IL-10 Targets Macrophage Metabolism

Total Page:16

File Type:pdf, Size:1020Kb

IL-10 Targets Macrophage Metabolism RESEARCH HIGHLIGHTS Nature Reviews Immunology | Published online 26 May 2017; doi:10.1038/nri.2017.57 wild-type metabolic phenotype IMMUNE REGULATION could not be restored in ATG5- deficient BMDMs by the addition IL‑10 targets macrophage of exogenous IL-10. Thus, the effect of IL-10 on mitochondrial function is highly dependent on autophagy. metabolism Activation of the mechanistic target of rapamycin (mTOR) complex 1 Although macrophages are Il10–/– BMDMs had a lower (mTORC1) promotes glycolysis and known to be the main target cells maximal respiratory capacity than inhibits autophagy, and mTORC1 of the anti-inflammatory cytokine accumulation wild-type cells after LPS stimulation, is thus a key potential target for interleukin-10 (IL-10), little is known of which indicates that lack of IL-10 the mitochondrial effects of IL-10. about the mechanisms by which could result in reduced mitochondrial Il10–/– BMDMs — and BMDMs IL-10 mediates its effects. Medzhitov dysfunctional fitness and hence a reduced OCR. lacking the transcription factor STAT3 and colleagues provide new evidence mitochondria The authors therefore investigated (which is activated downstream of that IL-10 regulates the metabolic in Il10–/– whether the altered metabolic profile the IL-10 receptor) — had increased –/– switch to glycolysis that occurs in BMDMs was of Il10 BMDMs also results from and prolonged mTORC1 activation macrophages that are activated abnormal mitochondrial function. after LPS stimulation compared with with lipo polysaccharide (LPS) and the result Il10–/– BMDMs had an increased mito- wild-type cells. Directly inhibiting prevents the accumulation of of impaired chondrial mass after LPS stimulation mTOR with rapamycin in LPS- dysfunctional mitochondria. mitophagy compared with wild-type BMDMs stimulated Il10–/– BMDMs resulted After LPS stimulation, Il10–/– due to the accumulation of dysfunc- in decreased accumulation of dys- bone marrow-derived macrophages tional mitochondria with loss of the functional mitochondria, decreased (BMDMs) had higher levels of mitochondrial membrane potential. ECAR and increased OCR. DDIT4 (a glycolysis and lower levels of oxida- This was associated with increased known negative regulator of mTOR) tive phosphorylation compared production of mitochondrial reactive was strongly induced by IL-10 in a with wild-type BMDMs, as oxygen species (ROS). STAT3-dependent manner during measured by a higher extracellular Further experiments showed that LPS stimulation, and Ddit4–/– BMDMs acidification rate (ECAR) and a this accumulation of dysfunctional had prolonged mTORC1 activation lower oxygen consumption rate mitochondria in Il10–/– BMDMs was during LPS stimulation, which could (OCR), respectively. In wild-type the result of impaired mitophagy (the not be reduced by exogenous IL-10. BMDMs, IL-10 inhibited glycolysis selective degradation of mitochondria The authors conclude that IL-10 by downregulating the translocation by autophagy). Il10–/– BMDMs had inhibits mTORC1 signalling in a of glucose transporter 1 (GLUT1) significantly lower levels of autophagy STAT3–DDIT4-dependent manner, from intracellular vesicles to the than control cells after LPS stimulation. which is required for the inhibition of cell surface. Furthermore, IL-10 BMDMs lacking autophagy protein 5 glycolysis and the autophagic clearance inhibited the expression of glycolytic (ATG5) had a decreased OCR, of dysfunctional, ROS-producing pathway genes. similar to Il10–/– BMDMs, but the mitochondria. As mitochondrial ROS can function as an endogenous DDIT4 second signal for inflammasome expression activation, BMDMs lacking IL-10, LPS STAT3 or DDIT4 had increased inflammasome-mediated caspase 1 TLR signalling cleavage and IL-1β production after LPS stimulation as a result of mTOR IL-10 STAT3 decreased mitophagy. Therefore, the mTORC1 activation inhibition expression activation mTORC1-mediated effects of IL-10 on macrophage metabolism have Mitophagy Switch from oxidative Inhibition Inhibition of crucial anti-inflammatory roles. phosphorylation to glycolysis of GLUT1 glycolytic gene Kirsty Minton translocation expression ORIGINAL ARTICLE Ip, W. K. E. et al. Anti‑inflammatory effect of IL‑10 mediated by Clearance of ROS- Glycolysis metabolic reprogramming of macrophages. producing mitochondria Science 356, 513–519 (2017) Macmillan Publishers Limited NATURE REVIEWS | IMMUNOLOGY www.nature.com/nri ©2017 Mac millan Publishers Li mited, part of Spri nger Nature. All ri ghts reserved. .
