1
|
Luo C, Li X, Yan H, Guo Q, Liu J, Li Y. Iron oxide nanoparticles induce ferroptosis under mild oxidative stress in vitro. Sci Rep 2024; 14:31383. [PMID: 39733146 DOI: 10.1038/s41598-024-82917-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 12/10/2024] [Indexed: 12/30/2024] Open
Abstract
Iron oxide nanoparticles (IONPs) have the potential to be utilized in a multitude of fields, including biomedicine. Consequently, the potential health risks associated with their use must be carefully considered. Most biosafety evaluations of IONPs have focused on examining the impact of the material's distinctive physicochemical attributes. However, the specific attributes of individual cells are frequently disregarded, particularly under the oxidative stress conditions. This may result in an underestimation of potential risk and impede the clinical translation of IONPs. The present study thus sought to evaluate the potential cytotoxicity and underlying mechanisms of IONPs in a pathological state characterized by mild oxidative stress. A cell model of mild oxidative stress was initially established in vitro. Subsequently, a series of indicators, including cell viability, live/dead ratio, mitochondrial membrane potential, and oxidative damage, were measured to assess the cytotoxicity of IONPs. Finally, a series of ferroptosis regulators were used to elucidate the involvement of ferroptosis. Preincubation with IONPs resulted in a significant reduction in cellular viability, morphological degeneration, elevated numbers of dead cells, impaired mitochondrial function, and increased oxidative damage under mild oxidative stress conditions in vitro. The cytotoxic effects of IONPs under mild oxidative stress are largely dependent on ROS and iron ions and are strongly associated with ferroptosis, which is based on the effects of ferroptosis regulators. The present in vitro study indicated that IONPs are toxic to cells under mild oxidative stress, which is linked to ferroptosis.
Collapse
Affiliation(s)
- Cheng Luo
- School of Medicine, Yichun University, Yichun, 336000, China
| | - Xuying Li
- School of Medicine, Yichun University, Yichun, 336000, China
| | - Hongyang Yan
- School of Medicine, Yichun University, Yichun, 336000, China
| | - Qitao Guo
- School of Medicine, Yichun University, Yichun, 336000, China
| | - Jiarong Liu
- School of Medicine, Yichun University, Yichun, 336000, China
| | - Yan Li
- School of Medicine, Yichun University, Yichun, 336000, China.
| |
Collapse
|
2
|
Lizardo LP, Elisa RB, Tania XR, Ulises TF, Brandon LQ, Regina MQ, Carlos CJ, Montserrat ZO, Francisco AH. Oxidative Stress, Lysosomal Permeability, and Mitochondrial-Derived Vesicles Induced in NL-20 Human Bronchial Cells Exposed to Benzo[ghi]Perylene. Toxicol In Vitro 2024; 104:105999. [PMID: 39701484 DOI: 10.1016/j.tiv.2024.105999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 10/01/2024] [Accepted: 12/13/2024] [Indexed: 12/21/2024]
Abstract
Benzo[ghi] perylene (b[ghi]p) is classified as non-carcinogenic to humans, and there are currently no occupational exposure models available to identify its effects. The aim of this work was to evaluate the effect of b[ghi]p on the lysosomes of NL-20 cells (a human bronchial cell line) exposed to 4.5 μM for 3 h. The effect was evaluated through an ultrastructural evaluation, morphological changes, and acridine orange staining of lysosomes. Superoxide was quantified; and SOD1, cathepsin B, LAMP1, galectin-3 and LC3α/β, and Rab7 expression was evaluated by immunocytochemistry. The expression of genes related to oxidative stress responses (NRF2, NQO1, HMOX1 and PRDX1) and genes related to autophagy (ULK1, ATG9, BCN1, VMP1, TMEM41B and p62) were quantified by RT-qPCR. The ultrastructural evaluation revealed an increase in autophagic vesicles and phagophores in cells exposed to b[ghi]p, as well as vesicles derived from mitochondria. Based on morphology, there were vesicles in the cytoplasm. B[ghi]p significantly decreased the number of lysosomes (p < 0.05), and NAC reverse this effect (p < 0.05). Superoxide production was observed from 30 min to 3 h (p < 0.05). Immunocytochemistry revealed increased galectin-3 and LC3α/β. All oxidative stress-related genes showed high expression (p < 0.05), and the expression of ATG9 gene was decreased (p < 0.05). These results demonstrate that b[ghi]p induces oxidative stress, responsible for producing the toxic effects in the lysosomes of NL-20 cells.
