exposure also caused the inactivation of STAT5, a primary transcriptional factor for milk production. Furthermore, p38 and JNK were activated by basolateral lactose exposure. The activation of p38 and JNK following anisomycin treatment reduced phosphorylated STAT5, and inhibitors of p38 blocked the reduction of phosphorylated STAT5 by basolateral lactose exposure. These findings suggest that lactose functions as a partial inhibitor for milk production but only when it directly makes contact with the basolateral membrane of MECs.
Collapse
Affiliation(s)
- Ken Kobayashi
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Hokkaido University, North 9, West 9, Sapporo, 060-8589, Japan.
| | - Haruka Wakasa
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Hokkaido University, North 9, West 9, Sapporo, 060-8589, Japan
| | - Liang Han
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Hokkaido University, North 9, West 9, Sapporo, 060-8589, Japan
| | - Taku Koyama
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Hokkaido University, North 9, West 9, Sapporo, 060-8589, Japan
| | - Yusaku Tsugami
- Laboratory of Animal Histophysiology, Graduate School of Integrated Science for Life Faculty of Applied Biological Science, Hiroshima University, 1-4-4Higashi-Hiroshima, Kagamiyama, 739-8528, Japan
| | - Takanori Nishimura
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Hokkaido University, North 9, West 9, Sapporo, 060-8589, Japan
| |
Collapse
|
17
|
Franklin JL, Amsler MO, Messina JL. Regulation of glucose responsive protein (GRP) gene expression by insulin. Cell Stress Chaperones 2022; 27:27-35. [PMID: 34755306 PMCID: PMC8821767 DOI: 10.1007/s12192-021-01243-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 09/08/2021] [Accepted: 10/15/2021] [Indexed: 11/28/2022] Open
Abstract
While screening for insulin-induced genes, we identified two members of a family of stress-induced genes referred to as glucose-regulated proteins (GRPs). GRPs are members of the stress-responsive superfamily of genes which also includes heat shock proteins (HSPs). The GRP proteins are not normally heat-inducible, but are overproduced when cells are starved of glucose. The two major GRP proteins, GRP78 and GRP94, are highly conserved among vertebrates. We have found that physiological concentrations of insulin stimulate the transcription of GRP78 and GRP94 in rat H4IIE hepatoma cells. The regulation of GRP78 transcription was rapid, with the first induction within minutes, and a further induction after several hours, and both occurred in the presence of glucose. GRP78 transcription was more greatly induced by insulin in the presence of SB202190, a specific p38-MAPK inhibitor. Transcription of GRP94 was also induced, but only after several hours. Calcimycin (A23187) and anisomycin were used to induce endoplasmic reticulum (ER)/cellular stress, and both induced GRP78 and GRP94 transcription.
Collapse
Affiliation(s)
- J Lee Franklin
- Department of Pathology, Division of Pathobiology and Molecular Medicine, University of Alabama at Birmingham, 1670 University Blvd., Volker Hall G019, Birmingham, AL, 35294-0019, USA
- Veterans Administration Medical Center, Birmingham, AL, 35294, USA
| | - Margaret O Amsler
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Joseph L Messina
- Department of Pathology, Division of Pathobiology and Molecular Medicine, University of Alabama at Birmingham, 1670 University Blvd., Volker Hall G019, Birmingham, AL, 35294-0019, USA.
- Veterans Administration Medical Center, Birmingham, AL, 35294, USA.
| |
Collapse
|
18
|
Kwong AJ, Pham TND, Oelschlager HE, Munshi HG, Scheidt KA. Rational Design, Optimization, and Biological Evaluation of Novel MEK4 Inhibitors against Pancreatic Adenocarcinoma. ACS Med Chem Lett 2021; 12:1559-1567. [PMID: 34676038 DOI: 10.1021/acsmedchemlett.1c00376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Indexed: 01/05/2023] Open
Abstract
Growth, division, and development of healthy cells relies on efficient response to environmental survival cues. The conserved mitogen-activated protein kinase (MAPK) family of pathways interface extracellular stimuli to intracellular processes for this purpose. Within these pathways, the MEK family has been identified as a target of interest due to its clinical relevance. Particularly, MEK4 has drawn recent attention for its indications in pancreatic and prostate cancers. Here, we report two potent MEK4 inhibitors demonstrating significant reduction of phospho-JNK and antiproliferative properties against pancreatic cancer cell lines. Furthermore, molecular inhibition of MEK4 pathway activates the MEK1/2 pathway, with the combination of MEK1/2 and MEK4 inhibitors demonstrating synergistic effects against pancreatic cancer cells. Our inhibitors provided insight into the crosstalk between MAPK pathways and new tools for elucidating the roles of MEK4 in disease states, findings which will pave the way for better understanding of the MAPK pathways and development of additional probes.
Collapse
Affiliation(s)
- Ada J. Kwong
- Department of Chemistry, Department of Pharmacology, Feinberg School of Medicine, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Thao N. D. Pham
- Department of Medicine, Feinberg School of Medicine, Northwestern University, 303 E. Superior Street, Chicago, Illinois 60611, United States
| | - Hannah E. Oelschlager
- Department of Chemistry, Department of Pharmacology, Feinberg School of Medicine, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Hidayatullah G. Munshi
- Department of Medicine, Feinberg School of Medicine, Northwestern University, 303 E. Superior Street, Chicago, Illinois 60611, United States
| | - Karl A. Scheidt
- Department of Chemistry, Department of Pharmacology, Feinberg School of Medicine, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| |
Collapse
|
19
|
Kočović DM, Bajuk-Bogdanović D, Pećinar I, Nedeljković BB, Daković M, Andjus PR. Assessment of cellular and molecular changes in the rat brain after gamma radiation and radioprotection by anisomycin. JOURNAL OF RADIATION RESEARCH 2021; 62:793-803. [PMID: 34062561 PMCID: PMC8438266 DOI: 10.1093/jrr/rrab045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 03/31/2021] [Indexed: 06/12/2023]
Abstract
The objective of the study was to describe cellular and molecular markers of radioprotection by anisomycin, focusing on the changes in rat brain tissue. Two-month-old Wistar rats were exposed to a 60Co radiation source at a dose of 6 Gy, with or without radioprotection with anisomycin (150 mg/kg) administered subcutaneously 30 min before or 3 or 6 h after irradiation. Survivors were analyzed 30 days after treatment. Astroglial and microglial responses were investigated based on the expression of glial markers assessed with immunohistochemistry, and quantitative changes in brain biomolecules were investigated by Raman microspectroscopy. In addition, blood plasma levels of pro-inflammatory (interleukin 6 and tumor necrosis factor α) and anti-inflammatory (interleukin 10) cytokines were assessed. We found that application of anisomycin either before or after irradiation significantly decreased the expression of the microglial marker Iba-1. We also found an increased intensity of Raman spectral bands related to nucleic acids, as well as an increased level of cytokines when anisomycin was applied after irradiation. This suggests that the radioprotective effects of anisomycin are by decreasing Iba-1 expression and stabilizing genetic material by increasing the level of nucleic acids.
Collapse
Affiliation(s)
- Dušica M Kočović
- Center for Laser Microscopy, Faculty of Biology, University of Belgrade, Studentski Trg 3, 11 000 Belgrade, Serbia
| | - Danica Bajuk-Bogdanović
- Faculty of Physical Chemistry, University of Belgrade, Studentski Trg 12-16, 11 000 Belgrade, Serbia
| | - Ilinka Pećinar
- Faculty of Agriculture, Department for Agrobotany, University of Belgrade, Nemanjina 6, 11 080 Belgrade, Serbia
| | - Biljana Božić Nedeljković
- Institute for Physiology and Biochemistry ``Jean Giaja'', Faculty of Biology, University of Belgrade, Studentski Trg 3, 11 000 Belgrade, Serbia
| | - Marko Daković
- Faculty of Physical Chemistry, University of Belgrade, Studentski Trg 12-16, 11 000 Belgrade, Serbia
| | - Pavle R Andjus
- Center for Laser Microscopy, Faculty of Biology, University of Belgrade, Studentski Trg 3, 11 000 Belgrade, Serbia
| |
Collapse
|
20
|
Carriba P, Davies AM. Signalling Pathways Mediating the Effects of CD40-Activated CD40L Reverse Signalling on Inhibitory Medium Spiny Neuron Neurite Growth. Cells 2021; 10:829. [PMID: 33917019 PMCID: PMC8067729 DOI: 10.3390/cells10040829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/15/2021] [Accepted: 04/02/2021] [Indexed: 01/16/2023] Open
Abstract
CD40-activated CD40L-mediated reverse signalling is a major physiological regulator of neurite growth from excitatory and inhibitory neurons in the developing central nervous system (CNS). Whereas in excitatory pyramidal neurons, CD40L reverse signalling promotes the growth and elaboration of dendrites and axons, in inhibitory GABAergic striatal medium spiny neurons (MSNs), it restricts neurite growth and branching. In pyramidal neurons, we previously reported that CD40L reverse signalling activates an interconnected and interdependent signalling network involving protein kinase C (PKC), extracellular regulated kinases 1 and 2 (ERK1/2), and c-Jun N-terminal kinase (JNK) signalling pathways that regulates dendrite and axon growth. Here, we have studied whether these signalling pathways also influence neurite growth from striatal inhibitory MSNs. To unequivocally activate CD40L reverse signalling, we treated MSN cultures from CD40-deficient mice with CD40-Fc. Here, we report that activation of CD40L reverse signalling in these cultures also increased the phosphorylation of PKC, ERK1/2, and JNK. Using pharmacological activators and inhibitors of these signalling pathways singularly and in combination, we have shown that, as in pyramidal neurons, these signalling pathways work in an interconnected and interdependent network to regulate the neurite growth, but their functions, relationships, and interdependencies are different from those observed in pyramidal neurons. Furthermore, immunoprecipitation studies showed that stimulation of CD40L reverse signalling recruits the catalytic fragment of Syk tyrosine kinase, but in contrast to pyramidal neurons, PKC does not participate in this recruitment. Our findings show that distinctive networks of three signalling pathways mediate the opposite effects of CD40L reverse signalling on neurite growth in excitatory and inhibitory neurons.
