201
|
Martínez-Zamora A, Meseguer S, Esteve JM, Villarroya M, Aguado C, Enríquez JA, Knecht E, Armengod ME. Defective Expression of the Mitochondrial-tRNA Modifying Enzyme GTPBP3 Triggers AMPK-Mediated Adaptive Responses Involving Complex I Assembly Factors, Uncoupling Protein 2, and the Mitochondrial Pyruvate Carrier. PLoS One 2015; 10:e0144273. [PMID: 26642043 PMCID: PMC4671719 DOI: 10.1371/journal.pone.0144273] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 11/16/2015] [Indexed: 12/22/2022] Open
Abstract
GTPBP3 is an evolutionary conserved protein presumably involved in mitochondrial tRNA (mt-tRNA) modification. In humans, GTPBP3 mutations cause hypertrophic cardiomyopathy with lactic acidosis, and have been associated with a defect in mitochondrial translation, yet the pathomechanism remains unclear. Here we use a GTPBP3 stable-silencing model (shGTPBP3 cells) for a further characterization of the phenotype conferred by the GTPBP3 defect. We experimentally show for the first time that GTPBP3 depletion is associated with an mt-tRNA hypomodification status, as mt-tRNAs from shGTPBP3 cells were more sensitive to digestion by angiogenin than tRNAs from control cells. Despite the effect of stable silencing of GTPBP3 on global mitochondrial translation being rather mild, the steady-state levels and activity of Complex I, and cellular ATP levels were 50% of those found in the controls. Notably, the ATPase activity of Complex V increased by about 40% in GTPBP3 depleted cells suggesting that mitochondria consume ATP to maintain the membrane potential. Moreover, shGTPBP3 cells exhibited enhanced antioxidant capacity and a nearly 2-fold increase in the uncoupling protein UCP2 levels. Our data indicate that stable silencing of GTPBP3 triggers an AMPK-dependent retrograde signaling pathway that down-regulates the expression of the NDUFAF3 and NDUFAF4 Complex I assembly factors and the mitochondrial pyruvate carrier (MPC), while up-regulating the expression of UCP2. We also found that genes involved in glycolysis and oxidation of fatty acids are up-regulated. These data are compatible with a model in which high UCP2 levels, together with a reduction in pyruvate transport due to the down-regulation of MPC, promote a shift from pyruvate to fatty acid oxidation, and to an uncoupling of glycolysis and oxidative phosphorylation. These metabolic alterations, and the low ATP levels, may negatively affect heart function.
Collapse
Affiliation(s)
- Ana Martínez-Zamora
- Laboratory of RNA Modification and Mitochondrial Diseases, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Salvador Meseguer
- Laboratory of RNA Modification and Mitochondrial Diseases, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Juan M. Esteve
- Laboratory of Intracellular Protein Degradation and Rare Diseases, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Magda Villarroya
- Laboratory of RNA Modification and Mitochondrial Diseases, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Carmen Aguado
- Laboratory of Intracellular Protein Degradation and Rare Diseases, Centro de Investigación Príncipe Felipe, Valencia, Spain
- Centro de Investigación Biomédica En Red de Enfermedades Raras (CIBERER), node U721, Valencia, Spain
| | - J. Antonio Enríquez
- Departamento de Desarrollo y Reparación Cardiovascular, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Zaragoza, Spain
| | - Erwin Knecht
- Laboratory of Intracellular Protein Degradation and Rare Diseases, Centro de Investigación Príncipe Felipe, Valencia, Spain
- Centro de Investigación Biomédica En Red de Enfermedades Raras (CIBERER), node U721, Valencia, Spain
| | - M.-Eugenia Armengod
- Laboratory of RNA Modification and Mitochondrial Diseases, Centro de Investigación Príncipe Felipe, Valencia, Spain
- Centro de Investigación Biomédica En Red de Enfermedades Raras (CIBERER), node U721, Valencia, Spain
- * E-mail:
| |
Collapse
|
202
|
Yu F, Chen Z, Wang B, Jin Z, Hou Y, Ma S, Liu X. The role of lysosome in cell death regulation. Tumour Biol 2015; 37:1427-36. [DOI: 10.1007/s13277-015-4516-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 11/25/2015] [Indexed: 02/01/2023] Open
|
203
|
Zhang L, Wang K, Lei Y, Li Q, Nice EC, Huang C. Redox signaling: Potential arbitrator of autophagy and apoptosis in therapeutic response. Free Radic Biol Med 2015; 89:452-65. [PMID: 26454086 DOI: 10.1016/j.freeradbiomed.2015.08.030] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 08/27/2015] [Accepted: 08/31/2015] [Indexed: 02/05/2023]
Abstract
Redox signaling plays important roles in the regulation of cell death and survival in response to cancer therapy. Autophagy and apoptosis are discrete cellular processes mediated by distinct groups of regulatory and executioner molecules, and both are thought to be cellular responses to various stress conditions including oxidative stress, therefore controlling cell fate. Basic levels of reactive oxygen species (ROS) may function as signals to promote cell proliferation and survival, whereas increase of ROS can induce autophagy and apoptosis by damaging cellular components. Growing evidence in recent years argues for ROS that below detrimental levels acting as intracellular signal transducers that regulate autophagy and apoptosis. ROS-regulated autophagy and apoptosis can cross-talk with each other. However, how redox signaling determines different cell fates by regulating autophagy and apoptosis remains unclear. In this review, we will focus on understanding the delicate molecular mechanism by which autophagy and apoptosis are finely orchestrated by redox signaling and discuss how this understanding can be used to develop strategies for the treatment of cancer.
Collapse
Affiliation(s)
- Lu Zhang
- State Key Laboratory for Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, P.R. China; Department of Neurology, the Affiliated Hospital of Hainan Medical College, Haikou, 570102, P.R. China
| | - Kui Wang
- State Key Laboratory for Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, P.R. China
| | - Yunlong Lei
- Department of Biochemistry and Molecular Biology, and Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, P.R. China
| | - Qifu Li
- Department of Neurology, the Affiliated Hospital of Hainan Medical College, Haikou, 570102, P.R. China
| | - Edouard Collins Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Canhua Huang
- State Key Laboratory for Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, P.R. China.
| |
Collapse
|
204
|
HDAC6 Regulates the Chaperone-Mediated Autophagy to Prevent Oxidative Damage in Injured Neurons after Experimental Spinal Cord Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:7263736. [PMID: 26649145 PMCID: PMC4663006 DOI: 10.1155/2016/7263736] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Revised: 07/10/2015] [Accepted: 07/22/2015] [Indexed: 11/17/2022]
Abstract
Hypoxia-ischemia- (HI-) induced oxidative stress plays a role in secondary pathocellular processes of acute spinal cord injury (SCI) due to HI from many kinds of mechanical trauma. Increasing evidence suggests that the histone deacetylase-6 (HDAC6) plays an important role in cell homeostasis in both physiological and abnormal, stressful, pathological conditions. This paper found that inhibition of HDAC6 accelerated reactive oxygen species (ROS) generation and cell apoptosis in response to the HI. Deficiency of HDAC6 hindered the chaperone-mediated autophagy (CMA) activity to resistance of HI-induced oxidative stress. Furthermore, this study provided the experimental evidence for the potential role of HDAC6 in the regulation of CMA by affecting HSP90 acetylation. Therefore, HDAC6 plays an important role in the function of CMA pathway under the HI stress induced by SCI and it may be a potential therapeutic target in acute SCI model.
Collapse
|
205
|
Abstract
Maintaining cellular redox status to allow cell signalling to occur requires modulation of both the controlled production of oxidants and the thiol-reducing networks to allow specific regulatory post-translational modification of protein thiols. The oxidative stress hypothesis captured the concept that overproduction of oxidants can be proteotoxic, but failed to predict the recent finding that hyperactivation of the KEAP1-NRF2 system also leads to proteotoxicity. Furthermore, sustained activation of thiol redox networks by KEAP1-NRF2 induces a reductive stress, by decreasing the lifetime of necessary oxidative post-translational modifications required for normal metabolism or cell signalling. In this context, it is now becoming clear why antioxidants or hyperactivation of antioxidant pathways with electrophilic therapeutics can be deleterious. Furthermore, it suggests that the autophagy-lysosomal pathway is particularly important in protecting the cell against redox-stress-induced proteotoxicity, since it can degrade redox-damaged proteins without causing aberrant changes to the redox network needed for metabolism or signalling. In this context, it is important to understand: (i) how NRF2-mediated redox signalling, or (ii) the autophagy-mediated antioxidant/reductant pathways sense cellular damage in the context of cellular pathogenesis. Recent studies indicate that the modification of protein thiols plays an important role in the regulation of both the KEAP1-NRF2 and autophagy pathways. In the present review, we discuss evidence demonstrating that the KEAP1-NRF2 pathway and autophagy act in concert to combat the deleterious effects of proteotoxicity. These findings are discussed with a special emphasis on their impact on cardiovascular disease and neurodegeneration.
Collapse
|
206
|
Marutani E, Yamada M, Ida T, Tokuda K, Ikeda K, Kai S, Shirozu K, Hayashida K, Kosugi S, Hanaoka K, Kaneki M, Akaike T, Ichinose F. Thiosulfate Mediates Cytoprotective Effects of Hydrogen Sulfide Against Neuronal Ischemia. J Am Heart Assoc 2015; 4:e002125. [PMID: 26546573 PMCID: PMC4845224 DOI: 10.1161/jaha.115.002125] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 09/22/2015] [Indexed: 12/30/2022]
Abstract
BACKGROUND Hydrogen sulfide (H2S) exhibits protective effects in various disease models including cerebral ischemia-reperfusion (I/R) injury. Nonetheless, mechanisms and identity of molecules responsible for neuroprotective effects of H2S remain incompletely defined. In the current study, we observed that thiosulfate, an oxidation product of H2S, mediates protective effects of an H2S donor compound sodium sulfide (Na2S) against neuronal I/R injury. METHODS AND RESULTS We observed that thiosulfate in cell culture medium is not only required but also sufficient to mediate cytoprotective effects of Na2S against oxygen glucose deprivation and reoxygenation of human neuroblastoma cell line (SH-SY5Y) and murine primary cortical neurons. Systemic administration of sodium thiosulfate (STS) improved survival and neurological function of mice subjected to global cerebral I/R injury. Beneficial effects of STS, as well as Na2S, were associated with marked increase of thiosulfate, but not H2S, in plasma and brain tissues. These results suggest that thiosulfate is a circulating "carrier" molecule of beneficial effects of H2S. Protective effects of thiosulfate were associated with inhibition of caspase-3 activity by persulfidation at Cys163 in caspase-3. We discovered that an SLC13 family protein, sodium sulfate cotransporter 2 (SLC13A4, NaS-2), facilitates transport of thiosulfate, but not sulfide, across the cell membrane, regulating intracellular concentrations and thus mediating cytoprotective effects of Na2S and STS. CONCLUSIONS The protective effects of H2S are mediated by thiosulfate that is transported across cell membrane by NaS-2 and exerts antiapoptotic effects via persulfidation of caspase-3. Given the established safety track record, thiosulfate may be therapeutic against ischemic brain injury.
