151
|
Viollet C, Davis DA, Tekeste SS, Reczko M, Ziegelbauer JM, Pezzella F, Ragoussis J, Yarchoan R. RNA Sequencing Reveals that Kaposi Sarcoma-Associated Herpesvirus Infection Mimics Hypoxia Gene Expression Signature. PLoS Pathog 2017; 13:e1006143. [PMID: 28046107 PMCID: PMC5234848 DOI: 10.1371/journal.ppat.1006143] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 01/13/2017] [Accepted: 12/19/2016] [Indexed: 01/09/2023] Open
Abstract
Kaposi sarcoma-associated herpesvirus (KSHV) causes several tumors and hyperproliferative disorders. Hypoxia and hypoxia-inducible factors (HIFs) activate latent and lytic KSHV genes, and several KSHV proteins increase the cellular levels of HIF. Here, we used RNA sequencing, qRT-PCR, Taqman assays, and pathway analysis to explore the miRNA and mRNA response of uninfected and KSHV-infected cells to hypoxia, to compare this with the genetic changes seen in chronic latent KSHV infection, and to explore the degree to which hypoxia and KSHV infection interact in modulating mRNA and miRNA expression. We found that the gene expression signatures for KSHV infection and hypoxia have a 34% overlap. Moreover, there were considerable similarities between the genes up-regulated by hypoxia in uninfected (SLK) and in KSHV-infected (SLKK) cells. hsa-miR-210, a HIF-target known to have pro-angiogenic and anti-apoptotic properties, was significantly up-regulated by both KSHV infection and hypoxia using Taqman assays. Interestingly, expression of KSHV-encoded miRNAs was not affected by hypoxia. These results demonstrate that KSHV harnesses a part of the hypoxic cellular response and that a substantial portion of hypoxia-induced changes in cellular gene expression are induced by KSHV infection. Therefore, targeting hypoxic pathways may be a useful way to develop therapeutic strategies for KSHV-related diseases.
Collapse
Affiliation(s)
- Coralie Viollet
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - David A. Davis
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Shewit S. Tekeste
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Martin Reczko
- Institute of Molecular Oncology, Alexander Fleming Biomedical Sciences Research Center, Vari, Greece
| | - Joseph M. Ziegelbauer
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Francesco Pezzella
- Nuffield Division of Clinical Laboratory Sciences, University of Oxford, Oxford, United Kingdom
| | - Jiannis Ragoussis
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Institute of Molecular Oncology, Alexander Fleming Biomedical Sciences Research Center, Vari, Greece
- McGill University and Génome Québec Innovation Centre, Montréal, Québec, Canada
- Department of Biochemistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Robert Yarchoan
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
152
|
Lipina C, Hundal HS. Is REDD1 a Metabolic Éminence Grise? Trends Endocrinol Metab 2016; 27:868-880. [PMID: 27613400 PMCID: PMC5119498 DOI: 10.1016/j.tem.2016.08.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 08/04/2016] [Accepted: 08/09/2016] [Indexed: 01/31/2023]
Abstract
Regulated in development and DNA damage response 1 (REDD1) has been functionally linked to the control of diverse cellular processes due, at least in part, to its ability to repress mammalian or mechanistic Target of Rapamycin (mTOR) Complex-1 (mTORC1), a key protein complex controlled by hormonal and nutrient cues. Notably, emerging evidence suggests that REDD1 also regulates several pathways involved in modulating energy balance and metabolism. Herein, we discuss evidence implicating REDD1 as a key modulator of insulin action and metabolic function, including its potential contribution to mitochondrial biology and pancreatic islet function. Collectively, the available evidence suggests that REDD1 has a more prominent role in energy homeostasis than was previously thought, and implicates REDD1 as a potential therapeutic target for treatment of metabolic disorders.
Collapse
Affiliation(s)
- Christopher Lipina
- Division of Cell Signalling and Immunology, Sir James Black Centre, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - Harinder S Hundal
- Division of Cell Signalling and Immunology, Sir James Black Centre, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK.
| |
Collapse
|
153
|
Zhao H, Rieger S, Abe K, Hewison M, Lisse TS. DNA Damage-Inducible Transcript 4 Is an Innate Surveillant of Hair Follicular Stress in Vitamin D Receptor Knockout Mice and a Regulator of Wound Re-Epithelialization. Int J Mol Sci 2016; 17:ijms17121984. [PMID: 27898044 PMCID: PMC5187784 DOI: 10.3390/ijms17121984] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 11/07/2016] [Accepted: 11/22/2016] [Indexed: 01/06/2023] Open
Abstract
Mice and human patients with impaired vitamin D receptor (VDR) signaling have normal developmental hair growth but display aberrant post-morphogenic hair cycle progression associated with alopecia. In addition, VDR–/– mice exhibit impaired cutaneous wound healing. We undertook experiments to determine whether the stress-inducible regulator of energy homeostasis, DNA damage-inducible transcript 4 (Ddit4), is involved in these processes. By analyzing hair cycle activation in vivo, we show that VDR−/− mice at day 14 exhibit increased Ddit4 expression within follicular stress compartments. At day 29, degenerating VDR−/− follicular keratinocytes, but not bulge stem cells, continue to exhibit an increase in Ddit4 expression. At day 47, when normal follicles and epidermis are quiescent and enriched for Ddit4, VDR−/− skin lacks Ddit4 expression. In a skin wound healing assay, the re-epithelialized epidermis in wildtype (WT) but not VDR−/− animals harbor a population of Ddit4- and Krt10-positive cells. Our study suggests that VDR regulates Ddit4 expression during epidermal homeostasis and the wound healing process, while elevated Ddit4 represents an early growth-arresting stress response within VDR−/− follicles.
Collapse
Affiliation(s)
- Hengguang Zhao
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Sandra Rieger
- Kathryn W. Davis Center for Regenerative Biology and Medicine, Mount Desert Island Biological Laboratory, 159 Old Bar Harbor Road, Salisbury Cove, ME 04672, USA.
| | - Koichiro Abe
- Department of Molecular Life Science, Tokai University School of Medicine, Isehara, Kanagawa 259-1193, Japan.
| | - Martin Hewison
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, The University of Birmingham, Birmingham B15 2TH, UK.
| | - Thomas S Lisse
- Kathryn W. Davis Center for Regenerative Biology and Medicine, Mount Desert Island Biological Laboratory, 159 Old Bar Harbor Road, Salisbury Cove, ME 04672, USA.
- The Jackson Laboratory, Bar Harbor, ME 04609, USA.
| |
Collapse
|
154
|
Daijo H, Hoshino Y, Kai S, Suzuki K, Nishi K, Matsuo Y, Harada H, Hirota K. Cigarette smoke reversibly activates hypoxia-inducible factor 1 in a reactive oxygen species-dependent manner. Sci Rep 2016; 6:34424. [PMID: 27680676 PMCID: PMC5041075 DOI: 10.1038/srep34424] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 09/13/2016] [Indexed: 11/12/2022] Open
Abstract
Cigarette smoke (CS) is a major contributor to the development of a large number of fatal and debilitating disorders. However, the precise molecular mechanisms underlying the effects of CS in lung disease are largely unknown. To elucidate these pathophysiological processes, we examined the in vitro and in vivo effects of CS extract (CSE) and CS on the transcription factor, hypoxia-inducible factor 1 (HIF-1). CSE induced concentration- and time-dependent accumulation of HIF-1α protein in human lung epithelial-like cells under non-hypoxic conditions. Genes upregulated by HIF-1, including vascular endothelial growth factor and regulated in development and DNA damage response 1, both of which are involved in smoking-induced emphysematous changes, were increased by CSE treatment under non-hypoxic conditions in vitro and in vivo. Further investigation revealed that reactive oxygen species were generated in cells exposed to CSE and were required for CSE-mediated induction of HIF-1α protein, as was activation of phosphoinositide 3-kinase and mitogen-activated protein kinase pathways. In conclusion, we demonstrated that CSE and CS induced HIF-1 activation in vitro and in vivo, respectively. The evidence warrants further investigation to indicate that HIF-1 plays an important role in CS-induced gene expression, which is deeply involved in pulmonary cellular stress and small airway remodelling.
Collapse
Affiliation(s)
- Hiroki Daijo
- Department of Anesthesia, Kyoto University Hospital, Kyoto, Japan
| | - Yuma Hoshino
- Department of Respiratory Medicine, Kyoto University Hospital, Kyoto, Japan
| | - Shinichi Kai
- Department of Anesthesiology, Kansai Medical University, Hirakata, Japan
| | - Kengo Suzuki
- Department of Anesthesiology, Kansai Medical University, Hirakata, Japan
| | - Kenichiro Nishi
- Department of Anesthesiology, Kansai Medical University, Hirakata, Japan
| | - Yoshiyuki Matsuo
- Department of Anesthesiology, Kansai Medical University, Hirakata, Japan
| | - Hiroshi Harada
- Laboratory of Cancer Cell Biology, Radiation Biology Center, Kyoto University, Kyoto, Japan.,Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency (JST), Saitama, Japan
| | - Kiichi Hirota
- Department of Anesthesiology, Kansai Medical University, Hirakata, Japan
| |
Collapse
|
155
|
Alvarez-Garcia O, Olmer M, Akagi R, Akasaki Y, Fisch KM, Shen T, Su AI, Lotz MK. Suppression of REDD1 in osteoarthritis cartilage, a novel mechanism for dysregulated mTOR signaling and defective autophagy. Osteoarthritis Cartilage 2016; 24:1639-47. [PMID: 27118398 PMCID: PMC4992644 DOI: 10.1016/j.joca.2016.04.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 04/09/2016] [Accepted: 04/18/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Aging is a main risk factor for the development of osteoarthritis (OA) and the molecular mechanisms underlying the aging-related changes in articular cartilage include increased mammalian target of rapamycin (mTOR) signaling and defective autophagy. REDD1 is an endogenous inhibitor of mTOR that regulates cellular stress responses. In this study we measured REDD1 expression in normal, aged and OA cartilage and assessed REDD1 function in human and mouse articular chondrocytes. METHODS REDD1 expression was analyzed in human and mouse articular cartilage by qPCR, western blotting, and immunohistochemistry. For functional studies, REDD1 and TXNIP knockdown or overexpression was performed in chondrocytes in the presence or absence of rapamycin and chloroquine, and mTOR signaling and autophagy were measured by western blotting. REDD1/TXNIP protein interaction was assessed by co-immunoprecipitation experiments. RESULTS Human and mouse cartilage from normal knee joints expressed high levels of REDD1. REDD1 expression was significantly reduced in aged and OA cartilage. In cultured chondrocytes, REDD1 knockdown increased whereas REDD1 overexpression decreased mTOR signaling. In addition, REDD1 activated autophagy by an mTOR independent mechanism that involved protein/protein interaction with TXNIP. The REDD1/TXNIP complex was required for autophagy activation in chondrocytes. CONCLUSION The present study shows that REDD1 is highly expressed in normal human articular cartilage and reduced during aging and OA. REDD1 in human chondrocytes negatively regulates mTOR activity and is essential for autophagy activation. Reduced REDD1 expression thus represents a novel mechanism for the increased mTOR activation and defective autophagy observed in OA.
Collapse
Affiliation(s)
- Oscar Alvarez-Garcia
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Merissa Olmer
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Ryuichiro Akagi
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA,Department of Orthopaedic Surgery, School of Medicine, Chiba University, 1-8-1, Inohana, Chuou, Chiba, 260-8677, Japan
| | - Yukio Akasaki
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Kathleen M. Fisch
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Tao Shen
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Andrew I. Su
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Martin K. Lotz
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
156
|
Regulation of hypoxia-inducible factor-1α, regulated in development and DNA damage response-1 and mammalian target of rapamycin in human placental BeWo cells under hypoxia. Placenta 2016; 45:24-31. [DOI: 10.1016/j.placenta.2016.07.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 07/04/2016] [Accepted: 07/12/2016] [Indexed: 12/17/2022]
|
157
|
Song C, Xu Q, Jiang K, Zhou G, Yu X, Wang L, Zhu Y, Fang L, Yu Z, Lee JD, Yu SC, Yang Q. Inhibition of BMK1 pathway suppresses cancer stem cells through BNIP3 and BNIP3L. Oncotarget 2016; 6:33279-89. [PMID: 26432836 PMCID: PMC4741765 DOI: 10.18632/oncotarget.5337] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 09/17/2015] [Indexed: 12/22/2022] Open
Abstract
Cancer stem cells (CSCs) possess many characteristics associated with stem cells and are believed to drive tumor initiation. Although targeting of CSCs offers great promise for the new generation of therapeutics, lack of the effective drugable target and appropriate pharmacological reagents significantly impedes the development of chemotherapies. Here, we show that the phosphorylation of BMK1 was significantly correlated with not only embryonic and induced pluripotent stem (iPS) cells, but also the CSCs. It was showed that activation of BMK1 by the expression of MEK5D enhanced the self-renew (sphere formation), proliferation (clone formation) and tumorigenic capacity of CSCs. While BMK1 inhibitor, XMD8-92, suppressed these capacities. RNA-seq and microarray analysis revealed that inhibition of BMK1 significantly enhanced the expression of BNIP3 and BNIP3L, which play important roles in cell death. Further study indicated that shRNA-mediated knock down of BNIP3 and BNIP3L impairs the BMK1 inhibitor, XMD8-92-induced suppression of sphere formation and clone formation of CSC. Collectively, these results not only indicate that BMK1 plays an important role in maintaining "stemness" of CSCs, but also implicate that BMK1 might be a potential drug target for CSCs.