Recommended publications
  • EGFR and Mtor As Therapeutic Targets in Glioblastoma
    www.oncotarget.com Oncotarget, 2019, Vol. 10, (No. 46), pp: 4721-4723 Editorial EGFR and mTOR as therapeutic targets in glioblastoma Michael W. Ronellenfitsch, Anna-Luisa Luger and Joachim P. Steinbach The quest for new and improved therapies for mammalian target of rapamycin complex 1 (mTORC1) glioblastoma (GB) has been mostly unsuccessful in signaling were found in the majority of GBs [3]. more than a decade despite significant efforts. The few Therefore, many hopes have rested on targeted therapies. exceptions include the optimization of classical alkylating However, the results from clinical trials have been largely chemotherapy by including lomustine in the first line disappointing [4]. Nevertheless, unplanned retrospective regimen for GB with a methylated MGMT promoter and subgroup analyses of the patient cohorts of negative tumor treating fields [1, 2]. The GB signaling network has clinical trials indicated that dysregulation or activation been well-characterized and genetic alterations resulting of signaling could be a predictive factor for susceptibility in activation of receptor tyrosine kinases and especially to pathway inhibition: Tumors with enhanced levels of epidermal growth factor receptor (EGFR) and downstream mTORC1 activation markers, including phosphorylated Figure 1: Scheme of EGFR signaling and DDIT4-mediated adaptive processes. Conditions of the glioblastoma microenvironment including hypoxia, alkylating therapy or irradiation trigger induction of DDIT4 which activates TSC1/2 to inhibit mTORC1 and can counteract epidermal growth factor receptor (EGFR)-mediated TSC1/2 inhibition. Inhibition of mTORC1 ultimately induces adaptive processes to cope with external stressors. www.oncotarget.com 4721 Oncotarget ribosomal protein S6 and phosphorylated mTOR itself, of effects. Depending on the half life and pharmacokinetics appeared to respond to pathway inhibition by the EGFR of the drugs, stepwise treatment algorithms could be an antibody nimotuzumab or the mTORC1 inhibitor option to prevent antagonistic effects.
    [Show full text]
  • Effects of Rapamycin on Social Interaction Deficits and Gene
    Kotajima-Murakami et al. Molecular Brain (2019) 12:3 https://doi.org/10.1186/s13041-018-0423-2 RESEARCH Open Access Effects of rapamycin on social interaction deficits and gene expression in mice exposed to valproic acid in utero Hiroko Kotajima-Murakami1,2, Toshiyuki Kobayashi3, Hirofumi Kashii1,4, Atsushi Sato1,5, Yoko Hagino1, Miho Tanaka1,6, Yasumasa Nishito7, Yukio Takamatsu7, Shigeo Uchino1,2 and Kazutaka Ikeda1* Abstract The mammalian target of rapamycin (mTOR) signaling pathway plays a crucial role in cell metabolism, growth, and proliferation. The overactivation of mTOR has been implicated in the pathogenesis of syndromic autism spectrum disorder (ASD), such as tuberous sclerosis complex (TSC). Treatment with the mTOR inhibitor rapamycin improved social interaction deficits in mouse models of TSC. Prenatal exposure to valproic acid (VPA) increases the incidence of ASD. Rodent pups that are exposed to VPA in utero have been used as an animal model of ASD. Activation of the mTOR signaling pathway was recently observed in rodents that were exposed to VPA in utero, and rapamycin ameliorated social interaction deficits. The present study investigated the effect of rapamycin on social interaction deficits in both adolescence and adulthood, and gene expressions in mice that were exposed to VPA in utero. We subcutaneously injected 600 mg/kg VPA in pregnant mice on gestational day 12.5 and used the pups as a model of ASD. The pups were intraperitoneally injected with rapamycin or an equal volume of vehicle once daily for 2 consecutive days. The social interaction test was conducted in the offspring after the last rapamycin administration at 5–6 weeks of ages (adolescence) or 10–11 weeks of age (adulthood).
