101
|
Sánchez-Álvarez M, Strippoli R, Donadelli M, Bazhin AV, Cordani M. Sestrins as a Therapeutic Bridge between ROS and Autophagy in Cancer. Cancers (Basel) 2019; 11:cancers11101415. [PMID: 31546746 PMCID: PMC6827145 DOI: 10.3390/cancers11101415] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/15/2019] [Accepted: 09/19/2019] [Indexed: 02/07/2023] Open
Abstract
The regulation of Reactive Oxygen Species (ROS) levels and the contribution therein from networks regulating cell metabolism, such as autophagy and the mTOR-dependent nutrient-sensing pathway, constitute major targets for selective therapeutic intervention against several types of tumors, due to their extensive rewiring in cancer cells as compared to healthy cells. Here, we discuss the sestrin family of proteins—homeostatic transducers of oxidative stress, and drivers of antioxidant and metabolic adaptation—as emerging targets for pharmacological intervention. These adaptive regulators lie at the intersection of those two priority nodes of interest in antitumor intervention—ROS control and the regulation of cell metabolism and autophagy—therefore, they hold the potential not only for the development of completely novel compounds, but also for leveraging on synergistic strategies with current options for tumor therapy and classification/stadiation to achieve personalized medicine.
Collapse
Affiliation(s)
- Miguel Sánchez-Álvarez
- Mechanoadaptation & Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC). Madrid 28029, Spain.
| | - Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, Rome 00161, Italy.
- Gene Expression Laboratory, National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, Rome 00161, Italy.
| | - Massimo Donadelli
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona 37134, Italy.
| | - Alexandr V Bazhin
- Department of General, Visceral and Transplantation Surgery, Ludwig-Maximilians University, Munich 81377, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, Munich 80366, Germany.
| | - Marco Cordani
- IMDEA Nanociencia, C/Faraday 9, Ciudad Universitaria de Cantoblanco, Madrid 28049, Spain..
| |
Collapse
|
102
|
Yang HC, Wu YH, Yen WC, Liu HY, Hwang TL, Stern A, Chiu DTY. The Redox Role of G6PD in Cell Growth, Cell Death, and Cancer. Cells 2019; 8:cells8091055. [PMID: 31500396 PMCID: PMC6770671 DOI: 10.3390/cells8091055] [Citation(s) in RCA: 158] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 09/02/2019] [Accepted: 09/07/2019] [Indexed: 02/07/2023] Open
Abstract
The generation of reducing equivalent NADPH via glucose-6-phosphate dehydrogenase (G6PD) is critical for the maintenance of redox homeostasis and reductive biosynthesis in cells. NADPH also plays key roles in cellular processes mediated by redox signaling. Insufficient G6PD activity predisposes cells to growth retardation and demise. Severely lacking G6PD impairs embryonic development and delays organismal growth. Altered G6PD activity is associated with pathophysiology, such as autophagy, insulin resistance, infection, inflammation, as well as diabetes and hypertension. Aberrant activation of G6PD leads to enhanced cell proliferation and adaptation in many types of cancers. The present review aims to update the existing knowledge concerning G6PD and emphasizes how G6PD modulates redox signaling and affects cell survival and demise, particularly in diseases such as cancer. Exploiting G6PD as a potential drug target against cancer is also discussed.
Collapse
Affiliation(s)
- Hung-Chi Yang
- Department of Medical Laboratory Science and Biotechnology, Yuanpei University of Medical Technology, Hsinchu, Taiwan.
| | - Yi-Hsuan Wu
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.
| | - Wei-Chen Yen
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
- Department of Medical Biotechnology and Laboratory Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Hui-Ya Liu
- Department of Medical Biotechnology and Laboratory Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Tsong-Long Hwang
- Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
- Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan.
- Department of Anaesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
- Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City, Taiwan.
- Research Center for Chinese Herbal Medicine, Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.
| | - Arnold Stern
- New York University School of Medicine, New York, NY, USA.
| | - Daniel Tsun-Yee Chiu
- Department of Medical Biotechnology and Laboratory Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
- Research Center for Chinese Herbal Medicine, Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.
- Department of Pediatric Hematology/Oncology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan.
- Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
103
|
Chakraborty S, Datta S, Ghosh S. Induction of autophagy under nitrosative stress: A complex regulatory interplay between SIRT1 and AMPK in MCF7 cells. Cell Signal 2019; 64:109411. [PMID: 31491460 DOI: 10.1016/j.cellsig.2019.109411] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 08/30/2019] [Accepted: 08/31/2019] [Indexed: 12/18/2022]
Abstract
Induction of nitrosative stress has been observed in various cancer types and in tumor environment. However, it is still unclear how cancer cells combat the effect of nitrosative stress. The main targets of nitrosative stress in cells are cellular lipids, proteins and DNA. Autophagy or self-cleaning generates energy for cell survival under stress conditions. In the present study we investigated the role of autophagy under nitrosative stress in MCF7, a breast cancer cell line. Interestingly, we observed induction of autophagy associated with cell death when MCF7 cells were treated with NO donor compound DETA-NONOate for eight hours. While investigating the mode of cell death under nitrosative stress in MCF7 cells, it was found that it was neither apoptotic nor necrotic. Moreover, nitrosative stress did not alter mitochondrial membrane potential and cellular redox status in MCF7 cells. But we observed an increase in NAD+/NADH and a drop in NADH level in MCF7 cells following NO donor treatment. Sirtuins having NAD+ dependent deacetylase activity, play an important role in cell survival mechanisms. So we further checked the status of SIRT1 under nitrosative stress in MCF7 cells. Surprisingly, we observed an induction of SIRT1, phospho-AMPK and p53 in MCF7 cells under nitrosative stress. Interestingly, autophagy markers were down regulated in MCF7 cells upon treatment with nicotinamide, an inhibitor of SIRT1 activity and dorsomorphin, a phospho-AMPK inhibitor when treated separately under nitrosative stress. To further confirm the role of SIRT1 in the induction of autophagy associated cell death, it was knocked down using si-RNA and nitrosative stress was applied. SIRT1 knock down led to increase in MCF7 cell viability along with down regulation of autophagic markers and phospho-AMPK as well as accumulation of acetylated p53. The increase in p53 controlled DRAM1 mRNA expression in MCF7 cells under nitrosative stress further confirmed a complex interplay between p53, SIRT1 and AMPK under nitrosative stress in MCF7 cells. Altogether our work for the first time suggests a complex inter-twined partnership between AMPK, SIRT1 and p53 in regulating autophagy in response to nitrosative stress in MCF7 cells.
Collapse
Affiliation(s)
- Subhamoy Chakraborty
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India
| | - Sampurna Datta
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India
| | - Sanjay Ghosh
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India.
| |
Collapse
|
104
|
Lévy E, El Banna N, Baïlle D, Heneman-Masurel A, Truchet S, Rezaei H, Huang ME, Béringue V, Martin D, Vernis L. Causative Links between Protein Aggregation and Oxidative Stress: A Review. Int J Mol Sci 2019; 20:ijms20163896. [PMID: 31405050 PMCID: PMC6719959 DOI: 10.3390/ijms20163896] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 07/30/2019] [Accepted: 08/01/2019] [Indexed: 01/17/2023] Open
Abstract
Compelling evidence supports a tight link between oxidative stress and protein aggregation processes, which are noticeably involved in the development of proteinopathies, such as Alzheimer’s disease, Parkinson’s disease, and prion disease. The literature is tremendously rich in studies that establish a functional link between both processes, revealing that oxidative stress can be either causative, or consecutive, to protein aggregation. Because oxidative stress monitoring is highly challenging and may often lead to artefactual results, cutting-edge technical tools have been developed recently in the redox field, improving the ability to measure oxidative perturbations in biological systems. This review aims at providing an update of the previously known functional links between oxidative stress and protein aggregation, thereby revisiting the long-established relationship between both processes.
Collapse
Affiliation(s)
- Elise Lévy
- Molecular Virology and Immunology Unit (VIM-UR892), INRA, Université Paris-Saclay, 78352 Jouy-en-Josas, France
- Institut Curie, PSL Research University, CNRS UMR3348, Université Paris-Sud, Université Paris-Saclay, 91400 Orsay, France
| | - Nadine El Banna
- Institut Curie, PSL Research University, CNRS UMR3348, Université Paris-Sud, Université Paris-Saclay, 91400 Orsay, France
| | - Dorothée Baïlle
- Institut Curie, PSL Research University, CNRS UMR3348, Université Paris-Sud, Université Paris-Saclay, 91400 Orsay, France
| | - Amélie Heneman-Masurel
- Institut Curie, PSL Research University, CNRS UMR3348, Université Paris-Sud, Université Paris-Saclay, 91400 Orsay, France
| | - Sandrine Truchet
- Molecular Virology and Immunology Unit (VIM-UR892), INRA, Université Paris-Saclay, 78352 Jouy-en-Josas, France
| | - Human Rezaei
- Molecular Virology and Immunology Unit (VIM-UR892), INRA, Université Paris-Saclay, 78352 Jouy-en-Josas, France
| | - Meng-Er Huang
- Institut Curie, PSL Research University, CNRS UMR3348, Université Paris-Sud, Université Paris-Saclay, 91400 Orsay, France
| | - Vincent Béringue
- Molecular Virology and Immunology Unit (VIM-UR892), INRA, Université Paris-Saclay, 78352 Jouy-en-Josas, France
| | - Davy Martin
- Molecular Virology and Immunology Unit (VIM-UR892), INRA, Université Paris-Saclay, 78352 Jouy-en-Josas, France.
| | - Laurence Vernis
- Institut Curie, PSL Research University, CNRS UMR3348, Université Paris-Sud, Université Paris-Saclay, 91400 Orsay, France.
| |
Collapse
|
105
|
Michaletti A, Mancini M, Smirnov A, Candi E, Melino G, Zolla L. Multi-omics profiling of calcium-induced human keratinocytes differentiation reveals modulation of unfolded protein response signaling pathways. Cell Cycle 2019; 18:2124-2140. [PMID: 31291818 DOI: 10.1080/15384101.2019.1642066] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
By proteomic, metabolomic and transcriptomic approaches we shed light on the molecular mechanism by which human keratinocytes undergo to terminal differentiation upon in vitro calcium treatment. Proteomic analysis revealed a selective induction of the ribosomal proteins RSSA, an inhibitor of cell proliferation and inducer of differentiation, HSP 60, a protein folding chaperone and GRP78, an unfolding protein response signal. Additionally, we observed an induction of EF1D, a transcription factor for genes that contain heat-shock responsive elements. Conversely, RAD23, a protein involved in regulating ER-associated protein degradation was down-regulated. All these modifications indicated an ER stress response, which in turn activated the unfolded protein response signaling pathway through ATF4, as confirmed both by the modulation of amino acids metabolism genes, such as XBP1, PDI and GPR78, and by the metabolomic analysis. Finally, we detected a reduction of PDI protein, as confirmed by the increase of oxidized glutathione. Metabolome analysis indicated that glycolysis failed to fuel the Krebs cycle, which continued to decrease during differentiation, at glance with the PPP pathway, allowing NADH production and glutathione reduction. Since unfolded protein response is linked to keratinization, these results may be useful for studying pathological mechanisms as well as potential treatments for different pathological conditions. Abbreviation: UPR, unfolded protein response; HEK, human epidermal keratinocytes; HKGS, human keratinocytes growth factor.
Collapse
Affiliation(s)
- Anna Michaletti
- a Department of Ecological and Biological Sciences (DEB), University of Tuscia , Viterbo , Italy
| | - Mara Mancini
- b Biochemistry Laboratory, Istituto Dermopatico dell'Immacolata, IDI-IRCCS , Rome , Italy
| | - Artem Smirnov
- c Department of Experimental Medicine, University of Rome "Tor Vergata" , Rome , Italy
| | - Eleonora Candi
- b Biochemistry Laboratory, Istituto Dermopatico dell'Immacolata, IDI-IRCCS , Rome , Italy.,c Department of Experimental Medicine, University of Rome "Tor Vergata" , Rome , Italy
| | - Gerry Melino
- c Department of Experimental Medicine, University of Rome "Tor Vergata" , Rome , Italy.,d MRC Toxicology Unit, Cambridge University , Leicester , UK
| | - Lello Zolla
- e Agriculture and Forest Sciences (DAFNE), University of Tuscia , Viterbo , Italy
| |
Collapse
|
106
|
Liu X, Wang M, Jiang T, He J, Fu X, Xu Y. IDO1 Maintains Pluripotency of Primed Human Embryonic Stem Cells by Promoting Glycolysis. Stem Cells 2019; 37:1158-1165. [PMID: 31145821 DOI: 10.1002/stem.3044] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 04/22/2019] [Accepted: 05/14/2019] [Indexed: 12/26/2022]
Abstract
Human embryonic stem cells (hESCs) depend on glycolysis for energy supply and pluripotency and switch to oxidative phosphorylation upon differentiation. The underlying mechanisms remain unclear. Here, we demonstrate that indoleamine 2,3-dioxygenase 1 (IDO1) is expressed in primed hESCs and its expression rapidly downregulated upon hESC differentiation. IDO1 is required to maintain pluripotency by suppressing mitochondria activity and promoting glycolysis through the increase of NAD+ /NADH ratio. The upregulation of IDO1 during hESC differentiation suppresses the differentiation of hESCs into certain lineages of cells such as cardiomyocytes, which depend on oxidative phosphorylation to satisfy their high energy demand. Therefore, IDO1 plays important roles in maintaining the pluripotency of hESCs. Stem Cells 2019;37:1158-1165.