Collapse
Affiliation(s)
- Paulina Carriba
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK
| | - Alun M Davies
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK
| |
Collapse
|
21
|
Banik I, Cheng PF, Dooley CM, Travnickova J, Merteroglu M, Dummer R, Patton EE, Busch-Nentwich EM, Levesque MP. NRAS Q61K melanoma tumor formation is reduced by p38-MAPK14 activation in zebrafish models and NRAS-mutated human melanoma cells. Pigment Cell Melanoma Res 2020; 34:150-162. [PMID: 32910840 DOI: 10.1111/pcmr.12925] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/02/2020] [Accepted: 09/02/2020] [Indexed: 01/05/2023]
Abstract
Oncogenic BRAF and NRAS mutations drive human melanoma initiation. We used transgenic zebrafish to model NRAS-mutant melanoma, and the rapid tumor onset allowed us to study candidate tumor suppressors. We identified P38α-MAPK14 as a potential tumor suppressor in The Cancer Genome Atlas melanoma cohort of NRAS-mutant melanomas, and overexpression significantly increased the time to tumor onset in transgenic zebrafish with NRAS-driven melanoma. Pharmacological activation of P38α-MAPK14 using anisomycin reduced in vitro viability of melanoma cultures, which we confirmed by stable overexpression of p38α. We observed that the viability of MEK inhibitor resistant melanoma cells could be reduced by combined treatment of anisomycin and MEK inhibition. Our study demonstrates that activating the p38α-MAPK14 pathway in the presence of oncogenic NRAS abrogates melanoma in vitro and in vivo. SIGNIFICANCE: The significance of our study is in the accountability of NRAS mutations in melanoma. We demonstrate here that activation of p38α-MAPK14 pathway can abrogate NRAS-mutant melanoma which is contrary to the previously published role of p38α-MAPK14 pathway in BRAF mutant melanoma. These results implicate that BRAF and NRAS-mutant melanoma may not be identical biologically. We also demonstrate the translational benefit of our study by using a small molecule compound-anisomycin (already in use for other diseases in clinical trials) to activate p38α-MAPK14 pathway.
Collapse
Affiliation(s)
- Ishani Banik
- University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Phil F Cheng
- University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Christopher M Dooley
- Wellcome Sanger Institute, Hinxton, Cambridge, UK.,Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Jana Travnickova
- MRC Human Genetics Unit and Cancer Research, UK Edinburgh Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Munise Merteroglu
- Wellcome Sanger Institute, Hinxton, Cambridge, UK.,Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge, UK
| | - Reinhard Dummer
- University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Elizabeth E Patton
- MRC Human Genetics Unit and Cancer Research, UK Edinburgh Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Elisabeth M Busch-Nentwich
- Wellcome Sanger Institute, Hinxton, Cambridge, UK.,Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
22
|
Carriba P, Davies AM. How CD40L reverse signaling regulates axon and dendrite growth. Cell Mol Life Sci 2020; 78:1065-1083. [PMID: 32506167 PMCID: PMC7897621 DOI: 10.1007/s00018-020-03563-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 05/01/2020] [Accepted: 05/25/2020] [Indexed: 12/18/2022]
Abstract
CD40-activated CD40L reverse signaling is a major physiological regulator of axon and dendrite growth from developing hippocampal pyramidal neurons. Here we have studied how CD40L-mediated reverse signaling promotes the growth of these processes. Cultures of hippocampal pyramidal neurons were established from Cd40-/- mouse embryos to eliminate endogenous CD40/CD40L signaling, and CD40L reverse signaling was stimulated by a CD40-Fc chimera. CD40L reverse signaling increased phosphorylation and hence activation of proteins in the PKC, ERK, and JNK signaling pathways. Pharmacological activators and inhibitors of these pathways revealed that whereas activation of JNK inhibited growth, activation of PKC and ERK1/ERK2 enhanced growth. Experiments using combinations of pharmacological reagents revealed that these signaling pathways regulate growth by functioning as an interconnected and interdependent network rather than acting in a simple linear sequence. Immunoprecipitation studies suggested that stimulation of CD40L reverse signaling generated a receptor complex comprising CD40L, PKCβ, and the Syk tyrosine kinase. Our studies have begun to elucidate the molecular network and interactions that promote axon and dendrite growth from developing hippocampal neurons following activation of CD40L reverse signaling.
Collapse
Affiliation(s)
- Paulina Carriba
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AX, Wales.
| | - Alun M Davies
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AX, Wales
| |
Collapse
|
23
|
Wei H, Ren Z, Tang L, Yao H, Li X, Wang C, Mu C, Shi C, Wang H. JNK signaling pathway regulates the development of ovaries and synthesis of vitellogenin (Vg) in the swimming crab Portunus trituberculatus. Cell Stress Chaperones 2020; 25:441-453. [PMID: 32172493 PMCID: PMC7193009 DOI: 10.1007/s12192-020-01085-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 03/02/2020] [Accepted: 03/04/2020] [Indexed: 12/16/2022] Open
Abstract
The development of Portunus trituberculatus egg cells is directly related to the nutritional status of the fertilized egg, which affects the key production stages of offspring hatching. Vitellogenin plays a key role in the nutrient supply required for the development of the egg cells. The c-Jun N-terminal kinase (JNK) is an important member of the mitogen-activated protein kinase (MAPK) superfamily and plays an important role in cell proliferation, transformation, differentiation, and apoptosis. At present, there are no reports on the involvement of the JNK signaling pathway in the reproductive regulation of P. trituberculatus. In this study, rapid amplification of complementary DNA ends amplification technology was used to clone the full length of JNK complementary DNA, which has a length of 2094 bp, including an open reading frame (ORF) of 1266 bp encoding a 421-amino acid protein. The protein includes the S_TKC conserved domain with a TPY phosphorylation site, which is a typical feature of the JNK gene family. Observing tissue sections found the oocytes in the inhibitor group developed slowly, while the oocytes in the activated group showed accelerated development. Meanwhile, Portunus trituberculatus JNK and vitellogenin (Vg) genes exhibited the same trend in the hepatopancreas and ovaries, and the expression of the SP600125 group was downregulated (P < 0.05), while the anisomycin group was upregulated (P < 0.05). In addition, JNK enzyme activity and vitellin (Vn) content in the ovarian tissue showed that the JNK activity of the SP600125 group decreased, while activity increased in the anisomycin group. The accumulation of Vn content in the SP600125 group decreased, and that in the anisomycin group increased. In summary, after injection with inhibitor or activator, the JNK signaling pathway of P. trituberculatus was inhibited or activated, the accumulation of Vn in the ovary was reduced or increased, and ovarian development was inhibited or accelerated, respectively. These results indicated that the JNK signaling pathway is involved in the regulation of Vg synthesis and ovarian development in P. trituberculatus. The results of this study further add to the knowledge of the breeding biology of P. trituberculatus and provide a theoretical reference for the optimization of breeding techniques in aquaculture production systems.
Collapse
Affiliation(s)
- Hongling Wei
- School of Marine Science, Ningbo University, Ningbo, 315211 Zhejiang China
| | - Zhiming Ren
- School of Marine Science, Ningbo University, Ningbo, 315211 Zhejiang China
| | - Lei Tang
- School of Marine Science, Ningbo University, Ningbo, 315211 Zhejiang China
| | - Hongzhi Yao
- School of Marine Science, Ningbo University, Ningbo, 315211 Zhejiang China
| | - Xing Li
- School of Marine Science, Ningbo University, Ningbo, 315211 Zhejiang China
| | - Chunlin Wang
- School of Marine Science, Ningbo University, Ningbo, 315211 Zhejiang China
- Key Laboratory of Applied Marine Biotechnology, Ministry of Education, Ningbo University, Ningbo, 315211 Zhejiang China
| | - Changkao Mu
- School of Marine Science, Ningbo University, Ningbo, 315211 Zhejiang China
- Key Laboratory of Applied Marine Biotechnology, Ministry of Education, Ningbo University, Ningbo, 315211 Zhejiang China
| | - Ce Shi
- School of Marine Science, Ningbo University, Ningbo, 315211 Zhejiang China
- Key Laboratory of Applied Marine Biotechnology, Ministry of Education, Ningbo University, Ningbo, 315211 Zhejiang China
| | - Huan Wang
- School of Marine Science, Ningbo University, Ningbo, 315211 Zhejiang China
- Key Laboratory of Applied Marine Biotechnology, Ministry of Education, Ningbo University, Ningbo, 315211 Zhejiang China
| |
Collapse
|
24
|
Yoon J, Cho Y, Kim KY, Yoon MJ, Lee HS, Jeon SD, Cho Y, Kim C, Kim MG. A JUN N-terminal kinase inhibitor induces ectodomain shedding of the cancer-associated membrane protease Prss14/epithin via protein kinase CβII. J Biol Chem 2020; 295:7168-7177. [PMID: 32241917 PMCID: PMC7242708 DOI: 10.1074/jbc.ra119.011206] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 03/04/2020] [Indexed: 12/28/2022] Open
Abstract
Serine protease 14 (Prss14)/epithin is a transmembrane serine protease that plays essential roles in tumor progression and metastasis and therefore is a promising target for managing cancer. Prss14/epithin shedding may underlie its activity in cancer and worsen outcomes; accordingly, a detailed understanding of the molecular mechanisms in Prss14/epithin shedding may inform the design of future cancer therapies. On the basis of our previous observation that an activator of PKC, phorbol 12-myristate 13-acetate (PMA), induces Prss14/epithin shedding, here we further investigated the intracellular signaling pathway involved in this process. While using mitogen-activated protein kinase inhibitors to investigate possible effectors of downstream PKC signaling, we unexpectedly found that an inhibitor of c-Jun N-terminal kinase (JNK), SP600125, induces Prss14/epithin shedding even in the absence of PMA. SP600125-induced shedding, like that stimulated by PMA, was mediated by tumor necrosis factor-α–converting enzyme. In contrast, a JNK activator, anisomycin, partially abolished the effects of SP600125 on Prss14/epithin shedding. Moreover, the results from loss-of-function experiments with specific inhibitors, short hairpin RNA–mediated knockdown, and overexpression of dominant-negative PKCβII variants indicated that PKCβII is a major player in JNK inhibition– and PMA-mediated Prss14/epithin shedding. SP600125 increased phosphorylation of PKCβII and tumor necrosis factor-α–converting enzyme and induced their translocation into the plasma membrane. Finally, in vitro cell invasion experiments and bioinformatics analysis of data in The Cancer Genome Atlas breast cancer database revealed that JNK and PKCβII are important for Prss14/epithin-mediated cancer progression. These results provide important information regarding strategies against tumor metastasis.
