251
|
Abstract
Lysosomes have emerged in the last decade as an immensely important intracellular site of Ca2+ storage and signalling. More recently there has been an increase in the number of new ion channels found to be functional on lysosomes and the potential roles that these signalling pathways might play in fundamental cellular processes are being uncovered. Defects in lysosomal function have been shown to result in changes in lysosomal Ca2+ homeostasis and ultimately can result in cell death. Several neurodegenerative diseases, from rare lysosomal storage diseases through to more common diseases of ageing, have recently been identified as having alterations in lysosomal Ca2+ homeostasis that may play an important role in neuronal excitotoxicity and ultimately cell death. This review will critically summarise these recent findings.
Collapse
Affiliation(s)
- Emyr Lloyd-Evans
- School of Biosciences, Sir Martin Evans Building, Cardiff University, Museum Avenue, Cardiff, CF10 3AX
| |
Collapse
|
252
|
Yang X, Jiang J, Yang X, Han J, Zheng Q. Licochalcone A induces T24 bladder cancer cell apoptosis by increasing intracellular calcium levels. Mol Med Rep 2016; 14:911-9. [PMID: 27221781 DOI: 10.3892/mmr.2016.5334] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 05/03/2016] [Indexed: 11/06/2022] Open
Abstract
Licochalcone A (LCA) has been reported to significantly inhibit cell proliferation, increase reactive oxygen species (ROS) levels, and induce apoptosis of T24 human bladder cancer cells via mitochondria and endoplasmic reticulum (ER) stress-triggered signaling pathways. Based on these findings, the present study aimed to investigate the mechanisms by which LCA induces apoptosis of T24 cells. Cultured T24 cells were treated with LCA, and cell viability was measured using the sulforhodamine B assay. Apoptosis was detected by flow cytometry with Annexin V/propidium iodide staining, and by fluorescent microscopy with Hoechst 33258 staining. The levels of intracellular free calcium ions were determined using Fluo-3 AM dye marker. Intracellular ROS levels were assessed using the 2',7'-dichlorodihydrofluorescein diacetate probe assay. The mitochondrial membrane potential was measured using 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethyl benzimidazole carbocyanine iodide. Furthermore, the mRNA expression levels of B‑cell lymphoma (Bcl)‑extra large, Bcl‑2‑associated X protein, Bcl‑2‑interacting mediator of cell death, apoptotic protease activating factor‑1 (Apaf‑1), calpain 2, cysteinyl aspartate specific proteinase (caspase)‑3, caspase‑4 and caspase‑9 were determined using reverse transcription semiquantitative and quantitative polymerase chain reaction analyses. Treatment with LCA inhibited proliferation and induced apoptosis of T24 cells, and increased intracellular Ca2+ levels and ROS production. Furthermore, LCA induced mitochondrial dysfunction, decreased mitochondrial membrane potential, and increased the mRNA expression levels of Apaf‑1, caspase‑9 and caspase‑3. Exposure of T24 cells to LCA also triggered calpain 2 and caspase‑4 activation, resulting in apoptosis. These findings indicated that LCA increased intracellular Ca2+ levels, which may be associated with mitochondrial dysfunction. In addition, the ER stress pathway may be considered an important mechanism by which LCA induces apoptosis of T24 bladder cancer cells.
Collapse
Affiliation(s)
- Xinhui Yang
- Department of Pharmacology, Key Laboratory of Xinjiang Endemic Phytomedicine Resources of Ministry of Education, School of Pharmacy, Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Jiangtao Jiang
- Department of Pharmacology, Key Laboratory of Xinjiang Endemic Phytomedicine Resources of Ministry of Education, School of Pharmacy, Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Xinyan Yang
- Department of Pharmacology, Key Laboratory of Xinjiang Endemic Phytomedicine Resources of Ministry of Education, School of Pharmacy, Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Jichun Han
- Department of Pharmacology, Key Laboratory of Xinjiang Endemic Phytomedicine Resources of Ministry of Education, School of Pharmacy, Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Qiusheng Zheng
- Department of Pharmacology, Key Laboratory of Xinjiang Endemic Phytomedicine Resources of Ministry of Education, School of Pharmacy, Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| |
Collapse
|
253
|
Guerrero CA, Acosta O. Inflammatory and oxidative stress in rotavirus infection. World J Virol 2016; 5:38-62. [PMID: 27175349 PMCID: PMC4861870 DOI: 10.5501/wjv.v5.i2.38] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 10/23/2015] [Accepted: 01/29/2016] [Indexed: 02/05/2023] Open
Abstract
Rotaviruses are the single leading cause of life-threatening diarrhea affecting children under 5 years of age. Rotavirus entry into the host cell seems to occur by sequential interactions between virion proteins and various cell surface molecules. The entry mechanisms seem to involve the contribution of cellular molecules having binding, chaperoning and oxido-reducing activities. It appears to be that the receptor usage and tropism of rotaviruses is determined by the species, cell line and rotavirus strain. Rotaviruses have evolved functions which can antagonize the host innate immune response, whereas are able to induce endoplasmic reticulum (ER) stress, oxidative stress and inflammatory signaling. A networking between ER stress, inflammation and oxidative stress is suggested, in which release of calcium from the ER increases the generation of mitochondrial reactive oxygen species (ROS) leading to toxic accumulation of ROS within ER and mitochondria. Sustained ER stress potentially stimulates inflammatory response through unfolded protein response pathways. However, the detailed characterization of the molecular mechanisms underpinning these rotavirus-induced stressful conditions is still lacking. The signaling events triggered by host recognition of virus-associated molecular patterns offers an opportunity for the development of novel therapeutic strategies aimed at interfering with rotavirus infection. The use of N-acetylcysteine, non-steroidal anti-inflammatory drugs and PPARγ agonists to inhibit rotavirus infection opens a new way for treating the rotavirus-induced diarrhea and complementing vaccines.
Collapse
|
254
|
Ali ES, Hua J, Wilson CH, Tallis GA, Zhou FH, Rychkov GY, Barritt GJ. The glucagon-like peptide-1 analogue exendin-4 reverses impaired intracellular Ca(2+) signalling in steatotic hepatocytes. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:2135-46. [PMID: 27178543 DOI: 10.1016/j.bbamcr.2016.05.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/20/2016] [Accepted: 05/04/2016] [Indexed: 02/07/2023]
Abstract
The release of Ca(2+) from the endoplasmic reticulum (ER) and subsequent replenishment of ER Ca(2+) by Ca(2+) entry through store-operated Ca(2+) channels (SOCE) play critical roles in the regulation of liver metabolism by adrenaline, glucagon and other hormones. Both ER Ca(2+) release and Ca(2+) entry are severely inhibited in steatotic hepatocytes. Exendin-4, a slowly-metabolised glucagon-like peptide-1 (GLP-1) analogue, is known to reduce liver glucose output and liver lipid, but the mechanisms involved are not well understood. The aim of this study was to determine whether exendin-4 alters intracellular Ca(2+) homeostasis in steatotic hepatocytes, and to evaluate the mechanisms involved. Exendin-4 completely reversed lipid-induced inhibition of SOCE in steatotic liver cells, but did not reverse lipid-induced inhibition of ER Ca(2+) release. The action of exendin-4 on Ca(2+) entry was rapid in onset and was mimicked by GLP-1 or dibutyryl cyclic AMP. In steatotic liver cells, exendin-4 caused a rapid decrease in lipid (half time 6.5min), inhibited the accumulation of lipid in liver cells incubated in the presence of palmitate plus the SOCE inhibitor BTP-2, and enhanced the formation of cyclic AMP. Hormone-stimulated accumulation of extracellular glucose in glycogen replete steatotic liver cells was inhibited compared to that in non-steatotic cells, and this effect of lipid was reversed by exendin-4. It is concluded that, in steatotic hepatocytes, exendin-4 reverses the lipid-induced inhibition of SOCE leading to restoration of hormone-regulated cytoplasmic Ca(2+) signalling. The mechanism may involve GLP-1 receptors, cyclic AMP, lipolysis, decreased diacylglycerol and decreased activity of protein kinase C.
Collapse
Affiliation(s)
- Eunüs S Ali
- Department of Medical Biochemistry and Centre for Neuroscience, School of Medicine, Flinders University, Adelaide, South Australia 5001, Australia
| | - Jin Hua
- Department of Medical Biochemistry and Centre for Neuroscience, School of Medicine, Flinders University, Adelaide, South Australia 5001, Australia
| | - Claire H Wilson
- Molecular Regulation Laboratory, Centre for Cancer Biology, Division of Health Sciences, University of South Australia, Adelaide, South Australia, 5001, Australia
| | - George A Tallis
- Medical Biochemistry, SA Pathology, Finders Medical Centre, Bedford Park, South Australia 5042, Australia
| | - Fiona H Zhou
- School of Medicine, The University of Adelaide, and South Australian Health and Medical Research Institute, Adelaide, South Australia 5005, Australia
| | - Grigori Y Rychkov
- School of Medicine, The University of Adelaide, and South Australian Health and Medical Research Institute, Adelaide, South Australia 5005, Australia
| | - Greg J Barritt
- Department of Medical Biochemistry and Centre for Neuroscience, School of Medicine, Flinders University, Adelaide, South Australia 5001, Australia.
| |
Collapse
|
255
|
Joshi AU, Kornfeld OS, Mochly-Rosen D. The entangled ER-mitochondrial axis as a potential therapeutic strategy in neurodegeneration: A tangled duo unchained. Cell Calcium 2016; 60:218-34. [PMID: 27212603 DOI: 10.1016/j.ceca.2016.04.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Revised: 04/28/2016] [Accepted: 04/28/2016] [Indexed: 12/12/2022]
Abstract
Endoplasmic reticulum (ER) and mitochondrial function have both been shown to be critical events in neurodegenerative diseases. The ER mediates protein folding, maturation, sorting as well acts as calcium storage. The unfolded protein response (UPR) is a stress response of the ER that is activated by the accumulation of misfolded proteins within the ER lumen. Although the molecular mechanisms underlying ER stress-induced apoptosis are not completely understood, increasing evidence suggests that ER and mitochondria cooperate to signal cell death. Similarly, calcium-mediated mitochondrial function and dynamics not only contribute to ATP generation and calcium buffering but are also a linchpin in mediating cell fate. Mitochondria and ER form structural and functional networks (mitochondria-associated ER membranes [MAMs]) essential to maintaining cellular homeostasis and determining cell fate under various pathophysiological conditions. Regulated Ca(2+) transfer from the ER to the mitochondria is important in maintaining control of pro-survival/pro-death pathways. In this review, we summarize the latest therapeutic strategies that target these essential organelles in the context of neurodegenerative diseases.
Collapse
Affiliation(s)
- Amit U Joshi
- Department of Chemical & Systems Biology, School of Medicine, Stanford University, CA, USA
| | - Opher S Kornfeld
- Department of Chemical & Systems Biology, School of Medicine, Stanford University, CA, USA
| | - Daria Mochly-Rosen
- Department of Chemical & Systems Biology, School of Medicine, Stanford University, CA, USA.
| |
Collapse
|
256
|
Ravera S, Dufour C, Cesaro S, Bottega R, Faleschini M, Cuccarolo P, Corsolini F, Usai C, Columbaro M, Cipolli M, Savoia A, Degan P, Cappelli E. Evaluation of energy metabolism and calcium homeostasis in cells affected by Shwachman-Diamond syndrome. Sci Rep 2016; 6:25441. [PMID: 27146429 PMCID: PMC4857091 DOI: 10.1038/srep25441] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 04/12/2016] [Indexed: 02/07/2023] Open
Abstract
Isomorphic mutation of the SBDS gene causes Shwachman-Diamond syndrome (SDS). SDS is a rare genetic bone marrow failure and cancer predisposition syndrome. SDS cells have ribosome biogenesis and their protein synthesis altered, which are two high-energy consuming cellular processes. The reported changes in reactive oxygen species production, endoplasmic reticulum stress response and reduced mitochondrial functionality suggest an energy production defect in SDS cells. In our work, we have demonstrated that SDS cells display a Complex IV activity impairment, which causes an oxidative phosphorylation metabolism defect, with a consequent decrease in ATP production. These data were confirmed by an increased glycolytic rate, which compensated for the energetic stress. Moreover, the signalling pathways involved in glycolysis activation also appeared more activated; i.e. we reported AMP-activated protein kinase hyper-phosphorylation. Notably, we also observed an increase in a mammalian target of rapamycin phosphorylation and high intracellular calcium concentration levels ([Ca(2+)]i), which probably represent new biochemical equilibrium modulation in SDS cells. Finally, the SDS cell response to leucine (Leu) was investigated, suggesting its possible use as a therapeutic adjuvant to be tested in clinical trials.