Collapse
Affiliation(s)
- Xin Liu
- Center for Regenerative and Translational Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China.,Division of Biological Sciences, University of California, San Diego, La Jolla, California, USA
| | - Meiyan Wang
- Division of Biological Sciences, University of California, San Diego, La Jolla, California, USA
| | - Tao Jiang
- Division of Biological Sciences, University of California, San Diego, La Jolla, California, USA.,The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, People's Republic of China
| | - Jingjin He
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, People's Republic of China
| | - Xuemei Fu
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, People's Republic of China
| | - Yang Xu
- Center for Regenerative and Translational Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China.,Division of Biological Sciences, University of California, San Diego, La Jolla, California, USA.,The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, People's Republic of China
| |
Collapse
|
107
|
Wu D, Zhang K, Hu P. The Role of Autophagy in Acute Myocardial Infarction. Front Pharmacol 2019; 10:551. [PMID: 31214022 PMCID: PMC6554699 DOI: 10.3389/fphar.2019.00551] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 05/01/2019] [Indexed: 12/14/2022] Open
Abstract
Acute myocardial infarction refers to a sudden death of cardiomyocytes, which leads to a large mortality worldwide. To attenuate acute myocardial infarction, strategies should be made to increase cardiomyocyte survival, improve postinfarcted cardiac function, and reverse the process of cardiac remodeling. Autophagy, a pivotal cellular response, has been widely studied and is known to be involved in various kinds of diseases. In the recent few years, the role of autophagy in diseases has been drawn increasing attention to by researchers. Here in this review, we mainly focus on the discussion of the effect of autophagy on the pathogenesis and progression of acute myocardial infarction under ischemic and ischemia/reperfusion injuries. Furthermore, several popular therapeutic agents and strategies taking advantage of autophagy will be described.
Collapse
Affiliation(s)
- Du Wu
- Department of Internal Medicine, The WuYun Mountain Sanatorium of Hangzhou, Hangzhou, China
| | - Kangfeng Zhang
- Department of Internal Medicine, The WuYun Mountain Sanatorium of Hangzhou, Hangzhou, China
| | - Pengfei Hu
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
108
|
Raffaele M, Pittalà V, Zingales V, Barbagallo I, Salerno L, Li Volti G, Romeo G, Carota G, Sorrenti V, Vanella L. Heme Oxygenase-1 Inhibition Sensitizes Human Prostate Cancer Cells towards Glucose Deprivation and Metformin-Mediated Cell Death. Int J Mol Sci 2019; 20:ijms20102593. [PMID: 31137785 PMCID: PMC6566853 DOI: 10.3390/ijms20102593] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 05/16/2019] [Accepted: 05/23/2019] [Indexed: 12/11/2022] Open
Abstract
High levels of heme oxygenase (HO)-1 have been frequently reported in different human cancers, playing a major role in drug resistance and regulation of cancer cell redox homeostasis. Metformin (MET), a drug widely used for type 2 diabetes, has recently gained interest for treating several cancers. Recent studies indicated that the anti-proliferative effects of metformin in cancer cells are highly dependent on glucose concentration. The present work was directed to determine whether use of a specific inhibitor of HO-1 activity, alone or in combination with metformin, affected metastatic prostate cancer cell viability under different concentrations of glucose. MTT assay and the xCELLigence system were used to evaluate cell viability and cell proliferation in DU145 human prostate cancer cells. Cell apoptosis and reactive oxygen species were analyzed by flow cytometry. The activity of HO-1 was inhibited using a selective imidazole-based inhibitor; genes associated with antioxidant systems and cell death were evaluated by qRT-PCR. Our study demonstrates that metformin suppressed prostate cancer growth in vitro and increased oxidative stress. Disrupting the antioxidant HO-1 activity, especially under low glucose concentration, could be an attractive approach to potentiate metformin antineoplastic effects and could provide a biochemical basis for developing HO-1-targeting drugs against solid tumors.
Collapse
Affiliation(s)
- Marco Raffaele
- Department of Drug Science, Biochemistry Section, University of Catania, 95125 Catania, Italy.
| | - Valeria Pittalà
- Department of Drug Science, Pharmaceutical Chemistry Section, University of Catania, 95125 Catania, Italy.
| | - Veronica Zingales
- Department of Drug Science, Biochemistry Section, University of Catania, 95125 Catania, Italy.
| | - Ignazio Barbagallo
- Department of Drug Science, Biochemistry Section, University of Catania, 95125 Catania, Italy.
| | - Loredana Salerno
- Department of Drug Science, Pharmaceutical Chemistry Section, University of Catania, 95125 Catania, Italy.
| | - Giovanni Li Volti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95125 Catania, Italy.
| | - Giuseppe Romeo
- Department of Drug Science, Pharmaceutical Chemistry Section, University of Catania, 95125 Catania, Italy.
| | - Giuseppe Carota
- Department of Drug Science, Biochemistry Section, University of Catania, 95125 Catania, Italy.
| | - Valeria Sorrenti
- Department of Drug Science, Biochemistry Section, University of Catania, 95125 Catania, Italy.
| | - Luca Vanella
- Department of Drug Science, Biochemistry Section, University of Catania, 95125 Catania, Italy.
| |
Collapse
|
109
|
Jiang L, Chen Q, Wu M, Ji T, Liu S, Zhu F, Shi D. Short-term high salt intake impairs hepatic mitochondrial bioenergetics and biosynthesis in SIRT3 knockout mice. Free Radic Res 2019; 53:387-396. [PMID: 31044629 DOI: 10.1080/10715762.2019.1580499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
High salt intake (HS) is an important factor in the development of many metabolic diseases. The liver is the metabolic center in the body. However, the effect of short-term HS on the liver mitochondria and its mechanism are still unclear. In this study, we investigated the effects of short-term HS on liver mitochondrial function. We found that HS reduced Sirtuin3 (SIRT3) protein level, increasing protein carbonylation in mice liver. HS intake decreased ATP production, mitochondrial transcription factor A (TFAM), and complex I level. SIRT3 knockout (SKO) mice exhibited similar results with HS-treated wild-type mice but with a less extent of carbonylation and ATP reduction. Our study shows that short-term HS led to increased hepatic oxidative state, impaired mitochondrial biosynthesis, and bioenergetics. HS-treated mice could still maintain hepatic glucose homeostasis by compensatory activation of Adenosine 5'-monophosphate-activated protein kinase (AMPK). However, in HS-treated SKO mice, AMPK was not activated, instead, the glycogen synthase activity increased, which caused an exceptionally increased glycogen accumulation. This study provides evidence that short-term HS intake could cause the early hepatic metabolic changes, highlighting the importance of controlling salt intake especially in those patients with defects in SIRT3. Highlights High salt intake down-regulates SIRT3 protein level and increases oxidation. High salt intake activates AMPK via AMP-dependent pathway. High salt intake impairs energy metabolism. High salt combined with SIRT3 knockout results in glycogen accumulation.
Collapse
Affiliation(s)
- Lihan Jiang
- a Department of Biochemistry and Molecular Biology , School of Basic Medical Sciences, Fudan University , Shanghai , People's Republic of China
| | - Qinghua Chen
- b Department of Food Hygiene and Nutrition , School of Public Health, Shanghai University of Traditionnal Chinese Medicine , Shanghai , People's Republic of China
| | - Meiling Wu
- a Department of Biochemistry and Molecular Biology , School of Basic Medical Sciences, Fudan University , Shanghai , People's Republic of China
| | - Tingting Ji
- c Department of Nephrology, Qingpu Branch , Zhongshan Hospital, Fudan University , Shanghai , People's Republic of China
| | - Shanlin Liu
- d Free Radical Regulation and Application Research Center, Fudan University , Shanghai , People's Republic of China
| | - Fengge Zhu
- e Department of Nephrology , Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases , Beijing , People's Republic of China
| | - Dongyun Shi
- a Department of Biochemistry and Molecular Biology , School of Basic Medical Sciences, Fudan University , Shanghai , People's Republic of China
| |
Collapse
|
110
|
Cordani M, Somoza Á. Targeting autophagy using metallic nanoparticles: a promising strategy for cancer treatment. Cell Mol Life Sci 2019; 76:1215-1242. [PMID: 30483817 PMCID: PMC6420884 DOI: 10.1007/s00018-018-2973-y] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 11/20/2018] [Indexed: 02/08/2023]
Abstract
Despite the extensive genetic and phenotypic variations present in the different tumors, they frequently share common metabolic alterations, such as autophagy. Autophagy is a self-degradative process in response to stresses by which damaged macromolecules and organelles are targeted by autophagic vesicles to lysosomes and then eliminated. It is known that autophagy dysfunctions can promote tumorigenesis and cancer development, but, interestingly, its overstimulation by cytotoxic drugs may also induce cell death and chemosensitivity. For this reason, the possibility to modulate autophagy may represent a valid therapeutic approach to treat different types of cancers and a variety of clinical trials, using autophagy modulators, are currently employed. On the other hand, recent progress in nanotechnology offers plenty of tools to fight cancer with innovative and efficient therapeutic agents by overcoming obstacles usually encountered with traditional drugs. Interestingly, nanomaterials can modulate autophagy and have been exploited as therapeutic agents against cancer. In this article, we summarize the most recent advances in the application of metallic nanostructures as potent modulators of autophagy process through multiple mechanisms, stressing their therapeutic implications in cancer diseases. For this reason, we believe that autophagy modulation with nanoparticle-based strategies would acquire clinical relevance in the near future, as a complementary therapy for the treatment of cancers and other diseases.
Collapse
Affiliation(s)
- Marco Cordani
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), CNB-CSIC-IMDEA Nanociencia Associated Unit "Unidad de Nanobiotecnología", Madrid, Spain.
- Institute for Advanced Studies in Nanoscience (IMDEA Nanociencia), Faraday 9, Office 129, Lab 137 Ciudad Universitaria de Cantoblanco, 28049, Madrid, Spain.
| | - Álvaro Somoza
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), CNB-CSIC-IMDEA Nanociencia Associated Unit "Unidad de Nanobiotecnología", Madrid, Spain.
- Institute for Advanced Studies in Nanoscience (IMDEA Nanociencia), Faraday 9, Office 129, Lab 137 Ciudad Universitaria de Cantoblanco, 28049, Madrid, Spain.
| |
Collapse
|
111
|
Wu JJ, Yuan XM, Huang C, An GY, Liao ZL, Liu GA, Chen RX. Farnesyl thiosalicylic acid prevents iNOS induction triggered by lipopolysaccharide via suppression of iNOS mRNA transcription in murine macrophages. Int Immunopharmacol 2019; 68:218-225. [PMID: 30658315 DOI: 10.1016/j.intimp.2018.12.066] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 12/12/2018] [Accepted: 12/31/2018] [Indexed: 02/06/2023]
Abstract
Inducible nitric oxide synthase (iNOS) is a molecule critical for the development of inflammation-associated disorders. Its induction should be tightly controlled in order to maintain cellular homeostasis. Upon lipopolysaccharide (LPS) stimulation, iNOS, in most settings, is induced by the activation of inhibitor of κB-α (IκB-α)-nuclear factor κB (NF-κB) signaling. Farnesyl thiosalicylic acid (FTS), a synthetic small molecule that is considered to detach Ras from the inner cell membrane, has been shown to exhibit numerous anti-inflammatory functions. However, it remains unclear whether and how it affects iNOS induction in macrophages. The present study addressed this issue in cultured macrophages and endotoxemic mice. Results showed that FTS pretreatment significantly prevented LPS-induced increases in iNOS protein and mRNA expression levels in murine cultured macrophages, which were confirmed in organs in vivo from endotoxemic mice, such as the liver and lung. Mechanistic studies revealed that FTS pretreatment did not affect IκB-α degradation and NF-κB activation in LPS-treated macrophages. The nuclear transport of the active NF-κB was also not affected by FTS. But FTS pretreatment reduced the binding of NF-κB to its DNA elements, and reduced NF-κB bindings to iNOS promoter inside LPS-treated macrophages. Finally, our results showed that FST pretreatment increased mouse survival rate compared to LPS alone treatment. Taken together, these results indicate that FTS attenuates iNOS induction in macrophages likely through inhibition of iNOS mRNA transcription, providing further insight into the molecular mechanism of action of FTS in inflammatory disorder therapy.
Collapse
Affiliation(s)
- Jing-Jing Wu
- Department of Cardiology, Suzhou Kowloon Hospital of Shanghai Jiaotong University School of Medicine, #118 Wansheng Street, Suzhou 215021, Jiangsu, China
| | - Xiao-Mei Yuan
- Heart Failure Center, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Chengdu, 610072, China.
| | - Chao Huang
- Department of Pharmacy, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu Province, China
| | - Guo-Yin An
- Department of Cardiology, Suzhou Kowloon Hospital of Shanghai Jiaotong University School of Medicine, #118 Wansheng Street, Suzhou 215021, Jiangsu, China
| | - Zhan-Ling Liao
- Department of Cardiology, Suzhou Kowloon Hospital of Shanghai Jiaotong University School of Medicine, #118 Wansheng Street, Suzhou 215021, Jiangsu, China
| | - Guang-An Liu
- Department of Cardiology, Suzhou Kowloon Hospital of Shanghai Jiaotong University School of Medicine, #118 Wansheng Street, Suzhou 215021, Jiangsu, China
| | - Run-Xiang Chen
- Department of Cardiology, Suzhou Kowloon Hospital of Shanghai Jiaotong University School of Medicine, #118 Wansheng Street, Suzhou 215021, Jiangsu, China.
| |
Collapse
|
112
|
Abstract
Autophagy is a lysosome-dependent catabolic process. Both extra- and intra-cellular components are engulfed in autophagic vacuoles and degraded to simple molecules, such as monosaccharides, fatty acids and amino acids. Then, these molecules can be further used to produce ATP through catabolic reactions and/or provide building blocks for the synthesis of essential proteins. Therefore, we consider autophagy a critical and fine-tuned process in maintaining energy homeostasis. The complicated relationships between autophagy and energy metabolism have raised broad interest and have been extensively studied. In this chapter, we summarize the relationships enabling autophagy to control or modulate energy metabolism and allowing metabolic pathways to regulate autophagy. Specifically, we review the correlations between autophagy and energy homeostasis in terms of oxidative phosphorylation, reactive oxygen species in mitochondria, glycolysis, metabolism of glycogen and protein, and so on. An understanding of the role of autophagy in energy homeostasis could help us better appreciate how autophagy determines cell fate under stressful conditions or pathological processes.