    [Show full text]
  • Transcriptional Changes in Huntington Disease Identified Using Genome
    Human Molecular Genetics, 2010, Vol. 19, No. 8 1438–1452 doi:10.1093/hmg/ddq018 Advance Access published on January 20, 2010 Transcriptional changes in Huntington disease identified using genome-wide expression profiling and cross-platform analysis Kristina Becanovic1, Mahmoud A. Pouladi1, Raymond S. Lim2, Alexandre Kuhn3, Paul Pavlidis2, Ruth Luthi-Carter3, Michael R. Hayden1 and Blair R. Leavitt1,Ã 1Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada V5Z 4H4 2Centre for High-throughput Biology and Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada V6T 1Z4 and 3Brain Mind Institute, E´ cole Polytechnique Fe´de´rale de Lausanne (EPFL), Station 15, CH1015 Lausanne, Switzerland Received September 10, 2009; Revised and Accepted January 18, 2010 Evaluation of transcriptional changes in the striatum may be an effective approach to understanding the natural history of changes in expression contributing to the pathogenesis of Huntington disease (HD). We have performed genome-wide expression profiling of the YAC128 transgenic mouse model of HD at 12 and 24 months of age using two platforms in parallel: Affymetrix and Illumina. The data from these two powerful platforms were integrated to create a combined rank list, thereby revealing the identity of additional genes that proved to be differentially expressed between YAC128 and control mice. Using this approach, we ident- ified 13 genes to be differentially expressed between YAC128 and controls which were validated by quanti- tative real-time PCR in independent cohorts of animals. In addition, we analyzed additional time points relevant to disease pathology: 3, 6 and 9 months of age.
    [Show full text]
  • An Integrative Transcriptome Study Reveals Ddit4/Redd1 As a Key Regulator of Cancer Cachexia in Rodent Models
    www.nature.com/cddis ARTICLE OPEN An integrative transcriptome study reveals Ddit4/Redd1 as a key regulator of cancer cachexia in rodent models 1,2,4 1,2,4 3 1,2 1,2 1,2 2 1,2 Mengyuan Niu ,LiLi , Zhonglan✉ Su , Lulu Wei✉ , Wenyuan Pu , Chen Zhao✉ , Yibing Ding , Junaid Wazir , Wangsen Cao 2, Shiyu Song 1,2 , Qian Gao 2 and Hongwei Wang 1,2 © The Author(s) 2021 Cancer cachexia is a multifactorial metabolic syndrome that causes up to 20% of cancer-related deaths. Muscle atrophy, the hallmark of cancer cachexia, strongly impairs the quality of life of cancer patients; however, the underlying pathological process is still poorly understood. Investigation of the disease pathogenesis largely relies on cachectic mouse models. In our study, the transcriptome of the cachectic gastrocnemius muscle in the C26 xenograft model was integrated and compared with that of 5 more different datasets. The bioinformatic analysis revealed pivotal gene ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways of the disease, and the key genes were validated. Construction of the protein-protein interaction network and the comparison of pathways enriched in cancer cachexia with 5 other muscle atrophy models revealed Ddit4 (DNA damage-inducible transcript 4), as a key protein in cancer cachexia. The higher expression of Ddit4 in cachectic muscle was further validated in animal models and cachectic cancer patients. Further study revealed that p38 induced the expression of Ddit4, which in turn inhibited the mTOR pathway in atrophic cells. Cell Death and Disease (2021)12:652 ; https://doi.org/10.1038/s41419-021-03932-0 INTRODUCTION the overall transcription profile between cancer or noncancer- Cancer cachexia is a multifactorial metabolic syndrome that affects induced muscle atrophy will be greatly helpful in revealing the approximately 50–80% of cancer patients, especially in the uniqueness of the disease.