Collapse
Affiliation(s)
- Joobyoung Yoon
- School of Biological Sciences, Seoul National University, Seoul 08826, Korea
| | - Youngkyung Cho
- School of Biological Sciences, Seoul National University, Seoul 08826, Korea.,Department of Life Sciences, Korea University, Seoul 02841, Korea
| | - Ki Yeon Kim
- Department of Biological Sciences, Inha University, Incheon 22212, Korea
| | - Min Ji Yoon
- Department of Life Sciences, Korea University, Seoul 02841, Korea
| | - Hyo Seon Lee
- School of Biological Sciences, Seoul National University, Seoul 08826, Korea
| | - Sangjun Davie Jeon
- Department of Biological Sciences, Inha University, Incheon 22212, Korea
| | - Yongcheol Cho
- Department of Life Sciences, Korea University, Seoul 02841, Korea
| | - Chungho Kim
- Department of Life Sciences, Korea University, Seoul 02841, Korea
| | - Moon Gyo Kim
- Department of Biological Sciences, Inha University, Incheon 22212, Korea
| |
Collapse
|
25
|
Mechanisms Linking Mitochondrial Dysfunction and Proteostasis Failure. Trends Cell Biol 2020; 30:317-328. [PMID: 32200806 DOI: 10.1016/j.tcb.2020.01.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 01/16/2020] [Accepted: 01/17/2020] [Indexed: 12/19/2022]
Abstract
Maintaining cellular protein homeostasis (proteostasis) is an essential task for all eukaryotes. Proteostasis failure worsens with aging and is considered a cause of and a therapeutic target for age-related diseases including neurodegenerative disorders. The cellular networks regulating proteostasis and the pathogenic events driving proteostasis failure in disease remain poorly understood. Model organism studies in yeast and Drosophila reveal an intriguing link between mitochondrial function and proteostasis. In this review we examine recent findings on mitochondrial outer membrane (MOM)-associated mRNA translation, how this process is sensitive to mitochondrial dysfunction and constantly surveyed by ribosome-associated quality control (RQC), and how defects in this process generate aberrant proteins with unusual C-terminal extensions (CTEs) that promote aggregation and drive proteostasis failure. We also discuss the implications for human diseases.
Collapse
|
26
|
Liu JC, Granieri L, Shrestha M, Wang DY, Vorobieva I, Rubie EA, Jones R, Ju Y, Pellecchia G, Jiang Z, Palmerini CA, Ben-David Y, Egan SE, Woodgett JR, Bader GD, Datti A, Zacksenhaus E. Identification of CDC25 as a Common Therapeutic Target for Triple-Negative Breast Cancer. Cell Rep 2019; 23:112-126. [PMID: 29617654 PMCID: PMC9357459 DOI: 10.1016/j.celrep.2018.03.039] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 01/31/2018] [Accepted: 03/11/2018] [Indexed: 12/18/2022] Open
Abstract
CDK4/6 inhibitors are effective against cancer cells expressing the tumor suppressor RB1, but not RB1-deficient cells, posing the challenge of how to target RB1 loss. In triple-negative breast cancer (TNBC), RB1 and PTEN are frequently inactivated together with TP53. We performed kinome/phosphatase inhibitor screens on primary mouse Rb/p53-, Pten/p53-, and human RB1/PTEN/TP53-deficient TNBC cell lines and identified CDC25 phosphatase as a common target. Pharmacological or genetic inhibition of CDC25 suppressed growth of RB1-deficient TNBC cells that are resistant to combined CDK4/6 plus CDK2 inhibition. Minimal cooperation was observed in vitro between CDC25 antagonists and CDK1, CDK2, or CDK4/6 inhibitors, but strong synergy with WEE1 inhibition was apparent. In accordance with increased PI3K signaling following long-term CDC25 inhibition, CDC25 and PI3K inhibitors effectively synergized to suppress TNBC growth both in vitro and in xenotransplantation models. These results provide a rationale for the development of CDC25-based therapies for diverse RB1/PTEN/TP53-deficient and -proficient TNBCs. Liu et al. report that inhibition of the protein phosphatase CDC25 kills diverse triple-negative breast cancer (TNBC) cells. Moreover, CDC25 antagonists cooperate with other drugs, such as PI3K inhibitors, to efficiently suppress growth of human TNBC engrafted into mice.
Collapse
Affiliation(s)
- Jeff C Liu
- Toronto General Research Institute - University Health Network, 67 College Street, Toronto, ON, Canada M5G 2M1
| | - Letizia Granieri
- Toronto General Research Institute - University Health Network, 67 College Street, Toronto, ON, Canada M5G 2M1; Department of Agriculture, Food, and Environmental Sciences, University of Perugia, Perugia, Italy
| | - Mariusz Shrestha
- Toronto General Research Institute - University Health Network, 67 College Street, Toronto, ON, Canada M5G 2M1; Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Dong-Yu Wang
- Toronto General Research Institute - University Health Network, 67 College Street, Toronto, ON, Canada M5G 2M1
| | - Ioulia Vorobieva
- Toronto General Research Institute - University Health Network, 67 College Street, Toronto, ON, Canada M5G 2M1; Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Elizabeth A Rubie
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, 600 University Avenue, Toronto, ON, Canada
| | - Rob Jones
- Toronto General Research Institute - University Health Network, 67 College Street, Toronto, ON, Canada M5G 2M1
| | - YoungJun Ju
- Toronto General Research Institute - University Health Network, 67 College Street, Toronto, ON, Canada M5G 2M1
| | - Giovanna Pellecchia
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada; The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON, Canada
| | - Zhe Jiang
- Toronto General Research Institute - University Health Network, 67 College Street, Toronto, ON, Canada M5G 2M1
| | - Carlo A Palmerini
- Department of Agriculture, Food, and Environmental Sciences, University of Perugia, Perugia, Italy
| | - Yaacov Ben-David
- The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang, Guizhou 550014, China; State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Sean E Egan
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Program in Cell Biology, The Peter Gilgan Center for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - James R Woodgett
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, 600 University Avenue, Toronto, ON, Canada
| | - Gary D Bader
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Alessandro Datti
- Department of Agriculture, Food, and Environmental Sciences, University of Perugia, Perugia, Italy; Network Biology Collaborative Centre, SMART Laboratory for High-Throughput Screening Programs, Mount Sinai Hospital, Toronto, ON, Canada
| | - Eldad Zacksenhaus
- Toronto General Research Institute - University Health Network, 67 College Street, Toronto, ON, Canada M5G 2M1; Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada; Department of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
27
|
Ke D, Ji L, Wang Y, Fu X, Chen J, Wang F, Zhao D, Xue Y, Lan X, Hou J. JNK1 regulates RANKL-induced osteoclastogenesis via activation of a novel Bcl-2-Beclin1-autophagy pathway. FASEB J 2019; 33:11082-11095. [PMID: 31295022 DOI: 10.1096/fj.201802597rr] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
JNK1 plays an important role in osteoclastogenesis in response to the osteoclastogenic cytokine receptor activator for nuclear factor-κB ligand (RANKL). JNK1 is widely accepted as an autophagy regulator under stress conditions. However, the role of JNK1-mediated autophagy in osteoclastogenesis remains largely unknown. In the current study, our data showed that JNK1 inhibition by a pharmacological inhibitor or RNA interference significantly reduced the autophagic response induced by RANKL in osteoclast precursors (OCPs) derived from bone marrow-derived macrophages. Overexpression of the key autophagy protein Beclin1 rescued autophagy deficiency and osteoclastogenesis in the presence of a JNK inhibitor (SP600125). In contrast, JNK activator (anisomycin)-induced autophagy was blocked by Beclin1 knockdown in OCPs. In addition, JNK1 inhibition increased apoptosis and blocked autophagy, whereas overexpression of Beclin1 reversed the enhanced apoptosis induced by JNK1 inhibition in OCPs. Furthermore, RANKL could induce the phosphorylation of Bcl-2, subsequently dissociating Beclin1 from the Bcl-2-Beclin1 complex, which could be blocked by JNK1 inhibition. Collectively, this study revealed that JNK1 regulated osteoclastogenesis by activating Bcl-2-Beclin1-autophagy signaling in addition to the classic c-Jun/activator protein 1 pathway, which provided the first evidence for the contribution of JNK1 signaling to OCP autophagy and the autophagic mechanism underlying JNK1-regulated osteoclastogenesis. An important osteoclastogenesis-regulating signaling pathway (JNK1-Bcl-2-Beclin1-autophagy activation) was identified, which provides novel potential targets for the clinical therapy of metabolic bone diseases.-Ke, D., Ji, L., Wang, Y., Fu, X., Chen, J., Wang, F., Zhao, D., Xue, Y., Lan, X., Hou, J. JNK1 regulates RANKL-induced osteoclastogenesis via activation of a novel Bcl-2-Beclin1-autophagy pathway.
Collapse
Affiliation(s)
- Dianshan Ke
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China.,Academy of Orthopedics in Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Lianmei Ji
- Department of Rheumatology and immunology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yu Wang
- Department of Orthopaedics, Chifeng Hospital, Chifeng, China
| | - Xiaomin Fu
- Division of Metabolism and Endocrinology, Pediatrics Department, John Hopkins University, Baltimore, Maryland, USA
| | - Jinyan Chen
- Fujian Academy of Medical Sciences, Institute for Immunology, Fuzhou, China
| | - Fan Wang
- Fujian Academy of Medical Sciences, Institute for Immunology, Fuzhou, China
| | - Dongbao Zhao
- Department of Rheumatology and immunology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Ying Xue
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China.,Department of Endocrinology, Fujian Provincial Hospital Key Laboratory of Endocrinology, Fuzhou, China
| | - Xuhua Lan
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China.,Department of Endocrinology, Fujian Provincial Hospital Key Laboratory of Endocrinology, Fuzhou, China
| | - Jianming Hou
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China.,Department of Endocrinology, Fujian Provincial Hospital Key Laboratory of Endocrinology, Fuzhou, China
| |
Collapse
|
28
|
Osorio-Gómez D, Saldivar-Mares KS, Perera-López A, McGaugh JL, Bermúdez-Rattoni F. Early memory consolidation window enables drug induced state-dependent memory. Neuropharmacology 2018; 146:84-94. [PMID: 30485798 DOI: 10.1016/j.neuropharm.2018.11.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 11/20/2018] [Accepted: 11/21/2018] [Indexed: 12/24/2022]
Abstract
It is well established that newly acquired information is stabilized over time by processes underlying memory consolidation, these events can be impaired by many drug treatments administered shortly after learning. The consolidation hypothesis has been challenged by a memory integration hypothesis, which suggests that the processes underlying new memories are vulnerable to incorporation of the neurobiological alterations induced by amnesic drugs generating a state-dependent memory. The present experiments investigated the effects of amnesic drugs infused into the insular cortex of male Wistar rats on memory for object recognition training. The findings provide evidence that infusions of several amnesic agents including a protein synthesis inhibitor, an RNA synthesis inhibitor, or an NMDA receptor antagonist administered both after a specific period of time and before retrieval induce state-dependent recognition memory. Additionally, when amnesic drugs were infused outside the early consolidation window, there was amnesia, but the amnesia was not state-dependent. Data suggest that amnesic agents can induce state-dependent memory when administered during the early consolidation window and only if the duration of the drug effect is long enough to become integrated to the memory trace. In consequence, there are boundary conditions in order to induce state-dependent memory.