Collapse
Affiliation(s)
- López-Pérez Lizardo
- Laboratorio de Investigación en Patología Experimental, Hospital Infantil de México Federico Gómez, Avenida Dr. Márquez 162, Colonia Doctores, Cuauhtémoc, 06720 Ciudad de México, Mexico.
| | - Roldán-Barreto Elisa
- Laboratorio de Investigación en Patología Experimental, Hospital Infantil de México Federico Gómez, Avenida Dr. Márquez 162, Colonia Doctores, Cuauhtémoc, 06720 Ciudad de México, Mexico; Posgrado en Ciencias Biológicas, Unidad de Posgrado, Edificio D, 1° Piso, Circuito de Posgrados, Ciudad Universitaria, Coyoacán, C.P, 04510 CDMX, Mexico.
| | - Xochiteotzin-Reyes Tania
- Laboratorio de Investigación en Patología Experimental, Hospital Infantil de México Federico Gómez, Avenida Dr. Márquez 162, Colonia Doctores, Cuauhtémoc, 06720 Ciudad de México, Mexico
| | - Torres-Flores Ulises
- Laboratorio de Investigación en Patología Experimental, Hospital Infantil de México Federico Gómez, Avenida Dr. Márquez 162, Colonia Doctores, Cuauhtémoc, 06720 Ciudad de México, Mexico
| | - Licea-Quintero Brandon
- Laboratorio de Investigación en Patología Experimental, Hospital Infantil de México Federico Gómez, Avenida Dr. Márquez 162, Colonia Doctores, Cuauhtémoc, 06720 Ciudad de México, Mexico.
| | - Monroy-Quintana Regina
- Laboratorio de Investigación en Patología Experimental, Hospital Infantil de México Federico Gómez, Avenida Dr. Márquez 162, Colonia Doctores, Cuauhtémoc, 06720 Ciudad de México, Mexico.
| | - Corona Juan Carlos
- Laboratorio de Investigación en Neurociencias, Hospital Infantil de México Federico Gómez, Avenida Dr. Márquez 162, Colonia Doctores, Cuauhtémoc, 06720 Ciudad de México, Mexico
| | - Zaragoza-Ojeda Montserrat
- Laboratorio de Investigación en Patología Experimental, Hospital Infantil de México Federico Gómez, Avenida Dr. Márquez 162, Colonia Doctores, Cuauhtémoc, 06720 Ciudad de México, Mexico
| | - Arenas-Huertero Francisco
- Laboratorio de Investigación en Patología Experimental, Hospital Infantil de México Federico Gómez, Avenida Dr. Márquez 162, Colonia Doctores, Cuauhtémoc, 06720 Ciudad de México, Mexico.
| |
Collapse
|
3
|
Lee HE, Jung M, Choi K, Jang JH, Hwang SK, Chae S, Lee JH, Mun JY. L-serine restored lysosomal failure in cells derived from patients with BPAN reducing iron accumulation with eliminating lipofuscin. Free Radic Biol Med 2024; 221:273-282. [PMID: 38740102 DOI: 10.1016/j.freeradbiomed.2024.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/30/2024] [Accepted: 05/08/2024] [Indexed: 05/16/2024]
Abstract
Defective mitochondria and autophagy, as well as accumulation of lipid and iron in WDR45 mutant fibroblasts, is related to beta-propeller protein-associated neurodegeneration (BPAN). In this study, we found that enlarged lysosomes in cells derived from patients with BPAN had low enzyme activity, and most of the enlarged lysosomes had an accumulation of iron and oxidized lipid. Cryo-electron tomography revealed elongated lipid accumulation, and spectrometry-based elemental analysis showed that lysosomal iron and oxygen accumulation superimposed with lipid aggregates. Lysosomal lipid aggregates superimposed with autofluorescence as free radical generator, lipofuscin. To eliminate free radical stress by iron accumulation in cells derived from patients with BPAN, we investigated the effects of the iron chelator, 2,2'-bipyridine (bipyridyl, BIP). To study whether the defects in patient-derived cells can be rescued by an iron chelator BIP, we tested whether the level of iron and reactive oxygen species (ROS) in the cells and genes related to oxidative stress were rescued BIP treatment. Although BIP treatment decreased some iron accumulation in the cytoplasm and mitochondria, the accumulation of iron in the lysosomes and levels of cellular ROS were unaffected. In addition, the change of specific RNA levels related to free radical stress in patient fibroblasts was not rescued by BIP. To alleviate free radical stress, we investigated whether l-serine can regulate abnormal structures in cells derived from patients with BPAN through the regulation of free radical stress. l-serine treatment alleviated increase of enlarged lysosomes and iron accumulation and rescued impaired lysosomal activity by reducing oxidized lipid accumulation in the lysosomes of the cells. Lamellated lipids in the lysosomes of the cells were identified as lipofuscin through correlative light and electron microscopy, and l-serine treatment reduced the increase of lipofuscin. These data suggest that l-serine reduces oxidative stress-mediated lysosomal lipid oxidation and iron accumulation by rescuing lysosomal activity.