    [Show full text]
  • DDIT4 Antibody (N-Term) Purified Rabbit Polyclonal Antibody (Pab) Catalog # AP6268A
    10320 Camino Santa Fe, Suite G San Diego, CA 92121 Tel: 858.875.1900 Fax: 858.622.0609 DDIT4 Antibody (N-term) Purified Rabbit Polyclonal Antibody (Pab) Catalog # AP6268A Specification DDIT4 Antibody (N-term) - Product Information Application WB, IHC-P,E Primary Accession Q9NX09 Reactivity Human Host Rabbit Clonality Polyclonal Isotype Rabbit Ig Antigen Region 20-49 DDIT4 Antibody (N-term) - Additional Information Gene ID 54541 Other Names DNA damage-inducible transcript 4 protein, HIF-1 responsive protein RTP801, Protein regulated in development and DNA damage Anti-DDIT4 Antibody (N-term) at 1:2000 response 1, REDD-1, DDIT4, REDD1, dilution + Hela whole cell lysate RTP801 Lysates/proteins at 20 µg per lane. Secondary Goat Anti-Rabbit IgG, (H+L), Target/Specificity Peroxidase conjugated at 1/10000 dilution. This DDIT4 antibody is generated from Predicted band size : 25 kDa rabbits immunized with a KLH conjugated Blocking/Dilution buffer: 5% NFDM/TBST. synthetic peptide between 20-49 amino acids from the N-terminal region of human DDIT4. Dilution WB~~1:2000 IHC-P~~1:25 Format Purified polyclonal antibody supplied in PBS with 0.09% (W/V) sodium azide. This antibody is purified through a protein A column, followed by peptide affinity purification. Storage Maintain refrigerated at 2-8°C for up to 2 weeks. For long term storage store at -20°C in small aliquots to prevent freeze-thaw cycles. AP6268a staining DDIT4 in human kidney tissue sections by Immunohistochemistry Precautions (IHC-P - paraformaldehyde-fixed, DDIT4 Antibody (N-term) is for research use paraffin-embedded sections). Tissue was Page 1/3 10320 Camino Santa Fe, Suite G San Diego, CA 92121 Tel: 858.875.1900 Fax: 858.622.0609 only and not for use in diagnostic or fixed with formaldehyde and blocked with 3% therapeutic procedures.
    [Show full text]
  • Downloaded from the UCSC PRO-Seq Library Preparation and Sequencing
    ARTICLE https://doi.org/10.1038/s41467-021-21687-2 OPEN Multi-omics analysis reveals contextual tumor suppressive and oncogenic gene modules within the acute hypoxic response ✉ ✉ Zdenek Andrysik1,2, Heather Bender1,2, Matthew D. Galbraith 1,2 & Joaquin M. Espinosa 1,2,3 Cellular adaptation to hypoxia is a hallmark of cancer, but the relative contribution of hypoxia- inducible factors (HIFs) versus other oxygen sensors to tumorigenesis is unclear. We employ 1234567890():,; a multi-omics pipeline including measurements of nascent RNA to characterize transcrip- tional changes upon acute hypoxia. We identify an immediate early transcriptional response that is strongly dependent on HIF1A and the kinase activity of its cofactor CDK8, includes indirect repression of MYC targets, and is highly conserved across cancer types. HIF1A drives this acute response via conserved high-occupancy enhancers. Genetic screen data indicates that, in normoxia, HIF1A displays strong cell-autonomous tumor suppressive effects through a gene module mediating mTOR inhibition. Conversely, in advanced malignancies, expression of a module of HIF1A targets involved in collagen remodeling is associated with poor prog- nosis across diverse cancer types. In this work, we provide a valuable resource for investi- gating context-dependent roles of HIF1A and its targets in cancer biology. 1 Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA. 2 Linda Crnic Institute for Down Syndrome, School of Medicine, University
    [Show full text]
  • Exploiting the Role of Hypoxia-Inducible Factor 1 and Pseudohypoxia in the Myelodysplastic Syndrome Pathophysiology
    International Journal of Molecular Sciences Article Exploiting the Role of Hypoxia-Inducible Factor 1 and Pseudohypoxia in the Myelodysplastic Syndrome Pathophysiology Ioanna E. Stergiou 1,† , Konstantinos Kambas 2,†, Aikaterini Poulaki 1,3 , Stavroula Giannouli 3,*, Theodora Katsila 4 , Aglaia Dimitrakopoulou 5, Veroniki Vidali 6 , Vasileios Mouchtouris 6 , Ismini Kloukina 7, Evangelia Xingi 8, Stamatis N. Pagakis 9, Lesley Probert 2 , George P. Patrinos 10 , Konstantinos Ritis 11, Athanasios G. Tzioufas 1 and Michael Voulgarelis 1,* 1 Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; [email protected] (I.E.S.); [email protected] (A.P.); [email protected] (A.G.T.) 2 Laboratory of Molecular Genetics, Department of Immunology, Hellenic Pasteur Institute, 11521 Athens, Greece; [email protected] (K.K.); [email protected] (L.P.) 3 Hematology Unit, Second Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece 4 Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; [email protected] 5 Laboratory of Flow Cytometry, Immunology Department, Laiko Hospital of Athens, 11526 Athens, Greece; [email protected] 6 Citation: Stergiou, I.E.; Kambas, K.; Laboratory of Synthesis of Natural Products and Bioorganic Chemistry, Institute of Nanoscience and Poulaki, A.; Giannouli, S.; Katsila, T.; Nanotechnology, National Center for Scientific Research “Demokritos”, 15341 Athens, Greece; Dimitrakopoulou, A.; Vidali, V.; [email protected] (V.V.); [email protected] (V.M.) 7 Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece; Mouchtouris, V.; Kloukina, I.; Xingi, [email protected] E.; et al.