Collapse
|
113
|
Wang K, Liu C, Hou Y, Zhou H, Wang X, Mai K, He G. Differential Apoptotic and Mitogenic Effects of Lectins in Zebrafish. Front Endocrinol (Lausanne) 2019; 10:356. [PMID: 31231312 PMCID: PMC6560201 DOI: 10.3389/fendo.2019.00356] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 05/20/2019] [Indexed: 01/06/2023] Open
Abstract
Plant lectins represent a major group of anti-nutritional factors that can be toxic to human and animals. However, the mechanisms by which lectins regulate cell fates are not well-understood. In the present study, the cellular and molecular impacts of three common lectins, agglutinins from wheat germ [wheat germ agglutinin (WGA)], soybean [soybean agglutinin (SBA)], and peanut [peanut agglutinin (PNA)] were examined in zebrafish embryo and liver cells. WGA and SBA were found to induce cell apoptosis both in vitro and in vivo, while PNA stimulated cell proliferation. WGA and SBA reduced levels of B cell lymphoma-2 (Bcl-2), phosphorylation of Bcl-2-associated death promoter (Bad), cyclin-dependent kinase 4 (Cdk4), and phosphorylation of the retinoblastoma (Rb). WGA and SBA also inhibited the activities of cell survival pathways including protein kinase B (Akt), extracellular signal-regulated protein kinases 1 and 2 (Erk1/2), and target of rapamycin (Tor). Furthermore, WGA and SBA shifted the cellular metabolism characterized by reduced expression of glucose-6-phosphate dehydrogenase (g6pd) and increased expression of glutamine synthetase (glul) and glutamate dehydrogenase (glud). However, PNA showed the opposite effects toward these molecular markers compared to those of WGA and SBA. Therefore, our results revealed some plant lectins (WGA and SBA) were toxic while the other (PNA) was mitogenic. Further characterization of the distinct functions of individual lectins should be valuable for both nutrition and other potential applications.
Collapse
Affiliation(s)
- Kaidi Wang
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Chengdong Liu
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Yiying Hou
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Huihui Zhou
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Xuan Wang
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Kangsen Mai
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Gen He
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
- *Correspondence: Gen He
| |
Collapse
|
114
|
Morsi M, Kobeissy F, Magdeldin S, Maher A, Aboelmagd O, Johar D, Bernstein L. A shared comparison of diabetes mellitus and neurodegenerative disorders. J Cell Biochem 2018; 120:14318-14325. [PMID: 30565720 DOI: 10.1002/jcb.28094] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 10/29/2018] [Indexed: 11/09/2022]
Abstract
Diabetes mellitus (DM), one of the most prevalent metabolic diseases in the world population, is associated with a number of comorbid conditions including obesity, pancreatic endocrine changes, and renal and cardio-cerebrovascular alterations, coupled with peripheral neuropathy and neurodegenerative disease, some of these disorders are bundled into metabolic syndrome. Type 1 DM (T1DM) is an autoimmune disease that destroys the insulin-secreting islet cells. Type 2 DM (T2DM) is diabetes that is associated with an imbalance in the glucagon/insulin homeostasis that leads to the formation of amyloid deposits in the brain, pancreatic islet cells, and possibly in the kidney glomerulus. There are several layers of molecular pathologic alterations that contribute to the DM metabolic pathophysiology and its associated neuropathic manifestations. In this review, we describe the general signature metabolic features of DM and the cross-talk with neurodegeneration. We will assess the underlying molecular key players associated with DM-induced neuropathic disorders that are associated with both T1DM and T2DM. In this context, we will highlight the role of tau and amyloid protein deposits in the brain as well in the pancreatic islet cells, and possibly in the kidney glomerulus. Furthermore, we will discuss the central role of mitochondria, oxidative stress, and the unfolded protein response in mediating the DM-associated neuropathic degeneration. This study will elucidate the relationship between DM and neurodegeneration which may account for the evolution of other neurodegenerative diseases, particularly Alzheimer's disease and Parkinson's disease as discussed later.
Collapse
Affiliation(s)
- Mahmoud Morsi
- Faculty of Medicine, Menoufia University, Shebin El-kom, Egypt
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Sameh Magdeldin
- Proteomics and Metabolomics Unit, Basic Research, Children's Cancer Hospital, Cairo, Egypt.,Physiology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Ahmed Maher
- Zoonotic Diseases Department, National Research Center, Dokki, Egypt
| | | | - Dina Johar
- Department of Biochemistry and Nutrition, Faculty of Women for Arts, Sciences and Education, Ain Shams University, Cairo, Egypt.,Department of Physiology and Pathophysiology, Faculty of Health Sciences, Rady College of Medicine, Max Rady University of Manitoba, Winnipeg, Canada
| | | |
Collapse
|
115
|
Bildirici I, Schaiff WT, Chen B, Morizane M, Oh SY, O’Brien M, Sonnenberg-Hirche C, Chu T, Barak Y, Nelson DM, Sadovsky Y. PLIN2 Is Essential for Trophoblastic Lipid Droplet Accumulation and Cell Survival During Hypoxia. Endocrinology 2018; 159:3937-3949. [PMID: 30351430 PMCID: PMC6240902 DOI: 10.1210/en.2018-00752] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 10/17/2018] [Indexed: 12/12/2022]
Abstract
Trophoblast hypoxia and injury, key components of placental dysfunction, are associated with fetal growth restriction and other complications of pregnancy. Accumulation of lipid droplets has been found in hypoxic nonplacental cells. Unique to pregnancy, lipid accumulation in the placenta might perturb lipid transport to the fetus. We tested the hypothesis that hypoxia leads to accumulation of lipid droplets in human trophoblasts and that trophoblastic PLIN proteins play a key role in this process. We found that hypoxia promotes the accumulation of lipid droplets in primary human trophoblasts. A similar accretion of lipid droplets was found in placental villi in vivo from pregnancies complicated by fetal growth restriction. In both situations, these changes were associated with an increased level of cellular triglycerides. Exposure of trophoblasts to hypoxia led to reduced fatty acid efflux and oxidation with no change in fatty acid uptake or synthesis. We further found that hypoxia markedly stimulated PLIN2 mRNA synthesis and protein expression, which colocalized to lipid droplets. Knockdown of PLIN2, but not PLIN3, enhanced trophoblast apoptotic death, and overexpression of PLIN2 promoted cell viability. Collectively, our data indicate that hypoxia enhances trophoblastic lipid retention in the form of lipid droplets and that PLIN2 plays a key role in this process and in trophoblast defense against apoptotic death. These findings also imply that this protective mechanism may lead to diminished trafficking of lipids to the developing fetus.
Collapse
Affiliation(s)
- Ibrahim Bildirici
- Department of Obstetrics and Gynecology, Washington University, St. Louis, Missouri
| | - W Timothy Schaiff
- Department of Obstetrics and Gynecology, Washington University, St. Louis, Missouri
| | - Baosheng Chen
- Department of Obstetrics and Gynecology, Washington University, St. Louis, Missouri
| | - Mayumi Morizane
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Soo-Young Oh
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Matthew O’Brien
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Tianjiao Chu
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yaacov Barak
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - D Michael Nelson
- Department of Obstetrics and Gynecology, Washington University, St. Louis, Missouri
| | - Yoel Sadovsky
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania
- Correspondence: Yoel Sadovsky, MD, Magee-Womens Research Institute, 204 Craft Avenue, Pittsburgh, Pennsylvania 15213. E-mail:
| |
Collapse
|
116
|
López-Grueso MJ, González-Ojeda R, Requejo-Aguilar R, McDonagh B, Fuentes-Almagro CA, Muntané J, Bárcena JA, Padilla CA. Thioredoxin and glutaredoxin regulate metabolism through different multiplex thiol switches. Redox Biol 2018; 21:101049. [PMID: 30639960 PMCID: PMC6327914 DOI: 10.1016/j.redox.2018.11.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 11/08/2018] [Accepted: 11/11/2018] [Indexed: 12/19/2022] Open
Abstract
The aim of the present study was to define the role of Trx and Grx on metabolic thiol redox regulation and identify their protein and metabolite targets. The hepatocarcinoma-derived HepG2 cell line under both normal and oxidative/nitrosative conditions by overexpression of NO synthase (NOS3) was used as experimental model. Grx1 or Trx1 silencing caused conspicuous changes in the redox proteome reflected by significant changes in the reduced/oxidized ratios of specific Cys's including several glycolytic enzymes. Cys91 of peroxiredoxin-6 (PRDX6) and Cys153 of phosphoglycerate mutase-1 (PGAM1), that are known to be involved in progression of tumor growth, are reported here for the first time as specific targets of Grx1. A group of proteins increased their CysRED/CysOX ratio upon Trx1 and/or Grx1 silencing, including caspase-3 Cys163, glyceraldehyde-3-phosphate dehydrogenase (GAPDH) Cys247 and triose-phosphate isomerase (TPI) Cys255 likely by enhancement of NOS3 auto-oxidation. The activities of several glycolytic enzymes were also significantly affected. Glycolysis metabolic flux increased upon Trx1 silencing, whereas silencing of Grx1 had the opposite effect. Diversion of metabolic fluxes toward synthesis of fatty acids and phospholipids was observed in siRNA-Grx1 treated cells, while siRNA-Trx1 treated cells showed elevated levels of various sphingomyelins and ceramides and signs of increased protein degradation. Glutathione synthesis was stimulated by both treatments. These data indicate that Trx and Grx have both, common and specific protein Cys redox targets and that down regulation of either redoxin has markedly different metabolic outcomes. They reflect the delicate sensitivity of redox equilibrium to changes in any of the elements involved and the difficulty of forecasting metabolic responses to redox environmental changes. Trx1 and Grx1 Cys redox targets are abundant among Glycolytic enzymes. PRDX6-Cys91 and PGAM-Cys153 are specific targets of Grx1. Down regulation of thioredoxin and glutaredoxin have different metabolic outcomes. Glutathione synthesis and membrane lipid composition are sensitive to Trx1 and Grx1 down regulation. Redoxins down regulation also induce target Cys reductive changes under NOS3 overexpression.
Collapse
Affiliation(s)
- M J López-Grueso
- Dept. Biochemistry and Molecular Biology, University of Córdoba, Córdoba, Spain; Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain
| | - R González-Ojeda
- Institute of Biomedicine of Seville (IBIS), IBiS/"Virgen del Rocío" University Hospital/CSIC/University of Seville, Seville, Spain
| | - R Requejo-Aguilar
- Dept. Biochemistry and Molecular Biology, University of Córdoba, Córdoba, Spain; Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain
| | - B McDonagh
- Dept. of Physiology, School of Medicine, NUI Galway, Ireland
| | | | - J Muntané
- Dept. of Physiology, School of Medicine, NUI Galway, Ireland
| | - J A Bárcena
- Dept. Biochemistry and Molecular Biology, University of Córdoba, Córdoba, Spain; Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain.
| | - C A Padilla
- Dept. Biochemistry and Molecular Biology, University of Córdoba, Córdoba, Spain; Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain
| |
Collapse
|
117
|
Nitric oxide mediated redox regulation of protein homeostasis. Cell Signal 2018; 53:348-356. [PMID: 30408515 DOI: 10.1016/j.cellsig.2018.10.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 10/27/2018] [Accepted: 10/29/2018] [Indexed: 12/12/2022]
Abstract
Nitric oxide is a versatile diffusible signaling molecule, whose biosynthesis by three NO synthases (NOS) is tightly regulated at transcriptional and posttranslational levels, availability of co-factors, and calcium binding. Above normal levels of NO have beneficial protective effects for example in the cardiovascular system, but also contribute to the pathophysiology in the context of inflammatory diseases, and to aging and neurodegeneration in the nervous system. The effect specificity relies on the functional and spatial specificity of the NOS isoenzymes, and on the duality of two major signaling mechanisms (i) activation of soluble guanylycylase (sGC)-dependent cGMP production and (ii) direct S-nitrosylation of redox sensitive cysteines of susceptible proteins. The present review summarizes the functional implications of S-nitrosylation in the context of proteostasis, and focuses on two NO target proteins, heat shock cognate of 70 kDa (Hsc70/HSPA8) and the ubiquitin 2 ligase (UBE2D), because both are modified on functionally critical cysteines and are key regulators of chaperone mediated and assisted autophagy and proteasomal protein degradation. SNO modifications of these candidates are associated with protein accumulations and adoption of a senescent phenotype of neuronal cells suggesting that S-nitrosylations of protein homeostatic machineries contribute to aging phenomena.
Collapse
|
118
|
Rungratanawanich W, Memo M, Uberti D. Redox Homeostasis and Natural Dietary Compounds: Focusing on Antioxidants of Rice ( Oryza sativa L.). Nutrients 2018; 10:nu10111605. [PMID: 30388764 PMCID: PMC6265930 DOI: 10.3390/nu10111605] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 10/25/2018] [Accepted: 10/26/2018] [Indexed: 12/11/2022] Open
Abstract
Redox homeostasis may be defined as the dynamic equilibrium between electrophiles and nucleophiles to maintain the optimum redox steady state. This mechanism involves complex reactions, including nuclear factor erythroid 2-related factor 2 (Nrf2) pathway, activated by oxidative stress in order to restore the redox balance. The ability to maintain the optimal redox homeostasis is fundamental for preserving physiological functions and preventing phenotypic shift toward pathological conditions. Here, we reviewed mechanisms involved in redox homeostasis and how certain natural compounds regulate the nucleophilic tone. In addition, we focused on the antioxidant properties of rice and particularly on its bioactive compound, γ-oryzanol. It is well known that γ-oryzanol exerts a variety of beneficial effects mediated by its antioxidant properties. Recently, γ-oryzanol was also found as a Nrf2 inducer, resulting in nucleophilic tone regulation and making rice a para-hormetic food.
Collapse
Affiliation(s)
- Wiramon Rungratanawanich
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
| | - Maurizio Memo
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
| | - Daniela Uberti
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
| |
Collapse
|
119
|
Ke T, Gonçalves FM, Gonçalves CL, Dos Santos AA, Rocha JBT, Farina M, Skalny A, Tsatsakis A, Bowman AB, Aschner M. Post-translational modifications in MeHg-induced neurotoxicity. Biochim Biophys Acta Mol Basis Dis 2018; 1865:2068-2081. [PMID: 30385410 DOI: 10.1016/j.bbadis.2018.10.024] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 10/16/2018] [Accepted: 10/19/2018] [Indexed: 12/29/2022]
Abstract
Mercury (Hg) exposure remains a major public health concern due to its widespread distribution in the environment. Organic mercurials, such as MeHg, have been extensively investigated especially because of their congenital effects. In this context, studies on the molecular mechanism of MeHg-induced neurotoxicity are pivotal to the understanding of its toxic effects and the development of preventive measures. Post-translational modifications (PTMs) of proteins, such as phosphorylation, ubiquitination, and acetylation are essential for the proper function of proteins and play important roles in the regulation of cellular homeostasis. The rapid and transient nature of many PTMs allows efficient signal transduction in response to stress. This review summarizes the current knowledge of PTMs in MeHg-induced neurotoxicity, including the most commonly PTMs, as well as PTMs induced by oxidative stress and PTMs of antioxidant proteins. Though PTMs represent an important molecular mechanism for maintaining cellular homeostasis and are involved in the neurotoxic effects of MeHg, we are far from understanding the complete picture on their role, and further research is warranted to increase our knowledge of PTMs in MeHg-induced neurotoxicity.