Collapse
Affiliation(s)
- Eizo Marutani
- Department of Anesthesia, Critical Care and Pain MedicineMassachusetts General Hospital and Harvard Medical SchoolBostonMA
| | - Marina Yamada
- Department of Anesthesia, Critical Care and Pain MedicineMassachusetts General Hospital and Harvard Medical SchoolBostonMA
- Shriners Hospitals for ChildrenBostonMA
| | - Tomoaki Ida
- Department of Environmental Health Sciences and Molecular ToxicologyTohoku University Graduate School of MedicineSendaiJapan
| | - Kentaro Tokuda
- Department of Anesthesia, Critical Care and Pain MedicineMassachusetts General Hospital and Harvard Medical SchoolBostonMA
| | - Kohei Ikeda
- Department of Anesthesia, Critical Care and Pain MedicineMassachusetts General Hospital and Harvard Medical SchoolBostonMA
| | - Shinichi Kai
- Department of Anesthesia, Critical Care and Pain MedicineMassachusetts General Hospital and Harvard Medical SchoolBostonMA
| | - Kazuhiro Shirozu
- Department of Anesthesia, Critical Care and Pain MedicineMassachusetts General Hospital and Harvard Medical SchoolBostonMA
| | - Kei Hayashida
- Department of Anesthesia, Critical Care and Pain MedicineMassachusetts General Hospital and Harvard Medical SchoolBostonMA
| | - Shizuko Kosugi
- Department of Anesthesia, Critical Care and Pain MedicineMassachusetts General Hospital and Harvard Medical SchoolBostonMA
| | - Kenjiro Hanaoka
- Graduate School of Pharmaceutical SciencesThe University of TokyoJapan
| | - Masao Kaneki
- Department of Anesthesia, Critical Care and Pain MedicineMassachusetts General Hospital and Harvard Medical SchoolBostonMA
- Shriners Hospitals for ChildrenBostonMA
| | - Takaaki Akaike
- Department of Environmental Health Sciences and Molecular ToxicologyTohoku University Graduate School of MedicineSendaiJapan
| | - Fumito Ichinose
- Department of Anesthesia, Critical Care and Pain MedicineMassachusetts General Hospital and Harvard Medical SchoolBostonMA
| |
Collapse
|
207
|
Moustapha A, Pérétout PA, Rainey NE, Sureau F, Geze M, Petit JM, Dewailly E, Slomianny C, Petit PX. Curcumin induces crosstalk between autophagy and apoptosis mediated by calcium release from the endoplasmic reticulum, lysosomal destabilization and mitochondrial events. Cell Death Discov 2015; 1:15017. [PMID: 27551451 PMCID: PMC4979459 DOI: 10.1038/cddiscovery.2015.17] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 06/23/2015] [Indexed: 12/22/2022] Open
Abstract
Curcumin, a major active component of turmeric (Curcuma longa, L.), has anticancer effects. In vitro studies suggest that curcumin inhibits cancer cell growth by activating apoptosis, but the mechanism underlying these effects is still unclear. Here, we investigated the mechanisms leading to apoptosis in curcumin-treated cells. Curcumin induced endoplasmic reticulum stress causing calcium release, with a destabilization of the mitochondrial compartment resulting in apoptosis. These events were also associated with lysosomal membrane permeabilization and of caspase-8 activation, mediated by cathepsins and calpains, leading to Bid cleavage. Truncated tBid disrupts mitochondrial homeostasis and enhance apoptosis. We followed the induction of autophagy, marked by the formation of autophagosomes, by staining with acridine orange in cells exposed curcumin. At this concentration, only the early events of apoptosis (initial mitochondrial destabilization with any other manifestations) were detectable. Western blotting demonstrated the conversion of LC3-I to LC3-II (light chain 3), a marker of active autophagosome formation. We also found that the production of reactive oxygen species and formation of autophagosomes following curcumin treatment was almost completely blocked by N-acetylcystein, the mitochondrial specific antioxidants MitoQ10 and SKQ1, the calcium chelators, EGTA-AM or BAPTA-AM, and the mitochondrial calcium uniporter inhibitor, ruthenium red. Curcumin-induced autophagy failed to rescue all cells and most cells underwent type II cell death following the initial autophagic processes. All together, these data imply a fail-secure mechanism regulated by autophagy in the action of curcumin, suggesting a therapeutic potential for curcumin. Offering a novel and effective strategy for the treatment of malignant cells.
Collapse
Affiliation(s)
- A Moustapha
- INSERM U1124 'Toxicologie, Pharmacologie et Signalisation Cellulaire', Université Paris-Descartes, Centre Universitaire des Saints-Pères , Paris, France
| | - P A Pérétout
- INSERM U1124 'Toxicologie, Pharmacologie et Signalisation Cellulaire', Université Paris-Descartes, Centre Universitaire des Saints-Pères , Paris, France
| | - N E Rainey
- INSERM U1124 'Toxicologie, Pharmacologie et Signalisation Cellulaire', Université Paris-Descartes, Centre Universitaire des Saints-Pères , Paris, France
| | - F Sureau
- Université Pierre et Marie Curie-Paris 6, Laboratoire Jean Perrin , Paris, France
| | - M Geze
- Muséum National d'Histoire Naturelles, CeMIM/USM 0504, 'Biologie Fonctionnelles des Protozoaires' 57 , Paris, France
| | - J-M Petit
- Muséum National d'Histoire Naturelles, UMR 7245 CNRS/MNHN 'Molécules de Communication et Adaptation des Micro-organismes' 57 , Paris, France
| | - E Dewailly
- Laboratoire de Physiologie cellulaire, INSERM U800, Université des Sciences et Techniques de Lille 1 , Villeneuve d'Ascq, France
| | - C Slomianny
- Laboratoire de Physiologie cellulaire, INSERM U800, Université des Sciences et Techniques de Lille 1 , Villeneuve d'Ascq, France
| | - P X Petit
- INSERM U1124 'Toxicologie, Pharmacologie et Signalisation Cellulaire', Université Paris-Descartes, Centre Universitaire des Saints-Pères , Paris, France
| |
Collapse
|
208
|
Wu J, Hafner C, Schramel JP, Kaun C, Krychtiuk KA, Wojta J, Boehme S, Ullrich R, Tretter EV, Markstaller K, Klein KU. Cyclic and constant hyperoxia cause inflammation, apoptosis and cell death in human umbilical vein endothelial cells. Acta Anaesthesiol Scand 2015; 60:492-501. [PMID: 26489399 DOI: 10.1111/aas.12646] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 09/02/2015] [Accepted: 09/09/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND Perioperative high-dose oxygen (O2 ) exposure can cause hyperoxia. While the effect of constant hyperoxia on the vascular endothelium has been investigated to some extent, the impact of cyclic hyperoxia largely remains unknown. We hypothesized that cyclic hyperoxia would induce more injury than constant hyperoxia to human umbilical vein endothelial cells (HUVECs). METHODS HUVECs were exposed to cyclic hyperoxia (5-95% O2 ) or constant hyperoxia (95% O2 ), normoxia (21% O2 ), and hypoxia (5% O2 ). Cell growth, viability (Annexin V/propidium iodide and 3-(4,5-dimethythiazol-2-yl)-2,5-diphenyl tetrazolium bromide, MTT) lactate dehydrogenase (LDH), release, cytokine (interleukin, IL and macrophage migration inhibitory factor, MIF) release, total antioxidant capacity (TAC), and superoxide dismutase activity (SOD) of cell lysate were assessed at baseline and 8, 24, and 72 h. A signal transduction pathway finder array for gene expression analysis was performed after 8 h. RESULTS Constant and cyclic hyperoxia-induced gradually detrimental effects on HUVECs. After 72 h, constant or cyclic hyperoxia exposure induced change in cytotoxic (LDH +12%, P = 0.026; apoptosis +121/61%, P < 0.01; alive cells -15%, P < 0.01; MTT -16/15%, P < 0.01), inflammatory (IL-6 +142/190%, P < 0.01; IL-8 +72/43%, P < 0.01; MIF +147/93%, P < 0.01), or redox-sensitive (SOD +278%, TAC-25% P < 0.01) markers. Gene expression analysis revealed that constant and cyclic hyperoxia exposure differently activates oxidative stress, nuclear factor kappa B, Notch, and peroxisome proliferator-activated receptor pathways. CONCLUSIONS Extreme hyperoxia exposure induces inflammation, apoptosis and cell death in HUVECs. Although our findings cannot be transferred to clinical settings, results suggest that hyperoxia exposure may cause vascular injury that could play a role in determining perioperative outcome.
Collapse
Affiliation(s)
- J. Wu
- Department of Anaesthesia; General Intensive Care and Pain Management; Medical University of Vienna; Vienna Austria
- Department of Anesthesiology; Union Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan China
| | - C. Hafner
- Department of Anaesthesia; General Intensive Care and Pain Management; Medical University of Vienna; Vienna Austria
| | - J. P. Schramel
- Unit of Anaesthesiology and Perioperative Intensive Care; University of Veterinary Medicine; Vienna Austria
| | - C. Kaun
- Department of Internal Medicine II; Medical University Vienna; Vienna Austria
- Core Facilities; Medical University of Vienna; Vienna Austria
| | - K. A. Krychtiuk
- Department of Internal Medicine II; Medical University Vienna; Vienna Austria
- Ludwig Boltzmann Cluster for Cardiovascular Research; Vienna Austria
| | - J. Wojta
- Department of Internal Medicine II; Medical University Vienna; Vienna Austria
- Core Facilities; Medical University of Vienna; Vienna Austria
- Ludwig Boltzmann Cluster for Cardiovascular Research; Vienna Austria
| | - S. Boehme
- Department of Anaesthesia; General Intensive Care and Pain Management; Medical University of Vienna; Vienna Austria
| | - R. Ullrich
- Department of Anaesthesia; General Intensive Care and Pain Management; Medical University of Vienna; Vienna Austria
| | - E. V. Tretter
- Department of Anaesthesia; General Intensive Care and Pain Management; Medical University of Vienna; Vienna Austria
| | - K. Markstaller
- Department of Anaesthesia; General Intensive Care and Pain Management; Medical University of Vienna; Vienna Austria
| | - K. U. Klein
- Department of Anaesthesia; General Intensive Care and Pain Management; Medical University of Vienna; Vienna Austria
| |
Collapse
|
209
|
Activity of BKM120 and BEZ235 against Lymphoma Cells. BIOMED RESEARCH INTERNATIONAL 2015; 2015:870918. [PMID: 26557706 PMCID: PMC4628710 DOI: 10.1155/2015/870918] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 07/27/2015] [Accepted: 08/19/2015] [Indexed: 12/23/2022]
Abstract
Non-Hodgkin lymphomas encompass a heterogeneous group of cancers, with 85–90% arising from B lymphocytes and the remainder deriving from T lymphocytes or NK lymphocytes. These tumors are molecularly and clinically heterogeneous, showing dramatically different responses and outcomes with standard therapies. Deregulated PI3K signaling is linked to oncogenesis and disease progression in hematologic malignancies and in a variety of solid tumors and apparently enhances resistance to antineoplastic therapy, resulting in a poor prognosis. Here, we have evaluated and compared the effects of the pan-PI3K inhibitor BKM120 and the dual PI3K/mTOR inhibitor BEZ235 on mantle, follicular, and T-cell lymphomas. Our results suggest that BKM120 and BEZ235 can effectively inhibit lymphoma cell proliferation by causing cell cycle arrest and can lead to cell death by inducing apoptosis and autophagy mediated by ROS accumulation. Despite great advances in lymphoma therapy after the introduction of monoclonal antibodies, many patients still die from disease progression. Therefore, novel treatment approaches are needed. BKM120 and BEZ235 alone and in combination are very effective against lymphoma cells in vitro. If further studies confirm their effectiveness in animal models, they may be promising candidates for development as new drugs.
Collapse
|
210
|
Huang C, Lu X, Tong L, Wang J, Zhang W, Jiang B, Yang R. Requirement for endogenous heat shock factor 1 in inducible nitric oxide synthase induction in murine microglia. J Neuroinflammation 2015; 12:189. [PMID: 26467650 PMCID: PMC4607096 DOI: 10.1186/s12974-015-0406-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 09/28/2015] [Indexed: 01/17/2023] Open
Abstract
Background Inducible nitric oxide synthase (iNOS) makes a great contribution to host defense and inflammation. In many settings, lipopolysaccharide (LPS) induces iNOS expression through activation of the inhibitor of κB-α (IκB-α)-nuclear factor-κB (NF-κB) cascade, whereas interferon-γ (IFN-γ) acts through Janus kinase (JAK)-signal transducer and activator of transcription 1 (STAT1) signals. Heat shock factor 1 (HSF1), a major regulator of heat shock protein transcription, has been shown to regulate the production of pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6), but it remains obscure whether and how HSF1 affects iNOS induction. Methods Western blot was used to measure the protein expression. The mRNA level was measured by real-time PCR. Silence of HSF1 was achieved by small interfering RNA. Nitric oxide (NO) content and NF-κB binding activity were assayed by commercial kits. Chromatin immunoprecipitation (ChIP) was used to measure the binding activity of NF-κB and STAT1 to iNOS promoters. Results HSF1 inhibition or knockdown prevented the LPS- and/or IFN-γ-stimulated iNOS protein expression in cultured microglia. HSF1 inhibition blocked iNOS mRNA transcription. These inhibitory effects of HSF1 inhibition on iNOS expression were confirmed in brain tissues from endotoxemic mice. Further analysis showed that HSF1 inhibition had no effect on IκB-α degradation and NF-κB or STAT1 phosphorylation in LPS/IFN-γ-stimulated cells. The nuclear transport of active NF-κB or STAT1 was also not affected by HSF1 inhibition, but HSF1 inhibition reduced the binding of NF-κB and STAT1 to their DNA elements. In addition, HSF1 inhibition reduced NF-κB and STAT1 bindings to iNOS promoter inside the LPS/IFN-γ-stimulated cells. Conclusions This preventing effect of HSF1 inhibition on iNOS mRNA transcription presents the necessary role of HSF1 in iNOS induction.