Collapse
Affiliation(s)
- Chengli Song
- Department of Oncology, The Second Affiliated Hospital of DaLian Medical University, Institute of Cancer Stem Cell, DaLian Medical University, Dalian, Liaoning 116044, China
| | - Qiang Xu
- Department of Oncology, The Second Affiliated Hospital of DaLian Medical University, Institute of Cancer Stem Cell, DaLian Medical University, Dalian, Liaoning 116044, China
| | - Kui Jiang
- Department of Oncology, The Second Affiliated Hospital of DaLian Medical University, Institute of Cancer Stem Cell, DaLian Medical University, Dalian, Liaoning 116044, China
| | - Guangyu Zhou
- Department of Oncology, The Second Affiliated Hospital of DaLian Medical University, Institute of Cancer Stem Cell, DaLian Medical University, Dalian, Liaoning 116044, China
| | - Xuebin Yu
- Department of Oncology, The Second Affiliated Hospital of DaLian Medical University, Institute of Cancer Stem Cell, DaLian Medical University, Dalian, Liaoning 116044, China
| | - Lina Wang
- Department of Oncology, The Second Affiliated Hospital of DaLian Medical University, Institute of Cancer Stem Cell, DaLian Medical University, Dalian, Liaoning 116044, China
| | - Yuting Zhu
- Department of Oncology, The Second Affiliated Hospital of DaLian Medical University, Institute of Cancer Stem Cell, DaLian Medical University, Dalian, Liaoning 116044, China
| | - Liping Fang
- Department of Oncology, The Second Affiliated Hospital of DaLian Medical University, Institute of Cancer Stem Cell, DaLian Medical University, Dalian, Liaoning 116044, China
| | - Zhe Yu
- Department of Oncology, The Second Affiliated Hospital of DaLian Medical University, Institute of Cancer Stem Cell, DaLian Medical University, Dalian, Liaoning 116044, China
| | - Jiing-Dwan Lee
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Shi-Cang Yu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Qingkai Yang
- Department of Oncology, The Second Affiliated Hospital of DaLian Medical University, Institute of Cancer Stem Cell, DaLian Medical University, Dalian, Liaoning 116044, China
| |
Collapse
|
158
|
Madan E, Dikshit B, Gowda SH, Srivastava C, Sarkar C, Chattopadhyay P, Sinha S, Chosdol K. FAT1 is a novel upstream regulator of HIF1α and invasion of high grade glioma. Int J Cancer 2016; 139:2570-82. [PMID: 27536856 PMCID: PMC6585695 DOI: 10.1002/ijc.30386] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 06/13/2016] [Accepted: 06/23/2016] [Indexed: 01/15/2023]
Abstract
The hypoxic microenvironment is an important contributor of glioblastoma (GBM) aggressiveness via HIF1α, while tumour inflammation is profoundly influenced by FAT Atypical Cadherin (FAT1). This study was designed to explore the functional interaction and significance of FAT1 and HIF1α under severe hypoxia‐mimicking tumour microenvironment in primary human tumours. We first identified a positive correlation of FAT1 with HIF1α and its target genes in GBM tumour specimens, revealing the significance of the FAT1‐HIF1α axis in glioma cells. We found that severe hypoxia leads to an increased expression of FAT1 and HIF1α in U87MG and U373MG cells. To reveal the relevance of FAT1 under hypoxic conditions, we depleted endogenous FAT1 under hypoxia and found a substantial reduction in the expression of HIF1α and its downstream target genes like CA9, GLUT1, VEGFA, MCT4, HK2, BNIP3 and REDD1, as well as a significant reduction in the invasiveness in GBM cells. At the molecular level, under hypoxia the FAT1 depletion‐associated reduction in HIF1α was due to compromised EGFR‐Akt signaling as well as increased VHL‐dependent proteasomal degradation of HIF1α. In brief, for the first time, these results reveal an upstream master regulatory role of FAT1 in the expression and role of HIF1α under hypoxic conditions and that FAT1‐HIF1α axis controls the invasiveness of GBM. Hence, FAT1 represents a novel potential therapeutic target for GBM. What's new? The hypoxic microenvironment is an important contributor of glioblastoma aggressiveness via HIF1α while tumor inflammation is profoundly influenced by FAT Atypical Cadherin (FAT1). This study explores the functional interaction of FAT1 and HIF1α in severe hypoxia‐mimicking tumor microenvironments. The results show that FAT1 upregulation is critical for enhancing and maintaining high HIFIα levels in tumors with severe hypoxia. FAT1 both increases HIFIα transcription and decreases HIFIα degradation in glioblastoma multiforme cell lines under hypoxic conditions. With FAT1 regulating the activity of HIF1α under hypoxic condition and the FAT1‐HIF1α axis controlling the invasiveness of glioblastoma, FAT1 represents a novel potential therapeutic target for glioblastomas.
Collapse
Affiliation(s)
- Evanka Madan
- Department of Biochemistry, AIIMS, New Delhi, India
| | | | | | | | | | | | - Subrata Sinha
- Department of Biochemistry, AIIMS, New Delhi, India. .,Director, National Brain Research Center, Manesar, Gurgaon, India.
| | | |
Collapse
|
159
|
Bridges between mitochondrial oxidative stress, ER stress and mTOR signaling in pancreatic β cells. Cell Signal 2016; 28:1099-104. [DOI: 10.1016/j.cellsig.2016.05.007] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 05/05/2016] [Accepted: 05/06/2016] [Indexed: 02/06/2023]
|
160
|
Gordon BS, Steiner JL, Williamson DL, Lang CH, Kimball SR. Emerging role for regulated in development and DNA damage 1 (REDD1) in the regulation of skeletal muscle metabolism. Am J Physiol Endocrinol Metab 2016; 311:E157-74. [PMID: 27189933 PMCID: PMC4967146 DOI: 10.1152/ajpendo.00059.2016] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 05/11/2016] [Indexed: 12/25/2022]
Abstract
Since its discovery, the protein regulated in development and DNA damage 1 (REDD1) has been implicated in the cellular response to various stressors. Most notably, its role as a repressor of signaling through the central metabolic regulator, the mechanistic target of rapamycin in complex 1 (mTORC1) has gained considerable attention. Not surprisingly, changes in REDD1 mRNA and protein have been observed in skeletal muscle under various physiological conditions (e.g., nutrient consumption and resistance exercise) and pathological conditions (e.g., sepsis, alcoholism, diabetes, obesity) suggesting a role for REDD1 in regulating mTORC1-dependent skeletal muscle protein metabolism. Our understanding of the causative role of REDD1 in skeletal muscle metabolism is increasing mostly due to the availability of genetically modified mice in which the REDD1 gene is disrupted. Results from such studies provide support for an important role for REDD1 in the regulation of mTORC1 as well as reveal unexplored functions of this protein in relation to other aspects of skeletal muscle metabolism. The goal of this work is to provide a comprehensive review of the role of REDD1 (and its paralog REDD2) in skeletal muscle during both physiological and pathological conditions.
Collapse
Affiliation(s)
- Bradley S Gordon
- Institute of Exercise Physiology and Wellness, The University of Central Florida, Orlando, Florida;
| | - Jennifer L Steiner
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and
| | - David L Williamson
- Department of Exercise and Nutrition Sciences, University at Buffalo, Buffalo, New York
| | - Charles H Lang
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and
| | - Scot R Kimball
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and
| |
Collapse
|
161
|
Guo J, Shi L, Gong X, Jiang M, Yin Y, Zhang X, Yin H, Li H, Emori C, Sugiura K, Eppig JJ, Su YQ. Oocyte-dependent activation of MTOR in cumulus cells controls the development and survival of cumulus-oocyte complexes. J Cell Sci 2016; 129:3091-103. [PMID: 27358481 DOI: 10.1242/jcs.182642] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 06/28/2016] [Indexed: 01/12/2023] Open
Abstract
Communication between oocytes and their companion somatic cells promotes the healthy development of ovarian follicles, which is crucial for producing oocytes that can be fertilized and are competent to support embryogenesis. However, how oocyte-derived signaling regulates these essential processes remains largely undefined. Here, we demonstrate that oocyte-derived paracrine factors, particularly GDF9 and GDF9-BMP15 heterodimer, promote the development and survival of cumulus-cell-oocyte complexes (COCs), partly by suppressing the expression of Ddit4l, a negative regulator of MTOR, and enabling the activation of MTOR signaling in cumulus cells. Cumulus cells expressed less Ddit4l mRNA and protein than mural granulosa cells, which is in striking contrast to the expression of phosphorylated RPS6 (a major downstream effector of MTOR). Knockdown of Ddit4l activated MTOR signaling in cumulus cells, whereas inhibition of MTOR in COCs compromised oocyte developmental competence and cumulus cell survival, with the latter likely to be attributable to specific changes in a subset of transcripts in the transcriptome of COCs. Therefore, oocyte suppression of Ddit4l expression allows for MTOR activation in cumulus cells, and this oocyte-dependent activation of MTOR signaling in cumulus cells controls the development and survival of COCs.
Collapse
Affiliation(s)
- Jing Guo
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, People's Republic of China
| | - Lanying Shi
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, People's Republic of China
| | - Xuhong Gong
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, People's Republic of China
| | - Mengjie Jiang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, People's Republic of China
| | - Yaoxue Yin
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, People's Republic of China
| | - Xiaoyun Zhang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, People's Republic of China
| | - Hong Yin
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, People's Republic of China
| | - Hui Li
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, People's Republic of China
| | - Chihiro Emori
- Department of Animal Resource Sciences, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Koji Sugiura
- Department of Animal Resource Sciences, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - John J Eppig
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - You-Qiang Su
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, People's Republic of China Key Laboratory of Model Animal Research, Nanjing Medical University, Nanjing, Jiangsu 211166, People's Republic of China
| |
Collapse
|
162
|
Sensing the Environment Through Sestrins: Implications for Cellular Metabolism. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 327:1-42. [PMID: 27692174 DOI: 10.1016/bs.ircmb.2016.05.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Sestrins are a family of stress-responsive genes that have evolved to attenuate damage induced by stress caused to the cell. By virtue of their antioxidant activity, protein products of Sestrin genes prevent the accumulation of reactive oxygen species within the cell, thereby attenuating the detrimental effects of oxidative stress. In parallel, Sestrins participate in several signaling pathways that control the activity of the target of rapamycin protein kinase (TOR). TOR is a crucial sensor of intracellular and extracellular conditions that promotes cell growth and anabolism when nutrients and growth factors are abundant. In addition to reacting to stress-inducing insults, Sestrins also monitor the changes in the availability of nutrients, which allows them to serve as a key checkpoint for the TOR-regulated signaling pathways. In this review, we will discuss how Sestrins integrate signals from numerous stress- and nutrient-responsive signaling pathways to orchestrate cellular metabolism and support cell viability.
Collapse
|
163
|
Caloric Restriction Normalizes Obesity-Induced Alterations on Regulators of Skeletal Muscle Growth Signaling. Lipids 2016; 51:905-12. [PMID: 27289530 DOI: 10.1007/s11745-016-4168-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 05/17/2016] [Indexed: 10/21/2022]
Abstract
The objective of this study was to establish the impact of caloric restriction on high fat diet-induced alterations on regulators of skeletal muscle growth. We hypothesized that caloric restriction would reverse the negative effects of high fat diet-induced obesity on REDD1 and mTOR-related signaling. Following an initial 8 week period of HF diet-induced obesity, caloric restriction (CR ~30 %) was employed while mice continued to consume either a low (LF) or high fat (HF) diet for 8 weeks. Western analysis of skeletal muscle showed that CR reduced (p < 0.05) the obesity-related effects on the lipogenic protein, SREBP1. Likewise, CR reduced (p < 0.05) the obesity-related effects on the hyperactivation of mTORC1 and ERK1/2 signaling to levels comparable to the LF mice. CR also reduced (p < 0.05) obesity-induced expression of negative regulators of growth, REDD1 and cleaved caspase 3. These findings have implications for on the reversibility of dysregulated growth signaling in obese skeletal muscle, using short-term caloric restriction.
Collapse
|
164
|
Chen Q, Zhou Y, Richards AM, Wang P. Up-regulation of miRNA-221 inhibits hypoxia/reoxygenation-induced autophagy through the DDIT4/mTORC1 and Tp53inp1/p62 pathways. Biochem Biophys Res Commun 2016; 474:168-174. [DOI: 10.1016/j.bbrc.2016.04.090] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 04/14/2016] [Indexed: 01/16/2023]
|
165
|
Pinno J, Bongartz H, Klepsch O, Wundrack N, Poli V, Schaper F, Dittrich A. Interleukin-6 influences stress-signalling by reducing the expression of the mTOR-inhibitor REDD1 in a STAT3-dependent manner. Cell Signal 2016; 28:907-16. [PMID: 27094713 DOI: 10.1016/j.cellsig.2016.04.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 04/05/2016] [Accepted: 04/09/2016] [Indexed: 11/30/2022]
Abstract
Interleukin 6 (IL-6) is a pleiotropic cytokine and a strong activator of Mammalian Target of Rapamycin (mTOR). In contrast, mTOR activity is negatively regulated by Regulated in Development and DNA Damage Responses 1 (REDD1). Expression of REDD1 is induced by cellular stressors such as glucocorticoids and DNA damaging agents. We show that the expression of basal as well as stress-induced REDD1 is reduced by IL-6. The reduction of REDD1 expression by IL-6 is independent of proteasomal or caspase-mediated degradation of REDD1 protein. Instead, induction of REDD1 mRNA is reduced by IL-6. The regulation of REDD1 expression by IL-6 is independent of Phosphatidylinositide-3-Kinase (PI3K) and Mitogen-Activated Protein Kinase (MAPK) signalling but depends on the expression and activation of Signal Transducer and Activator of Transcription 3 (STAT3). Furthermore, the reduction of basal REDD1 expression by IL-6 correlates with IL-6-induced activation of mTOR signalling. Inhibition of STAT3 activation blocks IL-6-induced mTOR activation. In summary, we present a novel STAT3-dependent mechanism of both IL-6-induced activation of mTOR and IL-6-dependent reversion of stress-induced inhibition of mTOR activity.