Collapse
Affiliation(s)
- Hye Eun Lee
- Neural Circuit Research Group, Korea Brain Research Institute, Daegu, South Korea; School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Minkyo Jung
- Neural Circuit Research Group, Korea Brain Research Institute, Daegu, South Korea
| | - Kiju Choi
- Division of Structural Biology, Baobab AiBIO, Incheon, South Korea
| | - Jae Hyuck Jang
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon, South Korea; Electron Microscopy Group for Materials Science, Korea Basic Science Institute, Daejeon, South Korea
| | - Su-Kyeong Hwang
- Department of Pediatrics, School of Medicine, Kyungpook National University, Daegu, South Korea; Astrogen Inc., Techno-Building 313, Kyungpook National University, Daegu, 41566, South Korea
| | - Sehyun Chae
- Division of Chemical Engineering and Bioengineering, College of Art, Culture and Engineering, Kangwon National University, Chuncheon, 24341, South Korea
| | - Jae-Hyeok Lee
- Department of Neurology, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, 50612, South Korea; Medical Research Institute, Pusan National University School of Medicine, Yangsan, 50612, South Korea.
| | - Ji Young Mun
- Neural Circuit Research Group, Korea Brain Research Institute, Daegu, South Korea.
| |
Collapse
|
4
|
Sharma H, Mossman K, Austin RC. Fatal attractions that trigger inflammation and drive atherosclerotic disease. Eur J Clin Invest 2024; 54:e14169. [PMID: 38287209 DOI: 10.1111/eci.14169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/14/2023] [Accepted: 01/09/2024] [Indexed: 01/31/2024]
Abstract
BACKGROUND Atherosclerosis is the salient, underlying cause of cardiovascular diseases, such as arrhythmia, coronary artery disease, cardiomyopathy, pulmonary embolism and myocardial infarction. In recent years, atherosclerosis pathophysiology has evolved from a lipid-based to an inflammation-centric ideology. METHODS This narrative review is comprised of review and original articles that were found through the PubMed search engine. The following search terms or amalgamation of terms were used: "cardiovascular disease," "atherosclerosis," "inflammation," "GRP78," "Hsp60," "oxidative low-density lipoproteins," "aldehyde dehydrogenase," "β2-glycoprotein," "lipoprotein lipase A," "human cytomegalovirus." "SARS-CoV-2," "chlamydia pneumonia," "autophagy," "thrombosis" and "therapeutics." RESULTS Emerging evidence supports the concept that atherosclerosis is associated with the interaction between cell surface expression of stress response chaperones, including GRP78 and Hsp60, and their respective autoantibodies. Moreover, various other autoantigens and their autoantibodies have displayed a compelling connection with the development of atherosclerosis, including oxidative low-density lipoproteins, aldehyde dehydrogenase, β2-glycoprotein and lipoprotein lipase A. Atherosclerosis progression is also concurrent with viral and bacterial activators of various diseases. This narrative review will focus on the contributions of human cytomegalovirus as well as SARS-CoV-2 and chlamydia pneumonia in atherosclerosis development. Notably, the interaction of an autoantigen with their respective autoantibodies or the presence of a foreign antigen can enhance inflammation development, which leads to atherosclerotic lesion progression. CONCLUSION We will highlight and discuss the complex role of the interaction between autoantigens and autoantibodies, and the presence of foreign antigens in the development of atherosclerotic lesions in relationship to pro-inflammatory responses.
Collapse
Affiliation(s)
- Hitesh Sharma
- Division of Nephrology, Department of Medicine, McMaster University, The Research Institute of St. Joe's Hamilton and the Hamilton Centre for Kidney Research, Hamilton, Ontario, Canada
| | - Karen Mossman
- Department of Medicine, Michael DeGroote Institute for Infectious Disease Research and the McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Richard C Austin
- Division of Nephrology, Department of Medicine, McMaster University, The Research Institute of St. Joe's Hamilton and the Hamilton Centre for Kidney Research, Hamilton, Ontario, Canada
| |
Collapse
|
5
|
Park SC, Lee YS, Cho KA, Kim SY, Lee YI, Lee SR, Lim IK. What matters in aging is signaling for responsiveness. Pharmacol Ther 2023; 252:108560. [PMID: 37952903 DOI: 10.1016/j.pharmthera.2023.108560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 10/03/2023] [Accepted: 11/06/2023] [Indexed: 11/14/2023]
Abstract
Biological responsiveness refers to the capacity of living organisms to adapt to changes in both their internal and external environments through physiological and behavioral mechanisms. One of the prominent aspects of aging is the decline in this responsiveness, which can lead to a deterioration in the processes required for maintenance, survival, and growth. The vital link between physiological responsiveness and the essential life processes lies within the signaling systems. To devise effective strategies for controlling the aging process, a comprehensive reevaluation of this connecting loop is imperative. This review aims to explore the impact of aging on signaling systems responsible for responsiveness and introduce a novel perspective on intervening in the aging process by restoring the compromised responsiveness. These innovative mechanistic approaches for modulating altered responsiveness hold the potential to illuminate the development of action plans aimed at controlling the aging process and treating age-related disorders.