Collapse
Affiliation(s)
- Daniel Osorio-Gómez
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, 04510, Mexico City, Mexico.
| | - Karina S Saldivar-Mares
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, 04510, Mexico City, Mexico
| | - Aldo Perera-López
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, 04510, Mexico City, Mexico
| | - James L McGaugh
- Department of Neurobiology and Behavior, Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA, 92697, USA
| | - Federico Bermúdez-Rattoni
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, 04510, Mexico City, Mexico
| |
Collapse
|
29
|
Astakhova AA, Chistyakov DV, Sergeeva MG, Reiser G. Regulation of the ARE-binding proteins, TTP (tristetraprolin) and HuR (human antigen R), in inflammatory response in astrocytes. Neurochem Int 2018; 118:82-90. [DOI: 10.1016/j.neuint.2018.04.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 04/20/2018] [Accepted: 04/23/2018] [Indexed: 01/06/2023]
|
30
|
Schöttelndreier D, Seeger K, Grassl GA, Winny MR, Lindner R, Genth H. Expression and (Lacking) Internalization of the Cell Surface Receptors of Clostridioides difficile Toxin B. Front Microbiol 2018; 9:1483. [PMID: 30022975 PMCID: PMC6039548 DOI: 10.3389/fmicb.2018.01483] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 06/14/2018] [Indexed: 12/30/2022] Open
Abstract
Toxin-producing strains of Clostridioides difficile and Clostridium perfringens cause infections of the gastrointestinal tract in humans and ruminants, with the toxins being major virulence factors, essential for the infection, and responsible for the onset of severe symptoms. C. difficile toxin A (TcdA) and toxin B (TcdB), and the large cytotoxin (TpeL) from C. perfringens are single chain bacterial protein toxins with an AB-like toxin structure. The C-terminal delivery domain mediates cell entry of the N-terminal glycosyltransferase domain by receptor-mediated endocytosis. Several cell surface proteins have been proposed to serve as toxin receptors, including chondroitin-sulfate proteoglycan 4 (CSPG4), poliovirus receptor-like 3 (PVRL3), and frizzled-1/2/7 (FZD1/2/7) for TcdB and LDL-receptor-related protein-1 (LRP1) for TpeL. The expression of the TcdB receptors was investigated in human intestinal organoids (HIOs) and in cultured cell lines. HIOs from four human donors exhibited a comparable profile of receptor expression, with PVRL3, LRP1, and FZD7 being expressed and CSPG4 and FZD2 not being expressed. In human epithelial Caco-2 cells and HT29 cells as well as in immortalized murine fibroblasts, either receptor FZD2/7, CSPG4, PVRL3, and LRP1 was expressed. The question whether the toxins take advantage of the normal turnover of their receptors (i.e., constitutive endocytosis and recycling) from the cell surface or whether the toxins activity induce the internalization of their receptors has not yet been addressed. For the analysis of receptor internalization, temperature-induced uptake of biotinylated toxin receptors into immortalized mouse embryonic fibroblasts (MEFs) and Caco-2 cells was exploited. Solely LRP1 exhibited constitutive endocytosis from the plasma membrane to the endosome, which might be abused by TpeL (and possibly TcdB as well) for cell entry. Furthermore, internalization of CSPG4, PVRL3, FZD2, and FZD7 was observed neither in MEFs nor in Caco-2 cells. FZD2/7, CSPG4, and PVRL3 did thus exhibit no constitutive recycling. The presence of TcdB and the p38 activation induced by anisomycin were not able to induce or enhance CSPG4 or PVRL3 uptake in MEFs. In conclusion, FZD2/7, CSPG4, and PVRL3 seem to serve as cell surface binding receptors rather than internalizing receptors of TcdB.
Collapse
Affiliation(s)
| | - Katrin Seeger
- Institute of Medical Microbiology and Hospital Epidemiology and DZIF Partner Site Hannover-Braunschweig, Hannover Medical School, Hanover, Germany
| | - Guntram A Grassl
- Institute of Medical Microbiology and Hospital Epidemiology and DZIF Partner Site Hannover-Braunschweig, Hannover Medical School, Hanover, Germany
| | - Markus R Winny
- Department of General, Visceral and Transplantation Surgery, Hannover Medical School, Hanover, Germany
| | - Robert Lindner
- Neuroanatomy and Cell Biology, Hannover Medical School, Hanover, Germany
| | - Harald Genth
- Institute for Toxicology, Hannover Medical School, Hanover, Germany
| |
Collapse
|
31
|
Liao JC, Wei ZX, Zhao C, Ma ZP, Cai DZ. Inhibition of osteoclastogenesis for periprosthetic osteolysis therapy through the suppression of p38 signaling by fraxetin. Int J Mol Med 2018; 42:1257-1264. [PMID: 29786751 PMCID: PMC6089765 DOI: 10.3892/ijmm.2018.3698] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Accepted: 05/14/2018] [Indexed: 01/13/2023] Open
Abstract
Periprosthetic osteolysis belongs to osteolytic diseases, which often occur due to an imbalance between osteoclast and osteoblast number or activity. Fraxetin, a natural plant extract, inhibits osteoblast apoptosis and has therapeutic potential for treating osteolytic diseases. However, data pertaining to the effects of fraxetin on osteoclasts are limited. In the present study, it was demonstrated that the inhibition of osteoclastogenesis by fraxetin had an important role on the therapy of titanium particle-induced osteolysis in vivo. In addition, fraxetin was demonstrated to suppress receptor activator of nuclear factor-κB ligand (RANKL)-mediated osteoclast differentiation and bone resorption in vitro in a dose-dependent manner. Fraxetin inhibited osteoclast differentiation and function through the suppression of p38 signaling and subsequently, the suppression of osteoclast-specific gene expression, including tartrate-resistant acid phosphatase, nuclear factor of activated T-cells, cytoplasmic 1, and cathepsin K. In conclusion, fraxetin administration may have potential as a treatment method for periprosthetic osteolysis and other osteolytic diseases.
Collapse
Affiliation(s)
- Jia-Cheng Liao
- Department of Orthopedics, Southern Medical University Affiliated People's Hospital of Longhua District Shenzhen, Shenzhen, Guangdong 518109, P.R. China
| | - Zhao-Xia Wei
- Department of Neurology, Liwan Hospital of The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510000, P.R. China
| | - Chang Zhao
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, P.R. China
| | - Zhong-Ping Ma
- Department of Orthopedics, The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 510030, P.R. China
| | - Dao-Zhang Cai
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, P.R. China
| |
Collapse
|
32
|
Yan J, Zhao W, Thomson JK, Gao X, DeMarco DM, Carrillo E, Chen B, Wu X, Ginsburg KS, Bakhos M, Bers DM, Anderson ME, Song LS, Fill M, Ai X. Stress Signaling JNK2 Crosstalk With CaMKII Underlies Enhanced Atrial Arrhythmogenesis. Circ Res 2018; 122:821-835. [PMID: 29352041 DOI: 10.1161/circresaha.117.312536] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
RATIONALE Atrial fibrillation (AF) is the most common arrhythmia, and advanced age is an inevitable and predominant AF risk factor. However, the mechanisms that couple aging and AF propensity remain unclear, making targeted therapeutic interventions unattainable. OBJECTIVE To explore the functional role of an important stress response JNK (c-Jun N-terminal kinase) in sarcoplasmic reticulum Ca2+ handling and consequently Ca2+-mediated atrial arrhythmias. METHODS AND RESULTS We used a series of cutting-edge electrophysiological and molecular techniques, exploited the power of transgenic mouse models to detail the molecular mechanism, and verified its clinical applicability in parallel studies on donor human hearts. We discovered that significantly increased activity of the stress response kinase JNK2 (JNK isoform 2) in the aged atria is involved in arrhythmic remodeling. The JNK-driven atrial proarrhythmic mechanism is supported by a pathway linking JNK, CaMKII (Ca2+/calmodulin-dependent kinase II), and sarcoplasmic reticulum Ca2+ release RyR2 (ryanodine receptor) channels. JNK2 activates CaMKII, a critical proarrhythmic molecule in cardiac muscle. In turn, activated CaMKII upregulates diastolic sarcoplasmic reticulum Ca2+ leak mediated by RyR2 channels. This leads to aberrant intracellular Ca2+ waves and enhanced AF propensity. In contrast, this mechanism is absent in young atria. In JNK challenged animal models, this is eliminated by JNK2 ablation or CaMKII inhibition. CONCLUSIONS We have identified JNK2-driven CaMKII activation as a novel mode of kinase crosstalk and a causal factor in atrial arrhythmic remodeling, making JNK2 a compelling new therapeutic target for AF prevention and treatment.
Collapse
Affiliation(s)
- Jiajie Yan
- From the Department of Physiology and Biophysics, Rush University, Chicago, IL (J.Y., W.Z., D.M.D., E.C., M.F., X.A.); Department of Cell and Molecular Physiology (J.Y., W.Z., J.K.T., X.G., D.M.D., E.C., X.W., X.A.) and Department of Thoracic and Cardiovascular Surgery (M.B.), Loyola University Chicago, Maywood, IL; Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City (B.C., L.-S.S.); Department of Pharmacology, University of California at Davis (K.S.G., D.M.B.); and Department of Internal Medicine, Johns Hopkins University, Baltimore, MD (M.E.A.)
| | - Weiwei Zhao
- From the Department of Physiology and Biophysics, Rush University, Chicago, IL (J.Y., W.Z., D.M.D., E.C., M.F., X.A.); Department of Cell and Molecular Physiology (J.Y., W.Z., J.K.T., X.G., D.M.D., E.C., X.W., X.A.) and Department of Thoracic and Cardiovascular Surgery (M.B.), Loyola University Chicago, Maywood, IL; Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City (B.C., L.-S.S.); Department of Pharmacology, University of California at Davis (K.S.G., D.M.B.); and Department of Internal Medicine, Johns Hopkins University, Baltimore, MD (M.E.A.)
| | - Justin K Thomson
- From the Department of Physiology and Biophysics, Rush University, Chicago, IL (J.Y., W.Z., D.M.D., E.C., M.F., X.A.); Department of Cell and Molecular Physiology (J.Y., W.Z., J.K.T., X.G., D.M.D., E.C., X.W., X.A.) and Department of Thoracic and Cardiovascular Surgery (M.B.), Loyola University Chicago, Maywood, IL; Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City (B.C., L.-S.S.); Department of Pharmacology, University of California at Davis (K.S.G., D.M.B.); and Department of Internal Medicine, Johns Hopkins University, Baltimore, MD (M.E.A.)
| | - Xianlong Gao
- From the Department of Physiology and Biophysics, Rush University, Chicago, IL (J.Y., W.Z., D.M.D., E.C., M.F., X.A.); Department of Cell and Molecular Physiology (J.Y., W.Z., J.K.T., X.G., D.M.D., E.C., X.W., X.A.) and Department of Thoracic and Cardiovascular Surgery (M.B.), Loyola University Chicago, Maywood, IL; Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City (B.C., L.-S.S.); Department of Pharmacology, University of California at Davis (K.S.G., D.M.B.); and Department of Internal Medicine, Johns Hopkins University, Baltimore, MD (M.E.A.)