Collapse
Affiliation(s)
- Jessica Pinno
- Institute of Biology, Department of Systems Biology, Otto-von-Guericke University, Universitätsplatz 2, Gebäude 28, 39106 Magdeburg, Germany.
| | - Hannes Bongartz
- Institute of Biology, Department of Systems Biology, Otto-von-Guericke University, Universitätsplatz 2, Gebäude 28, 39106 Magdeburg, Germany.
| | - Oliver Klepsch
- Institute of Biology, Department of Systems Biology, Otto-von-Guericke University, Universitätsplatz 2, Gebäude 28, 39106 Magdeburg, Germany.
| | - Nicole Wundrack
- Institute of Biology, Department of Systems Biology, Otto-von-Guericke University, Universitätsplatz 2, Gebäude 28, 39106 Magdeburg, Germany.
| | - Valeria Poli
- Molecular Biotechnology Center (MBC), Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy.
| | - Fred Schaper
- Institute of Biology, Department of Systems Biology, Otto-von-Guericke University, Universitätsplatz 2, Gebäude 28, 39106 Magdeburg, Germany.
| | - Anna Dittrich
- Institute of Biology, Department of Systems Biology, Otto-von-Guericke University, Universitätsplatz 2, Gebäude 28, 39106 Magdeburg, Germany.
| |
Collapse
|
166
|
Huang X, Yang K, Zhang Y, Wang Q, Li Y. Quinolinic acid induces cell apoptosis in PC12 cells through HIF-1-dependent RTP801 activation. Metab Brain Dis 2016; 31:435-44. [PMID: 26738727 DOI: 10.1007/s11011-015-9782-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 12/22/2015] [Indexed: 02/01/2023]
Abstract
Neurological disease comprises a series of disorders featuring brain dysfunction and neuronal cell death. Among the factors contributing to neuronal death, excitotoxicity induced by excitatory amino acids, such as glutamate, plays a critical role. However, the mechanisms about how the excitatory amino acids induce neuronal death remain elucidated. In this study, we investigated the role of HIF-1α (hypoxia inducible factor-1α) and RTP801 in cell apoptosis induced by quinolinic acid (QUIN), a glutamatergic agonist, in PC12 cells. We found that QUIN at 5 μM increased the expression of HIF-1α significantly with a peak at 24 h. After the treatment with QUIN (5-20 μM) for 24 h, the cells exhibited decreased viability and cell apoptosis with a concomitant increased expression of apoptosis related proteins. QUIN treatment also induced the generation of intracellular reactive oxygen species and RTP801 up-regulation in a HIF-1α-dependent manner that were inhibited by 2-methoxyestradiol, a HIF-1α inhibitor. Importantly, HIF-1 or RTP801 invalidation by siRNA rescued the cell apoptosis induced by QUIN or cobalt chloride, a chemical inducer of HIF-1. Taken together, these findings support the concept that neurotoxicity induced by QUIN is associated with HIF-1-dependent RTP801 activation and provide insight into the potential of RTP801 inhibitor in treatment of neurological disorders.
Collapse
Affiliation(s)
- Xiaojia Huang
- Department of Pharmacology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China.
| | - Kaiyong Yang
- Department of Pharmacology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Yi Zhang
- Department of Pharmacology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Qiang Wang
- Department of Preventive Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Yongjin Li
- Department of Pharmacology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| |
Collapse
|
167
|
Au A, Singh RP. A multimodal approach to diabetic macular edema. J Diabetes Complications 2016; 30:545-53. [PMID: 26853628 DOI: 10.1016/j.jdiacomp.2015.11.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 11/07/2015] [Accepted: 11/09/2015] [Indexed: 12/27/2022]
Abstract
Diabetic retinopathy is a common complication of uncontrolled diabetes. A complication is diabetic macular edema, which is the leading cause of blindness in patients with diabetic retinopathy. Historically, management of these conditions was laser photocoagulation with regulation of blood pressure, blood sugar, and cholesterol. The initial studies demonstrated that this treatment regimen prevented further visual deterioration but did not improve visual acuity. Novel studies identifying the presence of vascular endothelial growth factor (VEGF) in the eye with accompanying elucidation of diabetic pathophysiology allowed for the development of alternative therapies, namely antibodies against VEGF and corticosteroids. These two therapies revolutionized the management of diabetic macular edema by not only preventing vision loss, but also improving overall vision. In this review, we outline the major breakthroughs and underlying thought processes of the paradigm shifts that have occurred in management of these conditions. Further, we present how the evolving role of anti-inflammatory and anti-VEGF therapies, in a combinatorial approach, may provide further permutations to optimize treatment.
Collapse
Affiliation(s)
- Adrian Au
- Case Western Reserve School of Medicine, 2109 Adelbert Rd. Cleveland, OH 44106
| | - Rishi P Singh
- Cole Eye Institute, Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, OH 44106.
| |
Collapse
|
168
|
Grainger DL, Kutzler L, Rannels SL, Kimball SR. Validation of a commercially available anti-REDD1 antibody using RNA interference and REDD1-/- mouse embryonic fibroblasts. F1000Res 2016; 5:250. [PMID: 27335637 PMCID: PMC4893971 DOI: 10.12688/f1000research.7691.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/29/2016] [Indexed: 01/16/2023] Open
Abstract
REDD1 is a transcriptional target gene of p53 and HIF-1, and an inhibitor of mTOR (mechanistic target of rapamycin) complex 1 (mTORC1)-signaling through PP2A-dependent interaction, making it an important convergence point of both tumor suppression and cell growth pathways. In accordance with this positioning, REDD1 levels are transcriptionally upregulated in response to a variety of cellular stress factors such as nutrient deprivation, hypoxia and DNA damage. In the absence of such conditions, and in particular where growth factor signaling is activated, REDD1 expression is typically negligible; therefore, it is necessary to induce REDD1 prior to experimentation or detection in model systems. Here, we evaluated the performance of a commercially available polyclonal antibody recognizing REDD1 by Western blotting in the presence of thapsigargin, a pharmacological inducer of ER stress well known to upregulate REDD1 protein expression. Further, REDD1 antibody specificity was challenged in HEK-293 cells in the presence of RNA interference and with a REDD1
-/- mouse embryonic fibroblast knockout cell line. Results showed reproducibility and specificity of the antibody, which was upheld in the presence of thapsigargin treatment. We conclude that this antibody can be used to reliably detect REDD1 endogenous expression in samples of both human and mouse origin.
Collapse
Affiliation(s)
| | - Lydia Kutzler
- Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, PA, USA
| | - Sharon L Rannels
- Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, PA, USA
| | - Scot R Kimball
- Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, PA, USA
| |
Collapse
|
169
|
Novelle MG, Ali A, Diéguez C, Bernier M, de Cabo R. Metformin: A Hopeful Promise in Aging Research. Cold Spring Harb Perspect Med 2016; 6:a025932. [PMID: 26931809 DOI: 10.1101/cshperspect.a025932] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Even though the inevitable process of aging by itself cannot be considered a disease, it is directly linked to life span and is the driving force behind all age-related diseases. It is an undisputable fact that age-associated diseases are among the leading causes of death in the world, primarily in industrialized countries. During the last several years, an intensive search of antiaging treatments has led to the discovery of a variety of drugs that promote health span and/or life extension. The biguanide compound metformin is widely used for treating people with type 2 diabetes and appears to show protection against cancer, inflammation, and age-related pathologies. Here, we summarize the recent developments about metformin use in translational aging research and discuss its role as a potential geroprotector.
Collapse
Affiliation(s)
- Marta G Novelle
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, Maryland 21224 Research Center of Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain
| | - Ahmed Ali
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, Maryland 21224
| | - Carlos Diéguez
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain
| | - Michel Bernier
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, Maryland 21224
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, Maryland 21224
| |
Collapse
|
170
|
Kietzmann T, Mennerich D, Dimova EY. Hypoxia-Inducible Factors (HIFs) and Phosphorylation: Impact on Stability, Localization, and Transactivity. Front Cell Dev Biol 2016; 4:11. [PMID: 26942179 PMCID: PMC4763087 DOI: 10.3389/fcell.2016.00011] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 02/08/2016] [Indexed: 12/18/2022] Open
Abstract
The hypoxia-inducible factor α-subunits (HIFα) are key transcription factors in the mammalian response to oxygen deficiency. The HIFα regulation in response to hypoxia occurs primarily on the level of protein stability due to posttranslational hydroxylation and proteasomal degradation. However, HIF α-subunits also respond to various growth factors, hormones, or cytokines under normoxia indicating involvement of different kinase pathways in their regulation. Because these proteins participate in angiogenesis, glycolysis, programmed cell death, cancer, and ischemia, HIFα regulating kinases are attractive therapeutic targets. Although numerous kinases were reported to regulate HIFα indirectly, direct phosphorylation of HIFα affects HIFα stability, nuclear localization, and transactivity. Herein, we review the role of phosphorylation-dependent HIFα regulation with emphasis on protein stability, subcellular localization, and transactivation.
Collapse
Affiliation(s)
- Thomas Kietzmann
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of OuluFinland
| | | | | |
Collapse
|
171
|
Yun SM, Woo SH, Oh ST, Hong SE, Choe TB, Ye SK, Kim EK, Seong MK, Kim HA, Noh WC, Lee JK, Jin HO, Lee YH, Park IC. Melatonin enhances arsenic trioxide-induced cell death via sustained upregulation of Redd1 expression in breast cancer cells. Mol Cell Endocrinol 2016; 422:64-73. [PMID: 26607805 DOI: 10.1016/j.mce.2015.11.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 10/24/2015] [Accepted: 11/12/2015] [Indexed: 10/22/2022]
Abstract
Melatonin is implicated in various physiological functions, including anticancer activity. However, the mechanism(s) of its anticancer activity is not well understood. In the present study, we investigated the combined effects of melatonin and arsenic trioxide (ATO) on cell death in human breast cancer cells. Melatonin enhanced the ATO-induced apoptotic cell death via changes in the protein levels of Survivin, Bcl-2, and Bax, thus affecting cytochrome c release from the mitochondria to the cytosol. Interestingly, we found that the cell death induced by co-treatment with melatonin and ATO was mediated by sustained upregulation of Redd1, which was associated with increased production of reactive oxygen species (ROS). Combined treatment with melatonin and ATO induced the phosphorylation of JNK and p38 MAP kinase downstream from Redd1 expression. Rapamycin and S6K1 siRNA enhanced, while activation of mTORC1 by transfection with TSC2 siRNA suppressed the cell death induced by melatonin and ATO treatment. Taken together, our findings suggest that melatonin enhances ATO-induced apoptotic cell death via sustained upregulation of Redd1 expression and inhibition of mTORC1 upstream of the activation of the p38/JNK pathways in human breast cancer cells.
Collapse
Affiliation(s)
- Sun-Mi Yun
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 139-706, Republic of Korea
| | - Sang Hyeok Woo
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 139-706, Republic of Korea; KIRAMS Radiation Biobank, Korea Institute of Radiological & Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 139-709, Republic of Korea
| | - Sang Taek Oh
- Department of Radiation Oncology, College of Medicine, Yonsei University, Seoul 120-752, Republic of Korea
| | - Sung-Eun Hong
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 139-706, Republic of Korea
| | - Tae-Boo Choe
- Department of Microbiological Engineering, Kon-Kuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 143-701, Republic of Korea
| | - Sang-Kyu Ye
- Department of Pharmacology, Seoul National University College of Medicine, 103 Daehangno, Jongno-gu Seoul 110-799, Republic of Korea
| | - Eun-Kyu Kim
- Department of Surgery, Seoul National University Bundang Hospital, 82 Gumi-ro, 173 Beon-gil, Bundang-gu, Seongnam-si, Gyenggi-do 463-707, Republic of Korea
| | - Min Ki Seong
- Department of Surgery, Korea Cancer Center Hospital, Korea Institute of Radiological & Medical Sciences, 215-4 Gongneung-dong, Nowon-gu, Seoul 139-706, Republic of Korea
| | - Hyun-A Kim
- Department of Surgery, Korea Cancer Center Hospital, Korea Institute of Radiological & Medical Sciences, 215-4 Gongneung-dong, Nowon-gu, Seoul 139-706, Republic of Korea
| | - Woo Chul Noh
- Department of Surgery, Korea Cancer Center Hospital, Korea Institute of Radiological & Medical Sciences, 215-4 Gongneung-dong, Nowon-gu, Seoul 139-706, Republic of Korea
| | - Jin Kyung Lee
- KIRAMS Radiation Biobank, Korea Institute of Radiological & Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 139-709, Republic of Korea
| | - Hyeon-Ok Jin
- KIRAMS Radiation Biobank, Korea Institute of Radiological & Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 139-709, Republic of Korea
| | - Yun-Han Lee
- Department of Radiation Oncology, College of Medicine, Yonsei University, Seoul 120-752, Republic of Korea.
| | - In-Chul Park
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 139-706, Republic of Korea.
| |
Collapse
|
172
|
Effects of DDIT4 in Methamphetamine-Induced Autophagy and Apoptosis in Dopaminergic Neurons. Mol Neurobiol 2016; 54:1642-1660. [PMID: 26873849 DOI: 10.1007/s12035-015-9637-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 12/15/2015] [Indexed: 01/14/2023]
Abstract
Methamphetamine (METH) is an illicit psychoactive drug that can cause a variety of detrimental effects to the nervous system, especially dopaminergic pathways. We hypothesized that DNA damage-inducible transcript 4 (DDIT4) is involved in METH-induced dopaminergic neuronal autophagy and apoptosis. To test the hypothesis, we determined changes of DDIT4 protein expression and the level of autophagy in rat catecholaminergic PC12 cells and human dopaminergic SH-SY5Y cells, and in the hippocampus, prefrontal cortex, and striatum of Sprague Dawley rats exposed to METH. We also examined the effects of silencing DDIT4 expression on METH-induced dopaminergic neuronal autophagy using fluorescence microscopy and electron microscopy. Flow cytometry and Western blot were used to determine apoptosis and the expression of apoptotic markers (cleaved caspase-3 and cleaved PARP) after blocking DDIT4 expression in PC12 cells and SH-SY5Y cells with synthetic siRNA, as well as in the striatum of rats by injecting LV-shDDIT4 lentivirus using a stereotaxic positioning system. Our results showed that METH exposure increased DDIT4 expression that was accompanied with increased autophagy and apoptosis in PC12 cells (3 mM) and SH-SY5Y cells (2 mM), and in the hippocampus, prefrontal cortex, and striatum of rats. Inhibition of DDIT4 expression reduced METH-induced autophagy and apoptosis in vitro and in vivo. However, DDIT4-related effects were not observed at a low concentration of METH (1 μM). These results suggest that DDIT4 plays an essential role in METH-induced dopaminergic neuronal autophagy and apoptosis at higher doses and may be a potential gene target for therapeutics in high-dose METH-induced neurotoxicity.