Collapse
Affiliation(s)
- Sang Chul Park
- The Future Life & Society Research Center, Advanced Institute of Aging Science, Chonnam National University, Gwangju 61469, Republic of Korea.
| | - Young-Sam Lee
- Department of New Biology, DGIST, Daegu 42988, Republic of Korea; Well Aging Research Center, Division of Biotechnology, DGIST, Daegu 42988, Republic of Korea.
| | - Kyung A Cho
- Department of Biochemistry, Chonnam National University Medical School, Jeollanam-do 58128, Republic of Korea
| | - Sung Young Kim
- Department of Biochemistry, Konkuk University School of Medicine, Seoul 05029, Republic of Korea
| | - Yun-Il Lee
- Well Aging Research Center, Division of Biotechnology, DGIST, Daegu 42988, Republic of Korea; Interdisciplinary Engineering Major, Department of Interdisciplinary Studies, DGIST, Daegu 42988, Republic of Korea
| | - Seung-Rock Lee
- Department of Biochemistry, Chonnam National University Medical School, Jeollanam-do 58128, Republic of Korea; Department of Biomedical Sciences, Research Center for Aging and Geriatrics, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - In Kyoung Lim
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| |
Collapse
|
6
|
Hu J, Nieminen AL, Weemhoff JL, Jaeschke H, Murphy LG, Dent JA, Lemasters JJ. The mitochondrial calcium uniporter mediates mitochondrial Fe 2+ uptake and hepatotoxicity after acetaminophen. Toxicol Appl Pharmacol 2023; 479:116722. [PMID: 37848124 PMCID: PMC10872750 DOI: 10.1016/j.taap.2023.116722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/23/2023] [Accepted: 10/11/2023] [Indexed: 10/19/2023]
Abstract
Acetaminophen (APAP) overdose disrupts hepatocellular lysosomes, which release ferrous iron (Fe2+) that translocates into mitochondria putatively via the mitochondrial calcium uniporter (MCU) to induce oxidative/nitrative stress, the mitochondrial permeability transition (MPT), and hepatotoxicity. To investigate how MCU deficiency affects mitochondrial Fe2+ uptake and hepatotoxicity after APAP overdose, global MCU knockout (KO), hepatocyte specific (hs) MCU KO, and wildtype (WT) mice were treated with an overdose of APAP both in vivo and in vitro. Compared to strain-specific WT mice, serum ALT decreased by 88 and 56%, respectively, in global and hsMCU KO mice at 24 h after APAP (300 mg/kg). Hepatic necrosis also decreased by 84 and 56%. By contrast, when MCU was knocked out in Kupffer cells, ALT release and necrosis were unchanged after overdose APAP. Intravital multiphoton microscopy confirmed loss of viability and mitochondrial depolarization in pericentral hepatocytes of WT mice, which was decreased in MCU KO mice. CYP2E1 expression, hepatic APAP-protein adduct formation, and JNK activation revealed that APAP metabolism was equivalent between WT and MCU KO mice. In cultured hepatocytes after APAP, loss of cell viability decreased in hsMCU KO compared to WT hepatocytes. Using fructose plus glycine to prevent cell killing, mitochondrial Fe2+ increased progressively after APAP, as revealed with mitoferrofluor (MFF), a mitochondrial Fe2+ indicator. By contrast in hsMCU KO hepatocytes, mitochondrial Fe2+ uptake after APAP was suppressed. Rhod-2 measurements showed that Ca2+ did not increase in mitochondria after APAP in either WT or KO hepatocytes. In conclusion, MCU mediates uptake of Fe2+ into mitochondria after APAP and plays a central role in mitochondrial depolarization and cell death during APAP-induced hepatotoxicity.