Collapse
Affiliation(s)
- Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China. .,Key Laboratory of Inflammation and Molecular Drug Targets of Jiangsu Province, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China.
| | - Xu Lu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China. .,Key Laboratory of Inflammation and Molecular Drug Targets of Jiangsu Province, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China.
| | - Lijuan Tong
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China. .,Key Laboratory of Inflammation and Molecular Drug Targets of Jiangsu Province, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China.
| | - Jili Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China. .,Key Laboratory of Inflammation and Molecular Drug Targets of Jiangsu Province, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China.
| | - Wei Zhang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China. .,Key Laboratory of Inflammation and Molecular Drug Targets of Jiangsu Province, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China.
| | - Bo Jiang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China. .,Key Laboratory of Inflammation and Molecular Drug Targets of Jiangsu Province, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China.
| | - Rongrong Yang
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Jiangsu Province, #20Xisi Road, Nantong, Jiangsu Province, 226001, China.
| |
Collapse
|
211
|
Görlach A, Dimova EY, Petry A, Martínez-Ruiz A, Hernansanz-Agustín P, Rolo AP, Palmeira CM, Kietzmann T. Reactive oxygen species, nutrition, hypoxia and diseases: Problems solved? Redox Biol 2015; 6:372-385. [PMID: 26339717 PMCID: PMC4565025 DOI: 10.1016/j.redox.2015.08.016] [Citation(s) in RCA: 256] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 08/21/2015] [Accepted: 08/25/2015] [Indexed: 02/06/2023] Open
Abstract
Within the last twenty years the view on reactive oxygen species (ROS) has changed; they are no longer only considered to be harmful but also necessary for cellular communication and homeostasis in different organisms ranging from bacteria to mammals. In the latter, ROS were shown to modulate diverse physiological processes including the regulation of growth factor signaling, the hypoxic response, inflammation and the immune response. During the last 60–100 years the life style, at least in the Western world, has changed enormously. This became obvious with an increase in caloric intake, decreased energy expenditure as well as the appearance of alcoholism and smoking; These changes were shown to contribute to generation of ROS which are, at least in part, associated with the occurrence of several chronic diseases like adiposity, atherosclerosis, type II diabetes, and cancer. In this review we discuss aspects and problems on the role of intracellular ROS formation and nutrition with the link to diseases and their problematic therapeutical issues. Oxidative stress is linked to overnutrition, obesity and associated diseases or cancer. Reactive oxygen species (ROS) are crucially involved in modulation of signaling cascades. NOX proteins and hypoxia contribute to formation of ROS under different nutrient regimes. ROS are powerful post-transcriptional and epigenetic regulators. Treatment of obesity with antioxidants requires more, larger, and better monitored clinical trials.
Collapse
Affiliation(s)
- Agnes Görlach
- Experimental and Molecular Pediatric Cardiology, German Heart Center Munich, Technical University Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Elitsa Y Dimova
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Andreas Petry
- Experimental and Molecular Pediatric Cardiology, German Heart Center Munich, Technical University Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Antonio Martínez-Ruiz
- Servicio de Immunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa, Madrid, Spain
| | - Pablo Hernansanz-Agustín
- Servicio de Immunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa, Madrid, Spain; Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Anabela P Rolo
- Department of Life Sciences, University of Coimbra and Center for Neurosciences and Cell Biology, University of Coimbra, Portugal
| | - Carlos M Palmeira
- Department of Life Sciences, University of Coimbra and Center for Neurosciences and Cell Biology, University of Coimbra, Portugal
| | - Thomas Kietzmann
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, Oulu, Finland.
| |
Collapse
|
212
|
Allison RR. Radiobiological modifiers in clinical radiation oncology: current reality and future potential. Future Oncol 2015; 10:2359-79. [PMID: 25525845 DOI: 10.2217/fon.14.174] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Radiation therapy can successfully ablate tumors. However, the same ionization process that destroys a cancer can also permanently damage surrounding organs resulting in unwanted clinical morbidity. Therefore, modern radiation therapy attempts to minimize dose to normal tissue to prevent side effects. Still, as tumors and normal tissues intercalate, the risk of normal tissue injury often may prevent tumoricidal doses of radiation therapy to be delivered. This paper will review current outcomes and limitations of radiobiological modifiers that may selectively enhance the radiosensitivity of tumors as well as parallel techniques that may protect normal tissues from radiation injury. Future endeavors based in part upon newly elucidated genetic pathways will be highlighted.
Collapse
|
213
|
Jurkuvenaite A, Benavides GA, Komarova S, Doran SF, Johnson M, Aggarwal S, Zhang J, Darley-Usmar VM, Matalon S. Upregulation of autophagy decreases chlorine-induced mitochondrial injury and lung inflammation. Free Radic Biol Med 2015; 85:83-94. [PMID: 25881550 PMCID: PMC4508227 DOI: 10.1016/j.freeradbiomed.2015.03.039] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 03/06/2015] [Accepted: 03/24/2015] [Indexed: 12/14/2022]
Abstract
The mechanisms of toxicity during exposure of the airways to chlorinated biomolecules generated during the course of inflammation and to chlorine (Cl2) gas are poorly understood. We hypothesized that lung epithelial cell mitochondria are damaged by Cl2 exposure and activation of autophagy mitigates this injury. To address this, NCI-H441 (human lung adenocarcinoma epithelial) cells were exposed to Cl2 (100 ppm/15 min) and bioenergetics were assessed. One hour after Cl2, cellular bioenergetic function and mitochondrial membrane potential were decreased. These changes were associated with increased MitoSOX signal, and treatment with the mitochondrial redox modulator MitoQ attenuated these bioenergetic defects. At 6h postexposure, there was significant increase in autophagy, which was associated with an improvement of mitochondrial function. Pretreatment of H441 cells with trehalose (an autophagy activator) improved bioenergetic function, whereas 3-methyladenine (an autophagy inhibitor) resulted in increased bioenergetic dysfunction 1h after Cl2 exposure. These data indicate that Cl2 induces bioenergetic dysfunction, and autophagy plays a protective role in vitro. Addition of trehalose (2 vol%) to the drinking water of C57BL/6 mice for 6 weeks, but not 1 week, before Cl2 (400 ppm/30 min) decreased white blood cells in the bronchoalveolar lavage fluid at 6h after Cl2 by 70%. Acute administration of trehalose delivered through inhalation 24 and 1h before the exposure decreased alveolar permeability but not cell infiltration. These data indicate that Cl2 induces bioenergetic dysfunction associated with lung inflammation and suggests that autophagy plays a protective role.
Collapse
Affiliation(s)
- Asta Jurkuvenaite
- Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Pulmonary Injury and Repair Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Gloria A Benavides
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294,USA; Center for Free Radical Biology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Svetlana Komarova
- Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Stephen F Doran
- Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Michelle Johnson
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294,USA; Center for Free Radical Biology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Saurabh Aggarwal
- Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Pulmonary Injury and Repair Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Center for Free Radical Biology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jianhua Zhang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294,USA; Center for Free Radical Biology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Veterans Administration Medical Center, Birmingham, AL 35233, USA
| | - Victor M Darley-Usmar
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294,USA; Center for Free Radical Biology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Sadis Matalon
- Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Pulmonary Injury and Repair Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Center for Free Radical Biology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
214
|
Kumar B, Iqbal MA, Singh RK, Bamezai RNK. Resveratrol inhibits TIGAR to promote ROS induced apoptosis and autophagy. Biochimie 2015. [PMID: 26212201 DOI: 10.1016/j.biochi.2015.07.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Resveratrol has been shown to exhibit its anti-cancer effect through a variety of mechanisms. Here, TIGAR (TP53-Induced Glycolysis and Apoptosis Regulator) was identified as an important target of resveratrol for exhibiting ROS-dependent-consequences on apoptosis and autophagy. Resveratrol treatment decreased TIGAR protein irrespective of cell line used. Down-regulated TIGAR protein triggered a drop in reduced-glutathione levels which resulted in sustained ROS, responsible for apoptosis and autophagy. Over-expression and silencing experiments demonstrated the importance of TIGAR in affecting the ROS-dependent anti-cancer effects of resveratrol. Resveratrol treated cells exhibited autophagy to escape apoptosis, however, chloroquine treatment along with resveratrol, blocked protective autophagy and facilitated apoptosis. Collectively, results unravel the effects of resveratrol on TIGAR in mediating its ROS dependent influence and suggest a better combination therapy of resveratrol and chloroquine for probable cancer treatment.
Collapse
Affiliation(s)
- Bhupender Kumar
- National Centre of Applied Human Genetics, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Mohammad Askandar Iqbal
- National Centre of Applied Human Genetics, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Rajnish Kumar Singh
- National Centre of Applied Human Genetics, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Rameshwar N K Bamezai
- National Centre of Applied Human Genetics, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
215
|
Jilek JL, Sant KE, Cho KH, Reed MS, Pohl J, Hansen JM, Harris C. Ethanol Attenuates Histiotrophic Nutrition Pathways and Alters the Intracellular Redox Environment and Thiol Proteome during Rat Organogenesis. Toxicol Sci 2015; 147:475-89. [PMID: 26185205 DOI: 10.1093/toxsci/kfv145] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Ethanol (EtOH) is a reactive oxygen-generating teratogen involved in the etiology of structural and functional developmental defects. Embryonic nutrition, redox environment, and changes in the thiol proteome following EtOH exposures (1.56.0 mg/ml) were studied in rat whole embryo culture. Glutathione (GSH) and cysteine (Cys) concentrations with their respective intracellular redox potentials (Eh) were determined using high-performance liquid chromatography. EtOH reduced GSH and Cys concentrations in embryo (EMB) and visceral yolk sac (VYS) tissues, and also in yolk sac and amniotic fluids. These changes produced greater oxidation as indicated by increasingly positive Eh values. EtOH reduced histiotrophic nutrition pathway activities as measured by the clearance of fluorescin isothiocyanate (FITC)-albumin from culture media. A significant decrease in total FITC clearance was observed at all concentrations, reaching approximately 50% at the highest dose. EtOH-induced changes to the thiol proteome were measured in EMBs and VYSs using isotope-coded affinity tags. Decreased concentrations for specific proteins from cytoskeletal dynamics and endocytosis pathways (α-actinin, α-tubulin, cubilin, and actin-related protein 2); nuclear translocation (Ran and RanBP1); and maintenance of receptor-mediated endocytosis (cubilin) were observed. Kyoto encyclopedia of genes and genomes (KEGG) pathway analysis also identified a decrease in ribosomal proteins in both EMB and VYS. Results show that EtOH interferes with nutrient uptake to reduce availability of amino acids and micronutrients required by the conceptus. Intracellular antioxidants such as GSH and Cys are depleted following EtOH and Eh values increase. Thiol proteome analysis in the EMB and VYS show selectively altered actin/cytoskeleton, endocytosis, ribosome biogenesis and function, nuclear transport, and stress-related responses.
Collapse
Affiliation(s)
- Joseph L Jilek
- *Department of Environmental Health Sciences, University of Michigan, Ann Arbor, Michigan 48109
| | - Karilyn E Sant
- *Department of Environmental Health Sciences, University of Michigan, Ann Arbor, Michigan 48109
| | - Katherine H Cho
- *Department of Environmental Health Sciences, University of Michigan, Ann Arbor, Michigan 48109
| | - Matthew S Reed
- Biotechnology Core Facility Branch, Centers for Disease Control, Atlanta, Georgia 30333; and
| | - Jan Pohl
- Biotechnology Core Facility Branch, Centers for Disease Control, Atlanta, Georgia 30333; and
| | - Jason M Hansen
- Department of Physiology and Developmental Biology, College of Life Sciences, Brigham Young University, Provo, Utah 84602
| | - Craig Harris
- *Department of Environmental Health Sciences, University of Michigan, Ann Arbor, Michigan 48109;
| |
Collapse
|
216
|
Butterfield DA. Redox signaling in neurodegeneration. Neurobiol Dis 2015; 84:1-3. [PMID: 26171987 DOI: 10.1016/j.nbd.2015.07.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 07/06/2015] [Indexed: 12/20/2022] Open
Affiliation(s)
- D Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506-0055, USA.