Collapse
|
173
|
Choi HS, Ahn JH, Park JH, Won MH, Lee CH. Age-dependent changes in the protein expression levels of Redd1 and mTOR in the gerbil hippocampus during normal aging. Mol Med Rep 2016; 13:2409-14. [PMID: 26846432 PMCID: PMC4768963 DOI: 10.3892/mmr.2016.4835] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 12/30/2015] [Indexed: 11/30/2022] Open
Abstract
Redd1, also known as RTP801/Dig2/DDIT4, is a stress-induced protein and a negative regulator of mammalian target of rapamycin (mTOR). Redd1 is also closely associated with oxidative stress and DNA damage. In the present study, age-related changes in the protein expression levels of mTOR and Redd1 were investigated using immunohistochemistry and western blot in the gerbil hippocampus at postnatal month (PM) 3, 6, 12 and 24. No significant differences were identified in the levels of mTOR among the experimental groups, whereas, the levels of phosphorylated mTOR decreased with age. The protein expression levels of Redd1 were observed to gradually increase with age; in the PM 24 group, the level was significantly increased (~189.2%), compared with the PM 3 group. In addition, Redd1 immunoreactivity was significantly increased in the hippocampal principal neurons of the PM 24 group, including the pyramidal cells in the hippocampus proper and granule cells in the dentate gyrus, compared with the other experimental groups. These results demonstrated that the protein expression of Redd1 in the hippocampus was markedly increased during normal aging, indicating that the age-related increase in the expression of Redd1 may be closely associated with age-related hippocampal change.
Collapse
Affiliation(s)
- Hee-Soo Choi
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan, South Chungcheong 31116, Republic of Korea
| | - Ji Hyeon Ahn
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Joon Ha Park
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Choong-Hyun Lee
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan, South Chungcheong 31116, Republic of Korea
| |
Collapse
|
174
|
Iron depletion suppresses mTORC1-directed signalling in intestinal Caco-2 cells via induction of REDD1. Cell Signal 2016; 28:412-424. [PMID: 26827808 PMCID: PMC4804389 DOI: 10.1016/j.cellsig.2016.01.014] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 01/27/2016] [Accepted: 01/28/2016] [Indexed: 12/02/2022]
Abstract
Iron is an indispensable micronutrient that regulates many aspects of cell function, including growth and proliferation. These processes are critically dependent upon signalling via the mammalian or mechanistic target of rapamycin complex 1 (mTORC1). Herein, we test whether iron depletion induced by cell incubation with the iron chelator, deferoxamine (DFO), mediates its effects on cell growth through mTORC1-directed signalling and protein synthesis. We have used Caco-2 cells, a well-established in vitro model of human intestinal epithelia. Iron depletion increased expression of iron-regulated proteins (TfR, transferrin receptor and DMT1, divalent metal transporter, as predicted, but it also promoted a marked reduction in growth and proliferation of Caco-2 cells. This was strongly associated with suppressed mTORC1 signalling, as judged by reduced phosphorylation of mTOR substrates, S6K1 and 4E-BP1, and diminished protein synthesis. The reduction in mTORC1 signalling was tightly coupled with increased expression and accumulation of REDD1 (regulated in DNA damage and development 1) and reduced phosphorylation of Akt and TSC2. The increase in REDD1 abundance was rapidly reversed upon iron repletion of cells but was also attenuated by inhibitors of gene transcription, protein phosphatase 2A (PP2A) and by REDD1 siRNA — strategies that also antagonised the loss in mTORC1 signalling associated with iron depletion. Our findings implicate REDD1 and PP2A as crucial regulators of mTORC1 activity in iron-depleted cells and indicate that their modulation may help mitigate atrophy of the intestinal mucosa that may occur in response to iron deficiency. Cellular iron (Fe) depletion dramatically reduces growth of intestinal Caco-2 cells. mTORC1-directed signalling and protein synthesis are reduced in Fe-depleted cells. Fe deficiency induces expression and gain of REDD1 in a PP2A-dependent manner. PP2A inhibition blocks REDD1 gain and restores mTORC1 activity in Fe-depleted cells.
Collapse
|
175
|
Williamson DL, Dungan CM, Mahmoud AM, Mey JT, Blackburn BK, Haus JM. Aberrant REDD1-mTORC1 responses to insulin in skeletal muscle from Type 2 diabetics. Am J Physiol Regul Integr Comp Physiol 2015; 309:R855-63. [PMID: 26269521 PMCID: PMC4666944 DOI: 10.1152/ajpregu.00285.2015] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 08/04/2015] [Indexed: 11/22/2022]
Abstract
The objective of this study was to establish whether alterations in the REDD1-mTOR axis underlie skeletal muscle insensitivity to insulin in Type 2 diabetic (T2D), obese individuals. Vastus lateralis muscle biopsies were obtained from lean, control and obese, T2D subjects under basal and after a 2-h hyperinsulinemic (40 mU·m(-2)·min(-1))-euglycemic (5 mM) clamp. Muscle lysates were examined for total REDD1, and phosphorylated Akt, S6 kinase 1 (S6K1), 4E-BP1, ERK1/2, and MEK1/2 via Western blot analysis. Under basal conditions [(-) insulin], T2D muscle exhibited higher S6K1 and ERK1/2 and lower 4E-BP1 phosphorylation (P < 0.05), as well as elevations in blood cortisol, glucose, insulin, glycosylated hemoglobin (P < 0.05) vs. lean controls. Following insulin infusion, whole body glucose disposal rates (GDR; mg/kg/min) were lower (P < 0.05) in the T2D vs. the control group. The basal-to-insulin percent change in REDD1 expression was higher (P < 0.05) in muscle from the T2D vs. the control group. Whereas, the basal-to-insulin percent change in muscle Akt, S6K1, ERK1/2, and MEK1/2 phosphorylation was significantly lower (P < 0.05) in the T2D vs. the control group. Findings from this study propose a REDD1-regulated mechanism in T2D skeletal muscle that may contribute to whole body insulin resistance and may be a target to improve insulin action in insulin-resistant individuals.
Collapse
Affiliation(s)
- David L Williamson
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York; and
| | - Cory M Dungan
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York; and
| | - Abeer M Mahmoud
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois
| | - Jacob T Mey
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois
| | - Brian K Blackburn
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois
| | - Jacob M Haus
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
176
|
Wu X, Su J, Chen L, Ma B, Gu X, Zhu L. Ginkgolide B Protects Neurons from Ischemic Injury by Inhibiting the Expression of RTP801. Cell Mol Neurobiol 2015; 35:943-52. [PMID: 25869596 DOI: 10.1007/s10571-015-0189-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 03/25/2015] [Indexed: 01/28/2023]
Abstract
RTP801 (also known as REDD1), a stress-related protein, is induced by several environmental stresses such as ischemia and cigarette smoke. Although ischemia can dramatically up-regulate RTP801 expression in brain ischemia, up to now, the exact relation between RTP801 and neuronal death in ischemia is poorly understood. In the current study, using oxygen and glucose deprivation as an in vitro ischemic model in primary cultured cortical neurons, we found that the expression of RTP801 increased progressively with prolongation of ischemic duration, in which the expression of RTP801 is positively correlated with the release of lactate dehydrogenase (LDH) in neurons, and knockdown of RTP801 promoted neuronal survival in ischemia-reperfusion. It was further found that ginkgolide B (GB) could significantly increase cell viability and decrease LDH release, and at the same time reduce the levels of RTP801 mRNA and protein in neurons after ischemia and reperfusion. Moreover, GB-induced reduction in expression of RTP801 was blocked by application of LY294002, a specific inhibitor of phosphatidylinositol 3-kinase (PI3K). These results demonstrate that RTP801 could play a detrimental role on neurons in ischemia, and GB might protect neurons against ischemic injury by inhibiting RTP801 expression via PI3K pathway.
Collapse
Affiliation(s)
- Xiaomei Wu
- Department of Neurochemistry, Institute for Nautical Medicine, Nantong University, 9 Seyuan Road, Nantong, 226019, China
| | - Jianyou Su
- Medical Laboratory Center, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, China
| | - Lei Chen
- Department of Neurochemistry, Institute for Nautical Medicine, Nantong University, 9 Seyuan Road, Nantong, 226019, China
| | - Baodong Ma
- Department of Neurochemistry, Institute for Nautical Medicine, Nantong University, 9 Seyuan Road, Nantong, 226019, China
| | - Xiaosu Gu
- Department of Neurology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, China
| | - Li Zhu
- Department of Neurochemistry, Institute for Nautical Medicine, Nantong University, 9 Seyuan Road, Nantong, 226019, China.
| |
Collapse
|
177
|
Reuschel EL, Wang J, Shivers DK, Muthumani K, Weiner DB, Ma Z, Finkel TH. REDD1 Is Essential for Optimal T Cell Proliferation and Survival. PLoS One 2015; 10:e0136323. [PMID: 26301899 PMCID: PMC4547781 DOI: 10.1371/journal.pone.0136323] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 08/02/2015] [Indexed: 12/21/2022] Open
Abstract
REDD1 is a highly conserved stress response protein that is upregulated following many types of cellular stress, including hypoxia, DNA damage, energy stress, ER stress, and nutrient deprivation. Recently, REDD1 was shown to be involved in dexamethasone induced autophagy in murine thymocytes. However, we know little of REDD1’s function in mature T cells. Here we show for the first time that REDD1 is upregulated following T cell stimulation with PHA or CD3/CD28 beads. REDD1 knockout T cells exhibit a defect in proliferation and cell survival, although markers of activation appear normal. These findings demonstrate a previously unappreciated role for REDD1 in T cell function.
Collapse
Affiliation(s)
- Emma L. Reuschel
- Division of Rheumatology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
- Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - JiangFang Wang
- Division of Rheumatology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
- Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Debra K. Shivers
- Division of Rheumatology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Karuppiah Muthumani
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - David B. Weiner
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Zhengyu Ma
- Division of Rheumatology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
- Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Terri H. Finkel
- Division of Rheumatology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
- Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
178
|
Lee DK, Kim JH, Kim WS, Jeoung D, Lee H, Ha KS, Won MH, Kwon YG, Kim YM. Lipopolysaccharide induction of REDD1 is mediated by two distinct CREB-dependent mechanisms in macrophages. FEBS Lett 2015; 589:2859-65. [PMID: 26296313 DOI: 10.1016/j.febslet.2015.08.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 07/01/2015] [Accepted: 08/10/2015] [Indexed: 10/23/2022]
Abstract
REDD1 is induced by various cellular stresses; however, its expression in response to lipopolysaccharide (LPS) has not been clearly elucidated in immune cells. LPS stimulated CREB-dependent and NF-κB-independent REDD1 expression in macrophages. Early increases in CREB phosphorylation and REDD1 expression at 8h following LPS treatment were blocked by inhibition of p38MAPK and mitogen- and stress-activated protein kinase 1 (MSK1), but not PKA. However, delayed CREB-mediated REDD1 expression at 16h was suppressed by inhibition of cyclooxygenase-2 (COX-2) and PKA. It indicates that LPS induces REDD1 expression by two distinct CREB-mediated mechanisms, the early p38MAPK/MSK1 and the delayed COX-2/PGE2/PKA pathways.
Collapse
Affiliation(s)
- Dong-Keon Lee
- Departments of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do 200-701, South Korea
| | - Ji-Hee Kim
- Departments of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do 200-701, South Korea
| | - Wan-Sung Kim
- Departments of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do 200-701, South Korea
| | - Dooil Jeoung
- Departments of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, Gangwon-do 200-701, South Korea
| | - Hansoo Lee
- Life Sciences, College of Natural Sciences, Kangwon National University, Chuncheon, Gangwon-do 200-701, South Korea
| | - Kwon-Soo Ha
- Departments of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do 200-701, South Korea
| | - Moo-Ho Won
- Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do 200-701, South Korea
| | - Young-Guen Kwon
- Department of Biochemistry, College of Science and Biotechnology, Yonsei University, Seoul 120-749, South Korea
| | - Young-Myeong Kim
- Departments of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do 200-701, South Korea.
| |
Collapse
|
179
|
Ribosomal Protein S6 Phosphorylation: Four Decades of Research. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2015; 320:41-73. [PMID: 26614871 DOI: 10.1016/bs.ircmb.2015.07.006] [Citation(s) in RCA: 192] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The phosphorylation of ribosomal protein S6 (rpS6) has been described for the first time about four decades ago. Since then, numerous studies have shown that this modification occurs in response to a wide variety of stimuli on five evolutionarily conserved serine residues. However, despite a large body of information on the respective kinases and the signal transduction pathways, the physiological role of rpS6 phosphorylation remained obscure until genetic manipulations were applied in both yeast and mammals in an attempt to block this modification. Thus, studies based on both mice and cultured cells subjected to disruption of the genes encoding rpS6 and the respective kinases, as well as the substitution of the phosphorylatable serine residues in rpS6, have laid the ground for the elucidation of the multiple roles of this protein and its posttranslational modification. This review focuses primarily on newly identified kinases that phosphorylate rpS6, pathways that transduce various signals into rpS6 phosphorylation, and the recently established physiological functions of this modification. It should be noted, however, that despite the significant progress made in the last decade, the molecular mechanism(s) underlying the diverse effects of rpS6 phosphorylation on cellular and organismal physiology are still poorly understood.
Collapse
|
180
|
Baida G, Bhalla P, Kirsanov K, Lesovaya E, Yakubovskaya M, Yuen K, Guo S, Lavker RM, Readhead B, Dudley JT, Budunova I. REDD1 functions at the crossroads between the therapeutic and adverse effects of topical glucocorticoids. EMBO Mol Med 2015; 7:42-58. [PMID: 25504525 PMCID: PMC4309667 DOI: 10.15252/emmm.201404601] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Cutaneous atrophy is the major adverse effect of topical glucocorticoids; however, its molecular mechanisms are poorly understood. Here, we identify stress-inducible mTOR inhibitor REDD1 (regulated in development and DNA damage response 1) as a major molecular target of glucocorticoids, which mediates cutaneous atrophy. In REDD1 knockout (KO) mice, all skin compartments (epidermis, dermis, subcutaneous fat), epidermal stem, and progenitor cells were protected from atrophic effects of glucocorticoids. Moreover, REDD1 knockdown resulted in similar consequences in organotypic raft cultures of primary human keratinocytes. Expression profiling revealed that gene activation by glucocorticoids was strongly altered in REDD1 KO epidermis. In contrast, the down-regulation of genes involved in anti-inflammatory glucocorticoid response was strikingly similar in wild-type and REDD1 KO mice. Integrative bioinformatics analysis of our and published gene array data revealed similar changes of gene expression in epidermis and in muscle undergoing glucocorticoid-dependent and glucocorticoid-independent atrophy. Importantly, the lack of REDD1 did not diminish the anti-inflammatory effects of glucocorticoids in preclinical model. Our findings suggest that combining steroids with REDD1 inhibitors may yield a novel, safer glucocorticoid-based therapies.