Collapse
Affiliation(s)
- Jiangting Hu
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC, United States of America; Departments of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States of America
| | - Anna-Liisa Nieminen
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC, United States of America; Departments of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States of America; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States of America
| | - James L Weemhoff
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States of America
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States of America
| | - Laura G Murphy
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC, United States of America; Departments of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States of America
| | - Judith A Dent
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC, United States of America; Departments of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States of America
| | - John J Lemasters
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC, United States of America; Departments of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States of America; Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC, United States of America; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States of America.
| |
Collapse
|
7
|
Madsen MC, Podieh F, Overboom MC, Thijs A, den Heijer M, Hordijk PL. The effect of circulating iron on barrier integrity of primary human endothelial cells. Sci Rep 2023; 13:16857. [PMID: 37803072 PMCID: PMC10558552 DOI: 10.1038/s41598-023-44122-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 10/04/2023] [Indexed: 10/08/2023] Open
Abstract
Iron is hypothesized to be one of the contributors to cardiovascular disease and its levels in the circulation may correlate with cardiovascular risk. The aim of this study is to investigate the mechanisms that underlie the effects of iron on the barrier function of primary human endothelium. We used Human Umbilical Vein Endothelial Cells (HUVEC) to investigate the effects of Fe3+ using electric cell-substrate impedance sensing, microscopy, western blot and immunofluorescence microscopy. Exposure to Fe3+ caused EC elongation and upregulation of stress-induced proteins. Analysis of barrier function showed a dose-dependent drop in endothelial integrity, which was accompanied by Reactive Oxygen Species (ROS) production and could partly be prevented by ROS scavengers. Inhibition of contractility by the ROCK inhibitor Y27632, showed even more effective rescue of barrier integrity. Using western blot, we detected an increase in expression of the small GTPase RhoB, an inducer of EC contraction, and a small decrease in VE-cadherin, suggestive for an iron-induced stress response. Co-stimulation by TNFα and iron, used to investigate the role of low-grade inflammation, revealed an additive, negative effect on barrier integrity, concomitant with an upregulation of pro-inflammatory markers ICAM-1 and RhoB. Iron induces a response in HUVEC that leads to endothelial activation and a pro-inflammatory state measured by loss of barrier integrity which can be reversed by ROS scavengers, combined with inhibition of contractility. These data suggest that ROS-mediated damage of the vascular endothelium could contribute to the increased cardiovascular risk which is associated with elevated levels of circulating iron.
Collapse
Affiliation(s)
- M C Madsen
- Department of Physiology, Amsterdam UMC, De Boelelaan 1118, Amsterdam, 1081 HV, Netherlands.
- Department of Internal Medicine, Amsterdam UMC, Amsterdam, Netherlands.
- Center of Expertise on Gender Dysphoria, Amsterdam UMC, Amsterdam, Netherlands.
| | - F Podieh
- Department of Physiology, Amsterdam UMC, De Boelelaan 1118, Amsterdam, 1081 HV, Netherlands
| | - M C Overboom
- Department of Physiology, Amsterdam UMC, De Boelelaan 1118, Amsterdam, 1081 HV, Netherlands
| | - A Thijs
- Department of Internal Medicine, Amsterdam UMC, Amsterdam, Netherlands
| | - M den Heijer
- Department of Internal Medicine, Amsterdam UMC, Amsterdam, Netherlands
- Center of Expertise on Gender Dysphoria, Amsterdam UMC, Amsterdam, Netherlands
| | - P L Hordijk
- Department of Physiology, Amsterdam UMC, De Boelelaan 1118, Amsterdam, 1081 HV, Netherlands
| |
Collapse
|
8
|
Nousis L, Kanavaros P, Barbouti A. Oxidative Stress-Induced Cellular Senescence: Is Labile Iron the Connecting Link? Antioxidants (Basel) 2023; 12:1250. [PMID: 37371980 DOI: 10.3390/antiox12061250] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Cellular senescence, a cell state characterized by a generally irreversible cell cycle arrest, is implicated in various physiological processes and a wide range of age-related pathologies. Oxidative stress, a condition caused by an imbalance between the production and the elimination of reactive oxygen species (ROS) in cells and tissues, is a common driver of cellular senescence. ROS encompass free radicals and other molecules formed as byproducts of oxygen metabolism, which exhibit varying chemical reactivity. A prerequisite for the generation of strong oxidizing ROS that can damage macromolecules and impair cellular function is the availability of labile (redox-active) iron, which catalyzes the formation of highly reactive free radicals. Targeting labile iron has been proven an effective strategy to counteract the adverse effects of ROS, but evidence concerning cellular senescence is sparse. In the present review article, we discuss aspects of oxidative stress-induced cellular senescence, with special attention to the potential implication of labile iron.