Collapse
Affiliation(s)
- Silvia Ravera
- DIFAR-Biochemistry Lab., Department of Pharmacy, University of Genova, 16132 Genova, Italy
| | - Carlo Dufour
- Haematology Unit, Istituto Giannina Gaslini, 16148 Genova, Italy
| | - Simone Cesaro
- Oncoematologia Pediatrica, Azienda Ospedaleira universitaria Integrata, Verona, Italy
| | - Roberta Bottega
- Department of Medical Sciences, University of Trieste, Trieste, Italy
| | - Michela Faleschini
- Institute for Maternal and Child Health – IRCCS Burlo Garofolo, Trieste, Italy
| | - Paola Cuccarolo
- S. C. Mutagenesis, IRCCS AOU San Martino – IST (Istituto Nazionale per la Ricerca sul Cancro), CBA Torre A2, 16123 Genova, Italy
| | - Fabio Corsolini
- Centro Diagnostica Genetica e Biochimica Malattie Metaboliche, Istituto Giannina Gaslini, 16148 Genova, Italy
| | - Cesare Usai
- Institute of Biophysics, National Research Council, 16149 Genova, Italy
| | - Marta Columbaro
- SC Laboratory of Musculoskeletal Cell Biology, IOR, Bologna, Italy
| | - Marco Cipolli
- Cystic Fibrosis Centre, Azienda Ospedaliera Universitaria, Piazzale Stefani, 1-37126 Verona, Italy
| | - Anna Savoia
- Institute for Maternal and Child Health – IRCCS Burlo Garofolo, Trieste, Italy
- Department of Medical Sciences, University of Trieste, Trieste, Italy
| | - Paolo Degan
- S. C. Mutagenesis, IRCCS AOU San Martino – IST (Istituto Nazionale per la Ricerca sul Cancro), CBA Torre A2, 16123 Genova, Italy
| | - Enrico Cappelli
- Haematology Unit, Istituto Giannina Gaslini, 16148 Genova, Italy
| |
Collapse
|
257
|
La Rovere RML, Roest G, Bultynck G, Parys JB. Intracellular Ca(2+) signaling and Ca(2+) microdomains in the control of cell survival, apoptosis and autophagy. Cell Calcium 2016; 60:74-87. [PMID: 27157108 DOI: 10.1016/j.ceca.2016.04.005] [Citation(s) in RCA: 205] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Revised: 04/14/2016] [Accepted: 04/15/2016] [Indexed: 01/01/2023]
Abstract
The endoplasmic reticulum (ER), mitochondria and lysosomes are physically and/or functionally linked, establishing close contact sites between these organelles. As a consequence, Ca(2+) release events from the ER, the major intracellular Ca(2+)-storage organelle, have an immediate effect on the physiological function of mitochondria and lysosomes. Also, the lysosomes can act as a Ca(2+) source for Ca(2+) release into the cytosol, thereby influencing ER-based Ca(2+) signaling. Given the important role for mitochondria and lysosomes in cell survival, cell death and cell adaptation processes, it has become increasingly clear that Ca(2+) signals from or towards these organelles impact these processes. In this review, we discuss the most recent insights in the emerging role of Ca(2+) signaling in cellular survival by controlling basal mitochondrial bioenergetics and by regulating apoptosis, a mitochondrial process, and autophagy, a lysosomal process, in response to cell damage and cell stress.
Collapse
Affiliation(s)
- Rita M L La Rovere
- KU Leuven, Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, BE-3000 Leuven, Belgium
| | - Gemma Roest
- KU Leuven, Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, BE-3000 Leuven, Belgium
| | - Geert Bultynck
- KU Leuven, Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, BE-3000 Leuven, Belgium.
| | - Jan B Parys
- KU Leuven, Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, BE-3000 Leuven, Belgium.
| |
Collapse
|
258
|
Poletto V, Dragoni S, Lim D, Biggiogera M, Aronica A, Cinelli M, De Luca A, Rosti V, Porta C, Guerra G, Moccia F. Endoplasmic Reticulum Ca2+Handling and Apoptotic Resistance in Tumor-Derived Endothelial Colony Forming Cells. J Cell Biochem 2016; 117:2260-71. [DOI: 10.1002/jcb.25524] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 02/22/2016] [Indexed: 12/23/2022]
Affiliation(s)
- Valentina Poletto
- Center for the Study of Myelofibrosis, Biotechnology Research Laboratory; Fondazione IRCCS Policlinico San Matteo; Pavia 27100 Italy
| | - Silvia Dragoni
- Laboratory of General Physiology, Department of Biology and Biotechnology “L. Spallanzani”; University of Pavia; Pavia 27100 Italy
| | - Dmitry Lim
- Department of Pharmaceutical Sciences; Università del Piemonte Orientale “Amedeo Avogadro”; Novara 28100 Italy
| | - Marco Biggiogera
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology “L. Spallanzani”; University of Pavia; Pavia 27100 Italy
| | - Adele Aronica
- Center for the Study of Myelofibrosis, Biotechnology Research Laboratory; Fondazione IRCCS Policlinico San Matteo; Pavia 27100 Italy
| | - Mariapia Cinelli
- Department of Public Health; University of Naples “Federico II”; Naples 80131 Italy
| | - Antonio De Luca
- Department of Mental and Physical Health and Preventive Medicine, Section of Human Anatomy, Second University of Naples; Largo Madonna delle Grazie 1; Naples 80138 Italy
| | - Vittorio Rosti
- Center for the Study of Myelofibrosis, Biotechnology Research Laboratory; Fondazione IRCCS Policlinico San Matteo; Pavia 27100 Italy
| | - Camillo Porta
- Medical Oncology; Fondazione IRCCS Policlinico San Matteo; Pavia 27100 Italy
| | - Germano Guerra
- Department of Medicine and Health Sciences “Vincenzo Tiberio”; University of Molise; Campobasso 86100 Italy
| | - Francesco Moccia
- Laboratory of General Physiology, Department of Biology and Biotechnology “L. Spallanzani”; University of Pavia; Pavia 27100 Italy
| |
Collapse
|
259
|
Bcl-2 proteins and calcium signaling: complexity beneath the surface. Oncogene 2016; 35:5079-92. [DOI: 10.1038/onc.2016.31] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 01/12/2016] [Accepted: 01/12/2016] [Indexed: 12/12/2022]
|
260
|
Junctophilin-4, a component of the endoplasmic reticulum-plasma membrane junctions, regulates Ca2+ dynamics in T cells. Proc Natl Acad Sci U S A 2016; 113:2762-7. [PMID: 26929330 DOI: 10.1073/pnas.1524229113] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Orai1 and stromal interaction molecule 1 (STIM1) mediate store-operated Ca(2+) entry (SOCE) in immune cells. STIM1, an endoplasmic reticulum (ER) Ca(2+) sensor, detects store depletion and interacts with plasma membrane (PM)-resident Orai1 channels at the ER-PM junctions. However, the molecular composition of these junctions in T cells remains poorly understood. Here, we show that junctophilin-4 (JP4), a member of junctional proteins in excitable cells, is expressed in T cells and localized at the ER-PM junctions to regulate Ca(2+) signaling. Silencing or genetic manipulation of JP4 decreased ER Ca(2+) content and SOCE in T cells, impaired activation of the nuclear factor of activated T cells (NFAT) and extracellular signaling-related kinase (ERK) signaling pathways, and diminished expression of activation markers and cytokines. Mechanistically, JP4 directly interacted with STIM1 via its cytoplasmic domain and facilitated its recruitment into the junctions. Accordingly, expression of this cytoplasmic fragment of JP4 inhibited SOCE. Furthermore, JP4 also formed a complex with junctate, a Ca(2+)-sensing ER-resident protein, previously shown to mediate STIM1 recruitment into the junctions. We propose that the junctate-JP4 complex located at the junctions cooperatively interacts with STIM1 to maintain ER Ca(2+) homeostasis and mediate SOCE in T cells.
Collapse
|
261
|
Shen L, Wen N, Xia M, Zhang YU, Liu W, Xu YE, Sun L. Calcium efflux from the endoplasmic reticulum regulates cisplatin-induced apoptosis in human cervical cancer HeLa cells. Oncol Lett 2016; 11:2411-2419. [PMID: 27073489 PMCID: PMC4812401 DOI: 10.3892/ol.2016.4278] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 02/04/2016] [Indexed: 11/21/2022] Open
Abstract
The function of calcium efflux from the endoplasmic reticulum (ER) in cisplatin-induced apoptosis is not fully understood in cancer cells. The present study used western blot analysis, flow cytometry, immunofluorescence and 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide assay to investigate calcium signaling in human cervical cancer cells exposed to cisplatin. In the present study, treatment with cisplatin increased free Ca2+ levels in the cytoplasm and mitochondria of human cervical cancer HeLa cells, which further triggers the mitochondria-mediated and ER stress-associated apoptosis pathways. Notably, blocking calcium signaling using the calcium chelating agent bis-(o-aminophenoxy)ethane-N,N,N',N'-tetra-acetic acid acetoxymethyl ester inhibited cisplatin-induced apoptosis via downregulation of the calcium-dependent proteases, the calpains, and innate apoptosis proteins, such as caspsae-3, caspase-4 and C/EBP homologous protein (CHOP). In addition, use of the inositol triphosphate receptor inhibitor, 2-aminoethyl diphenylborinate, to inhibit calcium efflux from the ER resulted in similar effects. This data indicated that calcium efflux from the ER plays a significant role in cisplatin-induced apoptosis in human cervical cancer HeLa cells, which provides further mechanistic insights into the tumor cell-killing effect of cisplatin and potential therapeutic strategies to improve cisplatin chemotherapy.
Collapse
Affiliation(s)
- Luyan Shen
- Department of Pathophysiology, Basic College of Medicine, Jilin University, Changchun, Jilin 130021, P.R. China; Department of Obstetrics and Gynecology, Second Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Naiyan Wen
- Department of Pathophysiology, Basic College of Medicine, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Meihui Xia
- Department of Obstetrics and Gynecology, First Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Y U Zhang
- Department of Pathophysiology, Basic College of Medicine, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Weimin Liu
- Department of Interventional Radiology, Renmin Hospital, Jilin, Jilin 132013, P.R. China
| | - Y E Xu
- Medical Research Laboratory, Jilin Medical College, Jilin, Jilin 132013, P.R. China
| | - Liankun Sun
- Department of Pathophysiology, Basic College of Medicine, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
262
|
Bcl-2 and FKBP12 bind to IP3 and ryanodine receptors at overlapping sites: the complexity of protein-protein interactions for channel regulation. Biochem Soc Trans 2016; 43:396-404. [PMID: 26009182 DOI: 10.1042/bst20140298] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The 12- and 12.6-kDa FK506-binding proteins, FKBP12 (12-kDa FK506-binding protein) and FKBP12.6 (12.6-kDa FK506-binding protein), have been implicated in the binding to and the regulation of ryanodine receptors (RyRs) and inositol 1,4,5-trisphosphate receptors (IP3Rs), both tetrameric intracellular Ca2+-release channels. Whereas the amino acid sequences responsible for FKBP12 binding to RyRs are conserved in IP3Rs, FKBP12 binding to IP3Rs has been questioned and could not be observed in various experimental models. Nevertheless, conservation of these residues in the different IP3R isoforms and during evolution suggested that they could harbour an important regulatory site critical for IP3R-channel function. Recently, it has become clear that in IP3Rs, this site was targeted by B-cell lymphoma 2 (Bcl-2) via its Bcl-2 homology (BH)4 domain, thereby dampening IP3R-mediated Ca2+ flux and preventing pro-apoptotic Ca2+ signalling. Furthermore, vice versa, the presence of the corresponding site in RyRs implied that Bcl-2 proteins could associate with and regulate RyR channels. Recently, the existence of endogenous RyR-Bcl-2 complexes has been identified in primary hippocampal neurons. Like for IP3Rs, binding of Bcl-2 to RyRs also involved its BH4 domain and suppressed RyR-mediated Ca2+ release. We therefore propose that the originally identified FKBP12-binding site in IP3Rs is a region critical for controlling IP3R-mediated Ca2+ flux by recruiting Bcl-2 rather than FKBP12. Although we hypothesize that anti-apoptotic Bcl-2 proteins, but not FKBP12, are the main physiological inhibitors of IP3Rs, we cannot exclude that Bcl-2 could help engaging FKBP12 (or other FKBP isoforms) to the IP3R, potentially via calcineurin.