Collapse
Affiliation(s)
- Gleb Baida
- Department of Dermatology, Northwestern University, Chicago, IL, USA
| | - Pankaj Bhalla
- Department of Dermatology, Northwestern University, Chicago, IL, USA
| | | | | | | | - Kit Yuen
- Department of Dermatology, Northwestern University, Chicago, IL, USA
| | - Shuchi Guo
- Department of Dermatology, Northwestern University, Chicago, IL, USA
| | - Robert M Lavker
- Department of Dermatology, Northwestern University, Chicago, IL, USA
| | - Ben Readhead
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joel T Dudley
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Irina Budunova
- Department of Dermatology, Northwestern University, Chicago, IL, USA
| |
Collapse
|
181
|
Asker N, Carney Almroth B, Albertsson E, Coltellaro M, Bignell JP, Hanson N, Scarcelli V, Fagerholm B, Parkkonen J, Wijkmark E, Frenzilli G, Förlin L, Sturve J. A gene to organism approach--assessing the impact of environmental pollution in eelpout (Zoarces viviparus) females and larvae. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2015; 34:1511-1523. [PMID: 25663503 PMCID: PMC5008212 DOI: 10.1002/etc.2921] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 10/26/2014] [Accepted: 02/01/2015] [Indexed: 06/04/2023]
Abstract
A broad biomarker approach was applied to study the effects of marine pollution along the Swedish west coast using the teleost eelpout (Zoarces viviparus) as the sentinel species. Measurements were performed on different biological levels, from the molecular to the organismal, including measurements of messenger RNA (mRNA), proteins, cellular and tissue changes, and reproductive success. Results revealed that eelpout captured in Stenungsund had significantly higher hepatic ethoxyresorufin O-deethylase activity, high levels of both cytochrome P4501A and diablo homolog mRNA, and high prevalence of dead larvae and nuclear damage in erythrocytes. Eelpout collected in Göteborg harbor displayed extensive macrovesicular steatosis, whereby the majority of hepatocytes were affected throughout the liver, which could indicate an effect on lipid metabolism. Results also indicate that eelpouts collected at polluted sites might have an affected immune system, with lower mRNA expression of genes involved in the innate immune system and a higher number of lymphocytes. Biomarker assessment also was performed on livers dissected from unborn eelpout larvae collected from the ovary of the females. No significant differences were noted, which might indicate that the larvae to some extent are protected from effects of environmental pollutants. In conclusion, usage of the selected set of biological markers, covering responses from gene to organism, has demonstrated site-specific biomarker patterns that provided a broad and comprehensive picture of the impact of environmental stressors.
Collapse
Affiliation(s)
- Noomi Asker
- Department of Biological and Environmental Sciences, University of Gothenburg, Gothenburg, Sweden
| | - Bethanie Carney Almroth
- Department of Biological and Environmental Sciences, University of Gothenburg, Gothenburg, Sweden
| | - Eva Albertsson
- Department of Biological and Environmental Sciences, University of Gothenburg, Gothenburg, Sweden
| | | | - John Paul Bignell
- Centre for Environment, Fisheries and Aquaculture Science, Weymouth, Dorset, United Kingdom
| | - Niklas Hanson
- Department of Biological and Environmental Sciences, University of Gothenburg, Gothenburg, Sweden
| | - Vittoria Scarcelli
- Department of Clinic and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Björn Fagerholm
- Department of Aquatic Resources, Institute of Coastal Research, Swedish University of Agricultural Sciences, Väröbacka, Sweden
| | - Jari Parkkonen
- Department of Biological and Environmental Sciences, University of Gothenburg, Gothenburg, Sweden
| | - Emma Wijkmark
- Department of Mathematical Statistics, Chalmers University of Technology, Gothenburg, Sweden
| | - Giada Frenzilli
- Department of Clinic and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Lars Förlin
- Department of Biological and Environmental Sciences, University of Gothenburg, Gothenburg, Sweden
| | - Joachim Sturve
- Department of Biological and Environmental Sciences, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
182
|
Wilbe M, Kozyrev SV, Farias FHG, Bremer HD, Hedlund A, Pielberg GR, Seppälä EH, Gustafson U, Lohi H, Carlborg Ö, Andersson G, Hansson-Hamlin H, Lindblad-Toh K. Multiple Changes of Gene Expression and Function Reveal Genomic and Phenotypic Complexity in SLE-like Disease. PLoS Genet 2015; 11:e1005248. [PMID: 26057447 PMCID: PMC4461293 DOI: 10.1371/journal.pgen.1005248] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 04/27/2015] [Indexed: 02/07/2023] Open
Abstract
The complexity of clinical manifestations commonly observed in autoimmune disorders poses a major challenge to genetic studies of such diseases. Systemic lupus erythematosus (SLE) affects humans as well as other mammals, and is characterized by the presence of antinuclear antibodies (ANA) in patients’ sera and multiple disparate clinical features. Here we present evidence that particular sub-phenotypes of canine SLE-related disease, based on homogenous (ANAH) and speckled ANA (ANAS) staining pattern, and also steroid-responsive meningitis-arteritis (SRMA) are associated with different but overlapping sets of genes. In addition to association to certain MHC alleles and haplotypes, we identified 11 genes (WFDC3, HOMER2, VRK1, PTPN3, WHAMM, BANK1, AP3B2, DAPP1, LAMTOR3, DDIT4L and PPP3CA) located on five chromosomes that contain multiple risk haplotypes correlated with gene expression and disease sub-phenotypes in an intricate manner. Intriguingly, the association of BANK1 with both human and canine SLE appears to lead to similar changes in gene expression levels in both species. Our results suggest that molecular definition may help unravel the mechanisms of different clinical features common between and specific to various autoimmune disease phenotypes in dogs and humans. Autoimmune disorders display complex phenotypes with clinically diverse manifestations, which together with complex genetic inheritance and environmental factors triggering the disease may complicate the diagnosis and investigation of the disease mechanism. The use of dog breeds may facilitate the analysis of genetic factors based on genetic homogeneity within a breed. We performed genetic analysis of two diseases common in dogs, immune-mediated rheumatic disease (IMRD) and steroid-responsive meningitis-arteritis (SRMA) that are similar to human SLE and a group of vasulitides such as Kawasaki disease, Henoch-Schönlein purpura and Behçet’s disease, correspondingly. We identified eleven genes along with specific alleles and genotypes for the major histocompatibility complex II involved in susceptibility, and studied their expression. The genes shared between the two diseases may be involved in the common immune signaling pathways and hence account for the common clinical signs, whereas the phenotype-specific genes may be implicated in particular pathways active in certain tissues and organs, and thereby may be responsible for characteristic manifestations seen only in one of the diseases. Further, the similarity between human and dog SLE at the genetic and functional levels demonstrated by the association of the BANK1 gene in both species indicates the common cross-species mechanisms of autoimmunity and may help identification of novel disease genes and pathways.
Collapse
Affiliation(s)
- Maria Wilbe
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences (SLU), Uppsala, Sweden
| | - Sergey V. Kozyrev
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Fabiana H. G. Farias
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Hanna D. Bremer
- Department of Clinical Sciences, Swedish University of Agricultural Sciences (SLU), Uppsala, Sweden
| | - Anna Hedlund
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences (SLU), Uppsala, Sweden
| | - Gerli R. Pielberg
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Eija H. Seppälä
- Research Programs Unit, Molecular Neurology; Department of Veterinary Biosciences, University of Helsinki and Folkhälsan Research Center, Helsinki, Finland
| | - Ulla Gustafson
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences (SLU), Uppsala, Sweden
| | - Hannes Lohi
- Research Programs Unit, Molecular Neurology; Department of Veterinary Biosciences, University of Helsinki and Folkhälsan Research Center, Helsinki, Finland
| | - Örjan Carlborg
- Department of Clinical Sciences, Division of Computational Genetics, Swedish University of Agricultural Sciences (SLU), Uppsala, Sweden
| | - Göran Andersson
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences (SLU), Uppsala, Sweden
| | - Helene Hansson-Hamlin
- Department of Clinical Sciences, Swedish University of Agricultural Sciences (SLU), Uppsala, Sweden
- * E-mail: (HHH); (KLT)
| | - Kerstin Lindblad-Toh
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- Broad Institute, Cambridge, Cambridge, Massachusetts, United States of America
- * E-mail: (HHH); (KLT)
| |
Collapse
|
183
|
Benyoucef A, Calvo J, Renou L, Arcangeli ML, van den Heuvel A, Amsellem S, Mehrpour M, Larghero J, Soler E, Naguibneva I, Pflumio F. The SCL/TAL1 Transcription Factor Represses the Stress Protein DDiT4/REDD1 in Human Hematopoietic Stem/Progenitor Cells. Stem Cells 2015; 33:2268-79. [PMID: 25858676 DOI: 10.1002/stem.2028] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Accepted: 03/11/2015] [Indexed: 01/09/2023]
Abstract
Hematopoietic stem/progenitor cells (HSPCs) are regulated through numerous molecular mechanisms that have not been interconnected. The transcription factor stem cell leukemia/T-cell acute leukemia 1 (TAL1) controls human HSPC but its mechanism of action is not clarified. In this study, we show that knockdown (KD) or short-term conditional over-expression (OE) of TAL1 in human HSPC ex vivo, respectively, blocks and maintains hematopoietic potentials, affecting proliferation of human HSPC. Comparative gene expression analyses of TAL1/KD and TAL1/OE human HSPC revealed modifications of cell cycle regulators as well as previously described TAL1 target genes. Interestingly an inverse correlation between TAL1 and DNA damage-induced transcript 4 (DDiT4/REDD1), an inhibitor of the mammalian target of rapamycin (mTOR) pathway, is uncovered. Low phosphorylation levels of mTOR target proteins in TAL1/KD HSPC confirmed an interplay between mTOR pathway and TAL1 in correlation with TAL1-mediated effects of HSPC proliferation. Finally chromatin immunoprecipitation experiments performed in human HSPC showed that DDiT4 is a direct TAL1 target gene. Functional analyses showed that TAL1 represses DDiT4 expression in HSPCs. These results pinpoint DDiT4/REDD1 as a novel target gene regulated by TAL1 in human HSPC and establish for the first time a link between TAL1 and the mTOR pathway in human early hematopoietic cells. Stem Cells 2015;33:2268-2279.
Collapse
Affiliation(s)
- Aissa Benyoucef
- CEA, DSV-IRCM-SCSR-LSHL, UMR 967, équipe labellisée Ligue Nationale contre le Cancer, Fontenay-aux-Roses, Paris, France.,INSERM, U967, Fontenay-aux-Roses, Paris, France.,Université Paris Diderot, UMR 967, Fontenay-aux-Roses, Paris, France.,Université Paris-Sud, UMR 967, Fontenay-aux-Roses, Paris, France
| | - Julien Calvo
- CEA, DSV-IRCM-SCSR-LSHL, UMR 967, équipe labellisée Ligue Nationale contre le Cancer, Fontenay-aux-Roses, Paris, France.,INSERM, U967, Fontenay-aux-Roses, Paris, France.,Université Paris Diderot, UMR 967, Fontenay-aux-Roses, Paris, France.,Université Paris-Sud, UMR 967, Fontenay-aux-Roses, Paris, France
| | - Laurent Renou
- CEA, DSV-IRCM-SCSR-LSHL, UMR 967, équipe labellisée Ligue Nationale contre le Cancer, Fontenay-aux-Roses, Paris, France.,INSERM, U967, Fontenay-aux-Roses, Paris, France.,Université Paris Diderot, UMR 967, Fontenay-aux-Roses, Paris, France.,Université Paris-Sud, UMR 967, Fontenay-aux-Roses, Paris, France
| | - Marie-Laure Arcangeli
- CEA, DSV-IRCM-SCSR-LSHL, UMR 967, équipe labellisée Ligue Nationale contre le Cancer, Fontenay-aux-Roses, Paris, France.,INSERM, U967, Fontenay-aux-Roses, Paris, France.,Université Paris Diderot, UMR 967, Fontenay-aux-Roses, Paris, France.,Université Paris-Sud, UMR 967, Fontenay-aux-Roses, Paris, France
| | | | - Sophie Amsellem
- Centre d'Investigation Clinique-BioThérapie, Institut Gustave Roussy, Villejuif, Paris, France
| | - Maryam Mehrpour
- INSERM U1151-CNRS UMR 8253 Institut Necker Enfants-Malades (INEM), Université Paris Descartes, Paris, France
| | - Jerome Larghero
- Cell Therapy Unit and Clinical Investigation Center in Biotherapies, AP-HP, Saint-Louis Hospital, Paris, France
| | - Eric Soler
- INSERM, U967, Fontenay-aux-Roses, Paris, France.,Department of Cell Biology, Erasmus MC, Rotterdam, The Netherlands.,CEA, DSV-IRCM-SCSR-LHM, UMR967, Fontenay-aux-Roses, Paris, France
| | - Irina Naguibneva
- CEA, DSV-IRCM-SCSR-LSHL, UMR 967, équipe labellisée Ligue Nationale contre le Cancer, Fontenay-aux-Roses, Paris, France.,INSERM, U967, Fontenay-aux-Roses, Paris, France.,Université Paris Diderot, UMR 967, Fontenay-aux-Roses, Paris, France.,Université Paris-Sud, UMR 967, Fontenay-aux-Roses, Paris, France
| | - Francoise Pflumio
- CEA, DSV-IRCM-SCSR-LSHL, UMR 967, équipe labellisée Ligue Nationale contre le Cancer, Fontenay-aux-Roses, Paris, France.,INSERM, U967, Fontenay-aux-Roses, Paris, France.,Université Paris Diderot, UMR 967, Fontenay-aux-Roses, Paris, France.,Université Paris-Sud, UMR 967, Fontenay-aux-Roses, Paris, France
| |
Collapse
|
184
|
Zhang G, Zhang J, Shang D, Qi B, Chen H. Deoxycholic acid inhibited proliferation and induced apoptosis and necrosis by regulating the activity of transcription factors in rat pancreatic acinar cell line AR42J. In Vitro Cell Dev Biol Anim 2015; 51:851-6. [PMID: 25990271 DOI: 10.1007/s11626-015-9907-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Accepted: 04/01/2015] [Indexed: 12/24/2022]
Abstract
The objective of this study is to investigate the effect of deoxycholic acid (DCA) on rat pancreatic acinar cell line AR42J and the functional mechanisms of DCA on AR42J cells. AR42J cells were treated with various concentrations of DCA for 24 h and also treated with 0.4 mmol/L DCA for multiple times, and then, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was performed to detect the AR42J cell survival rate. Flow cytometric was used to detect the cell apoptosis and necrosis in AR42J cells treated with 0.4 mmol/L and 0.8 mmol/L DCA. The cells treated with phosphate buffer saline (PBS) were served as control. In addition, the DNA-binding activity assays of transcription factors (TFs) in nuclear proteins of cells treated with DCA were determined using Panomics Procarta Transcription Factor Assay Kit. The relative survival rates were markedly decreased (P < 0.05) in a dose- and time-dependent manner. Compared with control group, the cell apoptosis and necrosis ratio were both significantly elevated in 0.4 mmol/L DCA and 0.8 mmol/L DCA groups (P < 0.01). A significant increase (P < 0.05) in the activity of transcription factor 2 (ATF2), interferon-stimulated response element (ISRE), NKX-2.5, androgen receptor (AR), p53, and hypoxia-inducible factor-1 (HIF-1) was observed, and the activity of peroxisome proliferator-activated receptor (PPAR), activator protein 1 (AP1), and E2F1 was reduced (P < 0.05). In conclusion, DCA inhibited proliferation and induced apoptosis and necrosis in AR42J cells. The expression changes of related genes regulated by TFs might be the molecular mechanism of AR42J cell injury.