Collapse
Affiliation(s)
- Lambros Nousis
- Department of Hygiene and Epidemiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Panagiotis Kanavaros
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Alexandra Barbouti
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| |
Collapse
|
9
|
Li X, Zhu X, Wei Y. Autophagy in Atherosclerotic Plaque Cells: Targeting NLRP3 Inflammasome for Self-Rescue. Biomolecules 2022; 13:15. [PMID: 36671400 PMCID: PMC9855815 DOI: 10.3390/biom13010015] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Atherosclerosis (AS) is a lipid-driven disorder of the artery intima characterized by the equilibrium between inflammatory and regressive processes. A protein complex called NLRP3 inflammasome is involved in the release of mature interleukin-1β (IL-1β), which is connected to the initiation and progression of atherosclerosis. Autophagy, which includes macroautophagy, chaperone-mediated autophagy (CMA), and microautophagy, is generally recognized as the process by which cells transfer their constituents to lysosomes for digestion. Recent studies have suggested a connection between vascular inflammation and autophagy. This review summarizes the most recent studies and the underlying mechanisms associated with different autophagic pathways and NLRP3 inflammasomes in vascular inflammation, aiming to provide additional evidence for atherosclerosis research.
Collapse
Affiliation(s)
- Xuelian Li
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xianjie Zhu
- Department of Orthopaedic Surgery, Qingdao Municipal Hospital, Qingdao 266011, China
| | - Yumiao Wei
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
10
|
Song W, Bai L, Yang Y, Wang Y, Xu P, Zhao Y, Zhou X, Li X, Xue M. Long-Circulation and Brain Targeted Isoliquiritigenin Micelle Nanoparticles: Formation, Characterization, Tissue Distribution, Pharmacokinetics and Effects for Ischemic Stroke. Int J Nanomedicine 2022; 17:3655-3670. [PMID: 35999993 PMCID: PMC9393037 DOI: 10.2147/ijn.s368528] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 07/28/2022] [Indexed: 11/25/2022] Open
Abstract
Purpose We designed a novel isoliquiritigenin (ISL) loaded micelle prepared with DSPE-PEG2000 as the drug carrier modified with the brain-targeting polypeptide angiopep-2 to improve the poor water solubility and low bioavailability of ISL for the treatment of acute ischemic stroke. Methods Thin film evaporation was used to synthesize the ISL micelles (ISL-M) modified with angiopep-2 as the brain targeted ligands. The morphology of the micelles was observed by the TEM. The particle size and zeta potential were measured via the nanometer particle size analyzer. The drug loading, encapsulation and in vitro release rates of micelles were detected by the HPLC. The UPLC-ESI-MS/MS methods were used to measure the ISL concentrations of ISL in plasma and main tissues after intravenous administration, and compared the pharmacokinetics and tissue distributions between ISL and ISL-M. In the MCAO mice model, the protective effects of ISL and ISL-M were confirmed via the behavioral and molecular biology experiments. Results The results showed that the drug loading of ISL-M was 7.63 ± 2.62%, the encapsulation efficiency was 68.17 ± 6.23%, the particle size was 40.87 ± 4.82 nm, and the zeta potential was -34.23 ± 3.35 mV. The in vitro release experiments showed that ISL-M had good sustained-release effect and pH sensitivity. Compared with ISL monomers, the ISL-M could significantly prolong the in vivo circulation time of ISL and enhance the accumulation in the brain tissues. The ISL-M could ameliorate the brain injury induced by the MCAO mice via inhibition of cellular autophagy and neuronal apoptosis. There were no the cellular structural damages and other adverse effects for ISL-M on the main tissues and organs. Conclusion The ISL-M could serve as a promising and ideal drug candidate for the clinical application of ISL in the treatment of acute ischemic stroke.
Collapse
Affiliation(s)
- Weitong Song
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Lu Bai
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Yuya Yang
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Yongchao Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, People’s Republic of China
| | - Pingxiang Xu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Beijing Laboratory for Biomedical Detection Technology and Instrument, Beijing, People’s Republic of China
| | - Yuming Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Xuelin Zhou
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Beijing Laboratory for Biomedical Detection Technology and Instrument, Beijing, People’s Republic of China
| | - Xiaorong Li
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Beijing Laboratory for Biomedical Detection Technology and Instrument, Beijing, People’s Republic of China
| | - Ming Xue
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Beijing Laboratory for Biomedical Detection Technology and Instrument, Beijing, People’s Republic of China
| |
Collapse
|
11
|
Langeh U, Kumar V, Kumar A, Kumar P, Singh C, Singh A. Cellular and mitochondrial quality control mechanisms in maintaining homeostasis in ageing. Rejuvenation Res 2022; 25:208-222. [PMID: 35850516 DOI: 10.1089/rej.2022.0027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Aging is a natural process in all living organisms defined as destruction of cell function as a result of long-term accumulation of damages. Autophagy is a cellular house safeguard pathway which responsible for degrading damaged cellular organelles. Moreover, it maintains cellular homeostasis, control lifetime, and longevity. Damaged mitochondrial accumulation is a characteristic of aging which associated with neurodegeneration. Mitochondria functions as a principal energy source via supplying ATP through oxidative phosphorylation which serves as fuel for neuronal function. Mitophagy and mitochondrial specific autophagy plays an important role in maintenance of neuronal health via the removal of dysfunctional and aged mitochondria. The mitochondrial QC system involves different strategies for protecting against mitochondrial dysfunction and maintaining healthy mitochondria in cells. Mitochondrial function protection could be a strategy for the promotion of neuroprotection. Mitophagy, could be an effective target for drug discovery. Therefore, further detailed studies for mechanism of mitophagy will advance our mitochondrial phenotype knowledge and understanding to disease pathogenesis. This review mainly focuses on ageing mediated mechanism of autophagy and mitophagy for maintaining the cellular homeostasis and longevity.