Collapse
|
263
|
Shinde V, Kotla P, Strang C, Gorbatyuk M. Unfolded protein response-induced dysregulation of calcium homeostasis promotes retinal degeneration in rat models of autosomal dominant retinitis pigmentosa. Cell Death Dis 2016; 7:e2085. [PMID: 26844699 PMCID: PMC4670931 DOI: 10.1038/cddis.2015.325] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 09/15/2015] [Accepted: 09/30/2015] [Indexed: 12/12/2022]
Abstract
The molecular mechanism of autosomal dominant retinitis pigmentosa (ADRP) in rats is closely associated with a persistently activated unfolded protein response (UPR). If unchecked, the UPR might trigger apoptosis, leading to photoreceptor death. One of the UPR-activated cellular signaling culminating in apoptotic photoreceptor cell death is linked to an increase in intracellular Ca(2+). Therefore, we validated whether ADRP retinas experience a cytosolic Ca(2+) overload, and whether sustained UPR in the wild-type retina could promote retinal degeneration through Ca(2+)-mediated calpain activation. We performed an ex vivo experiment to measure intracellular Ca(2+) in ADRP retinas as well as to detect the expression levels of proteins that act as Ca(2+) sensors. In separate experiments with the subretinal injection of tunicamycin (UPR inducer) and a mixture of calcium ionophore (A231278) and thapsigargin (SERCA2b inhibitor) we assessed the consequences of a sustained UPR activation and increased intracellular Ca(2+) in the wild-type retina, respectively, by performing scotopic ERG, histological, and western blot analyses. Results of the study revealed that induced UPR in the retina activates calpain-mediated signaling, and increased intracellular Ca(2+) is capable of promoting retinal degeneration. A significant decline in ERG amplitudes at 6 weeks post treatment was associated with photoreceptor cell loss that occurred through calpain-activated CDK5-pJNK-Csp3/7 pathway. Similar calpain activation was found in ADRP rat retinas. A twofold increase in intracellular Ca(2+) and up- and downregulations of ER membrane-associated Ca(2+)-regulated IP3R channels and SERCA2b transporters were detected. Therefore, sustained UPR activation in the ADRP rat retinas could promote retinal degeneration through increased intracellular Ca(2+) and calpain-mediated apoptosis.
Collapse
Affiliation(s)
- V Shinde
- Department of Vision Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - P Kotla
- Department of Vision Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - C Strang
- Department of Vision Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - M Gorbatyuk
- Department of Vision Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
264
|
Ruiz M, Coderre L, Lachance D, Houde V, Martel C, Thompson Legault J, Gillis MA, Bouchard B, Daneault C, Carpentier AC, Gaestel M, Allen BG, Des Rosiers C. MK2 Deletion in Mice Prevents Diabetes-Induced Perturbations in Lipid Metabolism and Cardiac Dysfunction. Diabetes 2016; 65:381-92. [PMID: 26558681 DOI: 10.2337/db15-0238] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 11/05/2015] [Indexed: 11/13/2022]
Abstract
Heart disease remains a major complication of diabetes, and the identification of new therapeutic targets is essential. This study investigates the role of the protein kinase MK2, a p38 mitogen-activated protein kinase downstream target, in the development of diabetes-induced cardiomyopathy. Diabetes was induced in control (MK2(+/+)) and MK2-null (MK2(-/-)) mice using repeated injections of a low dose of streptozotocin (STZ). This protocol generated in MK2(+/+) mice a model of diabetes characterized by a 50% decrease in plasma insulin, hyperglycemia, and insulin resistance (IR), as well as major contractile dysfunction, which was associated with alterations in proteins involved in calcium handling. While MK2(-/-)-STZ mice remained hyperglycemic, they showed improved IR and none of the cardiac functional or molecular alterations. Further analyses highlighted marked lipid perturbations in MK2(+/+)-STZ mice, which encompass increased 1) circulating levels of free fatty acid, ketone bodies, and long-chain acylcarnitines and 2) cardiac triglyceride accumulation and ex vivo palmitate β-oxidation. MK2(-/-)-STZ mice were also protected against all these diabetes-induced lipid alterations. Our results demonstrate the benefits of MK2 deletion on diabetes-induced cardiac molecular and lipid metabolic changes, as well as contractile dysfunction. As a result, MK2 represents a new potential therapeutic target to prevent diabetes-induced cardiac dysfunction.
Collapse
Affiliation(s)
- Matthieu Ruiz
- Department of Nutrition, Université de Montréal, Montréal, Québec, Canada Research Center, Montreal Heart Institute, Montréal, Québec, Canada
| | - Lise Coderre
- Department of Nutrition, Université de Montréal, Montréal, Québec, Canada Research Center, Montreal Heart Institute, Montréal, Québec, Canada Department of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Dominic Lachance
- Department of Nutrition, Université de Montréal, Montréal, Québec, Canada Research Center, Montreal Heart Institute, Montréal, Québec, Canada
| | - Valérie Houde
- Department of Nutrition, Université de Montréal, Montréal, Québec, Canada Research Center, Montreal Heart Institute, Montréal, Québec, Canada
| | - Cécile Martel
- Research Center, Montreal Heart Institute, Montréal, Québec, Canada
| | - Julie Thompson Legault
- Department of Nutrition, Université de Montréal, Montréal, Québec, Canada Research Center, Montreal Heart Institute, Montréal, Québec, Canada
| | | | - Bertrand Bouchard
- Department of Nutrition, Université de Montréal, Montréal, Québec, Canada Research Center, Montreal Heart Institute, Montréal, Québec, Canada
| | - Caroline Daneault
- Department of Nutrition, Université de Montréal, Montréal, Québec, Canada Research Center, Montreal Heart Institute, Montréal, Québec, Canada
| | - André C Carpentier
- Department of Medicine, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Matthias Gaestel
- Institute of Biochemistry, Hannover Medical School, Hannover, Germany
| | - Bruce G Allen
- Research Center, Montreal Heart Institute, Montréal, Québec, Canada Department of Medicine, Université de Montréal, Montréal, Québec, Canada Department of Biochemistry, Université de Montréal, Montréal, Québec, Canada
| | - Christine Des Rosiers
- Department of Nutrition, Université de Montréal, Montréal, Québec, Canada Research Center, Montreal Heart Institute, Montréal, Québec, Canada Department of Medicine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
265
|
Duan J, Kodali VK, Gaffrey MJ, Guo J, Chu RK, Camp DG, Smith RD, Thrall BD, Qian WJ. Quantitative Profiling of Protein S-Glutathionylation Reveals Redox-Dependent Regulation of Macrophage Function during Nanoparticle-Induced Oxidative Stress. ACS NANO 2016; 10:524-38. [PMID: 26700264 PMCID: PMC4762218 DOI: 10.1021/acsnano.5b05524] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Engineered nanoparticles (ENPs) are increasingly utilized for commercial and medical applications; thus, understanding their potential adverse effects is an important societal issue. Herein, we investigated protein S-glutathionylation (SSG) as an underlying regulatory mechanism by which ENPs may alter macrophage innate immune functions, using a quantitative redox proteomics approach for site-specific measurement of SSG modifications. Three high-volume production ENPs (SiO2, Fe3O4, and CoO) were selected as representatives which induce low, moderate, and high propensity, respectively, to stimulate cellular reactive oxygen species (ROS) and disrupt macrophage function. The SSG modifications identified highlighted a broad set of redox sensitive proteins and specific Cys residues which correlated well with the overall level of cellular redox stress and impairment of macrophage phagocytic function (CoO > Fe3O4 ≫ SiO2). Moreover, our data revealed pathway-specific differences in susceptibility to SSG between ENPs which induce moderate versus high levels of ROS. Pathways regulating protein translation and protein stability indicative of ER stress responses and proteins involved in phagocytosis were among the most sensitive to SSG in response to ENPs that induce subcytoxic levels of redox stress. At higher levels of redox stress, the pattern of SSG modifications displayed reduced specificity and a broader set pathways involving classical stress responses and mitochondrial energetics (e.g., glycolysis) associated with apoptotic mechanisms. An important role for SSG in regulation of macrophage innate immune function was also confirmed by RNA silencing of glutaredoxin, a major enzyme which reverses SSG modifications. Our results provide unique insights into the protein signatures and pathways that serve as ROS sensors and may facilitate cellular adaption to ENPs, versus intracellular targets of ENP-induced oxidative stress that are linked to irreversible cell outcomes.
Collapse
Affiliation(s)
- Jicheng Duan
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - Vamsi K. Kodali
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - Matthew J. Gaffrey
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - Jia Guo
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - Rosalie K. Chu
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - David G. Camp
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - Richard D. Smith
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - Brian D. Thrall
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
- Corresponding Authors: .
| | - Wei-Jun Qian
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
- Corresponding Authors: .
| |
Collapse
|
266
|
Cho GW, Altamirano F, Hill JA. Chronic heart failure: Ca(2+), catabolism, and catastrophic cell death. Biochim Biophys Acta Mol Basis Dis 2016; 1862:763-777. [PMID: 26775029 DOI: 10.1016/j.bbadis.2016.01.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 12/28/2015] [Accepted: 01/06/2016] [Indexed: 12/12/2022]
Abstract
Robust successes have been achieved in recent years in conquering the acutely lethal manifestations of heart disease. Many patients who previously would have died now survive to enjoy happy and productive lives. Nevertheless, the devastating impact of heart disease continues unabated, as the spectrum of disease has evolved with new manifestations. In light of this ever-evolving challenge, insights that culminate in novel therapeutic targets are urgently needed. Here, we review fundamental mechanisms of heart failure, both with reduced (HFrEF) and preserved (HFpEF) ejection fraction. We discuss pathways that regulate cardiomyocyte remodeling and turnover, focusing on Ca(2+) signaling, autophagy, and apoptosis. In particular, we highlight recent insights pointing to novel connections among these events. We also explore mechanisms whereby potential therapeutic approaches targeting these processes may improve morbidity and mortality in the devastating syndrome of heart failure.
Collapse
Affiliation(s)
- Geoffrey W Cho
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Francisco Altamirano
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Joseph A Hill
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
267
|
Rashid HO, Yadav RK, Kim HR, Chae HJ. ER stress: Autophagy induction, inhibition and selection. Autophagy 2015; 11:1956-1977. [PMID: 26389781 DOI: 10.1080/15548627.2015.1091141] [Citation(s) in RCA: 573] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
An accumulation of unfolded or misfolded proteins in the endoplasmic reticulum (ER) leads to stress conditions. To mitigate such circumstances, stressed cells activate a homeostatic intracellular signaling network cumulatively called the unfolded protein response (UPR), which orchestrates the recuperation of ER function. Macroautophagy (hereafter autophagy), an intracellular lysosome-mediated bulk degradation pathway for recycling and eliminating wornout proteins, protein aggregates, and damaged organelles, has also emerged as an essential protective mechanism during ER stress. These 2 systems are dynamically interconnected, and recent investigations have revealed that ER stress can either stimulate or inhibit autophagy. However, the stress-associated molecular cues that control the changeover switch between induction and inhibition of autophagy are largely obscure. This review summarizes the crosstalk between ER stress and autophagy and their signaling networks mainly in mammalian-based systems. Additionally, we highlight current knowledge on selective autophagy and its connection to ER stress.
Collapse
Affiliation(s)
- Harun-Or Rashid
- a Department of Pharmacology ; Medical School; Chonbuk National University
| | - Raj Kumar Yadav
- a Department of Pharmacology ; Medical School; Chonbuk National University
| | - Hyung-Ryong Kim
- b Department of Dental Pharmacology ; College of Dentistry; Wonkwang University
| | - Han-Jung Chae
- a Department of Pharmacology ; Medical School; Chonbuk National University
| |
Collapse
|
268
|
Richter M, Vidovic N, Honrath B, Mahavadi P, Dodel R, Dolga AM, Culmsee C. Activation of SK2 channels preserves ER Ca²⁺ homeostasis and protects against ER stress-induced cell death. Cell Death Differ 2015; 23:814-27. [PMID: 26586570 DOI: 10.1038/cdd.2015.146] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 09/03/2015] [Accepted: 09/21/2015] [Indexed: 01/24/2023] Open
Abstract
Alteration of endoplasmic reticulum (ER) Ca(2+) homeostasis leads to excessive cytosolic Ca(2+) accumulation and delayed neuronal cell death in acute and chronic neurodegenerative disorders. While our recent studies established a protective role for SK channels against excessive intracellular Ca(2+) accumulation, their functional role in the ER has not been elucidated yet. We show here that SK2 channels are present in ER membranes of neuronal HT-22 cells, and that positive pharmacological modulation of SK2 channels with CyPPA protects against cell death induced by the ER stressors brefeldin A and tunicamycin. Calcium imaging of HT-22 neurons revealed that elevated cytosolic Ca(2+) levels and decreased ER Ca(2+) load during sustained ER stress could be largely prevented by SK2 channel activation. Interestingly, SK2 channel activation reduced the amount of the unfolded protein response transcription factor ATF4, but further enhanced the induction of CHOP. Using siRNA approaches we confirmed a detrimental role for ATF4 in ER stress, whereas CHOP regulation was dispensable for both, brefeldin A toxicity and CyPPA-mediated protection. Cell death induced by blocking Ca(2+) influx into the ER with the SERCA inhibitor thapsigargin was not prevented by CyPPA. Blocking the K(+) efflux via K(+)/H(+) exchangers with quinine inhibited CyPPA-mediated neuroprotection, suggesting an essential role of proton uptake and K(+) release in the SK channel-mediated neuroprotection. Our data demonstrate that ER SK2 channel activation preserves ER Ca(2+) uptake and retention which determines cell survival in conditions where sustained ER stress contributes to progressive neuronal death.