| |
Collapse
|
217
|
Huang C, Tong L, Lu X, Wang J, Yao W, Jiang B, Zhang W. Methylene Blue Attenuates iNOS Induction Through Suppression of Transcriptional Factor Binding Amid iNOS mRNA Transcription. J Cell Biochem 2015; 116:1730-40. [DOI: 10.1002/jcb.25132] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 02/06/2015] [Indexed: 12/26/2022]
Affiliation(s)
- Chao Huang
- Department of Pharmacology; School of Pharmacy; Nantong University; #19 Qixiu Road Nantong Jiangsu Province 226001 China
| | - Lijuan Tong
- Department of Pharmacology; School of Pharmacy; Nantong University; #19 Qixiu Road Nantong Jiangsu Province 226001 China
| | - Xu Lu
- Department of Pharmacology; School of Pharmacy; Nantong University; #19 Qixiu Road Nantong Jiangsu Province 226001 China
| | - Jia Wang
- Department of Pharmacology; School of Pharmacy; Nantong University; #19 Qixiu Road Nantong Jiangsu Province 226001 China
| | - Wenjuan Yao
- Department of Pharmacology; School of Pharmacy; Nantong University; #19 Qixiu Road Nantong Jiangsu Province 226001 China
| | - Bo Jiang
- Department of Pharmacology; School of Pharmacy; Nantong University; #19 Qixiu Road Nantong Jiangsu Province 226001 China
| | - Wei Zhang
- Department of Pharmacology; School of Pharmacy; Nantong University; #19 Qixiu Road Nantong Jiangsu Province 226001 China
| |
Collapse
|
218
|
Discovery of molecular mechanisms of lignan justicidin A using L1000 gene expression profiles and the Library of Integrated Network-based Cellular Signatures database. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.04.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
219
|
Yoo TH, Fornoni A. Nonimmunologic targets of immunosuppressive agents in podocytes. Kidney Res Clin Pract 2015; 34:69-75. [PMID: 26484025 PMCID: PMC4570600 DOI: 10.1016/j.krcp.2015.03.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 03/16/2015] [Accepted: 03/21/2015] [Indexed: 02/06/2023] Open
Abstract
Proteinuria is a characteristic finding in glomerular diseases and is closely associated with renal outcomes. In addition, therapeutic interventions that reduce proteinuria improve renal prognosis. Accumulating evidence has demonstrated that podocytes act as key modulators of glomerular injury and proteinuria. The podocyte, or glomerular visceral epithelial cell, is a highly specialized and differentiated cell that forms interdigitated foot processes with neighboring podocytes, which are bridged together by an extracellular structure known as the "slit diaphragm" (SD). The SD acts as a size- and charge-selective barrier to plasma protein. Derangement of SD structure or loss of SD-associated protein results in podocyte injury and proteinuria. During the past decades, several immune-modulating agents have been used for the treatment of glomerular diseases and for the reduction of proteinuria. Interestingly, recent studies have demonstrated that immunosuppressive agents can have a direct effect on the SD-associated proteins and stabilize actin cytoskeleton in podocyte and have therefore introduced the concept of nonimmunologic mechanism of renoprotection by immunomodulators. This review focuses on the evidence that immuno-modulating agents directly target podocytes.
Collapse
Affiliation(s)
- Tae-Hyun Yoo
- Department of Internal Medicine, College of Medicine, Yonsei University, Seoul, Korea
- Division of Nephrology and Hypertension, Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
- Peggy and Harold Katz Family Drug Discovery Center, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Alessia Fornoni
- Division of Nephrology and Hypertension, Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
- Peggy and Harold Katz Family Drug Discovery Center, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
220
|
Henry E, Fung N, Liu J, Drakakaki G, Coaker G. Beyond glycolysis: GAPDHs are multi-functional enzymes involved in regulation of ROS, autophagy, and plant immune responses. PLoS Genet 2015; 11:e1005199. [PMID: 25918875 PMCID: PMC4412566 DOI: 10.1371/journal.pgen.1005199] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 04/08/2015] [Indexed: 11/17/2022] Open
Abstract
Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is an important enzyme in energy metabolism with diverse cellular regulatory roles in vertebrates, but few reports have investigated the importance of plant GAPDH isoforms outside of their role in glycolysis. While animals possess one GAPDH isoform, plants possess multiple isoforms. In this study, cell biological and genetic approaches were used to investigate the role of GAPDHs during plant immune responses. Individual Arabidopsis GAPDH knockouts (KO lines) exhibited enhanced disease resistance phenotypes upon inoculation with the bacterial plant pathogen Pseudomonas syringae pv. tomato. KO lines exhibited accelerated programmed cell death and increased electrolyte leakage in response to effector triggered immunity. Furthermore, KO lines displayed increased basal ROS accumulation as visualized using the fluorescent probe H2DCFDA. The gapa1-2 and gapc1 KOs exhibited constitutive autophagy phenotypes in the absence of nutrient starvation. Due to the high sequence conservation between vertebrate and plant cytosolic GAPDH, our experiments focused on cytosolic GAPC1 cellular dynamics using a complemented GAPC1-GFP line. Confocal imaging coupled with an endocytic membrane marker (FM4-64) and endosomal trafficking inhibitors (BFA, Wortmannin) demonstrated cytosolic GAPC1 is localized to the plasma membrane and the endomembrane system, in addition to the cytosol and nucleus. After perception of bacterial flagellin, GAPC1 dynamically responded with a significant increase in size of fluorescent puncta and enhanced nuclear accumulation. Taken together, these results indicate that plant GAPDHs can affect multiple aspects of plant immunity in diverse sub-cellular compartments.
Collapse
Affiliation(s)
- Elizabeth Henry
- Department of Plant Pathology, University of California Davis, Davis, California, United States of America
| | - Nicholas Fung
- Department of Plant Pathology, University of California Davis, Davis, California, United States of America
| | - Jun Liu
- Department of Plant Pathology, University of California Davis, Davis, California, United States of America; Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Georgia Drakakaki
- Department of Plant Sciences, University of California Davis, Davis, California, United States of America
| | - Gitta Coaker
- Department of Plant Pathology, University of California Davis, Davis, California, United States of America
| |
Collapse
|
221
|
Kuo CY, Chiu YC, Lee AYL, Hwang TL. Mitochondrial Lon protease controls ROS-dependent apoptosis in cardiomyocyte under hypoxia. Mitochondrion 2015; 23:7-16. [PMID: 25922169 DOI: 10.1016/j.mito.2015.04.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 04/14/2015] [Accepted: 04/21/2015] [Indexed: 11/30/2022]
Abstract
Apoptosis of cardiomyocytes, under ischemic conditions, has been identified as an essential process in the progression of heart failure. Under hypoxic conditions, mitochondria can become a threat to the cell because of their capacity to generate reactive oxygen species (ROS). As ROS appear to have a critical role in heart failure, there has been considerable interest in identifying the candidate proteins involved and in developing strategies to reduce oxidative stress. Lon protease (Lon) is a multifunctional protein that mediates protein quality control and stress response in mitochondria. However, comprehensive and detailed studies, on the role of Lon in hypoxia-induced cardiomyocyte apoptosis, have yet to be carried out. In the present study, we demonstrated that hypoxia induced ROS-dependent cardiomyocyte apoptosis. Lon was upregulated in hypoxia-induced cardiomyocytes. Lon downregulation attenuated hypoxia-induced cardiomyocyte apoptosis through a reduction of ROS level. Moreover, overexpression of Lon stimulated ROS production and induced apoptosis under normoxic conditions in cardiomyocytes. Our results identify Lon as a novel regulator of cardiomyocyte fate and offers exciting new insights into the therapeutic potential of hypoxia-induced cardiomyocyte apoptosis.
Collapse
Affiliation(s)
- Chan-Yen Kuo
- Graduate Institute of Natural Products, School of Traditional Chinese Medicine, College of Medicine, and Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Taoyuan 33302, Taiwan; National Institute of Cancer Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Yi-Chieh Chiu
- National Institute of Cancer Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Alan Yueh-Luen Lee
- National Institute of Cancer Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan; Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan.
| | - Tsong-Long Hwang
- Graduate Institute of Natural Products, School of Traditional Chinese Medicine, College of Medicine, and Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Taoyuan 33302, Taiwan; Department of Cosmetic Science and Research Center for Industry of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33302, Taiwan; Immunology Consortium, Chang Gung Memorial Hospital, Kweishan, Taoyuan, Taiwan.
| |
Collapse
|
222
|
Jaafar SNT, Coelho AV, Sheehan D. Redox proteomic analysis ofmytilus edulisgills: effects of the pharmaceutical diclofenac on a non-target organism. Drug Test Anal 2015; 7:957-66. [DOI: 10.1002/dta.1786] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 02/20/2015] [Accepted: 02/20/2015] [Indexed: 11/08/2022]
Affiliation(s)
- Siti Nur Tahirah Jaafar
- Proteomics Research Group, School of Biochemistry and Cell Biology and Environmental Research Institute; University College Cork; Ireland
- Marine Biology Program, School of Marine Science and Environment; Universiti Malaysia Terengganu; Terengganu Malaysia
| | - Ana Varela Coelho
- Mass Spectrometry Laboratory, Analytical Services Unit, Institute of Chemical and Biological Technology (ITQB); New University of Lisbon; Avenida República - Quinta do Marquês 2784-505 Oeiras Portugal
| | - David Sheehan
- Proteomics Research Group, School of Biochemistry and Cell Biology and Environmental Research Institute; University College Cork; Ireland
| |
Collapse
|
223
|
Revelant G, Huber-Villaume S, Dunand S, Kirsch G, Schohn H, Hesse S. Synthesis and biological evaluation of novel 2-heteroarylimino-1,3-thiazolidin-4-ones as potential anti-tumor agents. Eur J Med Chem 2015; 94:102-12. [DOI: 10.1016/j.ejmech.2015.02.053] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 02/26/2015] [Accepted: 02/27/2015] [Indexed: 01/08/2023]
|
224
|
Lead toxicity induces autophagy to protect against cell death through mTORC1 pathway in cardiofibroblasts. Biosci Rep 2015; 35:BSR20140164. [PMID: 25686247 PMCID: PMC4381285 DOI: 10.1042/bsr20140164] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Heavy metals, such as lead (Pb2+), are usually accumulated in human bodies and impair human's health. Lead is a metal with many recognized adverse health side effects and yet the molecular processes underlying lead toxicity are still poorly understood. In the present study, we proposed to investigate the effects of lead toxicity in cultured cardiofibroblasts. After lead treatment, cultured cardiofibroblasts showed severe endoplasmic reticulum (ER) stress. However, the lead-treated cardiofibroblasts were not dramatically apoptotic. Further, we found that these cells determined to undergo autophagy through inhibiting mammalian target of rapamycin complex 1 (mTORC1) pathway. Moreover, inhibition of autophagy by 3-methyladenine (3-MA) may dramatically enhance lead toxicity in cardiofibroblasts and cause cell death. Our data establish that lead toxicity induces cell stress in cardiofibroblasts and protective autophagy is activated by inhibition of mTORC1 pathway. These findings describe a mechanism by which lead toxicity may promote the autophagy of cardiofibroblasts cells, which protects cells from cell stress. Our findings provide evidence that autophagy may help cells to survive under ER stress conditions in cardiofibroblasts and may set up an effective therapeutic strategy for heavy metal toxicity. Lead is a metal with many recognized adverse health side effects, and yet the molecular processes in cardiofibroblasts underlying lead toxicity are still poorly understood. Our current findings will help to understand the role of lead-mediated toxicity in cardiofibroblasts, indicating that autophagy serves a protective role in response to ER stress, which affords to set up an effective therapeutic strategy for the numerous diseases related to lead-toxicity.
Collapse
|
225
|
Niu NK, Wang ZL, Pan ST, Ding HQ, Au GHT, He ZX, Zhou ZW, Xiao G, Yang YX, Zhang X, Yang T, Chen XW, Qiu JX, Zhou SF. Pro-apoptotic and pro-autophagic effects of the Aurora kinase A inhibitor alisertib (MLN8237) on human osteosarcoma U-2 OS and MG-63 cells through the activation of mitochondria-mediated pathway and inhibition of p38 MAPK/PI3K/Akt/mTOR signaling pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:1555-84. [PMID: 25792811 PMCID: PMC4362906 DOI: 10.2147/dddt.s74197] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Osteosarcoma (OS) is the most common malignant bone tumor occurring mostly in children and adolescents between 10 and 20 years of age with poor response to current therapeutics. Alisertib (ALS, MLN8237) is a selective Aurora kinase A inhibitor that displays anticancer effects on several types of cancer. However, the role of ALS in the treatment of OS remains unknown. This study aimed to investigate the effects of ALS on the cell growth, apoptosis, autophagy, and epithelial to mesenchymal transition (EMT) and the underlying mechanisms in two human OS cell lines U-2 OS and MG-63. The results showed that ALS had potent growth inhibitory, pro-apoptotic, pro-autophagic, and EMT inhibitory effects on U-2 OS and MG-63 cells. ALS remarkably induced G2/M arrest and down-regulated the expression levels of cyclin-dependent kinases 1 and 2 and cyclin B1 in both U-2 OS and MG-63 cells. ALS markedly induced mitochondria-mediated apoptosis with a significant increase in the expression of key pro-apoptotic proteins and a decrease in main anti-apoptotic proteins. Furthermore, ALS promoted autophagic cell death via the inhibition of phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) and p38 mitogen-activated protein kinase (p38 MAPK) signaling pathways, and activation of 5′-AMP-dependent kinase (AMPK) signaling pathway. Inducers or inhibitors of apoptosis or autophagy simultaneously altered ALS-induced apoptotic and autophagic death in both U-2 OS and MG-63 cells, suggesting a crosstalk between these two primary modes of programmed cell death. Moreover, ALS suppressed EMT-like phenotypes with a marked increase in the expression of E-cadherin but a decrease in N-cadherin in U-2 OS and MG-63 cells. ALS treatment also induced reactive oxygen species (ROS) generation but inhibited the expression levels of sirtuin 1 and nuclear factor-erythroid-2-related factor 2 (Nrf2) in both cell lines. Taken together, these findings show that ALS promotes apoptosis and autophagy but inhibits EMT via PI3K/Akt/mTOR, p38 MAPK, and AMPK signaling pathways with involvement of ROS- and sirtuin 1-associated pathways in U-2 OS and MG-63 cells. ALS is a promising anticancer agent in OS treatment and further studies are needed to confirm its efficacy and safety in OS chemotherapy.