    [Show full text]
  • Downloaded Human (Grch37/Hg19; Nov 9, 2014) And
    bioRxiv preprint doi: https://doi.org/10.1101/177923; this version posted August 18, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. GIC: A computational method for predicting the essentiality of long noncoding lncRNAs Pan Zeng#, Ji Chen#, Yuan Zhou#, Jichun Yang*, and Qinghua Cui* Department of Biomedical Informatics, Department of Physiology and Pathophysiology, Centre for Noncoding RNA Medicine, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, 38 Xueyuan Rd, Beijing, 100191, China # These authors contributed equally to this work. * To whom the correspondence should be addressed: Qinghua Cui, email: [email protected] or Jichun Yang, email: [email protected] bioRxiv preprint doi: https://doi.org/10.1101/177923; this version posted August 18, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. ABSTRACT Measuring the essentiality of genes is critically important in biology and medicine. Some bioinformatic methods have been developed for this issue but none of them can be applied to long noncoding RNAs (lncRNAs), one big class of biological molecules. Here we developed a computational method, GIC (Gene Importance Calculator), which can predict the essentiality of both protein-coding genes and lncRNAs based on RNA sequence information. For identifying the essentiality of protein-coding genes, GIC is competitive with well-established computational scores. More important, GIC showed a high performance for predicting the essentiality of lncRNAs.
    [Show full text]
  • DDIT4 Gene Expression Is Switched on by a New HDAC4 Function in Ataxia Telangiectasia
    Received: 13 August 2019 | Revised: 12 October 2019 | Accepted: 6 November 2019 DOI: 10.1096/fj.201902039R RESEARCH ARTICLE DDIT4 gene expression is switched on by a new HDAC4 function in ataxia telangiectasia Anastasia Ricci | Luca Galluzzi | Mauro Magnani | Michele Menotta Department of Biomolecular Sciences, University of Urbino “Carlo Bo”, Urbino, Abstract Italy Ataxia telangiectasia (AT) is a rare, severe, and ineluctably progressive multisystemic neurodegenerative disease. Histone deacetylase 4 (HDAC4) nuclear accumulation Correspondence Anastasia Ricci, Department of has been related to neurodegeneration in AT. Since treatment with glucocorticoid an- Biomolecular Sciences, University of alogues has been shown to improve the neurological symptoms that characterize this Urbino “Carlo Bo”, Via Saffi 2, 61029 syndrome, the effects of dexamethasone on HDAC4 were investigated. In this paper, Urbino, Italy. Email: [email protected] we describe a novel nonepigenetic function of HDAC4 induced by dexamethasone, through which it can directly modulate HIF-1a activity and promote the upregulation Funding information of the DDIT4 gene and protein expression. This new HDAC4 transcription regula- EC | Horizon 2020 (EU Framework Programme for Research and Innovation), tion mechanism leads to a positive effect on autophagic flux, an AT-compromised Grant/Award Number: 667946; FanoAteneo biological pathway. This signaling was specifically induced by dexamethasone only in AT cell lines and can contribute in explaining the positive effects of
    [Show full text]
  • Up‐Regulation of DDIT4 Predicts Poor Prognosis in Acute Myeloid Leukaemia
    University of Groningen Up-regulation of DDIT4 predicts poor prognosis in acute myeloid leukaemia Cheng, Zhiheng; Dai, Yifeng; Pang, Yifan; Jiao, Yang; Liu, Yan; Cui, Longzhen; Quan, Liang; Qian, Tingting; Zeng, Tiansheng; Si, Chaozeng Published in: Journal of cellular and molecular medicine DOI: 10.1111/jcmm.14831 IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below. Document Version Publisher's PDF, also known as Version of record Publication date: 2019 Link to publication in University of Groningen/UMCG research database Citation for published version (APA): Cheng, Z., Dai, Y., Pang, Y., Jiao, Y., Liu, Y., Cui, L., Quan, L., Qian, T., Zeng, T., Si, C., Huang, W., Chen, J., Pang, Y., Ye, X., Shi, J., & Fu, L. (2019). Up-regulation of DDIT4 predicts poor prognosis in acute myeloid leukaemia. Journal of cellular and molecular medicine. https://doi.org/10.1111/jcmm.14831 Copyright Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons). Take-down policy If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim. Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.