Collapse
Affiliation(s)
- M Richter
- Institute for Pharmacology and Clinical Pharmacy, Philipps-University Marburg, Marburg, Germany.,Department of Neurology, Philipps-University Marburg, Marburg, Germany
| | - N Vidovic
- Department of Neurology, Philipps-University Marburg, Marburg, Germany
| | - B Honrath
- Institute for Pharmacology and Clinical Pharmacy, Philipps-University Marburg, Marburg, Germany
| | - P Mahavadi
- Department of Internal Medicine, Justus-Liebig-University, Giessen, Germany.,Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - R Dodel
- Department of Neurology, Philipps-University Marburg, Marburg, Germany
| | - A M Dolga
- Institute for Pharmacology and Clinical Pharmacy, Philipps-University Marburg, Marburg, Germany.,Faculty of Mathematics and Natural Sciences, Molecular Pharmacology - Groningen Research Institute of Pharmacy, Groningen, The Netherlands
| | - C Culmsee
- Institute for Pharmacology and Clinical Pharmacy, Philipps-University Marburg, Marburg, Germany
| |
Collapse
|
269
|
Cominacini L, Mozzini C, Garbin U, Pasini A, Stranieri C, Solani E, Vallerio P, Tinelli IA, Fratta Pasini A. Endoplasmic reticulum stress and Nrf2 signaling in cardiovascular diseases. Free Radic Biol Med 2015; 88:233-242. [PMID: 26051167 DOI: 10.1016/j.freeradbiomed.2015.05.027] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 05/14/2015] [Accepted: 05/17/2015] [Indexed: 12/30/2022]
Abstract
Various cellular perturbations implicated in the pathophysiology of human diseases, including cardiovascular and neurodegenerative diseases, diabetes mellitus, obesity, and liver diseases, can alter endoplasmic reticulum (ER) function and lead to the abnormal accumulation of misfolded proteins. This situation configures the so-called ER stress, a form of intracellular stress that occurs whenever the protein-folding capacity of the ER is overwhelmed. Reduction in blood flow as a result of atherosclerotic coronary artery disease causes tissue hypoxia, a condition that induces protein misfolding and ER stress. In addition, ER stress has an important role in cardiac hypertrophy mainly in the transition to heart failure (HF). ER transmembrane sensors detect the accumulation of unfolded proteins and activate transcriptional and translational pathways that deal with unfolded and misfolded proteins, known as the unfolded protein response (UPR). Once the UPR fails to control the level of unfolded and misfolded proteins in the ER, ER-initiated apoptotic signaling is induced. Furthermore, there is considerable evidence that implicates the presence of oxidative stress and subsequent related cellular damage as an initial cause of injury to the myocardium after ischemia/reperfusion (I/R) and in cardiac hypertrophy secondary to pressure overload. Oxidative stress is counterbalanced by complex antioxidant defense systems regulated by a series of multiple pathways, including the UPR, to ensure that the response to oxidants is adequate. Nuclear factor-E2-related factor (Nrf2) is an emerging regulator of cellular resistance to oxidants; Nrf2 is strictly interrelated with the UPR sensor called pancreatic endoplasmic reticulum kinase. A series of studies has shown that interventions against ER stress and Nrf2 activation reduce myocardial infarct size and cardiac hypertrophy in the transition to HF in animals exposed to I/R injury and pressure overload, respectively. Finally, recent data showed that Nrf2/antioxidant-response element pathway activation may be of importance also in ischemic preconditioning, a phenomenon in which the heart is subjected to one or more episodes of nonlethal myocardial I/R before the sustained coronary artery occlusion.
Collapse
Affiliation(s)
- Luciano Cominacini
- Section of Internal Medicine, Department of Medicine, University of Verona, 37134 Verona, Italy.
| | - Chiara Mozzini
- Section of Internal Medicine, Department of Medicine, University of Verona, 37134 Verona, Italy
| | - Ulisse Garbin
- Section of Internal Medicine, Department of Medicine, University of Verona, 37134 Verona, Italy
| | - Andrea Pasini
- Section of Internal Medicine, Department of Medicine, University of Verona, 37134 Verona, Italy
| | - Chiara Stranieri
- Section of Internal Medicine, Department of Medicine, University of Verona, 37134 Verona, Italy
| | - Erika Solani
- Section of Internal Medicine, Department of Medicine, University of Verona, 37134 Verona, Italy
| | - Paola Vallerio
- Section of Internal Medicine, Department of Medicine, University of Verona, 37134 Verona, Italy
| | | | - Anna Fratta Pasini
- Section of Internal Medicine, Department of Medicine, University of Verona, 37134 Verona, Italy
| |
Collapse
|
270
|
Marré ML, James EA, Piganelli JD. β cell ER stress and the implications for immunogenicity in type 1 diabetes. Front Cell Dev Biol 2015; 3:67. [PMID: 26579520 PMCID: PMC4621612 DOI: 10.3389/fcell.2015.00067] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 10/08/2015] [Indexed: 12/11/2022] Open
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disease characterized by hyperglycemia due to progressive immune-mediated destruction of insulin-producing pancreatic islet β cells. Although many elegant studies have identified β cell autoantigens that are targeted by the autoimmune response, the mechanisms by which these autoantigens are generated remain poorly understood. Normal β cell physiology includes a high demand for insulin production and secretion in response to dynamic glucose sensing. This secretory function predisposes β cells to significantly higher levels of endoplasmic reticulum (ER) stress compared to nonsecretory cells. In addition, many environmental triggers associated with T1D onset further augment this inherent ER stress in β cells. ER stress may increase abnormal post-translational modification (PTM) of endogenous β cell proteins. Indeed, in other autoimmune disorders such as celiac disease, systemic lupus erythematosus, multiple sclerosis, and rheumatoid arthritis, abnormally modified neo-antigens are presented by antigen presenting cells (APCs) in draining lymph nodes. In the context of genetic susceptibility to autoimmunity, presentation of neo-antigens activates auto-reactive T cells and pathology ensues. Therefore, the ER stress induced by normal β cell secretory physiology and environmental triggers may be sufficient to generate neo-antigens for the autoimmune response in T1D. This review summarizes what is currently known about ER stress and protein PTM in target organs of other autoimmune disease models, as well as the data supporting a role for ER stress-induced neo-antigen formation in β cells in T1D.
Collapse
Affiliation(s)
- Meghan L Marré
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Pittsburgh, PA, USA
| | - Eddie A James
- Benaroya Research Institute at Virginia Mason Seattle, WA, USA
| | - Jon D Piganelli
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Pittsburgh, PA, USA
| |
Collapse
|
271
|
Grinstead K, Joel S, Zingg JM, Dikici E, Daunert S. Enabling Aequorin for Biotechnology Applications Through Genetic Engineering. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2015:149-179. [PMID: 26475468 DOI: 10.1007/10_2015_336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In recent years, luminescent proteins have been studied for their potential application in a variety of detection systems. Bioluminescent proteins, which do not require an external excitation source, are especially well-suited as reporters in analytical detection. The photoprotein aequorin is a bioluminescent protein that can be engineered for use as a molecular reporter under a wide range of conditions while maintaining its sensitivity. Herein, the characteristics of aequorin as well as the engineering and production of aequorin variants and their impact on signal detection in biological systems are presented. The structural features and activity of aequorin, its benefits as a label for sensing and applications in highly sensitive detection, as well as in gaining insight into biological processes are discussed. Among those, focus has been placed on the highly sensitive calcium detection in vivo, in vitro DNA and small molecule sensing, and development of in vivo imaging technologies. Graphical Abstract.
Collapse
Affiliation(s)
- Kristen Grinstead
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Smita Joel
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Jean-Marc Zingg
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Emre Dikici
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Sylvia Daunert
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA.
| |
Collapse
|
272
|
Yang F, Luo J. Endoplasmic Reticulum Stress and Ethanol Neurotoxicity. Biomolecules 2015; 5:2538-53. [PMID: 26473940 PMCID: PMC4693246 DOI: 10.3390/biom5042538] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 09/02/2015] [Accepted: 09/21/2015] [Indexed: 12/21/2022] Open
Abstract
Ethanol abuse affects virtually all organ systems and the central nervous system (CNS) is particularly vulnerable to excessive ethanol exposure. Ethanol exposure causes profound damages to both the adult and developing brain. Prenatal ethanol exposure induces fetal alcohol spectrum disorders (FASD) which is associated with mental retardation and other behavioral deficits. A number of potential mechanisms have been proposed for ethanol-induced brain damage; these include the promotion of neuroinflammation, interference with signaling by neurotrophic factors, induction of oxidative stress, modulation of retinoid acid signaling, and thiamine deficiency. The endoplasmic reticulum (ER) regulates posttranslational protein processing and transport. The accumulation of unfolded or misfolded proteins in the ER lumen triggers ER stress and induces unfolded protein response (UPR) which are mediated by three transmembrane ER signaling proteins: pancreatic endoplasmic reticulum kinase (PERK), inositol-requiring enzyme 1 (IRE1), and activating transcription factor 6 (ATF6). UPR is initiated to protect cells from overwhelming ER protein loading. However, sustained ER stress may result in cell death. ER stress has been implied in various CNS injuries, including brain ischemia, traumatic brain injury, and aging-associated neurodegeneration, such as Alzheimer's disease (AD), Huntington's disease (HD), Amyotrophic lateral sclerosis (ALS), and Parkinson's disease (PD). However, effects of ethanol on ER stress in the CNS receive less attention. In this review, we discuss recent progress in the study of ER stress in ethanol-induced neurotoxicity. We also examine the potential mechanisms underlying ethanol-mediated ER stress and the interaction among ER stress, oxidative stress and autophagy in the context of ethanol neurotoxicity.
Collapse
Affiliation(s)
- Fanmuyi Yang
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, 132 Health Sciences Research Building, 1095 Veterans Drive, Lexington, KY 40536, USA.
| | - Jia Luo
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, 132 Health Sciences Research Building, 1095 Veterans Drive, Lexington, KY 40536, USA.
| |
Collapse
|
273
|
Kim AD, Madduma Hewage SRK, Piao MJ, Kang KA, Cho SJ, Hyun JW. Esculetin induces apoptosis in human colon cancer cells by inducing endoplasmic reticulum stress. Cell Biochem Funct 2015; 33:487-94. [DOI: 10.1002/cbf.3146] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 09/04/2015] [Accepted: 09/07/2015] [Indexed: 12/13/2022]
Affiliation(s)
| | | | - Mei Jing Piao
- School of Medicine; Jeju National University; Jeju Korea
| | - Kyoung Ah Kang
- School of Medicine; Jeju National University; Jeju Korea
| | - Suk Ju Cho
- School of Medicine; Jeju National University; Jeju Korea
| | - Jin Won Hyun
- School of Medicine; Jeju National University; Jeju Korea
| |
Collapse
|
274
|
The effect of bovine rotavirus and its nonstructural protein 4 on ER stress-mediated apoptosis in HeLa and HT-29 cells. Tumour Biol 2015; 37:3155-61. [PMID: 26427658 DOI: 10.1007/s13277-015-4097-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 09/14/2015] [Indexed: 02/08/2023] Open
Abstract
Endoplasmic reticulum (ER) plays important roles in multiple cellular processes as well as cell survival and apoptosis. Perturbation of ER functions leads to ER stress and unfolded protein response (UPR). The primary goal of this response is cell survival, but severe ER stress can trigger apoptosis signaling. In tumor cells, chronically activated UPR response provides tumor growth. So, apoptosis induced by the ER stress has been the target for anti-cancer therapy. In this in vitro study, we examined the apoptotic effect associated with ER stress of bovine rotavirus and its nonstructural protein 4 (NSP4) alone in two cancer cell lines. The plasmid pcDNA3.1 encoding NSP4 protein of bovine rotavirus transfected with lipofectamine 2000 into the HeLa and HT-29 cells for protein production. MTT, flow cytometry, and Western blot were used to evaluate the cell viability, apoptosis, and expression level of C/EBP-homologous protein (CHOP) and activated caspase-4. In parallel, the apoptotic effect of the bovine rotavirus associated with ER stress in the infected cells was examined too. The cytotoxic and apoptotic effect of NSP4 protein on the cells were statistically significant compared to the control groups. However, Western blot showed that the expression of the NSP4 protein by recombinant plasmid did not lead to high expression of CHOP and activation of caspase-4. Interestingly, rotavirus not only induced significant apoptosis but also caused an increase in CHOP expression and caspase-4 activation in the infected cells compared to control. As a result, NSP4 protein and bovine rotavirus can be considered a potential novel bio-therapeutic strategy for cancer treatment.