Collapse
Affiliation(s)
- Ning-Kui Niu
- Department of Spinal Surgery, General Hospital of Ningxia Medical University, Yinchuan, People's Republic of China ; Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA ; Department of Orthopedics, General Hospital of Tianjin Medical University, Tianjin, People's Republic of China
| | - Zi-Li Wang
- Department of Spinal Surgery, General Hospital of Ningxia Medical University, Yinchuan, People's Republic of China
| | - Shu-Ting Pan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA ; Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People's Republic of China
| | - Hui-Qiang Ding
- Department of Spinal Surgery, General Hospital of Ningxia Medical University, Yinchuan, People's Republic of China
| | - Giang H T Au
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Zhi-Xu He
- Guizhou Provincial Key Laboratory for Regenerative Medicine, Stem Cell and Tissue Engineering Research Center and Sino-US Joint Laboratory for Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Zhi-Wei Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA ; Guizhou Provincial Key Laboratory for Regenerative Medicine, Stem Cell and Tissue Engineering Research Center and Sino-US Joint Laboratory for Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Guozhi Xiao
- Department of Biochemistry, Medical Center, Rush University, Chicago, IL, USA
| | - Yin-Xue Yang
- Department of Colorectal Surgery, General Hospital of Ningxia Medical University, Yinchuan, People's Republic of China
| | - Xueji Zhang
- Research Center for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing, People's Republic of China
| | - Tianxin Yang
- Department of Internal Medicine, University of Utah and Salt Lake Veterans Affairs Medical Center, Salt Lake City, UT, USA
| | - Xiao-Wu Chen
- Department of General Surgery, The First People's Hospital of Shunde affiliated to Southern Medical University, Foshan, Guangdong, People's Republic of China
| | - Jia-Xuan Qiu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People's Republic of China
| | - Shu-Feng Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
| |
Collapse
|
226
|
Poole LB. The basics of thiols and cysteines in redox biology and chemistry. Free Radic Biol Med 2015; 80:148-57. [PMID: 25433365 PMCID: PMC4355186 DOI: 10.1016/j.freeradbiomed.2014.11.013] [Citation(s) in RCA: 651] [Impact Index Per Article: 65.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 10/20/2014] [Accepted: 11/17/2014] [Indexed: 02/06/2023]
Abstract
Cysteine is one of the least abundant amino acids, yet it is frequently found as a highly conserved residue within functional (regulatory, catalytic, or binding) sites in proteins. It is the unique chemistry of the thiol or thiolate group of cysteine that imparts to functional sites their specialized properties (e.g., nucleophilicity, high-affinity metal binding, and/or ability to form disulfide bonds). Highlighted in this review are some of the basic biophysical and biochemical properties of cysteine groups and the equations that apply to them, particularly with respect to pKa and redox potential. Also summarized are the types of low-molecular-weight thiols present in high concentrations in most cells, as well as the ways in which modifications of cysteinyl residues can impart or regulate molecular functions important to cellular processes, including signal transduction.
Collapse
Affiliation(s)
- Leslie B Poole
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA.
| |
Collapse
|
227
|
Wang X, Wang C, Sun YT, Sun CZ, Zhang Y, Wang XH, Zhao K. Taxol Produced from Endophytic Fungi Induces Apoptosis in Human Breast, Cervical and Ovarian Cancer Cells. Asian Pac J Cancer Prev 2015; 16:125-31. [DOI: 10.7314/apjcp.2015.16.1.125] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
228
|
Mitochondrial dependency in progression of acute myeloid leukemia. Mitochondrion 2015; 21:41-8. [PMID: 25640960 DOI: 10.1016/j.mito.2015.01.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 10/23/2014] [Accepted: 01/21/2015] [Indexed: 11/20/2022]
Abstract
Acute myeloid leukemia (AML) is a clonal hematopoietic malignant disorder which arises due to dysregulated differentiation, uncontrolled growth and inhibition of apoptosis leading to the accumulation of immature myeloid progenitor in the bone marrow. The heterogeneity of the disease at the molecular and cytogenetic level has led to the identification of several alteration of biological and clinical significance. One of the alterations which have gained attention in recent times is the altered energy and metabolic dependency of cancer originally proposed by Warburg. Mitochondria are important cell organelles regulating cellular energetic level, metabolism and apoptosis which in turn can affect cell proliferation and differentiation, the major manifestations of diseases like AML. In recent times the importance of mitochondrial generated ATP and mitochondrial localized metabolic pathways has been shown to play important role in the progression of AML. These studies have also demonstrated the clinical significance of mitochondrial targets for its effectiveness in combating relapsed or refractory AML. Here we review the importance of the mitochondrial dependency for the progression of AML and the emergence of the mitochondrial molecular targets which holds therapeutic importance.
Collapse
|
229
|
Teaching the basics of autophagy and mitophagy to redox biologists--mechanisms and experimental approaches. Redox Biol 2015; 4:242-59. [PMID: 25618581 PMCID: PMC4803799 DOI: 10.1016/j.redox.2015.01.003] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 12/24/2014] [Accepted: 01/01/2015] [Indexed: 02/08/2023] Open
Abstract
Autophagy is a lysosomal mediated degradation activity providing an essential mechanism for recycling cellular constituents, and clearance of excess or damaged lipids, proteins and organelles. Autophagy involves more than 30 proteins and is regulated by nutrient availability, and various stress sensing signaling pathways. This article provides an overview of the mechanisms and regulation of autophagy, its role in health and diseases, and methods for its measurement. Hopefully this teaching review together with the graphic illustrations will be helpful for instructors teaching graduate students who are interested in grasping the concepts and major research areas and introducing recent developments in the field. mTOR, Beclin–VPS34, LC3 homologs, and adaptor proteins in autophagy. Autophagosomal membranes may derive from multiple sources. Autophagosomal–lysosomal fusion contributes to the control of autophagic flux. Assess autophagy by autophagosomal and protein turnover, and morphological alterations. Autophagy adysfunction in cancer, aging, neurodegeneration and infection.
Collapse
|
230
|
Qiu JX, Zhou ZW, He ZX, Zhao RJ, Zhang X, Yang L, Zhou SF, Mao ZF. Plumbagin elicits differential proteomic responses mainly involving cell cycle, apoptosis, autophagy, and epithelial-to-mesenchymal transition pathways in human prostate cancer PC-3 and DU145 cells. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:349-417. [PMID: 25609920 PMCID: PMC4294653 DOI: 10.2147/dddt.s71677] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Plumbagin (PLB) has exhibited a potent anticancer effect in preclinical studies, but the molecular interactome remains elusive. This study aimed to compare the quantitative proteomic responses to PLB treatment in human prostate cancer PC-3 and DU145 cells using the approach of stable-isotope labeling by amino acids in cell culture (SILAC). The data were finally validated using Western blot assay. First, the bioinformatic analysis predicted that PLB could interact with 78 proteins that were involved in cell proliferation and apoptosis, immunity, and signal transduction. Our quantitative proteomic study using SILAC revealed that there were at least 1,225 and 267 proteins interacting with PLB and there were 341 and 107 signaling pathways and cellular functions potentially regulated by PLB in PC-3 and DU145 cells, respectively. These proteins and pathways played a critical role in the regulation of cell cycle, apoptosis, autophagy, epithelial to mesenchymal transition (EMT), and reactive oxygen species generation. The proteomic study showed substantial differences in response to PLB treatment between PC-3 and DU145 cells. PLB treatment significantly modulated the expression of critical proteins that regulate cell cycle, apoptosis, and EMT signaling pathways in PC-3 cells but not in DU145 cells. Consistently, our Western blotting analysis validated the bioinformatic and proteomic data and confirmed the modulating effects of PLB on important proteins that regulated cell cycle, apoptosis, autophagy, and EMT in PC-3 and DU145 cells. The data from the Western blot assay could not display significant differences between PC-3 and DU145 cells. These findings indicate that PLB elicits different proteomic responses in PC-3 and DU145 cells involving proteins and pathways that regulate cell cycle, apoptosis, autophagy, reactive oxygen species production, and antioxidation/oxidation homeostasis. This is the first systematic study with integrated computational, proteomic, and functional analyses revealing the networks of signaling pathways and differential proteomic responses to PLB treatment in prostate cancer cells. Quantitative proteomic analysis using SILAC represents an efficient and highly sensitive approach to identify the target networks of anticancer drugs like PLB, and the data may be used to discriminate the molecular and clinical subtypes, and to identify new therapeutic targets and biomarkers, for prostate cancer. Further studies are warranted to explore the potential of quantitative proteomic analysis in the identification of new targets and biomarkers for prostate cancer.
Collapse
Affiliation(s)
- Jia-Xuan Qiu
- School of Public Health, Wuhan University, Wuhan, Hubei, People's Republic of China ; Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People's Republic of China
| | - Zhi-Wei Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA ; Guizhou Provincial Key Laboratory for Regenerative Medicine, Stem Cell and Tissue Engineering Research Center and Sino-US Joint Laboratory for Medical Sciences, Guiyang Medical University, Guiyang, Guizhou, People's Republic of China
| | - Zhi-Xu He
- Guizhou Provincial Key Laboratory for Regenerative Medicine, Stem Cell and Tissue Engineering Research Center and Sino-US Joint Laboratory for Medical Sciences, Guiyang Medical University, Guiyang, Guizhou, People's Republic of China
| | - Ruan Jin Zhao
- Center for Traditional Chinese Medicine, Sarasota, FL, USA
| | - Xueji Zhang
- Research Center for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing, People's Republic of China
| | - Lun Yang
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Shu-Feng Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA ; Guizhou Provincial Key Laboratory for Regenerative Medicine, Stem Cell and Tissue Engineering Research Center and Sino-US Joint Laboratory for Medical Sciences, Guiyang Medical University, Guiyang, Guizhou, People's Republic of China
| | - Zong-Fu Mao
- School of Public Health, Wuhan University, Wuhan, Hubei, People's Republic of China
| |
Collapse
|
231
|
Sinha RA, Singh BK, Zhou J, Wu Y, Farah BL, Ohba K, Lesmana R, Gooding J, Bay BH, Yen PM. Thyroid hormone induction of mitochondrial activity is coupled to mitophagy via ROS-AMPK-ULK1 signaling. Autophagy 2015; 11:1341-1357. [PMID: 26103054 PMCID: PMC4590606 DOI: 10.1080/15548627.2015.1061849] [Citation(s) in RCA: 152] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Revised: 05/04/2015] [Accepted: 05/18/2015] [Indexed: 01/13/2023] Open
Abstract
Currently, there is limited understanding about hormonal regulation of mitochondrial turnover. Thyroid hormone (T3) increases oxidative phosphorylation (OXPHOS), which generates reactive oxygen species (ROS) that damage mitochondria. However, the mechanism for maintenance of mitochondrial activity and quality control by this hormone is not known. Here, we used both in vitro and in vivo hepatic cell models to demonstrate that induction of mitophagy by T3 is coupled to oxidative phosphorylation and ROS production. We show that T3 induction of ROS activates CAMKK2 (calcium/calmodulin-dependent protein kinase kinase 2, β) mediated phosphorylation of PRKAA1/AMPK (5' AMP-activated protein kinase), which in turn phosphorylates ULK1 (unc-51 like autophagy activating kinase 1) leading to its mitochondrial recruitment and initiation of mitophagy. Furthermore, loss of ULK1 in T3-treated cells impairs both mitophagy as well as OXPHOS without affecting T3 induced general autophagy/lipophagy. These findings demonstrate a novel ROS-AMPK-ULK1 mechanism that couples T3-induced mitochondrial turnover with activity, wherein mitophagy is necessary not only for removing damaged mitochondria but also for sustaining efficient OXPHOS.