    [Show full text]
  • Induces DNA Damage and Sensitizes Transformed Cells to Anticancer Agents
    Selective inhibition of histone deacetylase 6 (HDAC6) induces DNA damage and sensitizes transformed cells to anticancer agents Mandana Namdar, Gisela Perez, Lang Ngo, and Paul A. Marks1 Cell Biology Program, Sloan-Kettering Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, NY 10065 Contributed by Paul A. Marks, September 23, 2010 (sent for review June 17, 2010) Histone deacetylase 6 (HDAC6) is structurally and functionally HDAC. HDAC6 has two catalytic domains, a ubiquitin-binding unique among the 11 human zinc-dependent histone deacetylases. zinc-finger domain, and a dynein-binding domain, and selectively Here we show that chemical inhibition with the HDAC6-selective deacetylates nonhistone proteins such as tubulin, HSP90, cortac- inhibitor tubacin significantly enhances cell death induced by the tin, and peroxiredoxins (1, 11–13). Selective inhibition of HDAC6 topoisomerase II inhibitors etoposide and doxorubicin and the pan- can affect a number of cellular pathways important in tumori- HDAC inhibitor SAHA (vorinostat) in transformed cells (LNCaP, MCF- genesis. Hyperacetylation of HSP90 in response to HDAC6 in- fi 7), an effect not observed in normal cells (human foreskin broblast hibition reduces the chaperone association with its client proteins, cells). The inactive analogue of tubacin, nil-tubacin, does not resulting in polyubiquitination and proteasomal degradation of a sensitize transformed cells to these anticancer agents. Further, we number of HSP90 substrates (13). HDAC6 inhibition enhances show that down-regulation of HDAC6 expression by shRNA in α-tubulin acetylation, which stabilizes microtubules and is of- LNCaP cells enhances cell death induced by etoposide, doxorubicin, ten associated with reduced cell movement (14).
    [Show full text]
  • DDIT4/REDD1/RTP801 Is a Novel Negative Regulator of Schwann Cell Myelination
    The Journal of Neuroscience, September 18, 2013 • 33(38):15295–15305 • 15295 Cellular/Molecular DDIT4/REDD1/RTP801 Is a Novel Negative Regulator of Schwann Cell Myelination Roberta Noseda,1,2 Sophie Belin,1 Franc¸oise Piguet,1 Ilaria Vaccari,1 Stefania Scarlino,1 Paola Brambilla,3 Filippo Martinelli Boneschi,3 Maria Laura Feltri,4 Lawrence Wrabetz,4 Angelo Quattrini,3 Elena Feinstein,5 Richard L. Huganir,6 and Alessandra Bolino1 1Dulbecco Telethon Institute at Institute of Experimental Neurology (INSPE), San Raffaele Scientific Institute, 20132 Milan, Italy, 2UniSR, Vita Salute San Raffaele University, 20132 Milan, Italy, 3Institute of Experimental Neurology (INSPE), San Raffaele Scientific Institute, 20132 Milan, Italy, 4Hunter James Kelly Research Institute, University at Buffalo, State University of New York, Buffalo, New York 14203, 5Quark Pharmaceuticals, 70400 Ness Ziona, Israel, and 6The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205 Signals that promote myelination must be tightly modulated to adjust myelin thickness to the axonal diameter. In the peripheral nervous system, axonal neuregulin 1 type III promotes myelination by activating erbB2/B3 receptors and the PI3K/AKT/mTOR pathway in Schwann cells. Conversely, PTEN (phosphatase and tensin homolog on chromosome 10) dephosphorylates PtdIns(3,4,5)P3 and nega- tively regulates the AKT pathway and myelination. Recently, the DLG1/SAP97 scaffolding protein was described to interact with PTEN to enhance PIP3 dephosphorylation. Here we now report that nerves from mice with conditional inactivation of Dlg1 in Schwann cells display only a transient increase in myelin thickness during development, suggesting that DLG1 is a transient negative regulator of myelination. Instead, we identified DDIT4/RTP801/REDD1 as a sustained negative modulator of myelination.
    [Show full text]