Collapse
Affiliation(s)
- Tao Ke
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States.
| | - Filipe Marques Gonçalves
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Cinara Ludvig Gonçalves
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | | | - João B T Rocha
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, 97105900 Santa Maria, RS, Brazil
| | - Marcelo Farina
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, 88040900 Florianópolis, SC, Brazil
| | - Anatoly Skalny
- Yaroslavl State University, Sovetskaya St., 14, Yaroslavl 150000, Russia; Peoples' Friendship University of Russia (RUDN University), Miklukho-Maklaya St., 6, Moscow 105064, Russia; Orenburg State University, Pobedy Ave., 13, Orenburg 460352, Russia
| | - Aristidis Tsatsakis
- Center of Toxicology Science & Research, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN, United States.
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States.
| |
Collapse
|
120
|
Feidantsis K, Pörtner HO, Vlachonikola E, Antonopoulou E, Michaelidis B. Seasonal Changes in Metabolism and Cellular Stress Phenomena in the Gilthead Sea Bream (Sparus aurata). Physiol Biochem Zool 2018; 91:878-895. [PMID: 29553887 DOI: 10.1086/697170] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Seasonal temperature changes may take organisms to the upper and lower limit of their thermal range, with respective variations in their biochemical and metabolic profile. To elucidate these traits, we investigated metabolic and antioxidant patterns in tissues of sea bream Sparus aurata during seasonal acclimatization for 1 yr in the field. Metabolic patterns were assessed by determining lactate dehydrogenase, citrate synthase, and β-hydroxyacyl CoA dehydrogenase activities, their kinetic properties and plasma levels of glucose, lactate, and triglycerides and tissue succinate levels. Oxidative stress was assessed by determining antioxidant enzymes superoxide dismutase, catalase, and glutathione reductase activities and levels of thiobarbituric acid reactive substances. Xanthine oxidase (XO) activity was determined as another source of reactive oxygen species (ROS) production. Furthermore, we studied the antiapoptotic protein indicator Bcl-2 and the apoptotic protein indicators Bax, Bad, ubiquitin, and caspase as well as indexes of autophagy (LC3B II/LC3B I and SQSTM1/p62) in the liver and the heart to identify possible relationships between oxidative stress and cell death. The results indicate clear seasonal metabolic patterns involving oxidative stress during summer as well as winter. During cold acclimatization, lipid oxidation is induced, while during increased temperatures, warm-induced metabolic activation and carbohydrate oxidation are observed. Thus, oxidative stress seems to be more prominent during warming because of the increased aerobic metabolism. The seasonal profile of apoptosis and XO as another source of ROS matches the results obtained in the laboratory and are interpreted within the framework of oxygen- and capacity-limited thermal tolerance.
Collapse
|
121
|
Nitric oxide contributes to protein homeostasis by S-nitrosylations of the chaperone HSPA8 and the ubiquitin ligase UBE2D. Redox Biol 2018; 20:217-235. [PMID: 30368041 PMCID: PMC6202877 DOI: 10.1016/j.redox.2018.10.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 09/25/2018] [Accepted: 10/02/2018] [Indexed: 02/06/2023] Open
Abstract
Upregulations of neuronal nitric oxide synthase (nNOS) in the rodent brain have been associated with neuronal aging. To address underlying mechanisms we generated SH-SY5Y neuronal cells constitutively expressing nNOS at a level similar to mouse brain (nNOS+ versus MOCK). Initial experiments revealed S-nitrosylations (SNO) of key players of protein homeostasis: heat shock cognate HSC70/HSPA8 within its nucleotide-binding site, and UBE2D ubiquitin conjugating enzymes at the catalytic site cysteine. HSPA8 is involved in protein folding, organelle import/export and chaperone-mediated LAMP2a-dependent autophagy (CMA). A set of deep redox and full proteome analyses, plus analysis of autophagy, CMA and ubiquitination with rapamycin and starvation as stimuli confirmed the initial observations and revealed a substantial increase of SNO modifications in nNOS+ cells, in particular targeting protein networks involved in protein catabolism, ubiquitination, carbohydrate metabolism and cell cycle control. Importantly, NO-independent reversible oxidations similarly occurred in both cell lines. Functionally, nNOS caused an accumulation of proteins, including CMA substrates and loss of LAMP2a. UBE2D activity and proteasome activity were impaired, resulting in dysregulations of cell cycle checkpoint proteins. The observed changes of protein degradation pathways caused an expansion of the cytoplasm, large lysosomes, slowing of the cell cycle and suppression of proliferation suggesting a switch of the phenotype towards aging, supported by downregulations of neuronal progenitor markers but increase of senescence-associated proteins. Hence, upregulation of nNOS in neuronal cells imposes aging by SNOing of key players of ubiquitination, chaperones and of substrate proteins leading to interference with crucial steps of protein homeostasis.
Collapse
|
122
|
Zhou M, Xu W, Wang J, Yan J, Shi Y, Zhang C, Ge W, Wu J, Du P, Chen Y. Boosting mTOR-dependent autophagy via upstream TLR4-MyD88-MAPK signalling and downstream NF-κB pathway quenches intestinal inflammation and oxidative stress injury. EBioMedicine 2018; 35:345-360. [PMID: 30170968 PMCID: PMC6161481 DOI: 10.1016/j.ebiom.2018.08.035] [Citation(s) in RCA: 244] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 08/16/2018] [Accepted: 08/16/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND AIMS Defective autophagy has been proposed as an important event in a growing number of autoimmune and inflammatory diseases such as rheumatoid arthritis and lupus. However, the precise role of mechanistic target of rapamycin (mTOR)-dependent autophagy and its underlying regulatory mechanisms in the intestinal epithelium in response to inflammation and oxidative stress remain poorly understood. METHODS The levels of p-mTOR, LC3B, p62 and autophagy in mice and LPS-treated cells were examined by immunoblotting, immunohistochemistry, confocal microscopy and transmission electron microscopy (TEM). We evaluated the expression of IL-1β, IL-8, TNF-α, MDA, SOD and T-AOC by quantitative real time-polymerase chain reaction (qRT-PCR) and commercially available kits after silencing of mTOR and ATG5. In vivo modulation of mTOR and autophagy was achieved by using AZD8055, rapamycin and 3-methyladenine. Finally, to verify the involvement of TLR4 signalling and the NF-κB pathway in cells and active ulcerative colitis (UC) patients, immunofluorescence, qRT-PCR, immunoblotting and TEM were performed to determine TLR4 signalling relevance to autophagy and inflammation. RESULTS The mTOR-dependent autophagic flux impairment in a murine model of colitis, human intestinal epithelial cells and active UC patients is probably regulated by TLR4-MyD88-MAPK signalling and the NF-κB pathway. Silencing mTOR remarkably attenuated, whereas inhibiting ATG5 aggravated, LPS-induced inflammation and oxidative injury. Pharmacological administration of mTOR inhibitors and autophagy stimulators markedly ameliorated experimental colitis and oxidative stress in vivo. CONCLUSIONS Our findings not only shed light on the regulatory mechanism of mTOR-dependent autophagy, but also provided potential therapeutic targets for intestinal inflammatory diseases such as refractory inflammatory bowel disease.
Collapse
Affiliation(s)
- Mingxia Zhou
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China.
| | - Weimin Xu
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Jiazheng Wang
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China.
| | - Junkai Yan
- Shanghai Institute of Pediatric Research, Shanghai, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China.
| | - Yingying Shi
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China.
| | - Cong Zhang
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China.
| | - Wensong Ge
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Jin Wu
- Shanghai Institute of Pediatric Research, Shanghai, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China.
| | - Peng Du
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Yingwei Chen
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China.
| |
Collapse
|
123
|
Tsai YG, Wen YS, Wang JY, Yang KD, Sun HL, Liou JH, Lin CY. Complement regulatory protein CD46 induces autophagy against oxidative stress-mediated apoptosis in normal and asthmatic airway epithelium. Sci Rep 2018; 8:12973. [PMID: 30154478 PMCID: PMC6113329 DOI: 10.1038/s41598-018-31317-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 06/11/2018] [Indexed: 01/07/2023] Open
Abstract
Autophagy plays a major role in defending against oxidative stress in respiratory epithelial cells. The complement regulatory protein CD46 can enhance autophagy and decrease local complement activation at sites of inflammation. This study investigated the mechanism by which CD46 protects against oxidative stress-mediated apoptosis in respiratory epithelium in asthmatic patients. Nasal mucosa samples were obtained from 60 adults with mild asthma who received turbinectomy and 30 controls. A decreased expression of CD46 and increased apoptosis were noted in the damaged nasal epithelium from the asthmatic patients. Primary epithelial cells cultured with Dermatophagoides pteronyssinus 2 showed decreased CD46 and increased cleaved CASPASE-3A expressions. Crosslinking CD46 mAb could induce the formation of autophagosomes and LC3-II expression in primary epithelial cells. CD46 engagement could induce autophagy against hydrogen peroxide-induced epithelial cell death, whereas the autophagy inhibitor 3-methyladenine decreased this effect. In addition, CD46 engagement decreased the expressions of PRO-IL-1β and NLRP3, enhanced the expression of scaffold protein GOPC, and diminished hydrogen peroxide-induced 8-OHdG, IL-1β and IL-6 production. Silencing ATG5 in human lung epithelial A549 cells decreased CD46-activated autophagy with LC3-II. CD46 induced autophagy and decreased the oxidative stress-mediated apoptosis of respiratory epithelium, and this may offer a new therapeutic strategy to treat asthma.
Collapse
Affiliation(s)
- Yi-Giien Tsai
- Department of Pediatrics, Changhua Christian Children's Hospital, Changhua, Taiwan.,School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yung-Sung Wen
- Department of Otorhinolaryngology, Head and Neck Surgery, Changhua Christian Hospital, Changhua, Taiwan
| | - Jiu-Yao Wang
- Department of Pediatrics, College of Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Kuender D Yang
- Mackay Children's Hospital, and Institute of Biomedical Sciences, Mackay Medical College, Taipei, Taiwan
| | - Hai-Lun Sun
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Jia-Hung Liou
- Department of Pathology, Changhua Christian Hospital, Changhua, Taiwan
| | - Ching-Yuang Lin
- Clinical Immunological Center and College of Medicine, China Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
124
|
Oxidative stress-modulating drugs have preferential anticancer effects - involving the regulation of apoptosis, DNA damage, endoplasmic reticulum stress, autophagy, metabolism, and migration. Semin Cancer Biol 2018; 58:109-117. [PMID: 30149066 DOI: 10.1016/j.semcancer.2018.08.010] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 08/19/2018] [Accepted: 08/23/2018] [Indexed: 02/07/2023]
Abstract
To achieve preferential effects against cancer cells but less damage to normal cells is one of the main challenges of cancer research. In this review, we explore the roles and relationships of oxidative stress-mediated apoptosis, DNA damage, ER stress, autophagy, metabolism, and migration of ROS-modulating anticancer drugs. Understanding preferential anticancer effects in more detail will improve chemotherapeutic approaches that are based on ROS-modulating drugs in cancer treatments.
Collapse
|
125
|
Rekha KR, Inmozhi Sivakamasundari R. Geraniol Protects Against the Protein and Oxidative Stress Induced by Rotenone in an In Vitro Model of Parkinson's Disease. Neurochem Res 2018; 43:1947-1962. [PMID: 30141137 DOI: 10.1007/s11064-018-2617-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 07/21/2018] [Accepted: 08/17/2018] [Indexed: 12/27/2022]
Abstract
Dysfunction of autophagy, mitochondrial dynamics and endoplasmic reticulum (ER) stress are currently considered as major contributing factors in the pathogenesis of Parkinson's disease (PD). Accumulation of oxidatively damaged cytoplasmic organelles and unfolded proteins in the lumen of the ER causes ER stress and it is associated with dopaminergic cell death in PD. Rotenone is a pesticide that selectively kills dopaminergic neurons by a variety of mechanism, has been implicated in PD. Geraniol (GE; 3,7-dimethylocta-trans-2,6-dien-1-ol) is an acyclic monoterpene alcohol occurring in the essential oils of several aromatic plants. In this study, we investigated the protective effect of GE on rotenone-induced mitochondrial dysfunction dependent oxidative stress leads to cell death in SK-N-SH cells. In addition, we assessed the involvement of GE on rotenone-induced dysfunction in autophagy machinery via α-synuclein accumulation induced ER stress. We found that pre-treatment of GE enhanced cell viability, ameliorated intracellular redox, preserved mitochondrial membrane potential and improves the level of mitochondrial complex-1 in rotenone treated SK-N-SH cells. Furthermore, GE diminishes autophagy flux by reduced autophagy markers, and decreases ER stress by reducing α-synuclein expression in SK-N-SH cells. Our results demonstrate that GE possess its neuroprotective effect via reduced rotenone-induced oxidative stress by enhanced antioxidant status and maintain mitochondrial function. Furthermore, GE reduced ER stress and improved autophagy flux in the neuroblastomal SK-N-SH cells. The present study could suggest that GE a novel therapeutic avenue for clinical intervention in neurodegenerative diseases especially for PD.