Collapse
Affiliation(s)
- Rohit A Sinha
- Program of Cardiovascular and Metabolic Disorders; Duke-NUS Graduate Medical School; Singapore
| | - Brijesh K Singh
- Program of Cardiovascular and Metabolic Disorders; Duke-NUS Graduate Medical School; Singapore
| | - Jin Zhou
- Program of Cardiovascular and Metabolic Disorders; Duke-NUS Graduate Medical School; Singapore
| | - Yajun Wu
- Department of Anatomy; Yong Loo Lin School of Medicine; National University of Singapore; Singapore
| | - Benjamin L Farah
- Program of Cardiovascular and Metabolic Disorders; Duke-NUS Graduate Medical School; Singapore
| | - Kenji Ohba
- Program of Cardiovascular and Metabolic Disorders; Duke-NUS Graduate Medical School; Singapore
| | - Ronny Lesmana
- Program of Cardiovascular and Metabolic Disorders; Duke-NUS Graduate Medical School; Singapore
- Department of Physiology; Universitas Padjadjaran; Bandung, Indonesia
| | - Jessica Gooding
- Sarah W. Stedman Nutrition and Metabolism Center; Departments of Medicine and Pharmacology and Cancer Biology; Duke University Medical Center; Durham, NC USA
| | - Boon-Huat Bay
- Program of Cardiovascular and Metabolic Disorders; Duke-NUS Graduate Medical School; Singapore
| | - Paul M Yen
- Program of Cardiovascular and Metabolic Disorders; Duke-NUS Graduate Medical School; Singapore
- Department of Anatomy; Yong Loo Lin School of Medicine; National University of Singapore; Singapore
- Sarah W. Stedman Nutrition and Metabolism Center; Departments of Medicine and Pharmacology and Cancer Biology; Duke University Medical Center; Durham, NC USA
| |
Collapse
|
232
|
Shariatpanahi M, Khodagholi F, Ashabi G, Aghazadeh Khasraghi A, Azimi L, Abdollahi M, Ghahremani MH, Ostad SN, Noorbakhsh F, Sharifzadeh M. Ameliorating of Memory Impairment and Apoptosis in Amyloid β-Injected Rats Via Inhibition of Nitric Oxide Synthase: Possible Participation of Autophagy. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2015; 14:811-24. [PMID: 26330869 PMCID: PMC4518109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
It has been proposed that appearance of amyloid beta (Aβ) in hippocampus is one of the characteristic features of Alzheimer's disease (AD). The role of Nitric oxide (NO) in neurodegenerative disorders is controversy in different contexts. Here, we examined the effect of NO on spatial memory. For this purpose, we compared the effects of three different concentrations of L-NG-Nitroarginine Methyl Ester (L-NAME) as a nitric oxide synthase (NOS) inhibitor. We used Morris water maze (MWM) for evaluation of behavioral alterations. We also assessed the apoptosis and autophagy markers as two possible interfering pathways with NO signaling by western blot method. We found that in Aβ pretreated rats, intra-hippocampal injection of 1or 2 (μg/side) of L-NAME caused a significant reduction in escape latency and traveled distance comparing to Aβ-treatment group. Our molecular findings revealed that L-NAME could induce autophagy and attenuate apoptosis dose dependently. The protective role of autophagy and the deteriorative role of apoptosis is the hypothesis that can vindicate our findings. Thus using NOS inhibitors at low concentrations can be one of the therapeutic approaches in the future studies.
Collapse
Affiliation(s)
- Marjan Shariatpanahi
- Department of Toxicology and Pharmacology, Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Neuro Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Ghorbangol Ashabi
- Department of Physiology, School of Medicine, Physiology Research center, Ahvaz Jundishapour University of Medical Sciences, Ahvaz, Iran.
| | | | - Leila Azimi
- Department of Neuroscience, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Abdollahi
- Department of Toxicology and Pharmacology, Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Hossein Ghahremani
- Department of Toxicology and Pharmacology, Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Seyed Nasser Ostad
- Department of Toxicology and Pharmacology, Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Farshid Noorbakhsh
- Department of Immunology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Sharifzadeh
- Department of Toxicology and Pharmacology, Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Department of Neuroscience, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Corresponding author:
| |
Collapse
|
233
|
Zhang C, Li X, Liu Q. Sorbitol dehydrogenase inhibitor protects the liver from ischemia/reperfusion-induced injury via elevated glycolytic flux and enhanced sirtuin 1 activity. Mol Med Rep 2015; 11:283-288. [PMID: 25333577 DOI: 10.3892/mmr.2014.2715] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Accepted: 06/26/2014] [Indexed: 11/05/2022] Open
Abstract
Sorbitol dehydrogenase (SDH), a key enzyme of the polyol pathway, has recently been demonstrated to have an important role in mediating tissue ischemia/reperfusion (I/R) injury. The present study investigated how this enzyme may affect the ischemic liver and the mechanism underlying its effect. Firstly, C57BL/6 mice were subjected to oral administration of CP-470,711 (5 mg/kg body weight/day for five days) and 70% hepatic I/R. Next the present study further investigated the changes in liver function, histology, inflammation, apoptosis and necrosis; the cytosolic adenosine triphosphate (ATP) and nictotinamide adenine dinucleotide [NAD(H)] contents and the protein level of caspase 3 and sirtuin 1 (SIRT1). The data demonstrated that sorbitol dehydrogenase inhibitor (SDI)-administration significantly alleviated I/R-induced liver injury, palliated histological changes and lowered the level of hepatocyte apoptosis and necrosis. In addition, SDI-pretreatment in ischemic liver markedly maintained the cytosolic ATP and NAD(H) proportion, enhanced SIRT1 and suppressed the activation of caspase 3 at the protein level. The findings in the present study revealed that the flux through SDH may render the liver more vulnerable to I/R-induced injury and interventions targeting this enzyme may provide a novel adjunctive approach to protect from severe tissue injury following liver ischemia.
Collapse
Affiliation(s)
- Changhe Zhang
- Department of General Surgery, Taizhou People's Hospital, Taizhou, Jiangsu 225300, P.R. China
| | - Xiangcheng Li
- Department of Liver Transplantation, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Qinhong Liu
- Department of General Surgery, Taizhou People's Hospital, Taizhou, Jiangsu 225300, P.R. China
| |
Collapse
|
234
|
Wani W, Boyer-Guittaut M, Dodson M, Chatham J, Darley-Usmar V, Zhang J. Regulation of autophagy by protein post-translational modification. J Transl Med 2015; 95:14-25. [PMID: 25365205 PMCID: PMC4454381 DOI: 10.1038/labinvest.2014.131] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 08/14/2014] [Indexed: 12/11/2022] Open
Abstract
Autophagy is a lysosome-mediated intracellular protein degradation process that involves about 38 autophagy-related genes as well as key signaling pathways that sense cellular metabolic and redox status, and has an important role in quality control of macromolecules and organelles. As with other major cellular pathways, autophagy proteins are subjected to regulatory post-translational modification. Phosphorylation is so far the most intensively studied post-translational modification in the autophagy process, followed by ubiquitination and acetylation. An interesting and new area is also now emerging, which appears to complement these more traditional mechanisms, and includes O-GlcNAcylation and redox regulation at thiol residues. Identification of the full spectrum of post-translational modifications of autophagy proteins, and determination of their impact on autophagy will be crucial for a better understanding of autophagy regulation, its deficits in diseases, and how to exploit this process for disease therapies.
Collapse
Affiliation(s)
- Willayat Wani
- Center for Free Radical Biology, University of Alabama at Birmingham,Department of Pathology, University of Alabama at Birmingham
| | - Michaël Boyer-Guittaut
- Université de Franche-Comté, Laboratoire de Biochimie, EA3922, SFR IBCT FED4234, Sciences et Techniques, 16 route de Gray, 25030 Besançon Cedex, France
| | - Matthew Dodson
- Center for Free Radical Biology, University of Alabama at Birmingham,Department of Pathology, University of Alabama at Birmingham
| | - John Chatham
- Center for Free Radical Biology, University of Alabama at Birmingham,Department of Pathology, University of Alabama at Birmingham
| | - Victor Darley-Usmar
- Center for Free Radical Biology, University of Alabama at Birmingham,Department of Pathology, University of Alabama at Birmingham
| | - Jianhua Zhang
- Center for Free Radical Biology, University of Alabama at Birmingham,Department of Pathology, University of Alabama at Birmingham,Department of Veterans Affairs, Birmingham VA Medical Center
| |
Collapse
|
235
|
Upregulation of Unc-51-like kinase 1 by nitric oxide stabilizes SIRT1, independent of autophagy. PLoS One 2014; 9:e116165. [PMID: 25541949 PMCID: PMC4277463 DOI: 10.1371/journal.pone.0116165] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Accepted: 12/04/2014] [Indexed: 01/13/2023] Open
Abstract
SIRT1 is central to the lifespan and vascular health, but undergoes degradation that contributes to several medical conditions, including diabetes. How SIRT1 turnover is regulated remains unclear. However, emerging evidence suggests that endothelial nitric oxide synthase (eNOS) positively regulates SIRT1 protein expression. We recently identified NO as an endogenous inhibitor of 26S proteasome functionality with a cellular reporter system. Here we extended this finding to a novel pathway that regulates SIRT1 protein breakdown. In cycloheximide (CHX)-treated endothelial cells, NONOate, an NO donor, and A23187, an eNOS activator, significantly stabilized SIRT1 protein. Similarly, NO enhanced SIRT1 protein, but not mRNA expression, in CHX-free cells. NO also stabilized an autophagy-related protein unc-51 like kinase (ULK1), but did not restore SIRT1 protein levels in ULK1-siRNA-treated cells or in mouse embryonic fibroblasts (MEF) from Ulk1-/- mice. This suggests that ULK1 mediated the NO regulation of SIRT1. Furthermore, adenoviral overexpression of ULK1 increased SIRT1 protein expression, while ULK1 siRNA treatment decreased it. Rapamycin-induced autophagy did not mimic these effects, suggesting that the effects of ULK1 were autophagy-independent. Treatment with MG132, a proteasome inhibitor, or siRNA of β-TrCP1, an E3 ligase, prevented SIRT1 reduction induced by ULK1-siRNA. Mechanistically, ULK1 negatively regulated 26S proteasome functionality, which was at least partly mediated by O-linked-GlcNAc transferase (OGT), probably by increased O-GlcNAc modification of proteasomal subunit Rpt2. The NO-ULK1-SIRT1 axis was likely operative in the whole animal: both ULK1 and SIRT1 protein levels were significantly reduced in tissue homogenates in eNOS-knockout mice (lung) and in db/db mice where eNOS is downregulated (lung and heart). Taken together, the results show that NO stabilizes SIRT1 by regulating 26S proteasome functionality through ULK1 and OGT, but not autophagy, in endothelial cells.
Collapse
|
236
|
Abstract
In the past several years, it has been demonstrated that the reactive oxygen species (ROS) may act as intracellular signalling molecules to activate or inhibit specific signalling pathways and regulate physiological cellular functions. It is now well-established that ROS regulate autophagy, an intracellular degradation process. However, the signalling mechanisms through which ROS modulate autophagy in a regulated manner have only been minimally clarified. NADPH oxidase (Nox) enzymes are membrane-bound enzymatic complexes responsible for the dedicated generation of ROS. Different isoforms of Nox exist with different functions. Recent studies demonstrated that Nox-derived ROS can promote autophagy, with Nox2 and Nox4 representing the isoforms of Nox implicated thus far. Nox2- and Nox4-dependent autophagy plays an important role in the elimination of pathogens by phagocytes and in the regulation of vascular- and cancer-cell survival. Interestingly, we recently found that Nox is also important for autophagy regulation in cardiomyocytes. We found that Nox4, but not Nox2, promotes the activation of autophagy and survival in cardiomyocytes in response to nutrient deprivation and ischaemia through activation of the PERK (protein kinase RNA-like endoplasmic reticulum kinase) signalling pathway. In the present paper, we discuss the importance of Nox family proteins and ROS in the regulation of autophagy, with a particular focus on the role of Nox4 in the regulation of autophagy in the heart.