| | - Dominic M DeMarco
- From the Department of Physiology and Biophysics, Rush University, Chicago, IL (J.Y., W.Z., D.M.D., E.C., M.F., X.A.); Department of Cell and Molecular Physiology (J.Y., W.Z., J.K.T., X.G., D.M.D., E.C., X.W., X.A.) and Department of Thoracic and Cardiovascular Surgery (M.B.), Loyola University Chicago, Maywood, IL; Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City (B.C., L.-S.S.); Department of Pharmacology, University of California at Davis (K.S.G., D.M.B.); and Department of Internal Medicine, Johns Hopkins University, Baltimore, MD (M.E.A.)
| | - Elena Carrillo
- From the Department of Physiology and Biophysics, Rush University, Chicago, IL (J.Y., W.Z., D.M.D., E.C., M.F., X.A.); Department of Cell and Molecular Physiology (J.Y., W.Z., J.K.T., X.G., D.M.D., E.C., X.W., X.A.) and Department of Thoracic and Cardiovascular Surgery (M.B.), Loyola University Chicago, Maywood, IL; Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City (B.C., L.-S.S.); Department of Pharmacology, University of California at Davis (K.S.G., D.M.B.); and Department of Internal Medicine, Johns Hopkins University, Baltimore, MD (M.E.A.)
| | - Biyi Chen
- From the Department of Physiology and Biophysics, Rush University, Chicago, IL (J.Y., W.Z., D.M.D., E.C., M.F., X.A.); Department of Cell and Molecular Physiology (J.Y., W.Z., J.K.T., X.G., D.M.D., E.C., X.W., X.A.) and Department of Thoracic and Cardiovascular Surgery (M.B.), Loyola University Chicago, Maywood, IL; Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City (B.C., L.-S.S.); Department of Pharmacology, University of California at Davis (K.S.G., D.M.B.); and Department of Internal Medicine, Johns Hopkins University, Baltimore, MD (M.E.A.)
| | - Xiaomin Wu
- From the Department of Physiology and Biophysics, Rush University, Chicago, IL (J.Y., W.Z., D.M.D., E.C., M.F., X.A.); Department of Cell and Molecular Physiology (J.Y., W.Z., J.K.T., X.G., D.M.D., E.C., X.W., X.A.) and Department of Thoracic and Cardiovascular Surgery (M.B.), Loyola University Chicago, Maywood, IL; Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City (B.C., L.-S.S.); Department of Pharmacology, University of California at Davis (K.S.G., D.M.B.); and Department of Internal Medicine, Johns Hopkins University, Baltimore, MD (M.E.A.)
| | - Kenneth S Ginsburg
- From the Department of Physiology and Biophysics, Rush University, Chicago, IL (J.Y., W.Z., D.M.D., E.C., M.F., X.A.); Department of Cell and Molecular Physiology (J.Y., W.Z., J.K.T., X.G., D.M.D., E.C., X.W., X.A.) and Department of Thoracic and Cardiovascular Surgery (M.B.), Loyola University Chicago, Maywood, IL; Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City (B.C., L.-S.S.); Department of Pharmacology, University of California at Davis (K.S.G., D.M.B.); and Department of Internal Medicine, Johns Hopkins University, Baltimore, MD (M.E.A.)
| | - Mamdouh Bakhos
- From the Department of Physiology and Biophysics, Rush University, Chicago, IL (J.Y., W.Z., D.M.D., E.C., M.F., X.A.); Department of Cell and Molecular Physiology (J.Y., W.Z., J.K.T., X.G., D.M.D., E.C., X.W., X.A.) and Department of Thoracic and Cardiovascular Surgery (M.B.), Loyola University Chicago, Maywood, IL; Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City (B.C., L.-S.S.); Department of Pharmacology, University of California at Davis (K.S.G., D.M.B.); and Department of Internal Medicine, Johns Hopkins University, Baltimore, MD (M.E.A.)
| | - Donald M Bers
- From the Department of Physiology and Biophysics, Rush University, Chicago, IL (J.Y., W.Z., D.M.D., E.C., M.F., X.A.); Department of Cell and Molecular Physiology (J.Y., W.Z., J.K.T., X.G., D.M.D., E.C., X.W., X.A.) and Department of Thoracic and Cardiovascular Surgery (M.B.), Loyola University Chicago, Maywood, IL; Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City (B.C., L.-S.S.); Department of Pharmacology, University of California at Davis (K.S.G., D.M.B.); and Department of Internal Medicine, Johns Hopkins University, Baltimore, MD (M.E.A.)
| | - Mark E Anderson
- From the Department of Physiology and Biophysics, Rush University, Chicago, IL (J.Y., W.Z., D.M.D., E.C., M.F., X.A.); Department of Cell and Molecular Physiology (J.Y., W.Z., J.K.T., X.G., D.M.D., E.C., X.W., X.A.) and Department of Thoracic and Cardiovascular Surgery (M.B.), Loyola University Chicago, Maywood, IL; Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City (B.C., L.-S.S.); Department of Pharmacology, University of California at Davis (K.S.G., D.M.B.); and Department of Internal Medicine, Johns Hopkins University, Baltimore, MD (M.E.A.)
| | - Long-Sheng Song
- From the Department of Physiology and Biophysics, Rush University, Chicago, IL (J.Y., W.Z., D.M.D., E.C., M.F., X.A.); Department of Cell and Molecular Physiology (J.Y., W.Z., J.K.T., X.G., D.M.D., E.C., X.W., X.A.) and Department of Thoracic and Cardiovascular Surgery (M.B.), Loyola University Chicago, Maywood, IL; Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City (B.C., L.-S.S.); Department of Pharmacology, University of California at Davis (K.S.G., D.M.B.); and Department of Internal Medicine, Johns Hopkins University, Baltimore, MD (M.E.A.)
| | - Michael Fill
- From the Department of Physiology and Biophysics, Rush University, Chicago, IL (J.Y., W.Z., D.M.D., E.C., M.F., X.A.); Department of Cell and Molecular Physiology (J.Y., W.Z., J.K.T., X.G., D.M.D., E.C., X.W., X.A.) and Department of Thoracic and Cardiovascular Surgery (M.B.), Loyola University Chicago, Maywood, IL; Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City (B.C., L.-S.S.); Department of Pharmacology, University of California at Davis (K.S.G., D.M.B.); and Department of Internal Medicine, Johns Hopkins University, Baltimore, MD (M.E.A.)
| | - Xun Ai
- From the Department of Physiology and Biophysics, Rush University, Chicago, IL (J.Y., W.Z., D.M.D., E.C., M.F., X.A.); Department of Cell and Molecular Physiology (J.Y., W.Z., J.K.T., X.G., D.M.D., E.C., X.W., X.A.) and Department of Thoracic and Cardiovascular Surgery (M.B.), Loyola University Chicago, Maywood, IL; Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City (B.C., L.-S.S.); Department of Pharmacology, University of California at Davis (K.S.G., D.M.B.); and Department of Internal Medicine, Johns Hopkins University, Baltimore, MD (M.E.A.).
| |
Collapse
|
33
|
Schipp R, Varga J, Bátor J, Vecsernyés M, Árvai Z, Pap M, Szeberényi J. Partial p53-dependence of anisomycin-induced apoptosis in PC12 cells. Mol Cell Biochem 2017; 434:41-50. [PMID: 28432551 DOI: 10.1007/s11010-017-3035-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 04/12/2017] [Indexed: 01/25/2023]
Abstract
The bacterial antibiotic anisomycin is known to induce apoptosis by activating several mitogen-activated protein kinases and by inhibiting protein synthesis. In this study, the influence of p53 protein on the apoptosis-inducing effect of anisomycin was investigated. The effect of protein synthesis-inhibiting concentration of anisomycin on apoptotic events was analyzed using Western blot, DNA fragmentation, and cell viability assays in wild-type PC12 and in mutant p53 protein expressing p143p53PC12 cells. Anisomycin stimulated the main apoptotic pathways in both cell lines, but p143p53PC12 cells showed lower sensitivity to the drug than their wild-type counterparts. Anisomycin caused the activation of the main stress kinases, phosphorylation of the p53 protein and the eukaryotic initiation factor eIF2α, proteolytic cleavage of protein kinase R, Bid, caspase-9 and -3. Furthermore, anisomycin treatment led to the activation of TRAIL and caspase-8, two proteins involved in the extrinsic apoptotic pathway. All these changes were stronger and more sustained in wtPC12 cells. In the presence of the dominant inhibitory p53 protein, p53- dependent genes involved in the regulation of apoptosis may be less transcribed and this can lead to the decrease of apoptotic processes in p143p53PC12 cells.
Collapse
Affiliation(s)
- R Schipp
- Department of Medical Biology, Medical School, University of Pécs, Szigeti út 12, Pécs, 7624, Hungary.,Signal Transduction Research Group, Szentágothai Research Centre, Ifjúság útja 20, Pécs, 7624, Hungary
| | - J Varga
- Department of Medical Biology, Medical School, University of Pécs, Szigeti út 12, Pécs, 7624, Hungary.,Signal Transduction Research Group, Szentágothai Research Centre, Ifjúság útja 20, Pécs, 7624, Hungary
| | - J Bátor
- Department of Medical Biology, Medical School, University of Pécs, Szigeti út 12, Pécs, 7624, Hungary.,Signal Transduction Research Group, Szentágothai Research Centre, Ifjúság útja 20, Pécs, 7624, Hungary
| | - M Vecsernyés
- Department of Medical Biology, Medical School, University of Pécs, Szigeti út 12, Pécs, 7624, Hungary.,Signal Transduction Research Group, Szentágothai Research Centre, Ifjúság útja 20, Pécs, 7624, Hungary
| | - Z Árvai
- Department of Medical Biology, Medical School, University of Pécs, Szigeti út 12, Pécs, 7624, Hungary.,Signal Transduction Research Group, Szentágothai Research Centre, Ifjúság útja 20, Pécs, 7624, Hungary
| | - M Pap
- Department of Medical Biology, Medical School, University of Pécs, Szigeti út 12, Pécs, 7624, Hungary.,Signal Transduction Research Group, Szentágothai Research Centre, Ifjúság útja 20, Pécs, 7624, Hungary
| | - József Szeberényi
- Department of Medical Biology, Medical School, University of Pécs, Szigeti út 12, Pécs, 7624, Hungary. .,Signal Transduction Research Group, Szentágothai Research Centre, Ifjúság útja 20, Pécs, 7624, Hungary.
| |
Collapse
|
34
|
Schaaf K, Smith SR, Duverger A, Wagner F, Wolschendorf F, Westfall AO, Kutsch O, Sun J. Mycobacterium tuberculosis exploits the PPM1A signaling pathway to block host macrophage apoptosis. Sci Rep 2017; 7:42101. [PMID: 28176854 PMCID: PMC5296758 DOI: 10.1038/srep42101] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 12/30/2016] [Indexed: 12/19/2022] Open
Abstract
The ability to suppress host macrophage apoptosis is essential for M. tuberculosis (Mtb) to replicate intracellularly while protecting it from antibiotic treatment. We recently described that Mtb infection upregulated expression of the host phosphatase PPM1A, which impairs the antibacterial response of macrophages. Here we establish PPM1A as a checkpoint target used by Mtb to suppress macrophage apoptosis. Overproduction of PPM1A suppressed apoptosis of Mtb-infected macrophages by a mechanism that involves inactivation of the c-Jun N-terminal kinase (JNK). Targeted depletion of PPM1A by shRNA or inhibition of PPM1A activity by sanguinarine restored JNK activation, resulting in increased apoptosis of Mtb-infected macrophages. We also demonstrate that activation of JNK by subtoxic concentrations of anisomycin induced selective apoptotic killing of Mtb-infected human macrophages, which was completely blocked in the presence of a specific JNK inhibitor. Finally, selective killing of Mtb-infected macrophages and subsequent bacterial release enabled rifampicin to effectively kill Mtb at concentrations that were insufficient to act against intracellular Mtb, providing proof of principle for the efficacy of a "release and kill" strategy. Taken together, these findings suggest that drug-induced selective apoptosis of Mtb-infected macrophages is achievable.