Collapse
Affiliation(s)
- Karamkolly R Rekha
- Division of Biochemistry, Faculty of Medicine, Raja Muthaiah Medical College, Annamalai University, Annamalai Nagar, Tamilnadu, 608 002, India
| | - Ramu Inmozhi Sivakamasundari
- Division of Biochemistry, Faculty of Medicine, Raja Muthaiah Medical College, Annamalai University, Annamalai Nagar, Tamilnadu, 608 002, India.
| |
Collapse
|
126
|
Pascarella A, Terracciano C, Farina O, Lombardi L, Esposito T, Napolitano F, Franzese G, Panella G, Tuccillo F, la Marca G, Bernardini S, Boffo S, Giordano A, Di Iorio G, Melone MAB, Sampaolo S. Vacuolated PAS-positive lymphocytes as an hallmark of Pompe disease and other myopathies related to impaired autophagy. J Cell Physiol 2018; 233:5829-5837. [PMID: 29215735 DOI: 10.1002/jcp.26365] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 11/30/2017] [Indexed: 12/18/2022]
Abstract
Autosomal recessive Pompe disease is a lysosomal disorder caused by mutations of the acid-α-glucosidase (GAA) gene. Deficiency of GAA enzyme leads to glycogen accumulation and autophagy impairment in cardiac and skeletal muscles, but also in lymphocytes. Since an effective therapy is available, a rapid, sensitive, and specific test is crucial to early identify affected subjects. Number of lymphocytes containing PAS-positive vacuoles was evaluated on blood films from 72 consecutive adult patients with hyperckemia and/or muscle weakness, 13 genetically confirmed late-onset-Pompe-disease (LOPD) and 13 of their offspring. GAA activity, measured on dried blood spot (DBS) in all patients inversely correlated with number of PAS-positive lymphocytes. More than 4 PAS-positive lymphocytes were found in 11 out of the 72 patients (6 new diagnosis of LOPD, 3 different glycogen storage myopathies, 1 glucose-6-phosphate dehydrogenase deficiency, 1 caveolinopathy), in all 13 LOPD patients and in the 13 LOPD offspring. These latter resulted to have all a single GAA mutation but low GAA levels. Immunostaining with the autophagy markers LC3 and p62 confirmed the autophagic nature of lymphocytes vacuoles. ROC curve assessment of PAS-positive lymphocytes disclosed 100% of sensitivity and 94% of specificity in recognizing both compound heterozygous and heterozygous GAA carriers. The other myopathies with more than 4 PAS-positive lymphocytes appeared to be all related to impaired autophagy, which seems to be responsible of PAS-positive vacuolated lymphocytes formation. Quantification of PAS-positive lymphocytes in blood films is useful to identify autophagic vacuolar myopathies and should be routinely used as first level test for Pompe disease.
Collapse
Affiliation(s)
- Angelo Pascarella
- 2nd Division of Neurology, Department of Medicine, Surgery, Neurology, Metabolic and Aging Science, Reference Center for Neurological and Neuromuscular Rare Disease & Interuniversity Center for Research in Neurosciences, University of Campania "Luigi Vanvitelli", Naples, Italy
- Neurorehabilitation Unit and Research Lab. for Disorder of Consciousness, Maugeri ICS, Telese Terme, Italy
| | - Chiara Terracciano
- Division of Clinical Biochemistry, Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy
| | - Olimpia Farina
- 2nd Division of Neurology, Department of Medicine, Surgery, Neurology, Metabolic and Aging Science, Reference Center for Neurological and Neuromuscular Rare Disease & Interuniversity Center for Research in Neurosciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Luca Lombardi
- 2nd Division of Neurology, Department of Medicine, Surgery, Neurology, Metabolic and Aging Science, Reference Center for Neurological and Neuromuscular Rare Disease & Interuniversity Center for Research in Neurosciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Teresa Esposito
- Molecular Genetics and Genomics Laboratory, Institute of Genetics and Biophysics, "Adriano Buzzati Traverso", Italian National Research Council (CNR), Naples, Italy
- IRCCS INM Neuromed, Pozzilli, Italy
| | - Filomena Napolitano
- 2nd Division of Neurology, Department of Medicine, Surgery, Neurology, Metabolic and Aging Science, Reference Center for Neurological and Neuromuscular Rare Disease & Interuniversity Center for Research in Neurosciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giuseppina Franzese
- 2nd Division of Neurology, Department of Medicine, Surgery, Neurology, Metabolic and Aging Science, Reference Center for Neurological and Neuromuscular Rare Disease & Interuniversity Center for Research in Neurosciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giovanni Panella
- 2nd Division of Neurology, Department of Medicine, Surgery, Neurology, Metabolic and Aging Science, Reference Center for Neurological and Neuromuscular Rare Disease & Interuniversity Center for Research in Neurosciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Francesco Tuccillo
- 2nd Division of Neurology, Department of Medicine, Surgery, Neurology, Metabolic and Aging Science, Reference Center for Neurological and Neuromuscular Rare Disease & Interuniversity Center for Research in Neurosciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giancarlo la Marca
- Department of Experimental and Clinical Biomedical Sciences, University of Florence; Head, Newborn Screening, Clinical Chemistry and Pharmacology Lab, Meyer Offspring's Hospital, Florence, Italy
| | - Sergio Bernardini
- Division of Clinical Biochemistry, Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy
| | - Silvia Boffo
- Department of Biology, Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| | - Antonio Giordano
- Department of Biology, Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Giuseppe Di Iorio
- 2nd Division of Neurology, Department of Medicine, Surgery, Neurology, Metabolic and Aging Science, Reference Center for Neurological and Neuromuscular Rare Disease & Interuniversity Center for Research in Neurosciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Mariarosa A B Melone
- 2nd Division of Neurology, Department of Medicine, Surgery, Neurology, Metabolic and Aging Science, Reference Center for Neurological and Neuromuscular Rare Disease & Interuniversity Center for Research in Neurosciences, University of Campania "Luigi Vanvitelli", Naples, Italy
- Department of Biology, Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| | - Simone Sampaolo
- 2nd Division of Neurology, Department of Medicine, Surgery, Neurology, Metabolic and Aging Science, Reference Center for Neurological and Neuromuscular Rare Disease & Interuniversity Center for Research in Neurosciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
127
|
Moreno ML, Mérida S, Bosch-Morell F, Miranda M, Villar VM. Autophagy Dysfunction and Oxidative Stress, Two Related Mechanisms Implicated in Retinitis Pigmentosa. Front Physiol 2018; 9:1008. [PMID: 30093867 PMCID: PMC6070619 DOI: 10.3389/fphys.2018.01008] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 07/09/2018] [Indexed: 12/12/2022] Open
Abstract
Retinitis pigmentosa (RP) is one of the most common clinical subtypes of retinal degeneration (RD), and it is a neurodegenerative disease that could cause complete blindness in humans because it ultimately affects the photoreceptors viability. RP afflicts an estimated 1.5 million patients worldwide. The retina is highly susceptible to oxidative stress which can impair mitochondrial function. Many retina pathologies, such as diabetic retinopathy and secondary cone photoreceptor death in RP, have been related directly or indirectly with mitochondrial dysfunction. The possible role of autophagy in retina and cell differentiation is described and also the implications of autophagy dysregulation in RP. The present review shows the crucial role of autophagy in maintaining the retina homeostasis and possible therapeutic approaches for the treatment of RP.
Collapse
Affiliation(s)
- Mari-Luz Moreno
- Department of Basic Sciences, Universidad Católica de Valencia San Vicente Mártir, Valencia, Spain
| | - Salvador Mérida
- Departamento de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | - Francisco Bosch-Morell
- Departamento de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain.,Department of Medical Ophtalmology, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana, Valencia, Spain
| | - María Miranda
- Departamento de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | - Vincent M Villar
- Departamento de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| |
Collapse
|
128
|
Sawada N, Arany Z. Metabolic Regulation of Angiogenesis in Diabetes and Aging. Physiology (Bethesda) 2018; 32:290-307. [PMID: 28615313 DOI: 10.1152/physiol.00039.2016] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 02/24/2017] [Accepted: 04/05/2017] [Indexed: 12/16/2022] Open
Abstract
Impaired angiogenesis and endothelial dysfunction are hallmarks of diabetes and aging. Clinical efforts at promoting angiogenesis have largely focused on growth factor pathways, with mixed results. Recently, a new repertoire of endothelial intracellular molecules critical to endothelial metabolism has emerged as playing an important role in regulating angiogenesis. This review thus focuses on the emerging importance and therapeutic potential of these proteins and of endothelial bioenergetics in diabetes and aging.
Collapse
Affiliation(s)
- Naoki Sawada
- Department of Cell Biology and Molecular Medicine, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, New Jersey.,Department of Cell Biology and Molecular Medicine, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, New Jersey.,Division of Rheumatology, Endocrinology and Nephrology, Hokkaido University Graduate School of Medicine, Sapporo, Japan; and
| | - Zolt Arany
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
129
|
Lee DH, Kim D, Kim ST, Jeong S, Kim JL, Shim SM, Heo AJ, Song X, Guo ZS, Bartlett DL, Oh SC, Lee J, Saito Y, Kim BY, Kwon YT, Lee YJ. PARK7 modulates autophagic proteolysis through binding to the N-terminally arginylated form of the molecular chaperone HSPA5. Autophagy 2018; 14:1870-1885. [PMID: 29976090 PMCID: PMC6152518 DOI: 10.1080/15548627.2018.1491212] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 06/07/2018] [Indexed: 02/08/2023] Open
Abstract
Macroautophagy is induced under various stresses to remove cytotoxic materials, including misfolded proteins and their aggregates. These protein cargoes are collected by specific autophagic receptors such as SQSTM1/p62 (sequestosome 1) and delivered to phagophores for lysosomal degradation. To date, little is known about how cells sense and react to diverse stresses by inducing the activity of SQSTM1. Here, we show that the peroxiredoxin-like redox sensor PARK7/DJ-1 modulates the activity of SQSTM1 and the targeting of ubiquitin (Ub)-conjugated proteins to macroautophagy under oxidative stress caused by TNFSF10/TRAIL (tumor necrosis factor [ligand] superfamily, member 10). In this mechanism, TNFSF10 induces the N-terminal arginylation (Nt-arginylation) of the endoplasmic reticulum (ER)-residing molecular chaperone HSPA5/BiP/GRP78, leading to cytosolic accumulation of Nt-arginylated HSPA5 (R-HSPA5). In parallel, TNFSF10 induces the oxidation of PARK7. Oxidized PARK7 acts as a co-chaperone-like protein that binds the ER-derived chaperone R-HSPA5, a member of the HSPA/HSP70 family. By forming a complex with PARK7 (and possibly misfolded protein cargoes), R-HSPA5 binds SQSTM1 through its Nt-Arg, facilitating self-polymerization of SQSTM1 and the targeting of SQSTM1-cargo complexes to phagophores. The 3-way interaction among PARK7, R-HSPA5, and SQSTM1 is stabilized by the Nt-Arg residue of R-HSPA5. PARK7-deficient cells are impaired in the targeting of R-HSPA5 and SQSTM1 to phagophores and the removal of Ub-conjugated cargoes. Our results suggest that PARK7 functions as a co-chaperone for R-HSPA5 to modulate autophagic removal of misfolded protein cargoes generated by oxidative stress.
Collapse
Affiliation(s)
- Dae-Hee Lee
- Department of Oncology, Korea University Guro Hospital, Seoul, Republic of Korea
- Graduate School of Medicine, Korea University College of Medicine, Seoul, Republic of Korea
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daeho Kim
- Protein Metabolism Medical Research Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Department of Biophysics and Chemical Biology, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sung Tae Kim
- Protein Metabolism Medical Research Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Soyeon Jeong
- Department of Oncology, Korea University Guro Hospital, Seoul, Republic of Korea
| | - Jung Lim Kim
- Department of Oncology, Korea University Guro Hospital, Seoul, Republic of Korea
| | - Sang Mi Shim
- Protein Metabolism Medical Research Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Ah Jung Heo
- Protein Metabolism Medical Research Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Xinxin Song
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zong Sheng Guo
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - David L. Bartlett
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sang Cheul Oh
- Department of Oncology, Korea University Guro Hospital, Seoul, Republic of Korea
- Graduate School of Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Junho Lee
- Department of Biophysics and Chemical Biology, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
- The Institute of Molecular Biology and Genetics, Department of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Yoshiro Saito
- Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyoto, Japan
| | - Bo Yeon Kim
- World Class Institute, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si, Republic of Korea
| | - Yong Tae Kwon
- Protein Metabolism Medical Research Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Ischemic/Hypoxic Disease Institute, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Yong J. Lee
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
130
|
Janda E, Boi L, Carta AR. Microglial Phagocytosis and Its Regulation: A Therapeutic Target in Parkinson's Disease? Front Mol Neurosci 2018; 11:144. [PMID: 29755317 PMCID: PMC5934476 DOI: 10.3389/fnmol.2018.00144] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 04/09/2018] [Indexed: 12/12/2022] Open
Abstract
The role of phagocytosis in the neuroprotective function of microglia has been appreciated for a long time, but only more recently a dysregulation of this process has been recognized in Parkinson’s disease (PD). Indeed, microglia play several critical roles in central nervous system (CNS), such as clearance of dying neurons and pathogens as well as immunomodulation, and to fulfill these complex tasks they engage distinct phenotypes. Regulation of phenotypic plasticity and phagocytosis in microglia can be impaired by defects in molecular machinery regulating critical homeostatic mechanisms, including autophagy. Here, we briefly summarize current knowledge on molecular mechanisms of microglia phagocytosis, and the neuro-pathological role of microglia in PD. Then we focus more in detail on the possible functional role of microglial phagocytosis in the pathogenesis and progression of PD. Evidence in support of either a beneficial or deleterious role of phagocytosis in dopaminergic degeneration is reported. Altered expression of target-recognizing receptors and lysosomal receptor CD68, as well as the emerging determinant role of α-synuclein (α-SYN) in phagocytic function is discussed. We finally discuss the rationale to consider phagocytic processes as a therapeutic target to prevent or slow down dopaminergic degeneration.