Collapse
|
237
|
Plum S, Steinbach S, Abel L, Marcus K, Helling S, May C. Proteomics in neurodegenerative diseases: Methods for obtaining a closer look at the neuronal proteome. Proteomics Clin Appl 2014; 9:848-71. [DOI: 10.1002/prca.201400030] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 06/25/2014] [Accepted: 09/03/2014] [Indexed: 12/12/2022]
Affiliation(s)
- Sarah Plum
- Medizinisches Proteom-Center; Funktionelle Proteomik; Ruhr-Universität Bochum; Bochum Germany
| | - Simone Steinbach
- Medizinisches Proteom-Center; Medical Proteomics/Bioanalytics; Ruhr-Universität Bochum; Bochum Germany
| | - Laura Abel
- Medizinisches Proteom-Center; Medical Proteomics/Bioanalytics; Ruhr-Universität Bochum; Bochum Germany
| | - Katrin Marcus
- Medizinisches Proteom-Center; Funktionelle Proteomik; Ruhr-Universität Bochum; Bochum Germany
| | - Stefan Helling
- Medizinisches Proteom-Center; Funktionelle Proteomik; Ruhr-Universität Bochum; Bochum Germany
| | - Caroline May
- Medizinisches Proteom-Center; Medical Proteomics/Bioanalytics; Ruhr-Universität Bochum; Bochum Germany
| |
Collapse
|
238
|
Chen F, Sun ZW, Ye LF, Fu GS, Mou Y, Hu SJ. Lycopene protects against apoptosis in hypoxia/reoxygenation‑induced H9C2 myocardioblast cells through increased autophagy. Mol Med Rep 2014; 11:1358-65. [PMID: 25351505 DOI: 10.3892/mmr.2014.2771] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 09/24/2014] [Indexed: 11/06/2022] Open
Abstract
Lycopene (Ly), the most common type of antioxidant in the majority of diet types, provides tolerance to ischemia/reperfusion injury. However, the underlying mechanism of the protective effects observed following Ly administration remains poorly investigated. The aim of the current study was to investigate whether Ly prevents damage to hypoxia/reoxygenation (HR)‑induced H9C2 myocardioblasts in an autophagy‑dependent manner. The levels of autophagic markers were detected using western blotting, the level of apoptosis was detected using western blotting and flow cytometry. The activation of autophagy was impaired via knockdown of the expression of 'microtubule‑associated protein 1‑light chain 3β (MAP1LC3B)' and 'Beclin 1'. After 16 h hypoxia, followed by 2 h reoxygenation, the expression levels of the microtubule‑associated protein 1A/1B‑light chain 3 (LC3) and Βeclin 1 autophagic biomarkers, and cell viability were reduced, whereas the percentage of apoptotic cells, and the expression levels of the Bax/B‑cell lymphoma 2 (Bcl‑2) and active caspase‑3 apoptotic biomarkers were increased. Pre‑incubation of the cells with different Ly concentrations reversed the HR‑induced inhibition of autophagy and cell viability, and the HR‑induced elevation in apoptotic levels. The induction of autophagy was accompanied by reduced apoptosis, and decreased expression levels of Bax/Bcl‑2 and active caspase‑3. In addition, the impairment of autophagy by silencing the expression of MAP1LC3B and Beclin 1 accelerated HR‑induced H9C2 cell apoptosis and the Ly‑mediated protective effects disappeared. Furthermore, Bax/Bcl‑2 and active caspase‑3 expression levels were increased. Moreover, Ly‑induced autophagy was associated with increased adenosine monophosphate kinase (AMPK) phosphorylation. Suppressed AMPK phosphorylation using compound C terminates Ly‑mediated cytoprotective effects. Ly treatment improves cell viability and reduces apoptosis as a result of the activation of the adaptive autophagic response on HR‑induced H9C2 myocardioblasts. AMPK phosphorylation may be involved in the progression.
Collapse
Affiliation(s)
- Fei Chen
- Institution of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Ze-Wei Sun
- Institution of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Li-Fang Ye
- Institution of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Guo-Sheng Fu
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310012, P.R. China
| | - Yun Mou
- Department of Ultrasound, The Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Shen-Jiang Hu
- Institution of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| |
Collapse
|
239
|
S-adenosyl-l-methionine protection of acetaminophen mediated oxidative stress and identification of hepatic 4-hydroxynonenal protein adducts by mass spectrometry. Toxicol Appl Pharmacol 2014; 281:174-84. [PMID: 25246065 DOI: 10.1016/j.taap.2014.08.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 08/21/2014] [Accepted: 08/22/2014] [Indexed: 01/01/2023]
Abstract
Acetaminophen (APAP) hepatotoxicity is protected by S-adenosyl-l-methionine (SAMe) treatment 1hour (h) after APAP in C57/Bl6 mice. This study examined protein carbonylation as well as mitochondrial and cytosolic protein adduction by 4-hydroxynonenal (4-HNE) using mass spectrometry (MS) analysis. Additional studies investigated the leakage of mitochondrial proteins and 4-HNE adduction of these proteins. Male C57/Bl6 mice (n=5/group) were divided into the following groups and treated as indicated: Veh (15ml/kg water, ip), SAMe (1.25mmol/kg, ip), APAP (250mg/kg), and SAMe given 1h after APAP (S+A). APAP toxicity was confirmed by an increase (p<0.05) in plasma ALT (U/l) and liver weight/10g body weight relative to the Veh, SAMe and S+A groups 4h following APAP treatment. SAMe administered 1h post-APAP partially corrected APAP hepatotoxicity as ALT and liver weight/10g body weights were lower in the S+A group compared the APAP group. APAP induced leakage of the mitochondrial protein, carbamoyl phosphate synthase-1 (CPS-1) into the cytosol and which was reduced in the S+A group. SAMe further reduced the extent of APAP mediated 4-HNE adduction of CPS-1. MS analysis of hepatic and mitochondrial subcellular fractions identified proteins from APAP treated mice. Site specific 4-HNE adducts were identified on mitochondrial proteins sarcosine dehydrogenase and carbamoyl phosphate synthase-1 (CPS-1). In summary, APAP is associated with 4-HNE adduction of proteins as identified by MS analysis and that CPS-1 leakage was greater in APAP treated mice. SAMe reduced the extent of 4-HNE adduction of proteins as well as leakage of CPS-1.
Collapse
|
240
|
Allison DB, Antoine LH, Ballinger SW, Bamman MM, Biga P, Darley-Usmar VM, Fisher G, Gohlke JM, Halade GV, Hartman JL, Hunter GR, Messina JL, Nagy TR, Plaisance EP, Powell ML, Roth KA, Sandel MW, Schwartz TS, Smith DL, Sweatt JD, Tollefsbol TO, Watts SA, Yang Y, Zhang J, Austad SN. Aging and energetics' 'Top 40' future research opportunities 2010-2013. F1000Res 2014; 3:219. [PMID: 25324965 PMCID: PMC4197746 DOI: 10.12688/f1000research.5212.1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/08/2014] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND As part of a coordinated effort to expand our research activity at the interface of Aging and Energetics a team of investigators at The University of Alabama at Birmingham systematically assayed and catalogued the top research priorities identified in leading publications in that domain, believing the result would be useful to the scientific community at large. OBJECTIVE To identify research priorities and opportunities in the domain of aging and energetics as advocated in the 40 most cited papers related to aging and energetics in the last 4 years. DESIGN The investigators conducted a search for papers on aging and energetics in Scopus, ranked the resulting papers by number of times they were cited, and selected the ten most-cited papers in each of the four years that include 2010 to 2013, inclusive. RESULTS Ten research categories were identified from the 40 papers. These included: (1) Calorie restriction (CR) longevity response, (2) role of mTOR (mechanistic target of Rapamycin) and related factors in lifespan extension, (3) nutrient effects beyond energy (especially resveratrol, omega-3 fatty acids, and selected amino acids), 4) autophagy and increased longevity and health, (5) aging-associated predictors of chronic disease, (6) use and effects of mesenchymal stem cells (MSCs), (7) telomeres relative to aging and energetics, (8) accretion and effects of body fat, (9) the aging heart, and (10) mitochondria, reactive oxygen species, and cellular energetics. CONCLUSION The field is rich with exciting opportunities to build upon our existing knowledge about the relations among aspects of aging and aspects of energetics and to better understand the mechanisms which connect them.
Collapse
Affiliation(s)
- David B. Allison
- Office of Energetics, University of Alabama at Birmingham, Birmingham, USA
- School of Public Health, University of Alabama at Birmingham, Birmingham, USA
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Comprehensive Center for Healthy Aging, University of Alabama at Birmingham, Birmingham, USA
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, USA
| | - Lisa H. Antoine
- Office of Energetics, University of Alabama at Birmingham, Birmingham, USA
- School of Engineering, University of Alabama at Birmingham, Birmingham, USA
- Department of Environmental Health Sciences, University of Alabama at Birmingham, Birmingham, USA
| | - Scott W. Ballinger
- Department of Pathology, University of Alabama at Birmingham, Birmingham, USA
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, USA
| | - Marcas M. Bamman
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Cell, Developmental, & Integrative Biology, University of Alabama at Birmingham, Birmingham, USA
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, USA
- Birmingham VA Medical Center, Birmingham, USA
| | - Peggy Biga
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Biology, University of Alabama at Birmingham, Birmingham, USA
| | - Victor M. Darley-Usmar
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Pathology, University of Alabama at Birmingham, Birmingham, USA
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, USA
| | - Gordon Fisher
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Human Studies, University of Alabama at Birmingham, Birmingham, USA
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, USA
| | - Julia M. Gohlke
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Environmental Health Sciences, University of Alabama at Birmingham, Birmingham, USA
| | - Ganesh V. Halade
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Medicine – Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, USA
| | - John L. Hartman
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Genetics, University of Alabama at Birmingham, Birmingham, USA
| | - Gary R. Hunter
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Human Studies, University of Alabama at Birmingham, Birmingham, USA
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, USA
| | - Joseph L. Messina
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Pathology, University of Alabama at Birmingham, Birmingham, USA
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, USA
- Birmingham VA Medical Center, Birmingham, USA
| | - Tim R. Nagy
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, USA
- Comprehensive Center for Healthy Aging, University of Alabama at Birmingham, Birmingham, USA
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, USA
| | - Eric P. Plaisance
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Human Studies, University of Alabama at Birmingham, Birmingham, USA
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, USA
| | - Mickie L. Powell
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Biology, University of Alabama at Birmingham, Birmingham, USA
| | - Kevin A. Roth
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Biology, University of Alabama at Birmingham, Birmingham, USA
| | - Michael W. Sandel
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, USA
| | - Tonia S. Schwartz
- School of Public Health, University of Alabama at Birmingham, Birmingham, USA
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
| | - Daniel L. Smith
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Comprehensive Center for Healthy Aging, University of Alabama at Birmingham, Birmingham, USA
| | - J. David Sweatt
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, USA
| | - Trygve O. Tollefsbol
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Biology, University of Alabama at Birmingham, Birmingham, USA
| | - Stephen A. Watts
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Biology, University of Alabama at Birmingham, Birmingham, USA
| | - Yongbin Yang
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, USA
| | - Jianhua Zhang
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, USA
| | - Steven N. Austad
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Biology, University of Alabama at Birmingham, Birmingham, USA
- Comprehensive Center for Healthy Aging, University of Alabama at Birmingham, Birmingham, USA
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, USA
| |
Collapse
|
241
|
Abstract
Endothelial cell dysfunction is the hallmark of every cardiovascular disease/condition, including atherosclerosis and ischemia/reperfusion injury. Fluid shear stress acting on the vascular endothelium is known to regulate cell homeostasis. Altered hemodynamics is thought to play a causative role in endothelial dysfunction. The dysfunction is associated with/preceded by mitochondrial oxidative stress. Studies by our group and others have shown that the form and/or function of the mitochondrial network are affected when endothelial cells are exposed to shear stress in the absence or presence of additional physicochemical stimuli. The present review will summarize the current knowledge on the interconnections among intracellular Ca2+ - nitric oxide - mitochondrial reactive oxygen species, mitochondrial fusion/fission, autophagy/mitophagy, and cell apoptosis vs. survival. More specifically, it will list the evidence on potential regulation of the above intracellular species and processes by the fluid shear stress acting on the endothelium under either physiological flow conditions or during reperfusion (following a period of ischemia). Understanding how the local hemodynamics affects mitochondrial physiology and the cell redox state may lead to development of novel therapeutic strategies for prevention or treatment of the endothelial dysfunction and, hence, of cardiovascular disease.
Collapse
|
242
|
Giordano S, Dodson M, Ravi S, Redmann M, Ouyang X, Darley Usmar VM, Zhang J. Bioenergetic adaptation in response to autophagy regulators during rotenone exposure. J Neurochem 2014; 131:625-33. [PMID: 25081478 DOI: 10.1111/jnc.12844] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 07/27/2014] [Accepted: 07/28/2014] [Indexed: 12/12/2022]
Abstract
Parkinson's disease is the second most common neurodegenerative disorder with both mitochondrial dysfunction and insufficient autophagy playing a key role in its pathogenesis. Among the risk factors, exposure to the environmental neurotoxin rotenone increases the probability of developing Parkinson's disease. We previously reported that in differentiated SH-SY5Y cells, rotenone-induced cell death is directly related to inhibition of mitochondrial function. How rotenone at nM concentrations inhibits mitochondrial function, and whether it can engage the autophagy pathway necessary to remove damaged proteins and organelles, is unknown. We tested the hypothesis that autophagy plays a protective role against rotenone toxicity in primary neurons. We found that rotenone (10-100 nM) immediately inhibited cellular bioenergetics. Concentrations that decreased mitochondrial function at 2 h, caused cell death at 24 h with an LD50 of 10 nM. Overall, autophagic flux was decreased by 10 nM rotenone at both 2 and 24 h, but surprisingly mitophagy, or autophagy of the mitochondria, was increased at 24 h, suggesting that a mitochondrial-specific lysosomal degradation pathway may be activated. Up-regulation of autophagy by rapamycin protected against cell death while inhibition of autophagy by 3-methyladenine exacerbated cell death. Interestingly, while 3-methyladenine exacerbated the rotenone-dependent effects on bioenergetics, rapamycin did not prevent rotenone-induced mitochondrial dysfunction, but caused reprogramming of mitochondrial substrate usage associated with both complex I and complex II activities. Taken together, these data demonstrate that autophagy can play a protective role in primary neuron survival in response to rotenone; moreover, surviving neurons exhibit bioenergetic adaptations to this metabolic stressor.