Collapse
Affiliation(s)
- Urvashi Langeh
- ISF College of Pharmacy, 75126, Pharmacology, Moga, Punjab, India;
| | - Vishal Kumar
- ISF College of Pharmacy, 75126, Pharmacology, Moga, Punjab, India;
| | | | - Pradeep Kumar
- University of the Witwatersrand, 37707, Department of Pharmacy and Pharmacology, Johannesburg-Braamfontein, Gauteng, South Africa;
| | - Charan Singh
- ISF College of Pharmacy, 75126, Pharmacology, Moga, Punjab, India;
| | - Arti Singh
- ISF College of Pharmacy, 75126, Pharmacology, ISF College of Pharmacy, Department of Pharmacology, Moga, Moga, Punjab, India, 142001;
| |
Collapse
|
12
|
Zhan W, Tian W, Zhang W, Tian H, Sun T. ANGPTL4 attenuates palmitic acid-induced endothelial cell injury by increasing autophagy. Cell Signal 2022; 98:110410. [PMID: 35843572 DOI: 10.1016/j.cellsig.2022.110410] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/30/2022] [Accepted: 07/12/2022] [Indexed: 11/15/2022]
Abstract
ANGPTL4, a member of the angiopoietin-like protein family, is reported to be involved in angiogenesis regulation, lipid metabolism, glucose metabolism and redox reactions, among others. Our previous study showed that the plasma ANGPTL4 level was lower in coronary atherosclerotic heart disease (CAHD) and could be a useful predictor of coronary atherosclerosis. However, the molecular mechanism underlying the function of ANGPTL4 in atherosclerosis is poorly understood. In this study, we found that overexpression of ANGPTL4 in HUVECs enhanced cell proliferation and clone-forming ability in vitro, whereas knockdown of ANGPTL4 resulted in the opposite. The expression of ANGPTL4 was upregulated in palmitic acid (PA)-treated HUVECs. Overexpression of ANGPTL4 protected against PA-induced endothelial injury. Knockdown of ANGPTL4 exacerbated the effects of PA on HUVECs. Mechanistically, we demonstrated that ANGPTL4 promoted endothelial cell proliferation through the regulation of autophagy. Knockdown of ATG7 or 3-MA (an autophagy inhibitor) attenuated the effects of ANGPTL4 on endothelial cells. The serum level of ANGPTL4 was downregulated in atherosclerosis mice. Furthermore, the expression of ANGPTL4 was correlated with autophagy-related proteins in aortic tissues of atherosclerotic mice. ANGPTL4 promotes endothelial cell proliferation and suppresses PA-induced endothelial cell injury by increasing autophagy, which may protect against the development of atherosclerosis.
Collapse
Affiliation(s)
- Wanlin Zhan
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Wei Tian
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Wenlu Zhang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Hua Tian
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China.
| | - Ting Sun
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| |
Collapse
|
13
|
Li Z, Liu Z, Luo M, Li X, Chen H, Gong S, Zhang M, Zhang Y, Liu H, Li X. The pathological role of damaged organelles in renal tubular epithelial cells in the progression of acute kidney injury. Cell Death Dis 2022; 8:239. [PMID: 35501332 PMCID: PMC9061711 DOI: 10.1038/s41420-022-01034-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 12/14/2022]
Abstract
Acute kidney injury (AKI) is a common clinical condition associated with high morbidity and mortality. The pathogenesis of AKI has not been fully elucidated, with a lack of effective treatment. Renal tubular epithelial cells (TECs) play an important role in AKI, and their damage and repair largely determine the progression and prognosis of AKI. In recent decades, it has been found that the mitochondria, endoplasmic reticulum (ER), lysosomes, and other organelles in TECs are damaged to varying degrees in AKI, and that they can influence each other through various signaling mechanisms that affect the recovery of TECs. However, the association between these multifaceted signaling platforms, particularly between mitochondria and lysosomes during AKI remains unclear. This review summarizes the specific pathophysiological mechanisms of the main TECs organelles in the context of AKI, particularly the potential interactions among them, in order to provide insights into possible novel treatment strategies.