Collapse
Affiliation(s)
- Elzbieta Janda
- Department of Health Sciences, Magna Graecia University, Catanzaro, Italy
| | - Laura Boi
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Anna R Carta
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| |
Collapse
|
131
|
Datta S, Chakraborty S, Panja C, Ghosh S. Reactive nitrogen species control apoptosis and autophagy in K562 cells: implication of TAp73α induction in controlling autophagy. Free Radic Res 2018; 52:491-506. [DOI: 10.1080/10715762.2018.1449210] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Sampurna Datta
- Department of Biochemistry, University of Calcutta, Kolkata, India
| | | | - Chiranjit Panja
- Department of Biochemistry, University of Calcutta, Kolkata, India
| | - Sanjay Ghosh
- Department of Biochemistry, University of Calcutta, Kolkata, India
| |
Collapse
|
132
|
Santos AL, Sinha S, Lindner AB. The Good, the Bad, and the Ugly of ROS: New Insights on Aging and Aging-Related Diseases from Eukaryotic and Prokaryotic Model Organisms. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:1941285. [PMID: 29743972 PMCID: PMC5878877 DOI: 10.1155/2018/1941285] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 12/18/2017] [Accepted: 01/02/2018] [Indexed: 12/13/2022]
Abstract
Aging is associated with the accumulation of cellular damage over the course of a lifetime. This process is promoted in large part by reactive oxygen species (ROS) generated via cellular metabolic and respiratory pathways. Pharmacological, nonpharmacological, and genetic interventions have been used to target cellular and mitochondrial networks in an effort to decipher aging and age-related disorders. While ROS historically have been viewed as a detrimental byproduct of normal metabolism and associated with several pathologies, recent research has revealed a more complex and beneficial role of ROS in regulating metabolism, development, and lifespan. In this review, we summarize the recent advances in ROS research, focusing on both the beneficial and harmful roles of ROS, many of which are conserved across species from bacteria to humans, in various aspects of cellular physiology. These studies provide a new context for our understanding of the parts ROS play in health and disease. Moreover, we highlight the utility of bacterial models to elucidate the molecular pathways by which ROS mediate aging and aging-related diseases.
Collapse
Affiliation(s)
- Ana L. Santos
- Institut National de la Santé et de la Recherche Médicale, U1001 & Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Sanchari Sinha
- Defence Institute of Physiology and Allied Sciences, DRDO, New Delhi, India
| | - Ariel B. Lindner
- Institut National de la Santé et de la Recherche Médicale, U1001 & Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
133
|
Li J, Wang Q, Cai H, He Z, Wang H, Chen J, Zheng Z, Yin J, Liao Z, Xu H, Xiao J, Gong F. FGF1 improves functional recovery through inducing PRDX1 to regulate autophagy and anti-ROS after spinal cord injury. J Cell Mol Med 2018. [PMID: 29512938 PMCID: PMC5908106 DOI: 10.1111/jcmm.13566] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Fibroblast growth factor 1 (FGF1) is thought to exert protective and regenerative effects on neurons following spinal cord injury (SCI), although the mechanism of these effects is not well understood. The use of FGF1 as a therapeutic agent is limited by its lack of physicochemical stability and its limited capacity to cross the blood‐spinal cord barrier. Here, we demonstrated that overexpression of FGF1 in spinal cord following SCI significantly reduced tissue loss, protected neurons in the ventricornu, ameliorated pathological morphology of the lesion, dramatically improved tissue recovery via neuroprotection, and promoted axonal regeneration and remyelination both in vivo and in vivo. In addition, the autophagy and the expression levels of PRDX1 (an antioxidant protein) were induced by AAV‐FGF1 in PC12 cells after H2O2 treatment. Furthermore, the autophagy levels were not changed in PRDX1‐suppressing cells that were treated by AAV‐FGF1. Taken together, these results suggest that FGF1 improves functional recovery mainly through inducing PRDX1 expression to increase autophagy and anti‐ROS activity after SCI.
Collapse
Affiliation(s)
- Jiawei Li
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qingqing Wang
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hanxiao Cai
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zili He
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Haoli Wang
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jian Chen
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zengming Zheng
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jiayu Yin
- School of Pharmacy, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhiyong Liao
- School of Life and Environmental Science, Wenzhou University, Wenzhou, Zhejiang, China
| | - Huazi Xu
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jian Xiao
- School of Pharmacy, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Fanghua Gong
- School of Pharmacy, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
134
|
Zhang J, Culp ML, Craver JG, Darley-Usmar V. Mitochondrial function and autophagy: integrating proteotoxic, redox, and metabolic stress in Parkinson's disease. J Neurochem 2018; 144:691-709. [PMID: 29341130 PMCID: PMC5897151 DOI: 10.1111/jnc.14308] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 01/04/2018] [Accepted: 01/09/2018] [Indexed: 12/14/2022]
Abstract
Parkinson's disease (PD) is a movement disorder with widespread neurodegeneration in the brain. Significant oxidative, reductive, metabolic, and proteotoxic alterations have been observed in PD postmortem brains. The alterations of mitochondrial function resulting in decreased bioenergetic health is important and needs to be further examined to help develop biomarkers for PD severity and prognosis. It is now becoming clear that multiple hits on metabolic and signaling pathways are likely to exacerbate PD pathogenesis. Indeed, data obtained from genetic and genome association studies have implicated interactive contributions of genes controlling protein quality control and metabolism. For example, loss of key proteins that are responsible for clearance of dysfunctional mitochondria through a process called mitophagy has been found to cause PD, and a significant proportion of genes associated with PD encode proteins involved in the autophagy-lysosomal pathway. In this review, we highlight the evidence for the targeting of mitochondria by proteotoxic, redox and metabolic stress, and the role autophagic surveillance in maintenance of mitochondrial quality. Furthermore, we summarize the role of α-synuclein, leucine-rich repeat kinase 2, and tau in modulating mitochondrial function and autophagy. Among the stressors that can overwhelm the mitochondrial quality control mechanisms, we will discuss 4-hydroxynonenal and nitric oxide. The impact of autophagy is context depend and as such can have both beneficial and detrimental effects. Furthermore, we highlight the potential of targeting mitochondria and autophagic function as an integrated therapeutic strategy and the emerging contribution of the microbiome to PD susceptibility.
Collapse
Affiliation(s)
- 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
| | - M Lillian Culp
- Center for Free Radical Biology, University of Alabama at Birmingham
- Department of Pathology, University of Alabama at Birmingham
| | - Jason G Craver
- 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
| |
Collapse
|
135
|
Aumailley L, Roux-Dalvai F, Kelly I, Droit A, Lebel M. Vitamin C alters the amount of specific endoplasmic reticulum associated proteins involved in lipid metabolism in the liver of mice synthesizing a nonfunctional Werner syndrome (Wrn) mutant protein. PLoS One 2018; 13:e0193170. [PMID: 29494634 PMCID: PMC5832228 DOI: 10.1371/journal.pone.0193170] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 02/06/2018] [Indexed: 11/19/2022] Open
Abstract
Werner syndrome (WS) is a premature aging disorder caused by mutations in a protein containing both a DNA exonuclease and DNA helicase domain. Mice lacking the helicase domain of the Wrn protein orthologue exhibit transcriptomic and metabolic alterations, some of which are reversed by vitamin C. Recent studies on these animals indicated that the mutant protein is associated with enriched endoplasmic reticulum (ER) fractions of tissues resulting in an ER stress response. In this study, we identified proteins that exhibit actual level differences in the ER enriched fraction between the liver of wild type and Wrn mutant mice using quantitative proteomic profiling with label-free Liquid Chromatography-Tandem Mass Spectrometry (LC-MS/MS). Multiple Reaction Monitoring (MRM) and immunoblotting were performed to validate findings in a secondary independent cohort of wild type and Wrn mutant mice. DAVID 6.7 (NIH) was used for functional annotation analysis and indicated that the identified proteins exhibiting level changes between untreated wild type, Wrn mutant, and vitamin C treated Wrn mutant mice (ANOVA P–value < 0.05) were involved in fatty acid and steroid metabolism pathways (Bonferroni P-value = 0.0137). Finally, when we compared the transcriptomic and the proteomic data of our mouse cohorts only ~7% of the altered mRNA profiles encoding for ER gene products were consistent with their corresponding protein profiles measured by the label-free quantification methods. These results suggest that a great number of ER gene products are regulated at the post-transcriptional level in the liver of Wrn mutant mice exhibiting an ER stress response.
Collapse
Affiliation(s)
- Lucie Aumailley
- Centre de recherche du CHU de Québec, Faculty of Medicine, Université Laval, Quebec City Québec, Canada
| | - Florence Roux-Dalvai
- Proteomics Platform Center, Centre de recherche du CHU de Québec, Faculty of Medicine, Université Laval, Quebec City Québec, Canada
| | - Isabelle Kelly
- Proteomics Platform Center, Centre de recherche du CHU de Québec, Faculty of Medicine, Université Laval, Quebec City Québec, Canada
| | - Arnaud Droit
- Proteomics Platform Center, Centre de recherche du CHU de Québec, Faculty of Medicine, Université Laval, Quebec City Québec, Canada
| | - Michel Lebel
- Centre de recherche du CHU de Québec, Faculty of Medicine, Université Laval, Quebec City Québec, Canada
- * E-mail:
| |
Collapse
|
136
|
Dodson M, de la Vega MR, Harder B, Castro-Portuguez R, Rodrigues SD, Wong PK, Chapman E, Zhang DD. Low-level arsenic causes proteotoxic stress and not oxidative stress. Toxicol Appl Pharmacol 2018; 341:106-113. [PMID: 29408041 PMCID: PMC5929483 DOI: 10.1016/j.taap.2018.01.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 01/12/2018] [Accepted: 01/21/2018] [Indexed: 01/22/2023]
Abstract
Prolonged exposure to arsenic has been shown to increase the risk of developing a number of diseases, including cancer and type II diabetes. Arsenic is present throughout the environment in its inorganic forms, and the level of exposure varies greatly by geographical location. The current recommended maximum level of arsenic exposure by the EPA is 10μg/L, but levels>50-1000μg/L have been detected in some parts of Asia, the Middle East, and the Southwestern United States. One of the most important steps in developing treatment options for arsenic-linked pathologies is to understand the cellular pathways affected by low levels of arsenic. Here, we show that acute exposure to non-lethal, low-level arsenite, an environmentally relevant arsenical, inhibits the autophagy pathway. Furthermore, arsenite-induced autophagy inhibition initiates a transient, but moderate ER stress response. Significantly, low-level arsenite exposure does not exhibit an increase in oxidative stress. These findings indicate that compromised autophagy, and not enhanced oxidative stress occurs early during arsenite exposure, and that restoring the autophagy pathway and proper proteostasis could be a viable option for treating arsenic-linked diseases. As such, our study challenges the existing paradigm that oxidative stress is the main underlying cause of pathologies associated with environmental arsenic exposure.
Collapse
Affiliation(s)
- Matthew Dodson
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA, 85721
| | - Montserrat Rojo de la Vega
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA, 85721
| | - Bryan Harder
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA, 85721
| | - Raul Castro-Portuguez
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA, 85721
| | - Silvia D. Rodrigues
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA, 85721
| | - Pak Kin Wong
- Department of Biomedical Engineering, The Pennsylvania State University, University, Park, PA, USA, 16802
| | - Eli Chapman
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA, 85721
| | - Donna D. Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA, 85721,Arizona Cancer Center, University of Arizona, Tucson, AZ, USA, 85724,To whom correspondence should be addressed: Dr. Donna D. Zhang, College of Pharmacy, 1703 East Mabel Street, Tucson, AZ, 85721, , Phone number: 520-626-9918
| |
Collapse
|
137
|
Yan C, Luo L, Goto S, Urata Y, Guo CY, Doi H, Kitazato K, Li TS. Enhanced autophagy in colorectal cancer stem cells does not contribute to radio-resistance. Oncotarget 2018; 7:45112-45121. [PMID: 27129175 PMCID: PMC5216709 DOI: 10.18632/oncotarget.8972] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Accepted: 04/11/2016] [Indexed: 12/22/2022] Open
Abstract
Autophagy, an essential catabolic pathway of degrading cellular components within the lysosome, has been found to benefit the growth and therapeutic resistance of cancer cells. In this study, we investigated the role of autophagy in the radio-sensitivity of cancer stem cells. By separating CD44+/CD133+ cancer stem cells from parental HCT8 human colorectal cancer cells, we found a significantly higher level of autophagy in the CD44+/CD133+ cells than in the parental cells. Exposure to 5 Gy of γ-ray significantly damaged both CD44+/CD133+ cells and parental cells, but the radiation-induced damage did not differ between the groups. Unexpectedly, autophagy was not significantly induced by radiation exposure in the CD44+/CD133+ cells and parental cells. The inhibition of autophagy by the silencing of ATG7, a factor required for autophagy at the stage of autophagosome precursor synthesis, did not significantly change the growth and radiation-induced damage in both CD44+/CD133+ cells and parental cells. Although an enhanced basic level of autophagy was found in the CD44+/CD133+ cancer stem cells, our data suggest that the canonical autophagy in cancer cells plays few roles, if any, in radio-sensitivity.
Collapse
Affiliation(s)
- Chen Yan
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Lan Luo
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Shinji Goto
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yoshishige Urata
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Chang-Ying Guo
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Department of Thoracic Surgery, Jiangxi Cancer Hospital, Nanchang, PR China
| | - Hanako Doi
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kaio Kitazato
- Division of Molecular Pharmacology of Infectious Agents, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Tao-Sheng Li
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
138
|
Hypertrophied myocardium is vulnerable to ischemia/reperfusion injury and refractory to rapamycin-induced protection due to increased oxidative/nitrative stress. Clin Sci (Lond) 2018; 132:93-110. [PMID: 29175946 DOI: 10.1042/cs20171471] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 11/22/2017] [Accepted: 11/24/2017] [Indexed: 11/17/2022]
Abstract
Left ventricular hypertrophy (LVH) is causally related to increased morbidity and mortality following acute myocardial infarction (AMI) via still unknown mechanisms. Although rapamycin exerts cardioprotective effects against myocardial ischemia/reperfusion (MI/R) injury in normal animals, whether rapamycin-elicited cardioprotection is altered in the presence of LVH has yet to be determined. Pressure overload induced cardiac hypertrophied mice and sham-operated controls were exposed to AMI by coronary artery ligation, and treated with vehicle or rapamycin 10 min before reperfusion. Rapamycin produced marked cardioprotection in normal control mice, whereas pressure overload induced cardiac hypertrophied mice manifested enhanced myocardial injury, and was refractory to rapamycin-elicited cardioprotection evidenced by augmented infarct size, aggravated cardiomyocyte apoptosis, and worsening cardiac function. Rapamycin alleviated MI/R injury via ERK-dependent antioxidative pathways in normal mice, whereas cardiac hypertrophied mice manifested markedly exacerbated oxidative/nitrative stress after MI/R evidenced by the increased iNOS/gp91phox expression, superoxide production, total NO metabolites, and nitrotyrosine content. Moreover, scavenging superoxide or peroxynitrite by selective gp91phox assembly inhibitor gp91ds-tat or ONOO- scavenger EUK134 markedly ameliorated MI/R injury, as shown by reduced myocardial oxidative/nitrative stress, alleviated myocardial infarction, hindered cardiomyocyte apoptosis, and improved cardiac function in aortic-banded mice. However, no additional cardioprotective effects were achieved when we combined rapamycin and gp91ds-tat or EUK134 in ischemic/reperfused hearts with or without LVH. These results suggest that cardiac hypertrophy attenuated rapamycin-induced cardioprotection by increasing oxidative/nitrative stress and scavenging superoxide/peroxynitrite protects the hypertrophied heart from MI/R.