Collapse
|
275
|
Song L, Cui R, Yang Y, Wu X. Role of calcium channels in cellular antituberculosis effects: Potential of voltage-gated calcium-channel blockers in tuberculosis therapy. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2015; 48:471-6. [DOI: 10.1016/j.jmii.2014.08.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 06/13/2014] [Accepted: 08/07/2014] [Indexed: 11/26/2022]
|
276
|
Henderson MJ, Wires ES, Trychta KA, Yan X, Harvey BK. Monitoring Endoplasmic Reticulum Calcium Homeostasis Using a Gaussia Luciferase SERCaMP. J Vis Exp 2015. [PMID: 26383227 DOI: 10.3791/53199] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The endoplasmic reticulum (ER) contains the highest level of intracellular calcium, with concentrations approximately 5,000-fold greater than cytoplasmic levels. Tight control over ER calcium is imperative for protein folding, modification and trafficking. Perturbations to ER calcium can result in the activation of the unfolded protein response, a three-prong ER stress response mechanism, and contribute to pathogenesis in a variety of diseases. The ability to monitor ER calcium alterations during disease onset and progression is important in principle, yet challenging in practice. Currently available methods for monitoring ER calcium, such as calcium-dependent fluorescent dyes and proteins, have provided insight into ER calcium dynamics in cells, however these tools are not well suited for in vivo studies. Our lab has demonstrated that a modification to the carboxy-terminus of Gaussia luciferase confers secretion of the reporter in response to ER calcium depletion. The methods for using a luciferase based, secreted ER calcium monitoring protein (SERCaMP) for in vitro and in vivo applications are described herein. This video highlights hepatic injections, pharmacological manipulation of GLuc-SERCaMP, blood collection and processing, and assay parameters for longitudinal monitoring of ER calcium.
Collapse
Affiliation(s)
| | - Emily S Wires
- National Institute on Drug Abuse, National Institutes of Health
| | | | - Xiaokang Yan
- National Institute on Drug Abuse, National Institutes of Health
| | - Brandon K Harvey
- National Institute on Drug Abuse, National Institutes of Health;
| |
Collapse
|
277
|
Abstract
A number of chronic metabolic pathologies, including obesity, diabetes, cardiovascular disease, asthma, and cancer, cluster together to present the greatest threat to human health. As research in this field has advanced, it has become clear that unresolved metabolic inflammation, organelle dysfunction, and other cellular and metabolic stresses underlie the development of these chronic metabolic diseases. However, the relationship between these systems and pathological mechanisms is poorly understood. Here we discuss the role of cellular Ca(2+) homeostasis as a critical mechanism integrating the myriad of cellular and subcellular dysfunctional networks found in metabolic tissues such as liver and adipose tissue in the context of metabolic disease, particularly in obesity and diabetes.
Collapse
|
278
|
Wen L, Voronina S, Javed MA, Awais M, Szatmary P, Latawiec D, Chvanov M, Collier D, Huang W, Barrett J, Begg M, Stauderman K, Roos J, Grigoryev S, Ramos S, Rogers E, Whitten J, Velicelebi G, Dunn M, Tepikin AV, Criddle DN, Sutton R. Inhibitors of ORAI1 Prevent Cytosolic Calcium-Associated Injury of Human Pancreatic Acinar Cells and Acute Pancreatitis in 3 Mouse Models. Gastroenterology 2015; 149:481-92.e7. [PMID: 25917787 PMCID: PMC4556985 DOI: 10.1053/j.gastro.2015.04.015] [Citation(s) in RCA: 153] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 04/17/2015] [Accepted: 04/20/2015] [Indexed: 02/05/2023]
Abstract
BACKGROUND & AIMS Sustained activation of the cytosolic calcium concentration induces injury to pancreatic acinar cells and necrosis. The calcium release-activated calcium modulator ORAI1 is the most abundant Ca(2+) entry channel in pancreatic acinar cells; it sustains calcium overload in mice exposed to toxins that induce pancreatitis. We investigated the roles of ORAI1 in pancreatic acinar cell injury and the development of acute pancreatitis in mice. METHODS Mouse and human acinar cells, as well as HEK 293 cells transfected to express human ORAI1 with human stromal interaction molecule 1, were hyperstimulated or incubated with human bile acid, thapsigargin, or cyclopiazonic acid to induce calcium entry. GSK-7975A or CM_128 were added to some cells, which were analyzed by confocal and video microscopy and patch clamp recordings. Acute pancreatitis was induced in C57BL/6J mice by ductal injection of taurolithocholic acid 3-sulfate or intravenous' administration of cerulein or ethanol and palmitoleic acid. Some mice then were given GSK-7975A or CM_128, which inhibit ORAI1, at different time points to assess local and systemic effects. RESULTS GSK-7975A and CM_128 each separately inhibited toxin-induced activation of ORAI1 and/or activation of Ca(2+) currents after Ca(2+) release, in a concentration-dependent manner, in mouse and human pancreatic acinar cells (inhibition >90% of the levels observed in control cells). The ORAI1 inhibitors also prevented activation of the necrotic cell death pathway in mouse and human pancreatic acinar cells. GSK-7975A and CM_128 each inhibited all local and systemic features of acute pancreatitis in all 3 models, in dose- and time-dependent manners. The agents were significantly more effective, in a range of parameters, when given at 1 vs 6 hours after induction of pancreatitis. CONCLUSIONS Cytosolic calcium overload, mediated via ORAI1, contributes to the pathogenesis of acute pancreatitis. ORAI1 inhibitors might be developed for the treatment of patients with pancreatitis.
Collapse
Affiliation(s)
- Li Wen
- Pancreas Biomedical Research Unit, National Institute for Health Research Liverpool, Royal Liverpool University Hospital, Liverpool, United Kingdom; Department of Integrated Traditional and Western Medicine, Sichuan Provincial Pancreatitis Centre, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Svetlana Voronina
- Department of Molecular and Cellular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Muhammad A Javed
- Pancreas Biomedical Research Unit, National Institute for Health Research Liverpool, Royal Liverpool University Hospital, Liverpool, United Kingdom; Department of Molecular and Cellular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Muhammad Awais
- Pancreas Biomedical Research Unit, National Institute for Health Research Liverpool, Royal Liverpool University Hospital, Liverpool, United Kingdom
| | - Peter Szatmary
- Pancreas Biomedical Research Unit, National Institute for Health Research Liverpool, Royal Liverpool University Hospital, Liverpool, United Kingdom; Department of Molecular and Cellular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Diane Latawiec
- Pancreas Biomedical Research Unit, National Institute for Health Research Liverpool, Royal Liverpool University Hospital, Liverpool, United Kingdom
| | - Michael Chvanov
- Department of Molecular and Cellular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - David Collier
- Department of Molecular and Cellular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Wei Huang
- Pancreas Biomedical Research Unit, National Institute for Health Research Liverpool, Royal Liverpool University Hospital, Liverpool, United Kingdom
| | - John Barrett
- Respiratory Therapy Area Unit, Medicines Research Centre, GlaxoSmithKline, Stevenage, United Kingdom
| | - Malcolm Begg
- Respiratory Therapy Area Unit, Medicines Research Centre, GlaxoSmithKline, Stevenage, United Kingdom
| | | | | | | | | | | | | | | | | | - Alexei V Tepikin
- Department of Molecular and Cellular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - David N Criddle
- Department of Molecular and Cellular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Robert Sutton
- Pancreas Biomedical Research Unit, National Institute for Health Research Liverpool, Royal Liverpool University Hospital, Liverpool, United Kingdom.
| |
Collapse
|
279
|
Abstract
The endoplasmic reticulum (ER) supports many cellular processes and performs diverse functions, including protein synthesis, translocation across the membrane, integration into the membrane, folding, and posttranslational modifications including N-linked glycosylation; and regulation of Ca2+ homeostasis. In mammalian systems, the majority of proteins synthesized by the rough ER have N-linked glycans critical for protein maturation. The N-linked glycan is used as a quality control signal in the secretory protein pathway. A series of chaperones, folding enzymes, glucosidases, and carbohydrate transferases support glycoprotein synthesis and processing. Perturbation of ER-associated functions such as disturbed ER glycoprotein quality control, protein glycosylation and protein folding results in activation of an ER stress coping response. Collectively this ER stress coping response is termed the unfolded protein response (UPR), and occurs through the activation of complex cytoplasmic and nuclear signaling pathways. Cellular and ER homeostasis depends on balanced activity of the ER protein folding, quality control, and degradation pathways; as well as management of the ER stress coping response.
Collapse
|
280
|
Ma H, Butler MR, Thapa A, Belcher J, Yang F, Baehr W, Biel M, Michalakis S, Ding XQ. cGMP/Protein Kinase G Signaling Suppresses Inositol 1,4,5-Trisphosphate Receptor Phosphorylation and Promotes Endoplasmic Reticulum Stress in Photoreceptors of Cyclic Nucleotide-gated Channel-deficient Mice. J Biol Chem 2015; 290:20880-20892. [PMID: 26124274 DOI: 10.1074/jbc.m115.641159] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Indexed: 11/06/2022] Open
Abstract
Photoreceptor cyclic nucleotide-gated (CNG) channels play a pivotal role in phototransduction. Mutations in the cone CNG channel subunits CNGA3 and CNGB3 are associated with achromatopsia and cone dystrophies. We have shown endoplasmic reticulum (ER) stress-associated apoptotic cone death and increased phosphorylation of the ER Ca(2+) channel inositol 1,4,5-trisphosphate receptor 1 (IP3R1) in CNG channel-deficient mice. We also presented a remarkable elevation of cGMP and an increased activity of the cGMP-dependent protein kinase (protein kinase G, PKG) in CNG channel deficiency. This work investigated whether cGMP/PKG signaling regulates ER stress and IP3R1 phosphorylation in CNG channel-deficient cones. Treatment with PKG inhibitor and deletion of guanylate cyclase-1 (GC1), the enzyme producing cGMP in cones, were used to suppress cGMP/PKG signaling in cone-dominant Cnga3(-/-)/Nrl(-/-) mice. We found that treatment with PKG inhibitor or deletion of GC1 effectively reduced apoptotic cone death, increased expression levels of cone proteins, and decreased activation of Müller glial cells. Furthermore, we observed significantly increased phosphorylation of IP3R1 and reduced ER stress. Our findings demonstrate a role of cGMP/PKG signaling in ER stress and ER Ca(2+) channel regulation and provide insights into the mechanism of cone degeneration in CNG channel deficiency.
Collapse
Affiliation(s)
- Hongwei Ma
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Michael R Butler
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Arjun Thapa
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Josh Belcher
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Fan Yang
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Wolfgang Baehr
- John A. Moran Eye Center, University of Utah, Salt Lake City, Utah 84132
| | - Martin Biel
- Center for Integrated Protein Science Munich and Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Stylianos Michalakis
- Center for Integrated Protein Science Munich and Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Xi-Qin Ding
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104.
| |
Collapse
|
281
|
The Emerging Roles of Viroporins in ER Stress Response and Autophagy Induction during Virus Infection. Viruses 2015; 7:2834-57. [PMID: 26053926 PMCID: PMC4488716 DOI: 10.3390/v7062749] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 05/27/2015] [Accepted: 05/29/2015] [Indexed: 01/14/2023] Open
Abstract
Viroporins are small hydrophobic viral proteins that oligomerize to form aqueous pores on cellular membranes. Studies in recent years have demonstrated that viroporins serve important functions during virus replication and contribute to viral pathogenicity. A number of viroporins have also been shown to localize to the endoplasmic reticulum (ER) and/or its associated membranous organelles. In fact, replication of most RNA viruses is closely linked to the ER, and has been found to cause ER stress in the infected cells. On the other hand, autophagy is an evolutionarily conserved "self-eating" mechanism that is also observed in cells infected with RNA viruses. Both ER stress and autophagy are also known to modulate a wide variety of signaling pathways including pro-inflammatory and innate immune response, thereby constituting a major aspect of host-virus interactions. In this review, the potential involvement of viroporins in virus-induced ER stress and autophagy will be discussed.
Collapse
|
282
|
Knockdown of ERp44 leads to apoptosis via activation of ER stress in HeLa cells. Biochem Biophys Res Commun 2015; 463:606-11. [PMID: 26043695 DOI: 10.1016/j.bbrc.2015.05.106] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Accepted: 05/29/2015] [Indexed: 01/20/2023]
Abstract
ERp44, an endoplasmic reticulum (ER) resident protein, regulates intracellular Ca(2+) release and involves in the maturation of many proteins in mammalian cells. In this study, we investigated the effects and mechanism of ERp44 on cell apoptosis by using ERp44 knockdown stable HeLa cell lines. We found that ERp44 knockdown resulted in increases in cell apoptosis rate more than one fold higher than that of control; using serum starvation, caspase-3 protein level was significantly up-regulated in ERp44 knockdown cells compared to the control cells. Furthermore, we demonstrated that in response to serum starvation, the protein levels of CHOP and GRP78 were also largely raised in ERp44 knockdown cells. Moreover, caspase-12 was activated, which suggested cell apoptosis was induced by ER stress. Taken together, our results indicate that knockdown of ERp44 leads to cell apoptosis through the activation of ER stress.