Collapse
Affiliation(s)
- Kaitlyn Schaaf
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Samuel R. Smith
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Alexandra Duverger
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Frederic Wagner
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Frank Wolschendorf
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Andrew O. Westfall
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Olaf Kutsch
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jim Sun
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
35
|
Formosa LE, Hofer A, Tischner C, Wenz T, Ryan MT. Translation and Assembly of Radiolabeled Mitochondrial DNA-Encoded Protein Subunits from Cultured Cells and Isolated Mitochondria. Methods Mol Biol 2016; 1351:115-29. [PMID: 26530678 DOI: 10.1007/978-1-4939-3040-1_9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
In higher eukaryotes, the mitochondrial electron transport chain consists of five multi-subunit membrane complexes responsible for the generation of cellular ATP. Of these, four complexes are under dual genetic control as they contain subunits encoded by both the mitochondrial and nuclear genomes, thereby adding another layer of complexity to the puzzle of respiratory complex biogenesis. These subunits must be synthesized and assembled in a coordinated manner in order to ensure correct biogenesis of different respiratory complexes. Here, we describe techniques to (1) specifically radiolabel proteins encoded by mtDNA to monitor the rate of synthesis using pulse labeling methods, and (2) analyze the stability, assembly, and turnover of subunits using pulse-chase methods in cultured cells and isolated mitochondria.
Collapse
Affiliation(s)
- Luke E Formosa
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Building 77, Level 2, Clayton Campus, Melbourne, VIC, 3800, Australia
| | - Annette Hofer
- Institute for Genetics and Cluster of Excellence: Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Zülpicher Str. 47A, Cologne, 50674, Germany
| | - Christin Tischner
- Institute for Genetics and Cluster of Excellence: Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Zülpicher Str. 47A, Cologne, 50674, Germany
| | - Tina Wenz
- Institute for Genetics and Cluster of Excellence: Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Zülpicher Str. 47A, Cologne, 50674, Germany
| | - Michael T Ryan
- Department of Biochemistry and Molecular Biology, Monash University, Building 77, Level 2, Clayton Campus, Melbourne, VIC, 3800, Australia.
| |
Collapse
|
36
|
Abbas AK, Villers A, Ris L. Temporal phases of long-term potentiation (LTP): myth or fact? Rev Neurosci 2016; 26:507-46. [PMID: 25992512 DOI: 10.1515/revneuro-2014-0072] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 03/12/2015] [Indexed: 12/11/2022]
Abstract
Long-term potentiation (LTP) remains the most widely accepted model for learning and memory. In accordance with this belief, the temporal differentiation of LTP into early and late phases is accepted as reflecting the differentiation of short-term and long-term memory. Moreover, during the past 30 years, protein synthesis inhibitors have been used to separate the early, protein synthesis-independent (E-LTP) phase and the late, protein synthesis-dependent (L-LTP) phase. However, the role of these proteins has not been formally identified. Additionally, several reports failed to show an effect of protein synthesis inhibitors on LTP. In this review, a detailed analysis of extensive behavioral and electrophysiological data reveals that the presumed correspondence of LTP temporal phases to memory phases is neither experimentally nor theoretically consistent. Moreover, an overview of the time courses of E-LTP in hippocampal slices reveals a wide variability ranging from <1 h to more than 5 h. The existence of all these conflictual findings should lead to a new vision of LTP. We believe that the E-LTP vs. L-LTP distinction, established with protein synthesis inhibitor studies, reflects a false dichotomy. We suggest that the duration of LTP and its dependency on protein synthesis are related to the availability of a set of proteins at synapses and not to the de novo synthesis of plasticity-related proteins. This availability is determined by protein turnover kinetics, which is regulated by previous and ongoing electrical activities and by energy store availability.
Collapse
|
37
|
mTOR-Independent autophagy inducer trehalose rescues against insulin resistance-induced myocardial contractile anomalies: Role of p38 MAPK and Foxo1. Pharmacol Res 2016; 111:357-373. [PMID: 27363949 DOI: 10.1016/j.phrs.2016.06.024] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 05/02/2016] [Accepted: 06/26/2016] [Indexed: 12/22/2022]
Abstract
Insulin resistance is associated with cardiovascular diseases although the precise mechanisms remain elusive. Akt2, a critical member of the Akt family, plays an essential role in insulin signaling. This study was designed to examine the effect of trehalose, an mTOR-independent autophagy inducer, on myocardial function in an Akt2 knockout-induced insulin resistance model. Adult WT and Akt2 knockout (Akt2(-/-)) mice were administered trehalose (1mg/g/day, i.p.) for two days and were then given 2% trehalose in drinking water for two more months. Echocardiographic and myocardial mechanics, intracellular Ca(2+) properties, glucose tolerance, and autophagy were assessed. Apoptosis and ER stress were evaluated using TUNEL staining, Caspase 3 assay and Western blot. Autophagy and autophagy flux were examined with a focus on p38 mitogen activated protein kinase (MAPK), Forkhead box O (Foxo1) and Akt. Akt2 ablation impaired glucose tolerance, myocardial geometry and function accompanied with pronounced apoptosis, ER stress and dampened autophagy, the effects of which were ameliorated by trehalose treatment. Inhibition of lysosomal activity using bafilomycin A1 negated trehalose-induced induction of autophagy (LC3B-II and p62). Moreover, phosphorylation of p38 MAPK and Foxo1 were upregulated in Akt2(-/-) mice, the effect of which was attenuated by trehalose. Phosphorylation of Akt was suppressed in Akt2(-/-) mice and was unaffected by trehalose. In vitro findings revealed that the p38 MAPK activator anisomycin and the Foxo1 inhibitor (through phosphorylation) AS1842856 effectively masked trehalose-offered beneficial cardiomyocyte contractile response against Akt2 ablation. These data suggest that trehalose may rescue against insulin resistance-induced myocardial contractile defect and apoptosis, via autophagy associated with dephosphorylation of p38 MAPK and Foxo1 without affecting phosphorylation of Akt.
Collapse
|
38
|
Browne AJ, Göbel A, Thiele S, Hofbauer LC, Rauner M, Rachner TD. p38 MAPK regulates the Wnt inhibitor Dickkopf-1 in osteotropic prostate cancer cells. Cell Death Dis 2016; 7:e2119. [PMID: 26913608 PMCID: PMC4849158 DOI: 10.1038/cddis.2016.32] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 01/05/2016] [Accepted: 01/22/2016] [Indexed: 12/17/2022]
Abstract
The Wnt inhibitor Dickkopf-1 (DKK-1) has been associated with the occurrence of bone metastases in osteotropic prostate cancer by inhibiting osteoblastogenesis. P38 mitogen-activated protein kinase (MAPK) activity is also dysregulated in advanced prostate cancer. However, the impact of p38 MAPK signaling on DKK-1 remains unknown. Inhibition of p38 MAPK signaling in osteolytic PC3 cells by small molecule inhibitors (doramapimod, LY2228820 and SB202190) suppressed DKK-1 expression, whereas activation of p38 MAPK by anisomycin increased DKK-1. Further dissection by targeting individual p38 MAPK isoforms with siRNA revealed a stronger role for MAPK11 than MAPK14 and MAPK12 in the regulation of DKK-1. Moreover, prostate cancer cells with a predominantly osteolytic phenotype produced sufficient amounts of DKK-1 to inhibit Wnt3a-induced osteoblastic differentiation in C2C12 cells. This inhibition was blocked directly by neutralizing DKK-1 using a specific antibody and also indirectly by blocking p38 MAPK. Furthermore, tissue expression in human prostate cancer revealed a correlation between p38 MAPK and DKK-1 expression with higher expression in tumor compared with normal tissues. These results reveal that p38 MAPK regulates DKK-1 in prostate cancer and may present a potential target in osteolytic prostate cancers.
Collapse
Affiliation(s)
- A J Browne
- Division of Endocrinology and Metabolic Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| | - A Göbel
- Division of Endocrinology and Metabolic Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| | - S Thiele
- Division of Endocrinology and Metabolic Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| | - L C Hofbauer
- Division of Endocrinology and Metabolic Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - M Rauner
- Division of Endocrinology and Metabolic Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| | - T D Rachner
- Division of Endocrinology and Metabolic Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
39
|
Abstract
Stress-response kinases, the mitogen-activated protein kinases (MAPKs) are activated in response to the challenge of a myriad of stressors. c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinases (ERKs), and p38 MAPKs are the predominant members of the MAPK family in the heart. Extensive studies have revealed critical roles of activated MAPKs in the processes of cardiac injury and heart failure and many other cardiovascular diseases. Recently, emerging evidence suggests that MAPKs also promote the development of cardiac arrhythmias. Thus, understanding the functional impact of MAPKs in the heart could shed new light on the development of novel therapeutic approaches to improve cardiac function and prevent arrhythmia development in the patients. This review will summarize the recent findings on the role of MAPKs in cardiac remodeling and arrhythmia development and point to the critical need of future studies to further elucidate the fundamental mechanisms of MAPK activation and arrhythmia development in the heart.