Collapse
|
139
|
The Interrelation between Reactive Oxygen Species and Autophagy in Neurological Disorders. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:8495160. [PMID: 29391926 PMCID: PMC5748124 DOI: 10.1155/2017/8495160] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 10/30/2017] [Indexed: 01/08/2023]
Abstract
Neurological function deficits due to cerebral ischemia or neurodegenerative diseases such as Alzheimer's disease (AD) and Parkinson's disease (PD) have long been considered a thorny issue in clinical treatment. Recovery after neurologic impairment is fairly limited, which poses a major threat to health and quality of life. Accumulating evidences support that ROS and autophagy are both implicated in the onset and development of neurological disorders. Notably, oxidative stress triggered by excess of ROS not only puts the brain in a vulnerable state but also enhances the virulence of other pathogenic factors, just like mitochondrial dysfunction, which is described as the culprit of nerve cell damage. Nevertheless, autophagy is proposed as a subtle cellular defense mode against destructive stimulus by timely removal of damaged and cytotoxic substance. Emerging evidence suggests that the interplay of ROS and autophagy may establish a determinant role in the modulation of neuronal homeostasis. However, the underlying regulatory mechanisms are still largely unexplored. This review sets out to afford an overview of the crosstalk between ROS and autophagy and discusses relevant molecular mechanisms in cerebral ischemia, AD, and PD, so as to provide new insights into promising therapeutic targets for the abovementioned neurological conditions.
Collapse
|
140
|
Inhibition of autophagy-attenuated calcium oxalate crystal-induced renal tubular epithelial cell injury in vivo and in vitro. Oncotarget 2017; 9:4571-4582. [PMID: 29435125 PMCID: PMC5796996 DOI: 10.18632/oncotarget.23383] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 11/15/2017] [Indexed: 11/25/2022] Open
Abstract
Accumulating evidence suggests that autophagy is involved in the pathophysiological processes of kidney diseases. However, the role of autophagy in the formation of calcium oxalate (CaOx) nephrolithiasis remains unclear. In this study, we investigated the effects of autophagy on renal tubular epithelial cell injury induced by CaOx crystals in vivo and in vitro. We first observed that the expression levels of LC3-II and BECN1 and number of autophagic vacuoles were markedly increased in the renal tissue of CaOx stone patients. We subsequently found that exposure of HK-2 cells to CaOx crystals could increase LC3-II and BECN1 expression as well as the number of GFP-LC3 dots and autophagic vacuoles in a dose- and time-dependent manner. In addition, our results suggest that CaOx crystals induced autophagy, at least in part, via activation of the reactive oxygen species (ROS) pathway in HK-2 cells. Furthermore, inhibition of autophagy using 3-methyladenine or siRNA knockdown of BECN1 attenuated CaOx crystal-induced HK-2 cells injury. However, enhancing autophagic activity with rapamycin exerted an opposite effect. Taken together, our results demonstrate that autophagy is essential for CaOx crystal-induced renal tubular epithelial cell injury and that inhibition of autophagy could be a novel therapeutic strategy for CaOx nephrolithiasis.
Collapse
|
141
|
Wrutniak-Cabello C, Casas F, Cabello G. Mitochondrial T3 receptor and targets. Mol Cell Endocrinol 2017; 458:112-120. [PMID: 28167126 DOI: 10.1016/j.mce.2017.01.054] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 01/28/2017] [Accepted: 01/31/2017] [Indexed: 12/25/2022]
Abstract
The demonstration that TRα1 mRNA encodes a nuclear thyroid hormone receptor and two proteins imported into mitochondria with molecular masses of 43 and 28 kDa has brought new clues to better understand the pleiotropic influence of iodinated hormones. If p28 activity remains unknown, p43 binds to T3 responsive elements occurring in the organelle genome, and, in the T3 presence, stimulates mitochondrial transcription and the subsequent synthesis of mitochondrial encoded proteins. This influence increases mitochondrial activity and through changes in the mitochondrial/nuclear cross talk affects important nuclear target genes regulating cell proliferation and differentiation, oncogenesis, or apoptosis. In addition, this pathway influences muscle metabolic and contractile phenotype, as well as glycaemia regulation. Interestingly, according to the process considered, p43 exerts opposite or cooperative effects with the well-known T3 pathway, thus allowing a fine tuning of the physiological influence of this hormone.
Collapse
Affiliation(s)
- Chantal Wrutniak-Cabello
- INRA, UMR 866 Dynamique Musculaire et Métabolisme, 34060 Montpellier, France; Université de Montpellier, UMR 866 Dynamique Musculaire et Métabolisme, 34060 Montpellier, France.
| | - François Casas
- INRA, UMR 866 Dynamique Musculaire et Métabolisme, 34060 Montpellier, France; Université de Montpellier, UMR 866 Dynamique Musculaire et Métabolisme, 34060 Montpellier, France
| | - Gérard Cabello
- INRA, UMR 866 Dynamique Musculaire et Métabolisme, 34060 Montpellier, France; Université de Montpellier, UMR 866 Dynamique Musculaire et Métabolisme, 34060 Montpellier, France
| |
Collapse
|
142
|
Rech VC, Mezzomo NJ, Athaydes GA, Feksa LR, Figueiredo VC, Kessler A, Franceschi IDDE, Wannmacher CMD. Thiol/disulfide status regulates the activity of thiol-containing kinases related to energy homeostasis in rat kidney. AN ACAD BRAS CIENC 2017; 90:99-108. [PMID: 29236866 DOI: 10.1590/0001-3765201720160348] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 08/08/2016] [Indexed: 11/21/2022] Open
Abstract
Considering that thiol-containing enzymes like kinases are critical for several metabolic pathways and energy homeostasis, we investigated the effects of cystine dimethyl ester and/or cysteamine administration on kinases crucial for energy metabolism in the kidney of Wistar rats. Animals were injected twice a day with 1.6 µmol/g body weight cystine dimethyl ester and/or 0.26 µmol/g body weight cysteamine from the 16th to the 20th postpartum day and euthanized after 12 hours. Pyruvate kinase, adenylate kinase, creatine kinase activities and thiol/disulfide ratio were determined. Cystine dimethyl ester administration reduced thiol/disulfide ratio and inhibited the kinases activities. Cysteamine administration increased the thiol/disulfide ratio and co-administration with cystine dimethyl ester prevented the inhibition of the enzymes. Regression between the thiol/disulfide ratio, and the kinases activities were significant. These results suggest that redox status may regulate energy metabolism in the rat kidney. If thiol-containing enzymes inhibition and oxidative stress occur in patients with cystinosis, it is possible that lysosomal cystine depletion may not be the only beneficial effect of cysteamine administration, but also its antioxidant and thiol-protector effect.
Collapse
Affiliation(s)
- Virginia C Rech
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600, Anexo, Santa Cecília, 90035-003 Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Nanociências, Laboratório de Nanotecnologia, Centro Universitário Franciscano, Rua dos Andradas, 1614, 97010-032 Santa Maria, RS, Brazil
| | - Nathana J Mezzomo
- Programa de Pós-Graduação em Nanociências, Laboratório de Nanotecnologia, Centro Universitário Franciscano, Rua dos Andradas, 1614, 97010-032 Santa Maria, RS, Brazil
| | - Genaro A Athaydes
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600, Anexo, Santa Cecília, 90035-003 Porto Alegre, RS, Brazil
| | - Luciane R Feksa
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600, Anexo, Santa Cecília, 90035-003 Porto Alegre, RS, Brazil.,Instituto de Ciências da Saúde, Universidade Feevale, ERS-239, 2755, 93525-075 Novo Hamburgo, RS, Brazil
| | - Vandré C Figueiredo
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600, Anexo, Santa Cecília, 90035-003 Porto Alegre, RS, Brazil
| | - Adriana Kessler
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600, Anexo, Santa Cecília, 90035-003 Porto Alegre, RS, Brazil.,Faculdade de Enfermagem, Nutrição e Fisioterapia, Pontifícia Universidade Católica do Rio Grande do Sul, Av. Ipiranga, 6681, Prédio 12, Partenon, 90619-900 Porto Alegre, RS, Brazil
| | - Itiane D DE Franceschi
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600, Anexo, Santa Cecília, 90035-003 Porto Alegre, RS, Brazil
| | - Clovis M D Wannmacher
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600, Anexo, Santa Cecília, 90035-003 Porto Alegre, RS, Brazil
| |
Collapse
|
143
|
Moura-Martiniano NO, Machado-Ferreira E, Gazêta GS, Soares CAG. Relative transcription of autophagy-related genes in Amblyomma sculptum and Rhipicephalus microplus ticks. EXPERIMENTAL & APPLIED ACAROLOGY 2017; 73:401-428. [PMID: 29181673 DOI: 10.1007/s10493-017-0193-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 11/17/2017] [Indexed: 06/07/2023]
Abstract
Ticks endure stressful off-host periods and perform as vectors of a diversity of infectious agents, thus engaging pathways that expectedly demand for autophagy. Little is known of ticks' autophagy, a conserved eukaryotic machinery assisting in homeostasis processes that also participates in tissue-dependent metabolic functions. Here, the autophagy-related ATG4 (autophagin-1), ATG6 (beclin-1) and ATG8 (LC3) mRNAs from the human diseases vector Amblyomma sculptum and the cattle-tick Rhipicephalus microplus were identified. Comparative qPCR quantifications evidenced different transcriptional status for the ATG genes in the salivary glands (SG), ovaries and intestines of actively feeding ticks. These ATGs had increased relative transcription under nutrient-deprivation, as determined by validation tests with R. microplus embryo-derivative cells BME26 and A. sculptum SG explants incubations in HBSS. Starvation lead to 4-31.8× and ~ 60-500× increments on the ATGs mRNA loads in BME26 and A. sculptum SG explants, respectively. PI3K inhibitor 3MA treatment also affected ATGs expression in BME26. Some ATGs were more transcribed in the SGs than in the ovaries of cattle-ticks. Amblyomma sculptum/R. microplus interspecific comparisons showed that ATG4 and ATG6 were 0.18× less expressed in A. sculptum SGs, but ~ 10-100× more expressed in their ovaries when compared to R. microplus organs. ATG4 and ATG8a transcript loads were ~ 120× and ~ 40× higher, respectively, in A. sculptum intestines when compared to cattle-ticks of similar weight category. ATGs expression in A. sculptum intestines increased with tick weight, indicating Atgs contribution to intracellular blood digestion. Possible roles of the autophagy machinery and their organ-specific expression profile on vector biology are discussed.
Collapse
Affiliation(s)
- Nicole O Moura-Martiniano
- Laboratório de Genética Molecular de Eucariontes e Simbiontes, Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Erik Machado-Ferreira
- Laboratório de Genética Molecular de Eucariontes e Simbiontes, Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gilberto S Gazêta
- Laboratório de Referência Nacional em Vetores das Riquetsioses, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Carlos Augusto Gomes Soares
- Laboratório de Genética Molecular de Eucariontes e Simbiontes, Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
- , Ilha do Fundão, CCS, Bloco A, Lab. A2-120. Rua Professor Rodolpho Paulo Rocco S/N, Rio de Janeiro, RJ, 21941-617, Brazil.
| |
Collapse
|
144
|
Dodson M, Wani WY, Redmann M, Benavides GA, Johnson MS, Ouyang X, Cofield SS, Mitra K, Darley-Usmar V, Zhang J. Regulation of autophagy, mitochondrial dynamics, and cellular bioenergetics by 4-hydroxynonenal in primary neurons. Autophagy 2017; 13:1828-1840. [PMID: 28837411 DOI: 10.1080/15548627.2017.1356948] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
The production of reactive species contributes to the age-dependent accumulation of dysfunctional mitochondria and protein aggregates, all of which are associated with neurodegeneration. A putative mediator of these effects is the lipid peroxidation product 4-hydroxynonenal (4-HNE), which has been shown to inhibit mitochondrial function, and accumulate in the postmortem brains of patients with neurodegenerative diseases. This deterioration in mitochondrial quality could be due to direct effects on mitochondrial proteins, or through perturbation of the macroautophagy/autophagy pathway, which plays an essential role in removing damaged mitochondria. Here, we use a click chemistry-based approach to demonstrate that alkyne-4-HNE can adduct to specific mitochondrial and autophagy-related proteins. Furthermore, we found that at lower concentrations (5-10 μM), 4-HNE activates autophagy, whereas at higher concentrations (15 μM), autophagic flux is inhibited, correlating with the modification of key autophagy proteins at higher concentrations of alkyne-4-HNE. Increasing concentrations of 4-HNE also cause mitochondrial dysfunction by targeting complex V (the ATP synthase) in the electron transport chain, and induce significant changes in mitochondrial fission and fusion protein levels, which results in alterations to mitochondrial network length. Finally, inhibition of autophagy initiation using 3-methyladenine (3MA) also results in a significant decrease in mitochondrial function and network length. These data show that both the mitochondria and autophagy are critical targets of 4-HNE, and that the proteins targeted by 4-HNE may change based on its concentration, persistently driving cellular dysfunction.