Collapse
|
283
|
Kato H, Nishitoh H. Stress responses from the endoplasmic reticulum in cancer. Front Oncol 2015; 5:93. [PMID: 25941664 PMCID: PMC4403295 DOI: 10.3389/fonc.2015.00093] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 03/31/2015] [Indexed: 12/21/2022] Open
Abstract
The endoplasmic reticulum (ER) is a dynamic organelle that is essential for multiple cellular functions. During cellular stress conditions, including nutrient deprivation and dysregulation of protein synthesis, unfolded/misfolded proteins accumulate in the ER lumen, resulting in activation of the unfolded protein response (UPR). The UPR also contributes to the regulation of various intracellular signaling pathways such as calcium signaling and lipid signaling. More recently, the mitochondria-associated ER membrane (MAM), which is a site of close contact between the ER and mitochondria, has been shown to function as a platform for various intracellular stress responses including apoptotic signaling, inflammatory signaling, the autophagic response, and the UPR. Interestingly, in cancer, these signaling pathways from the ER are often dysregulated, contributing to cancer cell metabolism. Thus, the signaling pathway from the ER may be a novel therapeutic target for various cancers. In this review, we discuss recent research on the roles of stress responses from the ER, including the MAM.
Collapse
Affiliation(s)
- Hironori Kato
- Laboratory of Biochemistry and Molecular Biology, Department of Medical Sciences, University of Miyazaki , Miyazaki , Japan
| | - Hideki Nishitoh
- Laboratory of Biochemistry and Molecular Biology, Department of Medical Sciences, University of Miyazaki , Miyazaki , Japan
| |
Collapse
|
284
|
He WY, Chen GJ, Lai X, Wu F, Tang CS, Zhang AH. Expression levels of urotensin II are associated with endoplasmic reticulum stress in patients with severe preeclampsia. J Hum Hypertens 2015; 30:129-35. [DOI: 10.1038/jhh.2015.28] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 02/20/2015] [Accepted: 02/26/2015] [Indexed: 02/06/2023]
|
285
|
Chen S, Zhang Z, Wu Y, Shi Q, Yan H, Mei N, Tolleson WH, Guo L. Endoplasmic Reticulum Stress and Store-Operated Calcium Entry Contribute to Usnic Acid-Induced Toxicity in Hepatic Cells. Toxicol Sci 2015; 146:116-26. [PMID: 25870318 DOI: 10.1093/toxsci/kfv075] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The use of usnic acid as a weight loss agent is a safety concern due to reports of acute liver failure in humans. Previously we demonstrated that usnic acid induces apoptosis and cytotoxicity in hepatic HepG2 cells. We also demonstrated that usnic acid induces autophagy as a survival mechanism against its cytotoxicity. In this study, we investigated and characterized further molecular mechanisms underlying the toxicity of usnic acid in HepG2 cells. We found that usnic acid causes endoplasmic reticulum (ER) stress demonstrated by the increased expression of typical ER stress markers, including CHOP, ATF-4, p-eIF2α, and spliced XBP1. Usnic acid inhibited the secretion of Gaussia luciferase measured by an ER stress reporter assay. An ER stress inhibitor 4-phenylbutyrate attenuated usnic acid-induced apoptosis. Moreover, usnic acid significantly increased the cytosolic free Ca(2+) concentration. Usnic acid increased the expression of calcium release-activated calcium channel protein 1 (CRAM1 or ORAI1) and stromal interaction molecule 1, two key components of store-operated calcium entry (SOCE), which is the major Ca(2+) influx pathway in non-excitable cells, this finding was also confirmed in primary rat hepatocytes. Furthermore, knockdown of ORAI1 prevented ER stress and ATP depletion in response to usnic acid. In contrast, overexpression of ORAI1 increased ER stress and ATP depletion caused by usnic acid. Taken together, our results suggest that usnic acid disturbs calcium homeostasis, induces ER stress, and that usnic acid-induced cellular damage occurs at least partially via activation of the Ca(2+) channel of SOCE.
Collapse
Affiliation(s)
- Si Chen
- *Division of Biochemical Toxicology, Division of Genetic and Molecular Toxicology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079, Tianjin Medical University General Hospital, Tianjin 300052, China and Division of Systems Biology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079
| | - Zhuhong Zhang
- *Division of Biochemical Toxicology, Division of Genetic and Molecular Toxicology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079, Tianjin Medical University General Hospital, Tianjin 300052, China and Division of Systems Biology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079 *Division of Biochemical Toxicology, Division of Genetic and Molecular Toxicology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079, Tianjin Medical University General Hospital, Tianjin 300052, China and Division of Systems Biology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079
| | - Yuanfeng Wu
- *Division of Biochemical Toxicology, Division of Genetic and Molecular Toxicology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079, Tianjin Medical University General Hospital, Tianjin 300052, China and Division of Systems Biology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079
| | - Qiang Shi
- *Division of Biochemical Toxicology, Division of Genetic and Molecular Toxicology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079, Tianjin Medical University General Hospital, Tianjin 300052, China and Division of Systems Biology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079
| | - Hua Yan
- *Division of Biochemical Toxicology, Division of Genetic and Molecular Toxicology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079, Tianjin Medical University General Hospital, Tianjin 300052, China and Division of Systems Biology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079
| | - Nan Mei
- *Division of Biochemical Toxicology, Division of Genetic and Molecular Toxicology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079, Tianjin Medical University General Hospital, Tianjin 300052, China and Division of Systems Biology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079
| | - William H Tolleson
- *Division of Biochemical Toxicology, Division of Genetic and Molecular Toxicology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079, Tianjin Medical University General Hospital, Tianjin 300052, China and Division of Systems Biology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079
| | - Lei Guo
- *Division of Biochemical Toxicology, Division of Genetic and Molecular Toxicology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079, Tianjin Medical University General Hospital, Tianjin 300052, China and Division of Systems Biology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079
| |
Collapse
|
286
|
Nakanishi K, Kakiguchi K, Yonemura S, Nakano A, Morishima N. Transient Ca2+ depletion from the endoplasmic reticulum is critical for skeletal myoblast differentiation. FASEB J 2015; 29:2137-49. [PMID: 25678623 DOI: 10.1096/fj.14-261529] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 01/13/2015] [Indexed: 01/29/2023]
Abstract
Endoplasmic reticulum (ER) stress is a cellular condition in which unfolded proteins accumulate in the ER because of various but specific causes. Physiologic ER stress occurs transiently during myoblast differentiation, and although its cause remains unknown, it plays a critical role in myofiber formation. To examine the mechanism underlying ER stress, we monitored ER morphology during differentiation of murine myoblasts. Novel ER-derived structures transiently appeared prior to myoblast fusion both in vitro and in vivo. Electron microscopy studies revealed that these structures consisted of pseudoconcentric ER cisternae with narrow lumens. Similar structures specifically formed by pharmacologically induced ER Ca(2+) depletion, and inhibition of ER Ca(2+) efflux channels in differentiating myoblasts considerably suppressed ER-specific deformation and ER stress signaling. Thus, we named the novel structures stress-activated response to Ca(2+) depletion (SARC) bodies. Prior to SARC body formation, stromal interaction molecule 1 (STIM1), an ER Ca(2+) sensor protein, formed ER Ca(2+) depletion-specific clusters. Furthermore, myoblast differentiation manifested by myoblast fusion did not proceed under the same conditions as inhibition of ER Ca(2+) depletion. Altogether, these observations suggest that ER Ca(2+) depletion is a prerequisite for myoblast fusion, causing both physiologic ER stress signaling and SARC body formation.
Collapse
Affiliation(s)
- Keiko Nakanishi
- *Molecular Membrane Biology Laboratory, RIKEN Advanced Science Institute, Wako, Saitama, Japan; Electron Microscope Laboratory, RIKEN Center for Developmental Biology, Kobe, Hyogo, Japan; and Graduate School of Science, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Kisa Kakiguchi
- *Molecular Membrane Biology Laboratory, RIKEN Advanced Science Institute, Wako, Saitama, Japan; Electron Microscope Laboratory, RIKEN Center for Developmental Biology, Kobe, Hyogo, Japan; and Graduate School of Science, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Shigenobu Yonemura
- *Molecular Membrane Biology Laboratory, RIKEN Advanced Science Institute, Wako, Saitama, Japan; Electron Microscope Laboratory, RIKEN Center for Developmental Biology, Kobe, Hyogo, Japan; and Graduate School of Science, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Akihiko Nakano
- *Molecular Membrane Biology Laboratory, RIKEN Advanced Science Institute, Wako, Saitama, Japan; Electron Microscope Laboratory, RIKEN Center for Developmental Biology, Kobe, Hyogo, Japan; and Graduate School of Science, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Nobuhiro Morishima
- *Molecular Membrane Biology Laboratory, RIKEN Advanced Science Institute, Wako, Saitama, Japan; Electron Microscope Laboratory, RIKEN Center for Developmental Biology, Kobe, Hyogo, Japan; and Graduate School of Science, The University of Tokyo, Bunkyo, Tokyo, Japan
| |
Collapse
|
287
|
Zheng D, Wang G, Li S, Fan GC, Peng T. Calpain-1 induces endoplasmic reticulum stress in promoting cardiomyocyte apoptosis following hypoxia/reoxygenation. Biochim Biophys Acta Mol Basis Dis 2015; 1852:882-92. [PMID: 25660447 DOI: 10.1016/j.bbadis.2015.01.019] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 01/09/2015] [Accepted: 01/31/2015] [Indexed: 12/24/2022]
Abstract
Both calpain activation and endoplasmic reticulum (ER) stress are implicated in ischemic heart injury. However, the role of calpain in ER stress remains largely elusive. This study investigated whether calpain activation causes ER stress, thereby mediating cardiomyocyte apoptosis in an in vitro model of hypoxia/re-oxygenation (H/R). In neonatal mouse cardiomyocytes and rat cardiomyocyte-like H9c2 cells, up-regulation of calpain-1 sufficiently induced ER stress, c-Jun N-terminal protein kinase1/2 (JNK1/2) activation and apoptosis. Inhibition of ER stress or JNK1/2 prevented apoptosis induced by calpain-1. In an in vitro model of H/R-induced injury in cardiomyocytes, H/R was induced by a 24-hour hypoxia followed by a 24-hour re-oxygenation. H/R activated calpain-1, induced ER stress and JNK1/2 activation, and triggered apoptosis. Inhibition of calpain and ER stress blocked JNK1/2 activation and prevented H/R-induced apoptosis. Furthermore, blockade of JNK1/2 signaling inhibited apoptosis following H/R. The role of calpain in ER stress was also demonstrated in an in vivo model of ischemia/reperfusion using transgenic mice over-expressing calpastatin. In summary, calpain-1 induces ER stress and JNK1/2 activation, thereby mediating apoptosis in cardiomyocytes. Accordingly, inhibition of calpain prevents ER stress, JNK1/2 activation and apoptosis in H/R-induced cardiomyocytes. Thus, ER stress/JNK1/2 activation may represent an important mechanism linking calpain-1 to ischemic injury.