Collapse
|
40
|
Nie S, Xu J, Zhang C, Xu C, Liu M, Yu D. Salicortin inhibits osteoclast differentiation and bone resorption by down-regulating JNK and NF-κB/NFATc1 signaling pathways. Biochem Biophys Res Commun 2016; 470:61-67. [DOI: 10.1016/j.bbrc.2015.12.115] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 12/24/2015] [Indexed: 10/22/2022]
|
41
|
Gururaj S, Fleites J, Bhattacharjee A. Slack sodium-activated potassium channel membrane expression requires p38 mitogen-activated protein kinase phosphorylation. Neuropharmacology 2015; 103:279-89. [PMID: 26721627 DOI: 10.1016/j.neuropharm.2015.12.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 12/11/2015] [Accepted: 12/15/2015] [Indexed: 01/14/2023]
Abstract
p38 MAPK has long been understood as an inducible kinase under conditions of cellular stress, but there is now increasing evidence to support its role in the regulation of neuronal function. Several phosphorylation targets have been identified, an appreciable number of which are ion channels, implicating the possible involvement of p38 MAPK in neuronal excitability. The KNa channel Slack is an important protein to be studied as it is highly and ubiquitously expressed in DRG neurons and is important in the maintenance of their firing accommodation. We sought to examine if the Slack channel could be a substrate of p38 MAPK activity. First, we found that the Slack C-terminus contains two putative p38 MAPK phosphorylation sites that are highly conserved across species. Second, we show via electrophysiology experiments that KNa currents and further, Slack currents, are subject to tonic modulation by p38 MAPK. Third, biochemical approaches revealed that Slack channel regulation by p38 MAPK occurs through direct phosphorylation at the two putative sites of interaction, and mutating both sites prevented surface expression of Slack channels. Based on these results, we conclude that p38 MAPK is an obligate regulator of Slack channel function via the trafficking of channels into the membrane. The present study identifies Slack KNa channels as p38 MAPK substrates.
Collapse
Affiliation(s)
- Sushmitha Gururaj
- Department of Pharmacology and Toxicology, The State University of New York, Buffalo, USA
| | - John Fleites
- Program in Neuroscience, The State University of New York, Buffalo, USA
| | - Arin Bhattacharjee
- Department of Pharmacology and Toxicology, The State University of New York, Buffalo, USA; Program in Neuroscience, The State University of New York, Buffalo, USA.
| |
Collapse
|
42
|
Kudo K, Uchida T, Sawada M, Nakamura Y, Yoneda A, Fukami K. Phospholipase C δ1 in macrophages negatively regulates TLR4-induced proinflammatory cytokine production and Fcγ receptor-mediated phagocytosis. Adv Biol Regul 2015; 61:68-79. [PMID: 26643908 DOI: 10.1016/j.jbior.2015.11.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 11/14/2015] [Accepted: 11/14/2015] [Indexed: 12/26/2022]
Abstract
Macrophages are key players in the innate immune response. Turnover of phosphoinositides (PI), particularly phosphatidylinositol 4,5 bisphosphate (PI(4,5)P2), has been implicated in macrophage functions such as toll-like receptor (TLR)-mediated cytokine production and phagocytosis. However, PI metabolizing enzymes responsible for macrophage functions are not well defined. The phospholipase C (PLC) family of enzymes is critical in PI(4,5)P2 turnover. In this study, we investigated the role of PLCδ1, a prototype PLC, in macrophages on the expression of inflammation-associated genes and phagocytosis. Lipopolysaccharides (LPS) signal through TLR4 to produce proinflammatory cytokines such as interleukin (IL)-1β. LPS stimulation of both RAW264.7 murine macrophages and murine bone marrow-derived macrophages resulted in lower PLCδ1 mRNA and protein expression levels, compared to that in the control. Using chemical inhibitor compounds, we demonstrated that the up-regulation of p38 MAPK activity led to down-regulation of PLCδ1 mRNA expression in macrophages. PLCδ1 reduction by RNAi or gene deletion resulted in greater LPS-induced IL-1β expression than that observed in the control siRNA-treated cells, without increasing TLR4 cell surface expression. PLCδ1 also negatively regulated LPS-induced cell spreading. Analysis of Fcγ receptor-mediated phagocytosis demonstrated an increased phagocytosis index after PLCδ1 knockdown in RAW264.7 cells. Conversely, overexpression of PLCδ1 reduced phagocytosis whereas catalytic inactive PLCδ1 had no effect. Altered levels of PLCδ1 affected the binding of opsonized latex beads with cells, rather than the phagocytic activity. Taken together, the data suggest that PLCδ1 negatively regulates LPS-induced production of IL-1β and Fcγ receptor-mediated phagocytosis in macrophages.
Collapse
Affiliation(s)
- Kohya Kudo
- Laboratory of Genome and Biosignals, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Takafumi Uchida
- Laboratory of Genome and Biosignals, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Mayu Sawada
- Laboratory of Genome and Biosignals, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Yoshikazu Nakamura
- Laboratory of Genome and Biosignals, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan; AMED-CREST, AMED, Tokyo, Japan
| | - Atsuko Yoneda
- Laboratory of Genome and Biosignals, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan; AMED-CREST, AMED, Tokyo, Japan
| | - Kiyoko Fukami
- Laboratory of Genome and Biosignals, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan; AMED-CREST, AMED, Tokyo, Japan.
| |
Collapse
|
43
|
Ai X. SR calcium handling dysfunction, stress-response signaling pathways, and atrial fibrillation. Front Physiol 2015; 6:46. [PMID: 25745402 PMCID: PMC4333799 DOI: 10.3389/fphys.2015.00046] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 01/30/2015] [Indexed: 12/19/2022] Open
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia. It is associated with a markedly increased risk of premature death due to embolic stroke and also complicates co-existing cardiovascular diseases such as heart failure. The prevalence of AF increases dramatically with age, and aging has been shown to be an independent risk of AF. Due to an aging population in the world, a growing body of AF patients are suffering a diminished quality of life and causing an associated economic burden. However, effective pharmacologic treatments and prevention strategies are lacking due to a poor understanding of the molecular and electrophysiologic mechanisms of AF in the failing and/or aged heart. Recent studies suggest that altered atrial calcium handling contributes to the onset and maintenance of AF. Here we review the role of stress-response kinases and calcium handling dysfunction in AF genesis in the aged and failing heart.
Collapse
Affiliation(s)
- Xun Ai
- Department of Cell and Molecular Physiology, Loyola University Chicago Maywood, IL, USA
| |
Collapse
|
44
|
The inhibition of RANKL-induced osteoclastogenesis through the suppression of p38 signaling pathway by naringenin and attenuation of titanium-particle-induced osteolysis. Int J Mol Sci 2014; 15:21913-34. [PMID: 25464380 PMCID: PMC4284685 DOI: 10.3390/ijms151221913] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 10/30/2014] [Accepted: 11/24/2014] [Indexed: 12/25/2022] Open
Abstract
The aim of this study was to assess the effect of naringenin on osteoclastogenesis and titanium particle-induced osteolysis. Osteolysis from wear-induced particles and aseptic loosening are the most frequent late complications of total joint arthroplasty leading to revision of the prosthesis. Osteolysis during aseptic loosening is most likely due to increased bone resorption by osteoclasts. Through in vitro studies, we demonstrated that naringenin, a naturally occurring flavanone in grapefruit and tomatoes, exerts potent inhibitory effects on the ligand of the receptor activator of nuclear factor-κB (RANKL)-induced osteoclastogenesis and revealed that the mechanism of action of naringenin, which inhibited osteoclastogenesis by suppression of the p38 signaling pathway. Through in vivo studies, we proved that naringenin attenuated titanium particle-induced osteolysis in a mouse calvarial model. In general, we demonstrated that naringenin inhibited osteoclastogenesis via suppression of p38 signaling in vitro and attenuated titanium particle-induced osteolysis in vivo. This study also suggested that naringenin has significant potential for the treatment of osteolysis-related diseases caused by excessive osteoclast formation and activity.
Collapse
|
45
|
Kiran S, Oddi V, Ramakrishna G. Sirtuin 7 promotes cellular survival following genomic stress by attenuation of DNA damage, SAPK activation and p53 response. Exp Cell Res 2014; 331:123-141. [PMID: 25445786 DOI: 10.1016/j.yexcr.2014.11.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Revised: 10/29/2014] [Accepted: 11/04/2014] [Indexed: 01/10/2023]
Abstract
Maintaining the genomic integrity is a constant challenge in proliferating cells. Amongst various proteins involved in this process, Sirtuins play a key role in DNA damage repair mechanisms in yeast as well as mammals. In the present work we report the role of one of the least explored Sirtuin viz., SIRT7, under conditions of genomic stress when treated with doxorubicin. Knockdown of SIRT7 sensitized osteosarcoma (U2OS) cells to DNA damage induced cell death by doxorubicin. SIRT7 overexpression in NIH3T3 delayed cell cycle progression by causing delay in G1 to S transition. SIRT7 overexpressing cells when treated with low dose of doxorubicin (0.25 µM) showed delayed onset of senescence, lesser accumulation of DNA damage marker γH2AX and lowered levels of growth arrest markers viz., p53 and p21 when compared to doxorubicin treated control GFP expressing cells. Resistance to DNA damage following SIRT7 overexpression was also evident by EdU incorporation studies where cellular growth arrest was significantly delayed. When treated with higher dose of doxorubicin (>1 µM), SIRT7 conferred resistance to apoptosis by attenuating stress activated kinases (SAPK viz., p38 and JNK) and p53 response thereby shifting the cellular fate towards senescence. Interestingly, relocalization of SIRT7 from nucleolus to nucleoplasm together with its co-localization with SAPK was an important feature associated with DNA damage. SIRT7 mediated resistance to doxorubicin induced apoptosis and senescence was lost when p53 level was restored by nutlin treatment. Overall, we propose SIRT7 attenuates DNA damage, SAPK activation and p53 response thereby promoting cellular survival under conditions of genomic stress.
Collapse
Affiliation(s)
- Shashi Kiran
- Laboratory of Cancer Biology, Centre for DNA Fingerprinting and Diagnostics, Hyderabad, Telangana, 500001, India
| | - Vineesha Oddi
- Laboratory of Cancer Biology, Centre for DNA Fingerprinting and Diagnostics, Hyderabad, Telangana, 500001, India
| | - Gayatri Ramakrishna
- Laboratory of Cancer Biology, Centre for DNA Fingerprinting and Diagnostics, Hyderabad, Telangana, 500001, India; Laboratory of Cancer Cell Biology, Department of Research, Institute of Liver and Biliary Sciences, Delhi 110070, India.
| |
Collapse
|
46
|
Bunjobpol W, Dulloo I, Igarashi K, Concin N, Matsuo K, Sabapathy K. Suppression of acetylpolyamine oxidase by selected AP-1 members regulates DNp73 abundance: mechanistic insights for overcoming DNp73-mediated resistance to chemotherapeutic drugs. Cell Death Differ 2014; 21:1240-9. [PMID: 24722210 PMCID: PMC4085530 DOI: 10.1038/cdd.2014.41] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 02/17/2014] [Accepted: 02/28/2014] [Indexed: 12/18/2022] Open
Abstract
Enhanced resistance to chemotherapy has been correlated with high levels of Delta-Np73 (DNp73), an anti-apoptotic protein of the p53 tumor-suppressor family which inhibits the pro-apoptotic members such as p53 and TAp73. Although genotoxic drugs have been shown to induce DNp73 degradation, lack of mechanistic understanding of this process precludes strategies to enhance the targeting of DNp73 and improve treatment outcomes. Antizyme (Az) is a mediator of ubiquitin-independent protein degradation regulated by the polyamine biosynthesis pathway. We show here that acetylpolyamine oxidase (PAOX), a catabolic enzyme of this pathway, upregulates DNp73 levels by suppressing its degradation via the Az pathway. Conversely, downregulation of PAOX activity by siRNA-mediated knockdown or chemical inhibition leads to DNp73 degradation in an Az-dependent manner. PAOX expression is suppressed by several genotoxic drugs, via selected members of the activator protein-1 (AP-1) transcription factors, namely c-Jun, JunB and FosB, which are required for stress-mediated DNp73 degradation. Finally, chemical- and siRNA-mediated inhibition of PAOX significantly reversed the resistant phenotype of DNp73-overexpressing cancer cells to genotoxic drugs. Together, these data define a critical mechanism for the regulation of DNp73 abundance, and reveal that inhibition of PAOX could widen the therapeutic index of cytotoxic drugs and overcome DNp73-mediated chemoresistance in tumors.