Collapse
Affiliation(s)
- Guixin Zhang
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Zhongshan Road 222, Dalian, China, 116011.
| | - Jingwen Zhang
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Zhongshan Road 222, Dalian, China, 116011. .,Dalian Medical University, Dalian, China, 116044.
| | - Dong Shang
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Zhongshan Road 222, Dalian, China, 116011.
| | - Bing Qi
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Zhongshan Road 222, Dalian, China, 116011.
| | - Hailong Chen
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Zhongshan Road 222, Dalian, China, 116011.
| |
Collapse
|
185
|
Qiao S, Dennis M, Song X, Vadysirisack DD, Salunke D, Nash Z, Yang Z, Liesa M, Yoshioka J, Matsuzawa SI, Shirihai OS, Lee RT, Reed JC, Ellisen LW. A REDD1/TXNIP pro-oxidant complex regulates ATG4B activity to control stress-induced autophagy and sustain exercise capacity. Nat Commun 2015; 6:7014. [PMID: 25916556 PMCID: PMC4421852 DOI: 10.1038/ncomms8014] [Citation(s) in RCA: 150] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 03/23/2015] [Indexed: 12/12/2022] Open
Abstract
Macroautophagy (autophagy) is a critical cellular stress response; however, the signal transduction pathways controlling autophagy induction in response to stress are poorly understood. Here we reveal a new mechanism of autophagy control whose deregulation disrupts mitochondrial integrity and energy homeostasis in vivo. Stress conditions including hypoxia and exercise induce reactive oxygen species (ROS) through upregulation of a protein complex involving REDD1, an mTORC1 inhibitor and the pro-oxidant protein TXNIP. Decreased ROS in cells and tissues lacking either REDD1 or TXNIP increases catalytic activity of the redox-sensitive ATG4B cysteine endopeptidase, leading to enhanced LC3B delipidation and failed autophagy. Conversely, REDD1/TXNIP complex expression is sufficient to induce ROS, suppress ATG4B activity and activate autophagy. In Redd1−/− mice, deregulated ATG4B activity and disabled autophagic flux cause accumulation of defective mitochondria, leading to impaired oxidative phosphorylation, muscle ATP depletion and poor exercise capacity. Thus, ROS regulation through REDD1/TXNIP is physiological rheostat controlling stress-induced autophagy. Stress-induced macroautophagy is initiated by the induction of reactive oxygen species (ROS). Here Qiao et al. show that the mTOR inhibitor REDD1 in a complex with pro-oxidant protein TXNIP induces ROS formation, leading to ATG4B suppression and autophagy activation in a largely mTOR-independent manner.
Collapse
Affiliation(s)
- Shuxi Qiao
- 1] Massachusetts General Hospital Cancer Center, Boston, Massachusetts 02114, USA [2] Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Michael Dennis
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts 02114, USA
| | - Xiufeng Song
- 1] Massachusetts General Hospital Cancer Center, Boston, Massachusetts 02114, USA [2] Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Douangsone D Vadysirisack
- 1] Massachusetts General Hospital Cancer Center, Boston, Massachusetts 02114, USA [2] Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Devika Salunke
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts 02114, USA
| | - Zachary Nash
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts 02114, USA
| | - Zhifen Yang
- Sanford-Burnham Medical Research Institute, La Jolla, California 92037, USA
| | - Marc Liesa
- Department of Medicine, Evans Center, Mitochondria ARC, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - Jun Yoshioka
- 1] Harvard Medical School, Boston, Massachusetts 02115, USA [2] Regenerative Medicine Center, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | - Shu-Ichi Matsuzawa
- Sanford-Burnham Medical Research Institute, La Jolla, California 92037, USA
| | - Orian S Shirihai
- 1] Department of Medicine, Evans Center, Mitochondria ARC, Boston University School of Medicine, Boston, Massachusetts 02118, USA [2] Department of Clinical Biochemistry, Faculty of Medicine, Ben Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Richard T Lee
- 1] Harvard Medical School, Boston, Massachusetts 02115, USA [2] Regenerative Medicine Center, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | - John C Reed
- 1] Sanford-Burnham Medical Research Institute, La Jolla, California 92037, USA [2] Roche Pharmaceutical Research and Early Development, Basel 4070, Switzerland
| | - Leif W Ellisen
- 1] Massachusetts General Hospital Cancer Center, Boston, Massachusetts 02114, USA [2] Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
186
|
Martín-Flores N, Romaní-Aumedes J, Rué L, Canal M, Sanders P, Straccia M, Allen ND, Alberch J, Canals JM, Pérez-Navarro E, Malagelada C. RTP801 Is Involved in Mutant Huntingtin-Induced Cell Death. Mol Neurobiol 2015; 53:2857-2868. [PMID: 25876513 DOI: 10.1007/s12035-015-9166-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 03/30/2015] [Indexed: 01/16/2023]
Abstract
RTP801 expression is induced by cellular stress and has a pro-apoptotic function in non-proliferating differentiated cells such as neurons. In several neurodegenerative disorders, including Parkinson's disease and Alzheimer's disease, elevated levels of RTP801 have been observed, which suggests a role for RTP801 in neuronal death. Neuronal death is also a pathological hallmark in Huntington's disease (HD), an inherited neurodegenerative disorder caused by a CAG repeat expansion in the huntingtin gene. Currently, the exact mechanisms underlying mutant huntingtin (mhtt)-induced toxicity are still unclear. Here, we investigated whether RTP801 is involved in (mhtt)-induced cell death. Ectopic exon-1 mhtt elevated RTP801 mRNA and protein levels in nerve growth factor (NGF)-differentiated PC12 cells and in rat primary cortical neurons. In neuronal PC12 cells, mhtt also contributed to RTP801 protein elevation by reducing its proteasomal degradation rate, in addition to promoting RTP801 gene expression. Interestingly, silencing RTP801 expression with short hairpin RNAs (shRNAs) blocked mhtt-induced cell death in NGF-differentiated PC12 cells. However, RTP801 protein levels were not altered in the striatum of Hdh(Q7/Q111) and R6/1 mice, two HD models that display motor deficits but not neuronal death. Importantly, RTP801 protein levels were elevated in both neural telencephalic progenitors differentiated from HD patient-derived induced pluripotent stem cells and in the putamen and cerebellum of human HD postmortem brains. Taken together, our results suggest that RTP801 is a novel downstream effector of mhtt-induced toxicity and that it may be relevant to the human disease.
Collapse
Affiliation(s)
- Núria Martín-Flores
- Department of Pathological Anatomy, Pharmacology and Microbiology, Faculty of Medicine, University of Barcelona, Casanova 143, 08036, Barcelona, Catalonia, Spain
| | - Joan Romaní-Aumedes
- Department of Pathological Anatomy, Pharmacology and Microbiology, Faculty of Medicine, University of Barcelona, Casanova 143, 08036, Barcelona, Catalonia, Spain
| | - Laura Rué
- Department of Cell Biology, Immunology and Neurosciences, Faculty of Medicine, University of Barcelona, Casanova 143, 08036, Barcelona, Catalonia, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Mercè Canal
- Department of Pathological Anatomy, Pharmacology and Microbiology, Faculty of Medicine, University of Barcelona, Casanova 143, 08036, Barcelona, Catalonia, Spain
| | - Phil Sanders
- Department of Cell Biology, Immunology and Neurosciences, Faculty of Medicine, University of Barcelona, Casanova 143, 08036, Barcelona, Catalonia, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Marco Straccia
- Department of Cell Biology, Immunology and Neurosciences, Faculty of Medicine, University of Barcelona, Casanova 143, 08036, Barcelona, Catalonia, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Nicholas D Allen
- Divisions of Pathophysiology & Repair and Neuroscience, School of Biosciences, Cardiff University, Cardiff, CF10 3AX, UK
| | - Jordi Alberch
- Department of Cell Biology, Immunology and Neurosciences, Faculty of Medicine, University of Barcelona, Casanova 143, 08036, Barcelona, Catalonia, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Josep M Canals
- Department of Cell Biology, Immunology and Neurosciences, Faculty of Medicine, University of Barcelona, Casanova 143, 08036, Barcelona, Catalonia, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Esther Pérez-Navarro
- Department of Cell Biology, Immunology and Neurosciences, Faculty of Medicine, University of Barcelona, Casanova 143, 08036, Barcelona, Catalonia, Spain. .,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Catalonia, Spain. .,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| | - Cristina Malagelada
- Department of Pathological Anatomy, Pharmacology and Microbiology, Faculty of Medicine, University of Barcelona, Casanova 143, 08036, Barcelona, Catalonia, Spain.
| |
Collapse
|
187
|
Sesen J, Dahan P, Scotland SJ, Saland E, Dang VT, Lemarié A, Tyler BM, Brem H, Toulas C, Cohen-Jonathan Moyal E, Sarry JE, Skuli N. Metformin inhibits growth of human glioblastoma cells and enhances therapeutic response. PLoS One 2015; 10:e0123721. [PMID: 25867026 PMCID: PMC4395104 DOI: 10.1371/journal.pone.0123721] [Citation(s) in RCA: 147] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 03/05/2015] [Indexed: 01/08/2023] Open
Abstract
High-grade gliomas, glioblastomas (GB), are refractory to conventional treatment combining surgery, chemotherapy, mainly temozolomide, and radiotherapy. This highlights an urgent need to develop novel therapies and increase the efficacy of radio/chemotherapy for these very aggressive and malignant brain tumors. Recently, tumor metabolism became an interesting potential therapeutic target in various cancers. Accordingly, combining drugs targeting cell metabolism with appropriate chemotherapeutic agents or radiotherapy has become attractive. In light of these perspectives, we were particularly interested in the anti-cancer properties of a biguanide molecule used for type 2 diabetes treatment, metformin. In our present work, we demonstrate that metformin decreases mitochondrial-dependent ATP production and oxygen consumption and increases lactate and glycolytic ATP production. We show that metformin induces decreased proliferation, cell cycle arrest, autophagy, apoptosis and cell death in vitro with a concomitant activation of AMPK, Redd1 and inhibition of the mTOR pathway. Cell sensitivity to metformin also depends on the genetic and mutational backgrounds of the different GB cells used in this study, particularly their PTEN status. Interestingly, knockdown of AMPK and Redd1 with siRNA partially, but incompletely, abrogates the induction of apoptosis by metformin suggesting both AMPK/Redd1-dependent and –independent effects. However, the primary determinant of the effect of metformin on cell growth is the genetic and mutational backgrounds of the glioma cells. We further demonstrate that metformin treatment in combination with temozolomide and/or irradiation induces a synergistic anti-tumoral response in glioma cell lines. Xenografts performed in nude mice demonstrate in vivo that metformin delays tumor growth. As current treatments for GB commonly fail to cure, the need for more effective therapeutic options is overwhelming. Based on these results, metformin could represent a potential enhancer of the cytotoxic effects of temozolomide and/or radiotherapy.
Collapse
Affiliation(s)
- Julie Sesen
- INSERM U1037, Centre de Recherche en Cancérologie de Toulouse, Toulouse, France
| | - Perrine Dahan
- INSERM U1037, Centre de Recherche en Cancérologie de Toulouse, Toulouse, France
| | - Sarah J. Scotland
- INSERM U1037, Centre de Recherche en Cancérologie de Toulouse, Toulouse, France
- Department of Neurosurgery, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Estelle Saland
- INSERM U1037, Centre de Recherche en Cancérologie de Toulouse, Toulouse, France
| | - Van-Thi Dang
- INSERM U1037, Centre de Recherche en Cancérologie de Toulouse, Toulouse, France
| | - Anthony Lemarié
- INSERM U1037, Centre de Recherche en Cancérologie de Toulouse, Toulouse, France
| | - Betty M. Tyler
- Department of Neurosurgery, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Henry Brem
- Department of Neurosurgery, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Christine Toulas
- INSERM U1037, Centre de Recherche en Cancérologie de Toulouse, Toulouse, France
| | | | - Jean-Emmanuel Sarry
- INSERM U1037, Centre de Recherche en Cancérologie de Toulouse, Toulouse, France
| | - Nicolas Skuli
- INSERM U1037, Centre de Recherche en Cancérologie de Toulouse, Toulouse, France
- * E-mail:
| |
Collapse
|
188
|
Peng G, Liu Y. Hypoxia-inducible factors in cancer stem cells and inflammation. Trends Pharmacol Sci 2015; 36:374-83. [PMID: 25857287 DOI: 10.1016/j.tips.2015.03.003] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 03/05/2015] [Accepted: 03/10/2015] [Indexed: 10/23/2022]
Abstract
Hypoxia-inducible factors (HIF) mediate metabolic switches in cells in hypoxic environments, including those in both normal and malignant tissues with limited supplies of oxygen. Paradoxically, recent studies have shown that cancer stem cells (CSCs) and activated immune effector cells exhibit high HIF activity in normoxic environments and that HIF activity is critical in the maintenance of CSCs as well as the differentiation and function of inflammatory cells. Given that inflammation and CSCs are two major barriers to effective cancer therapy, targeting HIF may provide a new approach to developing such treatments.