Collapse
Affiliation(s)
- Matthew Dodson
- a Center for Free Radical Biology , University of Alabama at Birmingham , Birmingham , AL , USA.,b Department of Pathology , University of Alabama at Birmingham , Birmingham , AL , USA
| | - Willayat Y Wani
- a Center for Free Radical Biology , University of Alabama at Birmingham , Birmingham , AL , USA.,b Department of Pathology , University of Alabama at Birmingham , Birmingham , AL , USA
| | - Matthew Redmann
- a Center for Free Radical Biology , University of Alabama at Birmingham , Birmingham , AL , USA.,b Department of Pathology , University of Alabama at Birmingham , Birmingham , AL , USA
| | - Gloria A Benavides
- a Center for Free Radical Biology , University of Alabama at Birmingham , Birmingham , AL , USA.,b Department of Pathology , University of Alabama at Birmingham , Birmingham , AL , USA
| | - Michelle S Johnson
- a Center for Free Radical Biology , University of Alabama at Birmingham , Birmingham , AL , USA.,b Department of Pathology , University of Alabama at Birmingham , Birmingham , AL , USA
| | - Xiaosen Ouyang
- a Center for Free Radical Biology , University of Alabama at Birmingham , Birmingham , AL , USA.,b Department of Pathology , University of Alabama at Birmingham , Birmingham , AL , USA.,e Department of Veterans Affairs , Birmingham VA Medical Center , Birmingham , AL , USA
| | - Stacey S Cofield
- c Department of Biostatistics , University of Alabama at Birmingham , Birmingham , AL , USA
| | - Kasturi Mitra
- d Department of Genetics , University of Alabama at Birmingham , Birmingham , AL , USA
| | - Victor Darley-Usmar
- a Center for Free Radical Biology , University of Alabama at Birmingham , Birmingham , AL , USA.,b Department of Pathology , University of Alabama at Birmingham , Birmingham , AL , USA
| | - Jianhua Zhang
- a Center for Free Radical Biology , University of Alabama at Birmingham , Birmingham , AL , USA.,b Department of Pathology , University of Alabama at Birmingham , Birmingham , AL , USA.,e Department of Veterans Affairs , Birmingham VA Medical Center , Birmingham , AL , USA
| |
Collapse
|
145
|
Morsi M, Maher A, Aboelmagd O, Johar D, Bernstein L. A shared comparison of diabetes mellitus and neurodegenerative disorders. J Cell Biochem 2017; 119:1249-1256. [PMID: 28681964 DOI: 10.1002/jcb.26261] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 07/05/2017] [Indexed: 12/19/2022]
Abstract
Diabetes mellitus (DM) is one of the most common diseases in the world population, associated with obesity, pancreatic endocrine changes, cardiovascular disease, renal glomerular disease, cerebrovascular disease, peripheral neuropathy, neurodegenerative disease, retinal disease, sleep apnea, some of which are bundled into the metabolic syndrome. The main characteristic of this disease is hyperglycemia, and often with albuminuria. Nevertheless, the classic features, with ketoacidosis in the extreme, are only a first layer of description of this condition. The description of the islet cells of the endocrine pancreas was first described by Opie, and the discovery of insulin by tying off the exocrine pancreatic ducts followed. We later find that the β-cells secrete insulin and glucagon, which synchronously stimulate or suppress glycogenolysis, and that insulin is essential for glucose intake into the cell. There are yet two other layers for our understanding of diabetes and the effects of its dysfunction, which is the basis for understanding the system-wide expression of the disease. We describe the molecular basis for the central nervous system neuropathic diseases that are associated with both Type 1 DM (T1DM) and Type 2 DM (T2DM), but more so with T2DM. T2DM is an autoimmune disease that destroys the insulin secreting islet cells. T2DM is the diabetes that is associated with an imbalance in the glucagon/insulin homeostasis that leads to the formation of amyloid deposits in the brain, pancreatic islet cells, and possibly the kidney glomerulus.
Collapse
Affiliation(s)
- Mahmoud Morsi
- Faculty of Medicine, Menoufia University, Shebin El-kom, Menoufia, Egypt
| | - Ahmed Maher
- Zoonotic Diseases Department, National Research Center, Dokki, Giza, Egypt
| | - Omnia Aboelmagd
- Faculty of Medicine, CairoUniversity, Kasr Al-Ainy, Cairo, Egypt
| | - Dina Johar
- Department of Biochemistry and Nutrition, Ain Shams University Faculty of Women for Arts, Sciences and Education, Heliopolis, Cairo, Egypt.,Department of Physiology and Pathophysiology, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | | |
Collapse
|
146
|
Sun H, Liu Y, Zhang L, Shao X, Liu K, Ding Z, Liu X, Jiang C, Li H, Li H. Numb positively regulates autophagic flux via regulating lysosomal function. Biochem Biophys Res Commun 2017; 491:780-786. [PMID: 28720501 DOI: 10.1016/j.bbrc.2017.07.084] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 07/14/2017] [Indexed: 11/15/2022]
Abstract
Autophagy is a lysosome-dependent catabolic process involving in the degradation and recycling of unnecessary or damaged proteins and organelles. Emerging evidence indicates that autophagy dysfunction is closely related to various human diseases including cancer, aging, myopathies and neurodegenerative disorders. Here, using genetic knockdown, we uncover the role of Numb, an endocytic adaptor protein, in regulating the late steps of autophagy. We found that Numb depletion led to the accumulation of autophagic vacuole, as verified by RFP-LC3 staining combined with transmission electron microscopy. Further investigation indicated that Numb depletion impaired autophagic degradation through inhibiting the activities of lysosomal enzymes (Cathepsin D, β-glucuronidase and β-glucosidase). Moreover, Numb depletion induced elevation of lysosomal pH values and decrease of glycosylated lysosome-associated membrane proteins. We further observed that Rab7 activity was inhibited in Numb-depleted cells. Together, our findings revealed a novel function of Numb and its likely mechanism in regulation of autophagy events.
Collapse
Affiliation(s)
- Haiyan Sun
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Guangdong Key Laboratory of Nanomedicine, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yi Liu
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Guangdong Key Laboratory of Nanomedicine, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Lei Zhang
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Guangdong Key Laboratory of Nanomedicine, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Ximing Shao
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Guangdong Key Laboratory of Nanomedicine, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Ke Liu
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Guangdong Key Laboratory of Nanomedicine, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Zhihao Ding
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Guangdong Key Laboratory of Nanomedicine, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Xianming Liu
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Guangdong Key Laboratory of Nanomedicine, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China; College of Life Science and Technology, Jinan University, 601 West Huangpu Blvd., Guangzhou 510632, China
| | - Changan Jiang
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Guangdong Key Laboratory of Nanomedicine, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Huashun Li
- SARITEX Center for Stem Cell Engineering Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Advanced Institute of Translational Medicine, Shanghai 200123, China; ATCG Corporation, BioBay, Suzhou Industrial Park, Suzhou, Jiangsu 215123, China
| | - Hongchang Li
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Guangdong Key Laboratory of Nanomedicine, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China.
| |
Collapse
|
147
|
Singh A, Sen E. Reciprocal role of SIRT6 and Hexokinase 2 in the regulation of autophagy driven monocyte differentiation. Exp Cell Res 2017; 360:365-374. [PMID: 28935467 DOI: 10.1016/j.yexcr.2017.09.028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 09/15/2017] [Accepted: 09/16/2017] [Indexed: 12/16/2022]
Abstract
Emerging evidences suggest the impact of autophagy on differentiation but the underlying molecular links between metabolic restructuring and autophagy during monocyte differentiation remain elusive. An increase in PPARγ, HK2 and SIRT6 expression was observed upon PMA induced monocyte differentiation. While PPARγ positively regulated HK2 and SIRT6 expression, the latter served as a negative regulator of HK2. Changes in expression of these metabolic modelers were accompanied by decreased glucose uptake and increase in Chibby, a potent antagonist of β-catenin/Wnt pathway. Knockdown of Chibby abrogated PMA induced differentiation. While inhibition of HK2 either by Lonidamine or siRNA further elevated PMA induced Chibby, mitochondrial ROS, TIGAR and LC3II levels; siRNA mediated knock-down of SIRT6 exhibited contradictory effects as compared to HK2. Notably, inhibition of autophagy increased HK2, diminished Chibby level and CD33 expression. In addition, PMA induced expression of cytoskeletal architectural proteins, CXCR4, phagocytosis, acquisition of macrophage phenotypes and release of pro-inflammatory mediators was found to be HK2 dependent. Collectively, our findings highlight the previously unknown reciprocal influence of SIRT6 and HK2 in regulating autophagy driven monocyte differentiation.
Collapse
Affiliation(s)
- Ankita Singh
- National Brain Research Centre, Manesar, Haryana 122051, India
| | - Ellora Sen
- National Brain Research Centre, Manesar, Haryana 122051, India.
| |
Collapse
|
148
|
Wong HS, Dighe PA, Mezera V, Monternier PA, Brand MD. Production of superoxide and hydrogen peroxide from specific mitochondrial sites under different bioenergetic conditions. J Biol Chem 2017; 292:16804-16809. [PMID: 28842493 DOI: 10.1074/jbc.r117.789271] [Citation(s) in RCA: 327] [Impact Index Per Article: 40.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mitochondrial production of superoxide and hydrogen peroxide is potentially important in cell signaling and disease. Eleven distinct mitochondrial sites that differ markedly in capacity are known to leak electrons to oxygen to produce O2̇̄ and/or H2O2 We discuss their contributions to O2̇̄/H2O2 production under native conditions in mitochondria oxidizing different substrates and in conditions mimicking physical exercise and the changes in their capacities after caloric restriction. We review the use of S1QELs and S3QELs, suppressors of mitochondrial O2̇̄/H2O2 generation that do not inhibit oxidative phosphorylation, as tools to characterize the contributions of specific sites in situ and in vivo.
Collapse
Affiliation(s)
- Hoi-Shan Wong
- From the Buck Institute for Research on Aging, Novato, California 94945
| | - Pratiksha A Dighe
- From the Buck Institute for Research on Aging, Novato, California 94945
| | - Vojtech Mezera
- From the Buck Institute for Research on Aging, Novato, California 94945
| | | | - Martin D Brand
- From the Buck Institute for Research on Aging, Novato, California 94945
| |
Collapse
|
149
|
Anderson DC, Lapp SA, Barnwell JW, Galinski MR. A large scale Plasmodium vivax- Saimiri boliviensis trophozoite-schizont transition proteome. PLoS One 2017; 12:e0182561. [PMID: 28829774 PMCID: PMC5567661 DOI: 10.1371/journal.pone.0182561] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Accepted: 07/20/2017] [Indexed: 11/18/2022] Open
Abstract
Plasmodium vivax is a complex protozoan parasite with over 6,500 genes and stage-specific differential expression. Much of the unique biology of this pathogen remains unknown, including how it modifies and restructures the host reticulocyte. Using a recently published P. vivax reference genome, we report the proteome from two biological replicates of infected Saimiri boliviensis host reticulocytes undergoing transition from the late trophozoite to early schizont stages. Using five database search engines, we identified a total of 2000 P. vivax and 3487 S. boliviensis proteins, making this the most comprehensive P. vivax proteome to date. PlasmoDB GO-term enrichment analysis of proteins identified at least twice by a search engine highlighted core metabolic processes and molecular functions such as glycolysis, translation and protein folding, cell components such as ribosomes, proteasomes and the Golgi apparatus, and a number of vesicle and trafficking related clusters. Database for Annotation, Visualization and Integrated Discovery (DAVID) v6.8 enriched functional annotation clusters of S. boliviensis proteins highlighted vesicle and trafficking-related clusters, elements of the cytoskeleton, oxidative processes and response to oxidative stress, macromolecular complexes such as the proteasome and ribosome, metabolism, translation, and cell death. Host and parasite proteins potentially involved in cell adhesion were also identified. Over 25% of the P. vivax proteins have no functional annotation; this group includes 45 VIR members of the large PIR family. A number of host and pathogen proteins contained highly oxidized or nitrated residues, extending prior trophozoite-enriched stage observations from S. boliviensis infections, and supporting the possibility of oxidative stress in relation to the disease. This proteome significantly expands the size and complexity of the known P. vivax and Saimiri host iRBC proteomes, and provides in-depth data that will be valuable for ongoing research on this parasite’s biology and pathogenesis.
Collapse
Affiliation(s)
- D. C. Anderson
- Bioscience Division, SRI International, Harrisonburg, VA, United States of America
- * E-mail:
| | - Stacey A. Lapp
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States of America
| | - John W. Barnwell
- Malaria Branch, Division of Parasitic Diseases, Centers for Disease Control and Prevention, Atlanta, GA, United States of America
| | - Mary R. Galinski
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States of America
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, United States of America
| |
Collapse
|
150
|
Kang MK, Park SH, Kim YH, Lee EJ, Antika LD, Kim DY, Choi YJ, Kang YH. Chrysin ameliorates podocyte injury and slit diaphragm protein loss via inhibition of the PERK-eIF2α-ATF-CHOP pathway in diabetic mice. Acta Pharmacol Sin 2017; 38:1129-1140. [PMID: 28502979 DOI: 10.1038/aps.2017.30] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 02/20/2017] [Indexed: 12/17/2022]
Abstract
Glomerular epithelial podocytes are highly specialized cells that play a crucial role in maintaining normal function of the glomerular filtration barrier via their foot processes. Chrysin (5,7-dihydroxyflavone) is a natural flavonoid found in propolis and mushrooms that has anti-inflammatory, antioxidant and anticancer properties. This study aimed to evaluate the renoprotective effects of chrysin on podocyte apoptotic loss and slit diaphragm protein deficiency in high glucose-exposed podocytes and in db/db mouse kidneys. Exposure to high glucose (33 mmol/L) caused glomerular podocyte apoptosis in vitro, which was dose-dependently attenuated by nontoxic chrysin (1-20 μmol/L) through reduction of DNA fragmentation. Chrysin treatment dose-dependently restored the increased Bax/Bcl-2 ratio, and suppressed Apaf-1 induction and the elevated cytochrome c release in high glucose-exposed renal podocytes. In diabetic db/db mice, oral administration of chrysin (10 mg·kg-1·d-1, for 10 weeks) significantly attenuated proteinuria, and alleviated the abnormal alterations in glomerular ultrastructure, characterized by apoptotic podocytes and foot process effacement. In addition, this compound improved the induction of slit diaphragm proteins podocin/nephrin in the diabetic glomeruli. Exposure to high glucose elevated the unfolded protein response (UPR) to ER stress in renal podocytes, evidenced by up-regulation of PERK-eIF2α-ATF4-CHOP. Chrysin treatment blocked such ER stress responses pertinent to podocyte apoptosis and reduced synthesis of slit diaphragm proteins in vitro and in vivo. These observations demonstrate that targeting ER stress is an underlying mechanism of chrysin-mediated amelioration of diabetes-associated podocyte injury and dysfunction.
Collapse
|