Collapse
Affiliation(s)
- Samantha Giordano
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | | | | | | | | | | |
Collapse
|
243
|
Ursolic Acid-Regulated Energy Metabolism-Reliever or Propeller of Ultraviolet-Induced Oxidative Stress and DNA Damage? Proteomes 2014; 2:399-425. [PMID: 28250388 PMCID: PMC5302752 DOI: 10.3390/proteomes2030399] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Revised: 06/12/2014] [Accepted: 07/29/2014] [Indexed: 01/27/2023] Open
Abstract
Ultraviolet (UV) light is a leading cause of diseases, such as skin cancers and cataracts. A main process mediating UV-induced pathogenesis is the production of reactive oxygen species (ROS). Excessive ROS levels induce the formation of DNA adducts (e.g., pyrimidine dimers) and result in stalled DNA replication forks. In addition, ROS promotes phosphorylation of tyrosine kinase-coupled hormone receptors and alters downstream energy metabolism. With respect to the risk of UV-induced photocarcinogenesis and photodamage, the antitumoral and antioxidant functions of natural compounds become important for reducing UV-induced adverse effects. One important question in the field is what determines the differential sensitivity of various types of cells to UV light and how exogenous molecules, such as phytochemicals, protect normal cells from UV-inflicted damage while potentiating tumor cell death, presumably via interaction with intracellular target molecules and signaling pathways. Several endogenous molecules have emerged as possible players mediating UV-triggered DNA damage responses. Specifically, UV activates the PIKK (phosphatidylinositol 3-kinase-related kinase) family members, which include DNA-PKcs, ATM (ataxia telangiectasia mutated) and mTOR (mammalian target of rapamycin), whose signaling can be affected by energy metabolism; however, it remains unclear to what extent the activation of hormone receptors regulates PIKKs and whether this crosstalk occurs in all types of cells in response to UV. This review focuses on proteomic descriptions of the relationships between cellular photosensitivity and the phenotypic expression of the insulin/insulin-like growth receptor. It covers the cAMP-dependent pathways, which have recently been shown to regulate the DNA repair machinery through interactions with the PIKK family members. Finally, this review provides a strategic illustration of how UV-induced mitogenic activity is modulated by the insulin sensitizer, ursolic acid (UA), which results in the metabolic adaptation of normal cells against UV-induced ROS, and the metabolic switch of tumor cells subject to UV-induced damage. The multifaceted natural compound, UA, specifically inhibits photo-oxidative DNA damage in retinal pigment epithelial cells while enhancing that in skin melanoma. Considering the UA-mediated differential effects on cell bioenergetics, this article reviews the disparities in glucose metabolism between tumor and normal cells, along with (peroxisome proliferator-activated receptor-γ coactivator 1α)-dependent mitochondrial metabolism and redox (reduction-oxidation) control to demonstrate UA-induced synthetic lethality in tumor cells.
Collapse
|
244
|
Redmann M, Dodson M, Boyer-Guittaut M, Darley-Usmar V, Zhang J. Mitophagy mechanisms and role in human diseases. Int J Biochem Cell Biol 2014; 53:127-33. [PMID: 24842106 PMCID: PMC4111979 DOI: 10.1016/j.biocel.2014.05.010] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Revised: 04/10/2014] [Accepted: 05/11/2014] [Indexed: 11/21/2022]
Abstract
Mitophagy is a process of mitochondrial turnover through lysosomal mediated autophagy activities. This review will highlight recent studies that have identified mediators of mitophagy in response to starvation, loss of mitochondrial membrane potential or perturbation of mitochondrial integrity. Furthermore, we will review evidence of mitophagy dysfunction in various human diseases and discuss the potential for therapeutic interventions that target mitophagy processes.
Collapse
Affiliation(s)
- Matthew Redmann
- Center for Free Radical Biology, University of Alabama at Birmingham, USA; Department of Pathology, University of Alabama at Birmingham, USA
| | - Matthew Dodson
- Center for Free Radical Biology, University of Alabama at Birmingham, USA; Department of Pathology, University of Alabama at Birmingham, USA
| | - Michaël Boyer-Guittaut
- Université de Franche-Comté, Laboratoire de Biochimie, EA3922, SFR IBCT FED4234, Sciences et Techniques, 16 route de Gray, 25030 Besançon Cedex, France
| | - Victor Darley-Usmar
- Center for Free Radical Biology, University of Alabama at Birmingham, USA; Department of Pathology, University of Alabama at Birmingham, USA
| | - Jianhua Zhang
- Center for Free Radical Biology, University of Alabama at Birmingham, USA; Department of Pathology, University of Alabama at Birmingham, USA; Department of Veterans Affairs, Birmingham VA Medical Center, AL 35294, USA.
| |
Collapse
|
245
|
Lipid peroxidation: production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014. [PMID: 24999379 DOI: 10.1155/2014/360438,] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Lipid peroxidation can be described generally as a process under which oxidants such as free radicals attack lipids containing carbon-carbon double bond(s), especially polyunsaturated fatty acids (PUFAs). Over the last four decades, an extensive body of literature regarding lipid peroxidation has shown its important role in cell biology and human health. Since the early 1970s, the total published research articles on the topic of lipid peroxidation was 98 (1970-1974) and has been increasing at almost 135-fold, by up to 13165 in last 4 years (2010-2013). New discoveries about the involvement in cellular physiology and pathology, as well as the control of lipid peroxidation, continue to emerge every day. Given the enormity of this field, this review focuses on biochemical concepts of lipid peroxidation, production, metabolism, and signaling mechanisms of two main omega-6 fatty acids lipid peroxidation products: malondialdehyde (MDA) and, in particular, 4-hydroxy-2-nonenal (4-HNE), summarizing not only its physiological and protective function as signaling molecule stimulating gene expression and cell survival, but also its cytotoxic role inhibiting gene expression and promoting cell death. Finally, overviews of in vivo mammalian model systems used to study the lipid peroxidation process, and common pathological processes linked to MDA and 4-HNE are shown.
Collapse
|
246
|
Lipid peroxidation: production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014. [PMID: 24999379 DOI: 10.1155/2014/360438]] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Lipid peroxidation can be described generally as a process under which oxidants such as free radicals attack lipids containing carbon-carbon double bond(s), especially polyunsaturated fatty acids (PUFAs). Over the last four decades, an extensive body of literature regarding lipid peroxidation has shown its important role in cell biology and human health. Since the early 1970s, the total published research articles on the topic of lipid peroxidation was 98 (1970-1974) and has been increasing at almost 135-fold, by up to 13165 in last 4 years (2010-2013). New discoveries about the involvement in cellular physiology and pathology, as well as the control of lipid peroxidation, continue to emerge every day. Given the enormity of this field, this review focuses on biochemical concepts of lipid peroxidation, production, metabolism, and signaling mechanisms of two main omega-6 fatty acids lipid peroxidation products: malondialdehyde (MDA) and, in particular, 4-hydroxy-2-nonenal (4-HNE), summarizing not only its physiological and protective function as signaling molecule stimulating gene expression and cell survival, but also its cytotoxic role inhibiting gene expression and promoting cell death. Finally, overviews of in vivo mammalian model systems used to study the lipid peroxidation process, and common pathological processes linked to MDA and 4-HNE are shown.
Collapse
|
247
|
Lipid peroxidation: production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014. [PMID: 24999379 DOI: 10.1155/2014/360438\] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Lipid peroxidation can be described generally as a process under which oxidants such as free radicals attack lipids containing carbon-carbon double bond(s), especially polyunsaturated fatty acids (PUFAs). Over the last four decades, an extensive body of literature regarding lipid peroxidation has shown its important role in cell biology and human health. Since the early 1970s, the total published research articles on the topic of lipid peroxidation was 98 (1970-1974) and has been increasing at almost 135-fold, by up to 13165 in last 4 years (2010-2013). New discoveries about the involvement in cellular physiology and pathology, as well as the control of lipid peroxidation, continue to emerge every day. Given the enormity of this field, this review focuses on biochemical concepts of lipid peroxidation, production, metabolism, and signaling mechanisms of two main omega-6 fatty acids lipid peroxidation products: malondialdehyde (MDA) and, in particular, 4-hydroxy-2-nonenal (4-HNE), summarizing not only its physiological and protective function as signaling molecule stimulating gene expression and cell survival, but also its cytotoxic role inhibiting gene expression and promoting cell death. Finally, overviews of in vivo mammalian model systems used to study the lipid peroxidation process, and common pathological processes linked to MDA and 4-HNE are shown.
Collapse
|
248
|
Lipid peroxidation: production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014. [PMID: 24999379 DOI: 10.1155/2014/360438;] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Lipid peroxidation can be described generally as a process under which oxidants such as free radicals attack lipids containing carbon-carbon double bond(s), especially polyunsaturated fatty acids (PUFAs). Over the last four decades, an extensive body of literature regarding lipid peroxidation has shown its important role in cell biology and human health. Since the early 1970s, the total published research articles on the topic of lipid peroxidation was 98 (1970-1974) and has been increasing at almost 135-fold, by up to 13165 in last 4 years (2010-2013). New discoveries about the involvement in cellular physiology and pathology, as well as the control of lipid peroxidation, continue to emerge every day. Given the enormity of this field, this review focuses on biochemical concepts of lipid peroxidation, production, metabolism, and signaling mechanisms of two main omega-6 fatty acids lipid peroxidation products: malondialdehyde (MDA) and, in particular, 4-hydroxy-2-nonenal (4-HNE), summarizing not only its physiological and protective function as signaling molecule stimulating gene expression and cell survival, but also its cytotoxic role inhibiting gene expression and promoting cell death. Finally, overviews of in vivo mammalian model systems used to study the lipid peroxidation process, and common pathological processes linked to MDA and 4-HNE are shown.
Collapse
|
249
|
Lipid peroxidation: production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014. [PMID: 24999379 DOI: 10.1155/2014/360438"] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Lipid peroxidation can be described generally as a process under which oxidants such as free radicals attack lipids containing carbon-carbon double bond(s), especially polyunsaturated fatty acids (PUFAs). Over the last four decades, an extensive body of literature regarding lipid peroxidation has shown its important role in cell biology and human health. Since the early 1970s, the total published research articles on the topic of lipid peroxidation was 98 (1970-1974) and has been increasing at almost 135-fold, by up to 13165 in last 4 years (2010-2013). New discoveries about the involvement in cellular physiology and pathology, as well as the control of lipid peroxidation, continue to emerge every day. Given the enormity of this field, this review focuses on biochemical concepts of lipid peroxidation, production, metabolism, and signaling mechanisms of two main omega-6 fatty acids lipid peroxidation products: malondialdehyde (MDA) and, in particular, 4-hydroxy-2-nonenal (4-HNE), summarizing not only its physiological and protective function as signaling molecule stimulating gene expression and cell survival, but also its cytotoxic role inhibiting gene expression and promoting cell death. Finally, overviews of in vivo mammalian model systems used to study the lipid peroxidation process, and common pathological processes linked to MDA and 4-HNE are shown.
Collapse
|
250
|
Lipid peroxidation: production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014. [PMID: 24999379 DOI: 10.1155/2014/360438-- or] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Lipid peroxidation can be described generally as a process under which oxidants such as free radicals attack lipids containing carbon-carbon double bond(s), especially polyunsaturated fatty acids (PUFAs). Over the last four decades, an extensive body of literature regarding lipid peroxidation has shown its important role in cell biology and human health. Since the early 1970s, the total published research articles on the topic of lipid peroxidation was 98 (1970-1974) and has been increasing at almost 135-fold, by up to 13165 in last 4 years (2010-2013). New discoveries about the involvement in cellular physiology and pathology, as well as the control of lipid peroxidation, continue to emerge every day. Given the enormity of this field, this review focuses on biochemical concepts of lipid peroxidation, production, metabolism, and signaling mechanisms of two main omega-6 fatty acids lipid peroxidation products: malondialdehyde (MDA) and, in particular, 4-hydroxy-2-nonenal (4-HNE), summarizing not only its physiological and protective function as signaling molecule stimulating gene expression and cell survival, but also its cytotoxic role inhibiting gene expression and promoting cell death. Finally, overviews of in vivo mammalian model systems used to study the lipid peroxidation process, and common pathological processes linked to MDA and 4-HNE are shown.
Collapse
|