Collapse
Affiliation(s)
- Gong Peng
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
| | - Yang Liu
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China; Center for Cancer and Immunology Research, Children's Research Institute, Children's National Medical Center, Washington, DC, USA.
| |
Collapse
|
189
|
Desantis A, Bruno T, Catena V, De Nicola F, Goeman F, Iezzi S, Sorino C, Ponzoni M, Bossi G, Federico V, La Rosa F, Ricciardi MR, Lesma E, De Meo PD, Castrignanò T, Petrucci MT, Pisani F, Chesi M, Bergsagel PL, Floridi A, Tonon G, Passananti C, Blandino G, Fanciulli M. Che-1-induced inhibition of mTOR pathway enables stress-induced autophagy. EMBO J 2015; 34:1214-30. [PMID: 25770584 DOI: 10.15252/embj.201489920] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 02/16/2015] [Indexed: 01/13/2023] Open
Abstract
Mammalian target of rapamycin (mTOR) is a key protein kinase that regulates cell growth, metabolism, and autophagy to maintain cellular homeostasis. Its activity is inhibited by adverse conditions, including nutrient limitation, hypoxia, and DNA damage. In this study, we demonstrate that Che-1, a RNA polymerase II-binding protein activated by the DNA damage response, inhibits mTOR activity in response to stress conditions. We found that, under stress, Che-1 induces the expression of two important mTOR inhibitors, Redd1 and Deptor, and that this activity is required for sustaining stress-induced autophagy. Strikingly, Che-1 expression correlates with the progression of multiple myeloma and is required for cell growth and survival, a malignancy characterized by high autophagy response.
Collapse
Affiliation(s)
- Agata Desantis
- Epigenetics Laboratory, Molecular Medicine Area Regina Elena National Cancer Institute, Rome, Italy
| | - Tiziana Bruno
- Epigenetics Laboratory, Molecular Medicine Area Regina Elena National Cancer Institute, Rome, Italy
| | - Valeria Catena
- Epigenetics Laboratory, Molecular Medicine Area Regina Elena National Cancer Institute, Rome, Italy Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Francesca De Nicola
- Epigenetics Laboratory, Molecular Medicine Area Regina Elena National Cancer Institute, Rome, Italy
| | - Frauke Goeman
- Translational Oncogenomic Laboratory, Molecular Medicine Area Regina Elena National Cancer Institute, Rome, Italy
| | - Simona Iezzi
- Epigenetics Laboratory, Molecular Medicine Area Regina Elena National Cancer Institute, Rome, Italy
| | - Cristina Sorino
- Epigenetics Laboratory, Molecular Medicine Area Regina Elena National Cancer Institute, Rome, Italy
| | - Maurilio Ponzoni
- Pathology and Myeloma Units, Molecular Oncology Division, San Raffaele Scientific Institute, Milan, Italy
| | - Gianluca Bossi
- Molecular Oncogenesis Laboratory, Regina Elena National Cancer Institute, Rome, Italy
| | - Vincenzo Federico
- Division of Hematology, Department of Cellular Biotechnologies and Hematology, "Sapienza" University, Rome, Italy
| | - Francesca La Rosa
- Epigenetics Laboratory, Molecular Medicine Area Regina Elena National Cancer Institute, Rome, Italy
| | - Maria Rosaria Ricciardi
- Division of Hematology, Department of Cellular Biotechnologies and Hematology, "Sapienza" University, Rome, Italy
| | - Elena Lesma
- Laboratory of Pharmacology, Department of Health Sciences, University of Milan, Milan, Italy
| | | | | | - Maria Teresa Petrucci
- Division of Hematology, Department of Cellular Biotechnologies and Hematology, "Sapienza" University, Rome, Italy
| | - Francesco Pisani
- Hematology Laboratory, Regina Elena National Cancer Institute, Rome, Italy
| | - Marta Chesi
- Comprehensive Cancer Center, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - P Leif Bergsagel
- Comprehensive Cancer Center, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Aristide Floridi
- Epigenetics Laboratory, Molecular Medicine Area Regina Elena National Cancer Institute, Rome, Italy
| | - Giovanni Tonon
- Functional Genomics of Cancer Unit, Molecular Oncology Division, San Raffaele Scientific Institute, Milan, Italy
| | - Claudio Passananti
- Institute of Molecular Biology and Pathology, CNR Department of Molecular Medicine "Sapienza" University, Rome, Italy
| | - Giovanni Blandino
- Translational Oncogenomic Laboratory, Molecular Medicine Area Regina Elena National Cancer Institute, Rome, Italy
| | - Maurizio Fanciulli
- Epigenetics Laboratory, Molecular Medicine Area Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
190
|
Regazzetti C, Dumas K, Lacas-Gervais S, Pastor F, Peraldi P, Bonnafous S, Dugail I, Le Lay S, Valet P, Le Marchand-Brustel Y, Tran A, Gual P, Tanti JF, Cormont M, Giorgetti-Peraldi S. Hypoxia inhibits Cavin-1 and Cavin-2 expression and down-regulates caveolae in adipocytes. Endocrinology 2015; 156:789-801. [PMID: 25521582 DOI: 10.1210/en.2014-1656] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
During obesity, a hypoxic state develops within the adipose tissue, resulting in insulin resistance. To understand the underlying mechanism, we analyzed the involvement of caveolae because they play a crucial role in the activation of insulin receptors. In the present study, we demonstrate that in 3T3-L1 adipocytes, hypoxia induces the disappearance of caveolae and inhibits the expression of Cavin-1 and Cavin-2, two proteins necessary for the formation of caveolae. In mice, hypoxia induced by the ligature of the spermatic artery results in the decrease of cavin-1 and cavin-2 expression in the epididymal adipose tissue. Down-regulation of the expression of cavins in response to hypoxia is dependent on hypoxia-inducible factor-1. Indeed, the inhibition of hypoxia-inducible factor-1 restores the expression of cavins and caveolae formation. Expression of cavins regulates insulin signaling because the silencing of cavin-1 and cavin-2 impairs insulin signaling pathway. In human, cavin-1 and cavin-2 are decreased in the sc adipose tissue of obese diabetic patients compared with lean subjects. Moreover, the expression of cavin-2 correlates negatively with the homeostatic model assessment index of insulin resistance and glycated hemoglobin level. In conclusion, we propose a new mechanism in which hypoxia inhibits cavin-1 and cavin-2 expression, resulting in the disappearance of caveolae. This leads to the inhibition of insulin signaling and the establishment of insulin resistance.
Collapse
Affiliation(s)
- Claire Regazzetti
- INSERM Unité 1065 (C.R., K.D., F.P., Y.L.M.-B., J.-F.T., M.C., S.G.-P.), C3M, Mediterranean Research Centre for Molecular Medicine, Team 7 (Cellular and Molecular Physiopathology of Obesity and Diabetes), Unité de Formation et de Recherche (UFR) Medicine (C.R., K.D., F.P., P.P., S.B., Y.L.M.-B., A.T., P.G., J.-F.T., M.C., S.G.-P.), and INSERM Unité 1065 (S.B., A.T., P.G.), C3M, Mediterranean Research Centre for Molecular Medicine, Team 8 (Hepatic Complications in Obesity),University of Nice, Sophia Antipolis F-06204 Nice, France; Centre Commun de Microscopie Appliquée (S.L.-G.), University of Nice, Sophia Antipolis, UFR Sciences, Parc Valrose, F-06108 Nice, France; Unité Mixte de Recherche Centre National de la Recherche Scientifique 7277 (P.P.), Unité Mixte de Recherche INSERM Unité 1091, UFR Medicine, F-06107 Nice, France; Centre Hospitalier Universitaire de Nice, Digestive Center (S.B., A.T.), Nice F-06202, Cedex 3, France; INSERM Unité Mixte de Recherche S872 (I.D.), Centre de Recherche des Cordeliers, Eq8, F-75006 Paris, France; INSERM Unité 1063 (S.L.L.), Stress Oxydant et Pathologies Métaboliques, Institut de Biologie en Santé, F-49933 Angers, France; and INSERM Unité Mixte de Recherche 1048 (P.V.), Institut des Maladies Métaboliques et Cardiovasculaires, Université Paul Sabatier, F-31432 Toulouse, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
191
|
Shao C, Ahmad N, Hodges K, Kuang S, Ratliff T, Liu X. Inhibition of polo-like kinase 1 (Plk1) enhances the antineoplastic activity of metformin in prostate cancer. J Biol Chem 2015; 290:2024-33. [PMID: 25505174 PMCID: PMC4303657 DOI: 10.1074/jbc.m114.596817] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 12/08/2014] [Indexed: 01/21/2023] Open
Abstract
The widely used anti-diabetic drug metformin has been shown to exert strong antineoplastic actions in numerous tumor types, including prostate cancer (PCa). In this study, we show that BI2536, a specific Plk1 inhibitor, acted synergistically with metformin in inhibiting PCa cell proliferation. Furthermore, we also provide evidence that Plk1 inhibition makes PCa cells carrying WT p53 much more sensitive to low-dose metformin treatment. Mechanistically, we found that co-treatment with BI2536 and metformin induced p53-dependent apoptosis and further activated the p53/Redd-1 pathway. Moreover, we also show that BI2536 treatment inhibited metformin-induced glycolysis and glutamine anaplerosis, both of which are survival responses of cells against mitochondrial poisons. Finally, we confirmed the cell-based observations using both cultured cell-derived and patient-derived xenograft studies. Collectively, our findings support another promising therapeutic strategy by combining two well tolerated drugs against PCa proliferation and the progression of androgen-dependent PCa to the castration-resistant stage.
Collapse
Affiliation(s)
- Chen Shao
- From the Departments of Biochemistry and
| | - Nihal Ahmad
- the Department of Dermatology, University of Wisconsin, Madison, Wisconsin 53706, and
| | - Kurt Hodges
- the Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | | | - Tim Ratliff
- the Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907
| | - Xiaoqi Liu
- From the Departments of Biochemistry and the Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907,
| |
Collapse
|
192
|
Insight into the role of mTOR and metabolism in T cells reveals new potential approaches to preventing graft rejection. Curr Opin Organ Transplant 2015; 19:363-71. [PMID: 24991977 DOI: 10.1097/mot.0000000000000098] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW In this review, we discuss the recent advances with regard to the mammalian target of rapamycin (mTOR) signaling pathway and focus on how this pathway modulates immune responses. Overall, these insights provide important clues in terms of strategically integrating mTOR and metabolic inhibitors into transplantation rejection protocols. RECENT FINDINGS mTOR is regulated by environmental cues and activates diverse downstream pathways to guide cell growth and fate. What has emerged from recent studies is that mechanistically mTOR directs T cell differentiation and function in part by regulating metabolic programs. Such findings not only inform us with regard to the metabolic demands of effector and memory T cells but also elucidate metabolic pathways that might be targeted to selectively regulate immune responses. SUMMARY Initial studies focused on the ability of the mTOR inhibitor rapamycin to suppress immune responses by inhibiting T cell proliferation. Since then, both pharmacologic and genetic studies have revealed a central role for mTOR in regulating T cell activation, differentiation, and function independent of proliferation. Specifically, it has become clear that mTOR plays an important role in regulating the metabolic machinery necessary for effector, regulatory, and memory T cell generation. As such, direct inhibition of metabolism may emerge as a potent and selective means of preventing graft rejection. This review will discuss new insights regarding the ability of downstream signaling pathways, including mTOR-dependent metabolic pathways in regulating T cell responses. Finally, we will discuss these new insights in the context of developing novel immunoregulatory regimens for transplantation.
Collapse
|
193
|
Pal SK, He M, Tong T, Wu H, Liu X, Lau C, Wang JH, Warden C, Wu X, Signoretti S, Choueiri TK, Karam JA, Jones JO. RNA-seq reveals aurora kinase-driven mTOR pathway activation in patients with sarcomatoid metastatic renal cell carcinoma. Mol Cancer Res 2015; 13:130-7. [PMID: 25183163 PMCID: PMC4608366 DOI: 10.1158/1541-7786.mcr-14-0352] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
UNLABELLED Sarcomatoid metastatic renal cell carcinoma (mRCC) is associated with a poor prognosis, and the biology of the disease has been inadequately characterized. RNA sequencing (RNA-seq) was performed on adjacent benign, clear cell, and sarcomatoid components from clinical specimens with sarcomatoid mRCC. M phase and cell-cycle pathways were enriched in sarcomatoid versus adjacent clear cell components, suggesting greater cell proliferation. The expression of aurora kinase A (AURKA) was increased as part of these pathways, and its increased expression was validated by quantitative PCR (qPCR). Immunohistochemical (IHC) analysis revealed that AURKA levels were increased in sarcomatoid tissue compared with their benign or clear cell parts. The increase in AURKA correlated with increased mTOR pathway activity, as evidenced by increased expression of phosphorylated mTOR (S2448) and ribosomal protein S6K (T389). When AURKA was stably expressed in a RCC cell line (Renca), it resulted in increased expression and activity of mTOR, suggesting that overexpression of AURKA can activate the mTOR pathway. These results warrant the analysis of a larger clinical cohort and suggest that targeting AURKA and/or mTOR in patients with sarcomatoid mRCC should be explored. IMPLICATIONS Comparative RNA-seq of adjacent sarcomatoid and clear cell histology of RCC indicates a proliferative phenotype and increased AURKA-dependent activation of mTOR signaling in sarcomatoid RCC, which could be targeted by available agents.
Collapse
Affiliation(s)
- Sumanta K Pal
- Department of Medical Oncology and Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Miaoling He
- Department of Molecular Pharmacology, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Tommy Tong
- Department of Pathology, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Huiqing Wu
- Department of Pathology, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Xueli Liu
- Department of Biostatistics, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Clayton Lau
- Department of Surgery, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Jin-Hui Wang
- Functional Genomics, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Charles Warden
- Functional Genomics, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Xiwei Wu
- Functional Genomics, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Sabina Signoretti
- Department of Pathology, Dana Farber Cancer Center, Boston, Massachusetts
| | - Toni K Choueiri
- Department of Medical Oncology, Dana Farber Cancer Center, Boston, Massachusetts
| | - Jose A Karam
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jeremy O Jones
- Department of Molecular Pharmacology, City of Hope Comprehensive Cancer Center, Duarte, California.
| |
Collapse
|
194
|
Liang RY, Tu HF, Tan X, Yeh YS, Chueh PJ, Chuang SM. A gene signature for gold nanoparticle-exposed human cell lines. Toxicol Res (Camb) 2015. [DOI: 10.1039/c4tx00181h] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A unique four-gene signature for AuNP exposure was identified using the cDNA microarray and evaluated by qPCR and biological assays in mammalian cell lines.