Collapse
Affiliation(s)
- W Bunjobpol
- Laboratory of Molecular Carcinogenesis, Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre, 11, Hospital Drive, Singapore, Singapore
| | - I Dulloo
- Laboratory of Molecular Carcinogenesis, Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre, 11, Hospital Drive, Singapore, Singapore
| | - K Igarashi
- Amine Pharma Research Institute, Innovation Plaza at Chiba University, 1-8-15Inohana, Chiba, Japan
| | - N Concin
- Department of Obstetrics and Gynecology, Innsbruck Medical University, Anichstrasse 35, Innsbruck, Austria
| | - K Matsuo
- Department of Microbiology and Immunology, School of Medicine, Keio University, 35 Shinanomachi, Tokyo, Japan
| | - K Sabapathy
- Laboratory of Molecular Carcinogenesis, Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre, 11, Hospital Drive, Singapore, Singapore
- Cancer and Stem Cell Biology Program, Duke-NUS Graduate Medical School, 8, College Road, Singapore, Singapore
- Department of Biochemistry, National University of Singapore, 8, Medical Drive, Singapore, Singapore
| |
Collapse
|
47
|
Tian B, Jiang T, Shao Z, Zhai Z, Li H, Fan Q, Liu X, Ouyang Z, Tang T, Jiang Q, Zheng M, Dai K, Qin A, Yu Y, Zhu Z. The prevention of titanium-particle-induced osteolysis by OA-14 through the suppression of the p38 signaling pathway and inhibition of osteoclastogenesis. Biomaterials 2014; 35:8937-50. [PMID: 25086794 DOI: 10.1016/j.biomaterials.2014.06.055] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 06/29/2014] [Indexed: 12/29/2022]
Abstract
Wear-particle-induced osteolysis leads to prosthesis loosening, which is one of the most common causes of joint-implant failure, a problem that must be fixed using revision surgery. Thus, a potential treatment for prosthetic loosening is focused on inhibiting osteoclastic bone resorption, which prevents wear-particle-induced osteolysis. In this study, we synthesized a compound named OA-14 (N-(3- (dodecylcarbamoyl)phenyl)-1H-indole-2-carboxamide) and examined how OA-14 affects titanium (Ti)-particle-induced osteolysis and osteoclastogenesis. We report that OA-14 treatment protected against Ti-particle-induced osteolysis in a mouse calvarial model. Interestingly, the number of tartrate-resistant acid phosphatase-positive osteoclasts decreased after treatment with OA-14 in vivo, which suggested that OA-14 inhibits osteoclast formation. To test this hypothesis, we conducted in vitro studies, and our results revealed that OA-14 markedly diminished osteoclast differentiation and osteoclast-specific gene expression in a dose- and time-dependent manner. Moreover, OA-14 suppressed osteoclastic bone resorption and F-actin ring formation. Furthermore, we determined that OA-14 inhibited osteoclastogenesis by specifically blocking the p38-Mitf-c-fos-NFATc1 signaling cascade induced by RANKL (ligand of receptor activator of nuclear factor κB). Collectively, our results suggest that the compound OA-14 can be safely used for treating particle-induced peri-implant osteolysis and other diseases caused by excessive osteoclast formation and function.
Collapse
Affiliation(s)
- Bo Tian
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Tao Jiang
- The Center of Diagnosis and Treatment for Joint Disease, Drum Tower Clinical Medical College of Nanjing Medical University, Jiangsu, PR China
| | - Zhanying Shao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, PR China
| | - Zanjing Zhai
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Haowei Li
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Qiming Fan
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Xuqiang Liu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Zhengxiao Ouyang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Tingting Tang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Qing Jiang
- The Center of Diagnosis and Treatment for Joint Disease, Drum Tower Clinical Medical College of Nanjing Medical University, Jiangsu, PR China
| | - Minghao Zheng
- Centre for Orthopaedic Research, School of Surgery, The University of Western Australia, Perth, Australia
| | - Kerong Dai
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - An Qin
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; Centre for Orthopaedic Research, School of Surgery, The University of Western Australia, Perth, Australia.
| | - Yongping Yu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, PR China.
| | - Zhenan Zhu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| |
Collapse
|
48
|
Liu X, Qu X, Wu C, Zhai Z, Tian B, Li H, Ouyang Z, Xu X, Wang W, Fan Q, Tang T, Qin A, Dai K. The effect of enoxacin on osteoclastogenesis and reduction of titanium particle-induced osteolysis via suppression of JNK signaling pathway. Biomaterials 2014; 35:5721-30. [DOI: 10.1016/j.biomaterials.2014.04.006] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 04/01/2014] [Indexed: 02/07/2023]
|
49
|
Ueno-Shuto K, Kato K, Tasaki Y, Sato M, Sato K, Uchida Y, Sakai H, Ono T, Suico MA, Mitsutake K, Tokutomi N, Kai H, Shuto T. Lipopolysaccharide decreases single immunoglobulin interleukin-1 receptor-related molecule (SIGIRR) expression by suppressing specificity protein 1 (Sp1) via the Toll-like receptor 4 (TLR4)-p38 pathway in monocytes and neutrophils. J Biol Chem 2014; 289:18097-109. [PMID: 24821721 DOI: 10.1074/jbc.m113.532093] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Single immunoglobulin interleukin-1 receptor-related molecule (SIGIRR) is one of the immunoglobulin-like membrane proteins that is crucial for negative regulation of toll-like receptor 4 (TLR4) and interleukin-1 receptor. Despite the importance of understanding its expression and function, knowledge is limited on the regulatory mechanism in the epithelial tissues, such as the liver, lung, and gut, where its predominant expression is originally described. Here, we found expression of SIGIRR in non-epithelial innate immune cells, including primary peripheral blood monocytes, polymorphonuclear neutrophils, monocytic RAW264 cells, and neutrophilic-differentiated HL-60 cells. Consistent with previous findings in epithelial tissues, SIGIRR gene and protein expression were also down-regulated by LPS treatment in a time-dependent manner in primary blood monocytes and polymorphonuclear neutrophils. A reduction was also observed in RAW264 and differentiated HL-60 cells. Notably, exogenous introduction of the dominant negative form of TLR4 and siRNA of p38 resulted in inhibition of LPS-induced SIGIRR down-regulation, whereas treatment with p38 activator anisomycin showed a dose-dependent decrease in SIGIRR expression, suggesting TLR4-p38 signal as a critical pathway for LPS-induced SIGIRR down-regulation. Finally, reporter gene and chromatin immunoprecipitation assays demonstrated that Sp1 is a key factor that directly binds to the proximal promoter of SIGIRR gene and consequently regulates basal SIGIRR expression, which is negatively regulated by the LPS-dependent TLR4-p38 pathway. In summary, the data precisely demonstrate how LPS down-regulates SIGIRR expression and provide a role of LPS signal that counteracts Sp1-dependent basal promoter activation of SIGIRR gene via TLR4-p38 pathway in non-epithelial innate immune cells.
Collapse
Affiliation(s)
- Keiko Ueno-Shuto
- From the Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan, the Laboratory of Pharmacology, Division of Life Science, Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto 860-0082, Japan
| | - Kosuke Kato
- the Department of Physiology and Lung Center, Temple University School of Medicine, Philadelphia, Pennsylvania 19140, and
| | - Yukihiro Tasaki
- From the Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Miki Sato
- From the Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Keizo Sato
- the School of Pharmacy, Kyushu University of Health and Welfare, Nobeoka, Miyazaki 882-8508, Japan
| | - Yuji Uchida
- the Laboratory of Pharmacology, Division of Life Science, Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto 860-0082, Japan
| | - Hiromichi Sakai
- From the Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Tomomi Ono
- From the Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Mary Ann Suico
- From the Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Kazunori Mitsutake
- From the Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Naofumi Tokutomi
- the Laboratory of Pharmacology, Division of Life Science, Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto 860-0082, Japan
| | - Hirofumi Kai
- From the Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Tsuyoshi Shuto
- From the Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan,
| |
Collapse
|
50
|
Curcumol suppresses RANKL-induced osteoclast formation by attenuating the JNK signaling pathway. Biochem Biophys Res Commun 2014; 447:364-70. [PMID: 24732351 DOI: 10.1016/j.bbrc.2014.04.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 04/06/2014] [Indexed: 11/24/2022]
Abstract
Osteoclasts, derived from hemopoietic progenitors of the monocyte/macrophage lineage, have a unique role in bone resorption, and are considered a potential therapeutic target in the treatment of such pathologic bone diseases as osteoporosis, rheumatoid arthritis, and periodontitis. In the present study, we demonstrate that curcumol, one of the major components of the essential oil of Rhizoma Curcumae, exhibits an inhibitory effect on receptor activator of nuclear factor kappaB ligand (RANKL)-induced osteoclast differentiation with both bone marrow-derived macrophages and RAW264.7 cells in a dose-dependent manner. In addition, RANKL-induced mRNA expression of osteoclast-specific genes, such as tartrate-resistant acid phosphatase, calcitonin receptor, and cathepsin K, is prominently reduced in the presence of curcumol. Furthermore, the molecular mechanism of action was investigated, and curcumol inhibited osteoclastogenesis by specifically impairing RANKL-induced c-Jun N-terminal kinase (JNK)/activator protein-1 (AP-1) signaling, which was further identified in rescue studies by means of anisomycin, a JNK signaling-specific activator. Taken together, these findings suggest that curcumol suppresses RANKL-induced osteoclast differentiation through the JNK/AP-1 signaling pathway, and may be useful as a therapeutic treatment for bone resorption-associated diseases.
Collapse
|