Collapse
Affiliation(s)
- Dong Zheng
- Institutes of Biology and Medical Science, Soochow University, Suzhou 215123, China; Critical Illness Research, Lawson Health Research Institute, Canada; Department of Medicine, University of Western Ontario, London, Ontario N6A 4G5, Canada; Institute of Cardiovascular Science, Soochow University, Suzhou 215008, China
| | - Grace Wang
- Department of Pathology, University of Western Ontario, London, Ontario N6A 4G5, Canada
| | - Shuai Li
- Critical Illness Research, Lawson Health Research Institute, Canada; Department of Medicine, University of Western Ontario, London, Ontario N6A 4G5, Canada; Department of Pathology, University of Western Ontario, London, Ontario N6A 4G5, Canada
| | - Guo-Chang Fan
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati 45267, OH, USA
| | - Tianqing Peng
- Institutes of Biology and Medical Science, Soochow University, Suzhou 215123, China; Critical Illness Research, Lawson Health Research Institute, Canada; Department of Medicine, University of Western Ontario, London, Ontario N6A 4G5, Canada; Department of Pathology, University of Western Ontario, London, Ontario N6A 4G5, Canada; Institute of Cardiovascular Science, Soochow University, Suzhou 215008, China.
| |
Collapse
|
288
|
Zhang S, Li C, Gao J, Qiu X, Cui Z. [Application of the Ca2+ indicator fluo-3 and fluo-4 in the process of H2O2 induced apoptosis of A549 cell]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2015; 17:197-202. [PMID: 24667255 PMCID: PMC6019359 DOI: 10.3779/j.issn.1009-3419.2014.03.03] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND OBJECTIVE Lung cancer is a common malignant tumor all over the world, and Ca(2+) is a critical regulator for apoptosis of cancer cells. The monitoring of cytoplastic Ca(2+) level in real-time will contribute to further investigate the molecular mechanisms of apoptosis mediated by Ca2+ in lung cancer cells. To evaluate the Ca(2+) indicator fluo-3 and fluo-4 in the process of H2O2 induced the apoptosis of lung adenocarcinoma A549 cells. The cytoplastic Ca(2+) concentration ([Ca(2+)]i) was determined in real-time, and the correlations between [Ca(2+)]i and cell apoptosis were investigated. The differences in fluorescence intensity and measured value were compared between the two Ca(2+) indicators. METHODS Cells were loaded with the Ca(2+) indicator fluo-3 or fluo-4 for 1 h, and then stimulated with 50 mM H2O2. Laser scanning confocal microscope was applied to perform real-time monitoring on the variation of [Ca(2+)]i in selected cells. DAPI staining was used to observe apoptosis in H2O2 treated cells. RESULTS Our results showed that the fluorescence intensity of fluo-4 was stronger than that of fluo-3 in the same condition of dye concentration, loading time and image acquisition parameters before or after H2O2 stimulation. The cytoplastic [Ca(2+)]i was rapidly elevated in H2O2 stimulated A549 cells. The range of [Ca(2+)]i in selected cells loaded with fluo-3 was 112.2 nM-1,069.6 nM, and that in selected cells loaded with fluo-4 was 7.6 nM-505.4 nM. Moreover, the apoptotic rate was significantly increased in H2O2 treated cells, compared with untreated ones (P<0.01). CONCLUSION In summary, H2O2 promoted Ca(2+) release in A549 cells, and induced cell apoptosis. Ca(2+) indicator fluo-4 was probably more applicable to measure [Ca(2+)]i in cells with less content of Ca(2+).
Collapse
Affiliation(s)
- Siyang Zhang
- Center of Laboratory Technology and Experimental Medicine, China Medical University,
Shenyang 110001, China
| | - Chunyan Li
- Center of Laboratory Technology and Experimental Medicine, China Medical University,
Shenyang 110001, China
| | - Jian Gao
- Center of Laboratory Technology and Experimental Medicine, China Medical University,
Shenyang 110001, China
| | - Xueshan Qiu
- Department of Pathology, China Medical University,
Shenyang 110001, China
| | - Zeshi Cui
- Center of Laboratory Technology and Experimental Medicine, China Medical University,
Shenyang 110001, China
| |
Collapse
|
289
|
Gani AR, Uppala JK, Ramaiah KVA. Tauroursodeoxycholic acid prevents stress induced aggregation of proteins in vitro and promotes PERK activation in HepG2 cells. Arch Biochem Biophys 2015; 568:8-15. [PMID: 25579883 DOI: 10.1016/j.abb.2014.12.031] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 12/27/2014] [Accepted: 12/30/2014] [Indexed: 12/15/2022]
Abstract
Tauroursodeoxycholic acid (TUDCA) a bile salt and chemical chaperone reduces stress-induced aggregation of proteins; activates PERK [PKR (RNA-dependent protein kinase)-like ER (endoplasmic reticulum) kinase] or EIF2AK3, one of the hall marks of ER stress induced unfolded protein response (UPR) in human hepatoblastoma HepG2 cells; prevents heat and dithiothreitol (DTT) induced aggregation of BSA (bovine serum albumin), and reduces ANS (1-anilino-naphthalene-8-sulfonate) bound BSA fluorescence in vitro. TUDCA inactivates heat treated, but not the native EcoR1 enzyme, and reduces heat-induced aggregation and activity of COX-1 (cyclooxygenase enzyme-1) in vitro. These findings suggest that TUDCA binds to the hydrophobic regions of proteins and prevents their subsequent aggregation. This may stabilize unfolded proteins that can mount UPR or facilitate their degradation through cellular degradation pathways.
Collapse
Affiliation(s)
- Amina R Gani
- Department of Biochemistry, University of Hyderabad, Hyderabad 500046, India
| | | | - Kolluru V A Ramaiah
- Department of Biochemistry, University of Hyderabad, Hyderabad 500046, India.
| |
Collapse
|
290
|
Ryan AJ, Larson-Casey JL, He C, Murthy S, Carter AB. Asbestos-induced disruption of calcium homeostasis induces endoplasmic reticulum stress in macrophages. J Biol Chem 2014; 289:33391-403. [PMID: 25324550 PMCID: PMC4246095 DOI: 10.1074/jbc.m114.579870] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 10/10/2014] [Indexed: 01/05/2023] Open
Abstract
Although the mechanisms for fibrosis development remain largely unknown, recent evidence indicates that endoplasmic reticulum (ER) stress and activation of the unfolded protein response (UPR) may act as an important fibrotic stimulus in diseased lungs. ER stress is observed in lungs of patients with idiopathic pulmonary fibrosis. In this study we evaluated if ER stress and the UPR was present in macrophages exposed to chrysotile asbestos and if ER stress in macrophages was associated with asbestos-induced pulmonary fibrosis. Macrophages exposed to chrysotile had elevated transcript levels of several ER stress genes. Macrophages loaded with the Ca(2+)-sensitive dye Fura2-AM showed that cytosolic Ca(2+) increased significantly within minutes after chrysotile exposure and remained elevated for a prolonged time. Chrysotile-induced increases in cytosolic Ca(2+) were partially inhibited by either anisomycin, an inhibitor of passive Ca(2+) leak from the ER, or 1,2-bis(2-aminophenoxyl)ethane-N,N,N',N'-tetraacetic acid (BAPTA-AM), an intracellular Ca(2+) chelator known to deplete ER Ca(2+) stores. Anisomycin inhibited X-box-binding protein 1 (XBP1) mRNA splicing and reduced immunoglobulin-binding protein (BiP) levels, whereas BAPTA-AM increased XBP1 splicing and BiP expression, suggesting that ER calcium depletion may be one factor contributing to ER stress in cells exposed to chrysotile. To evaluate ER stress in vivo, asbestos-exposed mice showed fibrosis development, and alveolar macrophages from fibrotic mice showed increased expression of BiP. Bronchoalveolar macrophages from asbestosis patients showed increased expression of several ER stress genes compared with normal subjects. These findings suggest that alveolar macrophages undergo ER stress, which is associated with fibrosis development.
Collapse
Affiliation(s)
| | - Jennifer L Larson-Casey
- Radiation Oncology and Program in Free Radical and Radiation Biology, Carver College of Medicine
| | - Chao He
- Radiation Oncology and Program in Free Radical and Radiation Biology, Carver College of Medicine
| | | | - A Brent Carter
- From the Departments of Internal Medicine, Radiation Oncology and Program in Free Radical and Radiation Biology, Carver College of Medicine, Human Toxicology, College of Public Health, University of Iowa, and Iowa City Veterans Administration Center, Iowa City, Iowa 52242
| |
Collapse
|
291
|
Gilon P, Chae HY, Rutter GA, Ravier MA. Calcium signaling in pancreatic β-cells in health and in Type 2 diabetes. Cell Calcium 2014; 56:340-61. [DOI: 10.1016/j.ceca.2014.09.001] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 08/26/2014] [Accepted: 09/01/2014] [Indexed: 12/24/2022]
|
292
|
Ermakova YG, Bilan DS, Matlashov ME, Mishina NM, Markvicheva KN, Subach OM, Subach FV, Bogeski I, Hoth M, Enikolopov G, Belousov VV. Red fluorescent genetically encoded indicator for intracellular hydrogen peroxide. Nat Commun 2014; 5:5222. [PMID: 25330925 PMCID: PMC4553041 DOI: 10.1038/ncomms6222] [Citation(s) in RCA: 191] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 09/09/2014] [Indexed: 01/10/2023] Open
Abstract
Reactive oxygen species (ROS) are conserved regulators of numerous cellular functions, and overproduction of ROS is a hallmark of various pathological processes. Genetically encoded fluorescent probes are unique tools to study ROS production in living systems of different scale and complexity. However, the currently available recombinant redox sensors have green emission, which overlaps with the spectra of many other probes. Expanding the spectral range of recombinant in vivo ROS probes would enable multiparametric in vivo ROS detection. Here we present the first genetically encoded red fluorescent sensor for hydrogen peroxide detection, HyPerRed. The performance of this sensor is similar to its green analogues. We demonstrate the utility of the sensor by tracing low concentrations of H2O2 produced in the cytoplasm of cultured cells upon growth factor stimulation. Moreover, using HyPerRed we detect local and transient H2O2 production in the mitochondrial matrix upon inhibition of the endoplasmic reticulum Ca(2+) uptake.
Collapse
Affiliation(s)
- Yulia G Ermakova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia
| | - Dmitry S Bilan
- 1] Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia [2] NBIC, Moscow Institute of Physics and Technology, 123182 Moscow, Russia
| | - Mikhail E Matlashov
- 1] Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia [2] NBIC, Moscow Institute of Physics and Technology, 123182 Moscow, Russia
| | - Natalia M Mishina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia
| | | | - Oksana M Subach
- NBIC, Moscow Institute of Physics and Technology, 123182 Moscow, Russia
| | - Fedor V Subach
- NBIC, Moscow Institute of Physics and Technology, 123182 Moscow, Russia
| | - Ivan Bogeski
- Department of Biophysics, School of Medicine, Saarland University, Homburg 66421, Germany
| | - Markus Hoth
- Department of Biophysics, School of Medicine, Saarland University, Homburg 66421, Germany
| | - Grigori Enikolopov
- 1] NBIC, Moscow Institute of Physics and Technology, 123182 Moscow, Russia [2] Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Vsevolod V Belousov
- 1] Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia [2] NBIC, Moscow Institute of Physics and Technology, 123182 Moscow, Russia
| |
Collapse
|
293
|
Wu Y, Yang M, Fan J, Peng Y, Deng L, Ding Y, Yang R, Zhou J, Miao D, Fu Q. Deficiency of osteoblastic Arl6ip5 impaired osteoblast differentiation and enhanced osteoclastogenesis via disturbance of ER calcium homeostasis and induction of ER stress-mediated apoptosis. Cell Death Dis 2014; 5:e1464. [PMID: 25321471 PMCID: PMC4237252 DOI: 10.1038/cddis.2014.427] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 08/29/2014] [Accepted: 09/04/2014] [Indexed: 12/15/2022]
Abstract
ADP-ribosylation-like factor 6 interacting protein 5 (Arl6ip5), which belongs to the prenylated rab-acceptor-family, has an important role in exocytic protein trafficking, glutathione metabolism and involves in cancer progression. However, its expression pattern and functional role in bone are unknown. Here we demonstrate that Arl6ip5 knock-out mice (Arl6ip5 (Δ2/Δ2)) show marked decrease of bone mineral density, trabecular bone volume and trabecular thickness. Histomorphometric studies reveal that bone formation parameters are decreased but bone resorption parameters and mRNA level of osteoclast-specific markers are increased in Arl6ip5(Δ2/Δ2) mice. In osteoblast, we demonstrate that Arl6ip5 abundantly expresses in osteoblastic cells and is regulated by bone metabolism-related hormones and growth factors. In vitro analysis reveals that osteoblast proliferation and differentiation are impaired in Arl6ip5 knocked-down and deficient primary osteoblast. Arl6ip5 is also found to function as an ER calcium regulator and control calmodulin signaling for osteoblast proliferation. Moreover, Arl6ip5 insufficiency in osteoblast induces ER stress and enhances ER stress-mediated apoptosis. CCAAT/enhancer-binding protein homologous protein (Chop) is involved in the regulation of apoptosis and differentiation in Arl6ip5 knocked-down osteoblasts. For osteoclastogenesis, Arl6ip5 insufficiency in osteoclast precursors has no effect on osteoclast formation. However, knocked-down osteoblastic Arl6ip5 induces receptor activator of nuclear factor-κB ligand (RANKL) expression and enhances osteoclastogenesis. In addition, ER stress and Chop are involved in the RANKL expression in Arl6ip5 knocked-down osteoblasts. In conclusion, we demonstrate that Arl6ip5 is a novel regulator of bone formation in osteoblasts.