Collapse
Affiliation(s)
- Ruei-Yue Liang
- Institute of Biomedical Sciences
- National Chung Hsing University
- Taichung
- Taiwan
| | - Hsin-Fang Tu
- Bachelor Program of Biotechnology
- National Chung Hsing University
- Taichung
- Taiwan
| | - Xiaotong Tan
- Institute of Biomedical Sciences
- National Chung Hsing University
- Taichung
- Taiwan
| | - Yu-Shan Yeh
- Center for Measurement Standards (CMS)
- Industrial Technology Research Institute (ITRI)
- Hsinchu
- Taiwan
| | - Pin Ju Chueh
- Institute of Biomedical Sciences
- National Chung Hsing University
- Taichung
- Taiwan
| | - Show-Mei Chuang
- Institute of Biomedical Sciences
- National Chung Hsing University
- Taichung
- Taiwan
| |
Collapse
|
195
|
Dennis MD, Kimball SR, Fort PE, Jefferson LS. Regulated in development and DNA damage 1 is necessary for hyperglycemia-induced vascular endothelial growth factor expression in the retina of diabetic rodents. J Biol Chem 2014; 290:3865-74. [PMID: 25548280 DOI: 10.1074/jbc.m114.623058] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) is considered a major role player in the pathogenesis of diabetic retinopathy, yet the mechanisms regulating its expression are not fully understood. Our laboratory previously demonstrated that diabetes-induced VEGF expression in the retina was dependent on the repressor of mRNA translation 4E-BP1. Interaction of 4E-BP1 with the cap-binding protein eIF4E regulates protein expression by controlling the selection of mRNAs for translation. The process is regulated by the master kinase mTOR in complex 1 (mTORC1), which phosphorylates 4E-BP1, thus promoting its disassociation from eIF4E. In the present study, we investigated the role of the Akt/mTORC1 repressor REDD1 (regulated in development and DNA damage) in diabetes-induced VEGF expression. REDD1 expression was induced by hyperglycemia in the retina of diabetic rodents and by hyperglycemic conditions in Müller cells concomitant with increased VEGF expression. In Müller cells, hyperglycemic conditions attenuated global rates of protein synthesis and cap-dependent mRNA translation concomitant with up-regulated cap-independent VEGF mRNA translation, as assessed by a bicistronic luciferase reporter assay. Hyperglycemic conditions also attenuated mTORC1 signaling and enhanced 4E-BP1 binding to eIF4E. Furthermore, ectopic expression of REDD1 in Müller cells was sufficient to promote both increased 4E-BP1 binding to eIF4E and VEGF expression. Whereas the retina of wild-type mice exhibited increased expression of VEGF and tumor necrosis factor alpha (TNF-α) 4 weeks after streptozotocin administration, the retina of REDD1 knock-out mice failed to do so. Overall, the results demonstrate that REDD1 contributes to the pathogenesis of diabetes in the retina by mediating the pathogenic effects of hyperglycemia.
Collapse
Affiliation(s)
- Michael D Dennis
- From the Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033 and
| | - Scot R Kimball
- From the Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033 and
| | - Patrice E Fort
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan 48105
| | - Leonard S Jefferson
- From the Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033 and
| |
Collapse
|
196
|
Hayasaka M, Tsunekawa H, Yoshinaga M, Murakami T. Endurance exercise induces REDD1 expression and transiently decreases mTORC1 signaling in rat skeletal muscle. Physiol Rep 2014; 2:2/12/e12254. [PMID: 25539833 PMCID: PMC4332227 DOI: 10.14814/phy2.12254] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Working muscle conserves adenosine triphosphate (ATP) for muscle contraction by attenuating protein synthesis through several different pathways. Regulated in development and DNA damage response 1 (REDD1) is one candidate protein that can itself attenuate muscle protein synthesis during muscle contraction. In this study, we investigated whether endurance exercise induces REDD1 expression in association with decreased mammalian target of rapamycin (mTOR) complex I (mTORC1) signaling and global protein synthesis in rat skeletal muscle. After overnight fasting, rats ran on a treadmill at a speed of 28 m/min for 60 min, and were killed before and immediately, 1, 3, 6, 12, and 24 h after exercise. REDD1 mRNA and corresponding protein levels increased rapidly immediately after exercise, and gradually decreased back to the basal level over a period of 6 h in the gastrocnemius muscle. Phosphorylation of mTOR Ser2448 and S6K1 Thr389 increased with the exercise, but diminished in 1–3 h into the recovery period after cessation of exercise. The rate of protein synthesis, as determined by the surface sensing of translation (SUnSET) method, was not altered by exercise in fasted muscle. These results suggest that REDD1 attenuates exercise‐induced mTORC1 signaling. This may be one mechanism responsible for blunting muscle protein synthesis during exercise and in the early postexercise recovery period. We show that REDD1 expression is rapidly induced by an acute bout of endurance exercise in association with a decrease in mTORC1 signaling in rat muscle. The rate of mixed‐muscle protein synthesis was, however, not altered by exercise in fasted state. These results suggest that REDD1‐induced suppression of mTORC1 signaling may be one mechanism to blunt muscle protein synthesis during exercise and postexercise early recovery period.
Collapse
Affiliation(s)
- Miki Hayasaka
- Department of Nutrition, Shigakkan University, Yokone-Machi, Ohbu, Japan
| | - Haruka Tsunekawa
- Department of Nutrition, Shigakkan University, Yokone-Machi, Ohbu, Japan
| | - Mariko Yoshinaga
- Department of Nutrition, Shigakkan University, Yokone-Machi, Ohbu, Japan
| | - Taro Murakami
- Department of Nutrition, Shigakkan University, Yokone-Machi, Ohbu, Japan
| |
Collapse
|
197
|
Bandarra D, Biddlestone J, Mudie S, Müller HAJ, Rocha S. HIF-1α restricts NF-κB-dependent gene expression to control innate immunity signals. Dis Model Mech 2014; 8:169-81. [PMID: 25510503 PMCID: PMC4314782 DOI: 10.1242/dmm.017285] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Hypoxia and inflammation are intimately linked. It is known that nuclear factor κB (NF-κB) regulates the hypoxia-inducible factor (HIF) system, but little is known about how HIF regulates NF-κB. Here, we show that HIF-1α represses NF-κB-dependent gene expression. HIF-1α depletion results in increased NF-κB transcriptional activity both in mammalian cells and in the model organism Drosophila melanogaster. HIF-1α depletion enhances the NF-κB response, and this required not only the TAK-IKK complex, but also CDK6. Loss of HIF-1α results in an increased angiogenic response in mammalian cancer cells and increased mortality in Drosophila following infection. These results indicate that HIF-1α is required to restrain the NF-κB response, and thus prevents excessive and damaging pro-inflammatory responses.
Collapse
Affiliation(s)
- Daniel Bandarra
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, Dow Street, DD1 5EH, UK
| | - John Biddlestone
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, Dow Street, DD1 5EH, UK
| | - Sharon Mudie
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, Dow Street, DD1 5EH, UK
| | - H-Arno J Müller
- Division of Cell and Developmental Biology, College of Life Sciences, University of Dundee, Dow Street, DD1 5EH, UK
| | - Sonia Rocha
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, Dow Street, DD1 5EH, UK.
| |
Collapse
|
198
|
Gene expression alterations in chronic hypoxic MCF7 breast cancer cell line. Genomics 2014; 104:477-81. [DOI: 10.1016/j.ygeno.2014.10.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Revised: 09/10/2014] [Accepted: 10/24/2014] [Indexed: 01/27/2023]
|
199
|
Britto FA, Begue G, Rossano B, Docquier A, Vernus B, Sar C, Ferry A, Bonnieu A, Ollendorff V, Favier FB. REDD1 deletion prevents dexamethasone-induced skeletal muscle atrophy. Am J Physiol Endocrinol Metab 2014; 307:E983-93. [PMID: 25315696 DOI: 10.1152/ajpendo.00234.2014] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
REDD1 (regulated in development and DNA damage response 1) has been proposed to inhibit the mechanistic target of rapamycin complex 1 (mTORC1) during in vitro hypoxia. REDD1 expression is low under basal conditions but is highly increased in response to several catabolic stresses, like hypoxia and glucocorticoids. However, REDD1 function seems to be tissue and stress dependent, and its role in skeletal muscle in vivo has been poorly characterized. Here, we investigated the effect of REDD1 deletion on skeletal muscle mass, protein synthesis, proteolysis, and mTORC1 signaling pathway under basal conditions and after glucocorticoid administration. Whereas skeletal muscle mass and typology were unchanged between wild-type (WT) and REDD1-null mice, oral gavage with dexamethasone (DEX) for 7 days reduced tibialis anterior and gastrocnemius muscle weights as well as tibialis anterior fiber size only in WT. Similarly, REDD1 deletion prevented the inhibition of protein synthesis and mTORC1 activity (assessed by S6, 4E-BP1, and ULK1 phosphorylation) observed in gastrocnemius muscle of WT mice following single DEX administration for 5 h. However, our results suggest that REDD1-mediated inhibition of mTORC1 in skeletal muscle is not related to the modulation of the binding between TSC2 and 14-3-3. In contrast, our data highlight a new mechanism involved in mTORC1 inhibition linking REDD1, Akt, and PRAS40. Altogether, these results demonstrated in vivo that REDD1 is required for glucocorticoid-induced inhibition of protein synthesis via mTORC1 downregulation. Inhibition of REDD1 may thus be a strategy to limit muscle loss in glucocorticoid-mediated atrophy.
Collapse
Affiliation(s)
- Florian A Britto
- Institut National de la Recherche Agronomique, UMR 866 Dynamique Musculaire et Métabolisme, F-34000 Montpellier, France; Université Montpellier 1, F-34000 Montpellier, France; Université Montpellier 2, F-34000 Montpellier, France
| | - Gwenaelle Begue
- Institut National de la Recherche Agronomique, UMR 866 Dynamique Musculaire et Métabolisme, F-34000 Montpellier, France; Université Montpellier 1, F-34000 Montpellier, France; Université Montpellier 2, F-34000 Montpellier, France
| | - Bernadette Rossano
- Institut National de la Recherche Agronomique, UMR 866 Dynamique Musculaire et Métabolisme, F-34000 Montpellier, France; Université Montpellier 1, F-34000 Montpellier, France; Université Montpellier 2, F-34000 Montpellier, France
| | - Aurélie Docquier
- Institut National de la Recherche Agronomique, UMR 866 Dynamique Musculaire et Métabolisme, F-34000 Montpellier, France; Université Montpellier 1, F-34000 Montpellier, France; Université Montpellier 2, F-34000 Montpellier, France
| | - Barbara Vernus
- Institut National de la Recherche Agronomique, UMR 866 Dynamique Musculaire et Métabolisme, F-34000 Montpellier, France; Université Montpellier 1, F-34000 Montpellier, France; Université Montpellier 2, F-34000 Montpellier, France
| | - Chamroeun Sar
- Institut National de la Sante et de la Recherche Medicale (INSERM) U 583, Institut de Neuroscience de Montpellier, France
| | - Arnaud Ferry
- Institut de Myologie, INSERM, U974, Centre National de la Recherche Scientifique UMR 7215, Université Pierre et Marie Curie, Paris, France; and Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Anne Bonnieu
- Institut National de la Recherche Agronomique, UMR 866 Dynamique Musculaire et Métabolisme, F-34000 Montpellier, France; Université Montpellier 1, F-34000 Montpellier, France; Université Montpellier 2, F-34000 Montpellier, France
| | - Vincent Ollendorff
- Institut National de la Recherche Agronomique, UMR 866 Dynamique Musculaire et Métabolisme, F-34000 Montpellier, France; Université Montpellier 1, F-34000 Montpellier, France; Université Montpellier 2, F-34000 Montpellier, France
| | - François B Favier
- Institut National de la Recherche Agronomique, UMR 866 Dynamique Musculaire et Métabolisme, F-34000 Montpellier, France; Université Montpellier 1, F-34000 Montpellier, France; Université Montpellier 2, F-34000 Montpellier, France;
| |
Collapse
|
200
|
Leprivier G, Rotblat B, Khan D, Jan E, Sorensen PH. Stress-mediated translational control in cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1849:845-60. [PMID: 25464034 DOI: 10.1016/j.bbagrm.2014.11.002] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 10/31/2014] [Accepted: 11/04/2014] [Indexed: 12/22/2022]
Abstract
Tumor cells are continually subjected to diverse stress conditions of the tumor microenvironment, including hypoxia, nutrient deprivation, and oxidative or genotoxic stress. Tumor cells must evolve adaptive mechanisms to survive these conditions to ultimately drive tumor progression. Tight control of mRNA translation is critical for this response and the adaptation of tumor cells to such stress forms. This proceeds though a translational reprogramming process which restrains overall translation activity to preserve energy and nutrients, but which also stimulates the selective synthesis of major stress adaptor proteins. Here we present the different regulatory signaling pathways which coordinate mRNA translation in the response to different stress forms, including those regulating eIF2α, mTORC1 and eEF2K, and we explain how tumor cells hijack these pathways for survival under stress. Finally, mechanisms for selective mRNA translation under stress, including the utilization of upstream open reading frames (uORFs) and internal ribosome entry sites (IRESes) are discussed in the context of cell stress. This article is part of a Special Issue entitled: Translation and Cancer.
Collapse
Affiliation(s)
- Gabriel Leprivier
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia V5Z 1L4, Canada; Department of Pathology, University of British Columbia, Vancouver, British Columbia V6T 2B5, Canada
| | - Barak Rotblat
- Department of Life Science, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Debjit Khan
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia V5Z 1L4, Canada; Department of Pathology, University of British Columbia, Vancouver, British Columbia V6T 2B5, Canada
| | - Eric Jan
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T1Z3, Canada
| | - Poul H Sorensen
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia V5Z 1L4, Canada; Department of Pathology, University of British Columbia, Vancouver, British Columbia V6T 2B5, Canada.
| |
Collapse
|