Collapse
Affiliation(s)
- Y Wu
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China
| | - M Yang
- Department of Radiotherapy, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - J Fan
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China
| | - Y Peng
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China
| | - L Deng
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China
| | - Y Ding
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China
| | - R Yang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China
| | - J Zhou
- Department of Molecular Cell Biology and Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - D Miao
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology, and Embryology, Nanjing, China
| | - Q Fu
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China
| |
Collapse
|
294
|
|
295
|
Tripathi R, Benz N, Culleton B, Trouvé P, Férec C. Biophysical characterisation of calumenin as a charged F508del-CFTR folding modulator. PLoS One 2014; 9:e104970. [PMID: 25120007 PMCID: PMC4132023 DOI: 10.1371/journal.pone.0104970] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 07/16/2014] [Indexed: 11/19/2022] Open
Abstract
The cystic fibrosis transmembrane regulator (CFTR) is a cyclic-AMP dependent chloride channel expressed at the apical surface of epithelial cells lining various organs such as the respiratory tract. Defective processing and functioning of this protein caused by mutations in the CFTR gene results in loss of ionic balance, defective mucus clearance, increased proliferation of biofilms and inflammation of human airways observed in cystic fibrosis (CF) patients. The process by which CFTR folds and matures under the influence of various chaperones in the secretory pathway remains incompletely understood. Recently, calumenin, a secretory protein, belonging to the CREC family of low affinity calcium binding proteins has been identified as a putative CFTR chaperone whose biophysical properties and functions remain uncharacterized. We compared hydropathy, instability, charge, unfoldability, disorder and aggregation propensity of calumenin and other CREC family members with CFTR associated chaperones and calcium binding proteins, wild-type and mutant CFTR proteins and intrinsically disordered proteins (IDPs). We observed that calumenin, along with other CREC proteins, was significantly more charged and less folded compared to CFTR associated chaperones. Moreover like IDPs, calumenin and other CREC proteins were found to be less hydrophobic and aggregation prone. Phylogenetic analysis revealed a close link between calumenin and other CREC proteins indicating how evolution might have shaped their similar biophysical properties. Experimentally, calumenin was observed to significantly reduce F508del-CFTR aggregation in a manner similar to AavLEA1, a well-characterized IDP. Fluorescence microscopy based imaging analysis also revealed altered trafficking of calumenin in bronchial cells expressing F508del-CFTR, indicating its direct role in the pathophysiology of CF. In conclusion, calumenin is characterized as a charged protein exhibiting close similarity with IDPs and is hypothesized to regulate F508del-CFTR folding by electrostatic effects. This work provides useful insights for designing optimized synthetic structural correctors of CFTR mutant proteins in the future.
Collapse
Affiliation(s)
- Rashmi Tripathi
- INSERM UMR1078, Brest, France
- Université de Bretagne Occidentale, Faculté de Medecine et des sciences de la santé, Brest, France
- * E-mail:
| | - Nathalie Benz
- INSERM UMR1078, Brest, France
- Association Gaétan Saleün, Brest, France
| | - Bridget Culleton
- Hôpital Morvan, Laboratoire de Génétique Moléculaire et d‘Histocompatibilité, Brest, France
| | | | - Claude Férec
- INSERM UMR1078, Brest, France
- Université de Bretagne Occidentale, Faculté de Medecine et des sciences de la santé, Brest, France
- Hôpital Morvan, Laboratoire de Génétique Moléculaire et d‘Histocompatibilité, Brest, France
- Etablissement Français du Sang-Bretagne, Brest, France
| |
Collapse
|
296
|
Li H, Chang G, Wang J, Wang L, Jin W, Lin Y, Yan Y, Wang R, Gao W, Ma L, Li Q, Pang T. Cariporide sensitizes leukemic cells to tumor necrosis factor related apoptosis-inducing ligand by up-regulation of death receptor 5 via endoplasmic reticulum stress-CCAAT/enhancer binding protein homologous protein dependent mechanism. Leuk Lymphoma 2014; 55:2135-40. [PMID: 24188478 DOI: 10.3109/10428194.2013.861064] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CCAAT/enhancer binding protein homologous protein (CHOP) expression increases when Na(+)-H(+) exchanger 1 (NHE1) is inhibited. Endoplasmic reticulum (ER) stress has been shown to trigger tumor cell death through CHOP. We therefore hypothesized that NHE1 activity correlates with ER stress and confers pharmaceutical potential to NHE1 inhibitor as an anti-tumor agent. The present study showed that treatment with the NHE1 inhibitor cariporide led to ER stress-induced up-regulation of the death receptor 5 (DR5) which is mediated by CHOP at the transcriptional level. We also determined that ER stress-induced Janus kinase (JNK) activation was responsible for the modulation of CHOP. Combining cariporide with tumor necrosis factor related apoptosis-inducing ligand (TRAIL) led to a significantly enhanced level of apoptosis that was abrogated by siRNA silencing of CHOP. This study provides a potential mechanistic rationale for the use of NHE1 inhibitor in combination with DR5 agonists to induce apoptosis in leukemia.
Collapse
Affiliation(s)
- Huawen Li
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College , Tianjin , China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
297
|
Feng X, Krogh KA, Wu CY, Lin YW, Tsai HC, Thayer SA, Wei LN. Receptor-interacting protein 140 attenuates endoplasmic reticulum stress in neurons and protects against cell death. Nat Commun 2014; 5:4487. [PMID: 25066731 PMCID: PMC4200015 DOI: 10.1038/ncomms5487] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 06/23/2014] [Indexed: 12/29/2022] Open
Abstract
Inositol 1, 4, 5-trisphosphate receptor (IP3R)-mediated Ca(2+) release from the endoplasmic reticulum (ER) triggers many physiological responses in neurons, and when uncontrolled can cause ER stress that contributes to neurological disease. Here we show that the unfolded protein response (UPR) in neurons induces rapid translocation of nuclear receptor-interacting protein 140 (RIP140) to the cytoplasm. In the cytoplasm, RIP140 localizes to the ER by binding to the IP3R. The carboxyl-terminal RD4 domain of RIP140 interacts with the carboxyl-terminal gate-keeping domain of the IP3R. This molecular interaction disrupts the IP3R's 'head-tail' interaction, thereby suppressing channel opening and attenuating IP3R-mediated Ca(2+) release. This contributes to a rapid suppression of the ER stress response and provides protection from apoptosis in both hippocampal neurons in vitro and in an animal model of ER stress. Thus, RIP140 translocation to the cytoplasm is an early response to ER stress and provides protection against neuronal death.
Collapse
Affiliation(s)
- Xudong Feng
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Kelly A. Krogh
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Cheng-Ying Wu
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Yi-Wei Lin
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Hong-Chieh Tsai
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
- Department of Neurosurgery, Chang-Gung Memorial Hospital and University, Tao-Yuan, Taiwan, R.O.C
| | - Stanley A. Thayer
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Li-Na Wei
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| |
Collapse
|
298
|
Banerjee C, Singh A, Das TK, Raman R, Shrivastava A, Mazumder S. Ameliorating ER-stress attenuates Aeromonas hydrophila-induced mitochondrial dysfunctioning and caspase mediated HKM apoptosis in Clarias batrachus. Sci Rep 2014; 4:5820. [PMID: 25059203 PMCID: PMC5376045 DOI: 10.1038/srep05820] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 06/16/2014] [Indexed: 02/07/2023] Open
Abstract
Endoplasmic reticulum (ER)-stress and unfolding protein response (UPR) has not been implied in Aeromonas hydrophila-pathogenicity. We report increased expression of the ER-stress markers: CHOP, BiP and phospho-eIF2α in A. hydrophila-infected headkidney macrophages (HKM) in Clarias batrachus. Pre-treatment with ER-stress inhibitor, 4-PBA alleviated ER-stress and HKM apoptosis suggesting ER-UPR critical for the process. The ER-Ca(2+) released via inositol-triphosphate and ryanodine receptors induced calpain-2 mediated superoxide ion generation and consequent NF-κB activation. Inhibiting NF-κB activation attenuated NO production suggesting the pro-apoptotic role of NF-κB on HKM pathology. Calpain-2 activated caspase-12 to intensify the apoptotic cascade through mitochondrial-membrane potential (ψm) dissipation and caspase-9 activation. Altered mitochondrial ultra-structure consequent to ER-Ca(2+) uptake via uniporters reduced ψm and released cytochrome C. Nitric oxide induced the cGMP/PKG-dependent activation of caspase-8 and truncated-Bid formation. Both the caspases converge onto caspase-3 to execute HKM apoptosis. These findings offer a possible molecular explanation for A. hydrophila pathogenicity.
Collapse
Affiliation(s)
- Chaitali Banerjee
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi 110 007, India
| | - Ambika Singh
- Gut Biology Laboratory, Department of Zoology, University of Delhi, Delhi 110 007, India
| | - Taposh Kumar Das
- Department of Anatomy, All India Institute of Medical Sciences, Delhi 110 029, India
| | - Rajagopal Raman
- Gut Biology Laboratory, Department of Zoology, University of Delhi, Delhi 110 007, India
| | - Anju Shrivastava
- Cell Signalling and Molecular Immunology Laboratory, Department of Zoology, University of Delhi, Delhi 110 007, India
| | - Shibnath Mazumder
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi 110 007, India
| |
Collapse
|
299
|
Henderson MJ, Wires ES, Trychta KA, Richie CT, Harvey BK. SERCaMP: a carboxy-terminal protein modification that enables monitoring of ER calcium homeostasis. Mol Biol Cell 2014; 25:2828-39. [PMID: 25031430 PMCID: PMC4161517 DOI: 10.1091/mbc.e14-06-1141] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Depletion of ER calcium can lead to cell death and is implicated in numerous diseases. Secreted ER calcium-monitoring proteins (SERCaMPs) are secreted in response to ER calcium depletion. SERCaMPs identified ER calcium depletion in primary neurons exposed to glutamate, hyperthermia, and coxibs and in rat liver after a single exposure to thapsigargin. Endoplasmic reticulum (ER) calcium homeostasis is disrupted in diverse pathologies, including neurodegeneration, cardiovascular diseases, and diabetes. Temporally defining calcium dysregulation during disease progression, however, has been challenging. Here we describe secreted ER calcium-monitoring proteins (SERCaMPs), which allow for longitudinal monitoring of ER calcium homeostasis. We identified a carboxy-terminal modification that is sufficient to confer release of a protein specifically in response to ER calcium depletion. A Gaussia luciferase (GLuc)–based SERCaMP provides a simple and sensitive method to monitor ER calcium homeostasis in vitro or in vivo by analyzing culture medium or blood. GLuc-SERCaMPs revealed ER calcium depletion in rat primary neurons exposed to various ER stressors. In vivo, ER calcium disruption in rat liver was monitored over several days by repeated sampling of blood. Our results suggest that SERCaMPs will have broad applications for the long-term monitoring of ER calcium homeostasis and the development of therapeutic approaches to counteract ER calcium dysregulation.
Collapse
Affiliation(s)
- Mark J Henderson
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224
| | - Emily S Wires
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224
| | - Kathleen A Trychta
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224
| | - Christopher T Richie
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224
| | - Brandon K Harvey
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224
| |
Collapse
|
300
|
Selenite cataracts: activation of endoplasmic reticulum stress and loss of Nrf2/Keap1-dependent stress protection. Biochim Biophys Acta Mol Basis Dis 2014; 1842:1794-805. [PMID: 24997453 DOI: 10.1016/j.bbadis.2014.06.028] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 06/19/2014] [Accepted: 06/24/2014] [Indexed: 12/21/2022]
Abstract
Cataract-induced by sodium selenite in suckling rats is one of the suitable animal models to study the basic mechanism of human cataract formation. The aim of this present investigation is to study the endoplasmic reticulum (ER) stress-mediated activation of unfolded protein response (UPR), overproduction of reactive oxygen species (ROS), and suppression of Nrf2/Keap1-dependent antioxidant protection through endoplasmic reticulum-associated degradation (ERAD) pathway and Keap1 promoter DNA demethylation in human lens epithelial cells (HLECs) treated with sodium selenite. Lenses enucleated from sodium selenite injected rats generated overproduction of ROS in lens epithelial cells and newly formed lens fiber cells resulting in massive lens epithelial cells death after 1-5days. All these lenses developed nuclear cataracts after 4-5days. Sodium selenite treated HLECs induced ER stress and activated the UPR leading to release of Ca(2+) from ER, ROS overproduction and finally HLECs death. Sodium selenite also activated the mRNA expressions of passive DNA demethylation pathway enzymes such as Dnmt1, Dnmt3a, and Dnmt3b, and active DNA demethylation pathway enzyme, Tet1 leading to DNA demethylation in the Keap1 promoter of HLECs. This demethylated Keap1 promoter results in overexpression of Keap1 mRNA and protein. Overexpression Keap1 protein suppresses the Nrf2 protein through ERAD leading to suppression of Nrf2/Keap1 dependent antioxidant protection in the HLECs treated with sodium selenite. As an outcome, the cellular redox status is altered towards lens oxidation and results in cataract formation.
Collapse
|