Collapse
Affiliation(s)
- Zixian Li
- Institute of Nephrology, and Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Zejian Liu
- Institute of Nephrology, and Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Mianna Luo
- Institute of Nephrology, and Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Xingyu Li
- Institute of Nephrology, and Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Huixia Chen
- Institute of Nephrology, and Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Siqiao Gong
- Institute of Nephrology, and Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Minjie Zhang
- Institute of Nephrology, and Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Yaozhi Zhang
- Institute of Nephrology, and Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Huafeng Liu
- Institute of Nephrology, and Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
| | - Xiaoyu Li
- Institute of Nephrology, and Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
| |
Collapse
|
14
|
Ma J, Yu Q, Han L. The effect of postmortem pH decline rate on caspase-3 activation and tenderness of bovine skeletal muscle during aging. J Food Biochem 2022; 46:e14215. [PMID: 35484879 DOI: 10.1111/jfbc.14215] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/30/2022] [Accepted: 04/05/2022] [Indexed: 01/13/2023]
Abstract
This study aimed to investigate the effect of postmortem pH decline rate on caspase-3 activity and bovine muscle tenderness during aging. Protein denaturation, reactive oxygen species (ROS) levels, mitochondrial apoptosis factors, and shear force were assessed in bovine muscles with different pH decline rates. The results showed that, compared with the slow group, the fast pH decline group had a 1.79% and 1.39% higher sarcoplasmic protein denaturation at 6 and 12 h, respectively (p < .05), and a significantly or extremely significantly higher ROS levels at 6-24 (p < .05, p < .01). Moreover, the fast group had a 14.05%, 22.39%,18.34%, and 25.28% of higher mitochondrial dysfunction at 6, 12, 24, and 72 h, respectively (p < .05); a 16.71%, 23.39%, 17.05%, and 26.61% of lower cytochrome c reduction levels at 6, 12, 24, and 120 h, respectively (p < .05); a significantly increased caspase-3 activity and proportion of apoptotic nuclei at 12-168 and 24-168 h, respectively (p < .05); and a 5.70%, 7.24%, 12.16%, 10.10% and 10.49% decreased shear force at 12, 24, 72,120, and 168 h, respectively (p < .05). These results demonstrated that the fast postmortem pH decline enhanced caspase-3 activation and bovine muscle tenderization by mitochondrial dysfunction-induced apoptosis during aging. PRACTICAL APPLICATIONS: Beef tenderness has long been one of the most important concerns for consumers and the meat industry. To date, the postmortem aging process has been an effective way to improve the tenderness of chilled beef. However, changes in many of the elements in a cattle's muscle after slaughter might actually determine the final tenderness of the meat. The present study suggested that the fast postmortem pH decline could promote the activation of caspase-3 and improve the tenderness of beef during aging. This finding can provide a basis for the meat processing industry to produce beef with high tenderness. In the future, beef tenderness could even be improved by adjusting the glycolytic rate and pH of muscle for a short time after slaughter.
Collapse
Affiliation(s)
- Jibing Ma
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou, China
| | - Qunli Yu
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou, China
| | - Ling Han
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou, China
| |
Collapse
|
15
|
Hua Y, Zhang J, Liu Q, Su J, Zhao Y, Zheng G, Yang Z, Zhuo D, Ma C, Fan G. The Induction of Endothelial Autophagy and Its Role in the Development of Atherosclerosis. Front Cardiovasc Med 2022; 9:831847. [PMID: 35402552 PMCID: PMC8983858 DOI: 10.3389/fcvm.2022.831847] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 02/08/2022] [Indexed: 12/29/2022] Open
Abstract
Increasing attention is now being paid to the important role played by autophagic flux in maintaining normal blood vessel walls. Endothelial cell dysfunction initiates the development of atherosclerosis. In the endothelium, a variety of critical triggers ranging from shear stress to circulating blood lipids promote autophagy. Furthermore, emerging evidence links autophagy to a range of important physiological functions such as redox homeostasis, lipid metabolism, and the secretion of vasomodulatory substances that determine the life and death of endothelial cells. Thus, the promotion of autophagy in endothelial cells may have the potential for treating atherosclerosis. This paper reviews the role of endothelial cells in the pathogenesis of atherosclerosis and explores the molecular mechanisms involved in atherosclerosis development.
Collapse
Affiliation(s)
- Yunqing Hua
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jing Zhang
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qianqian Liu
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jing Su
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yun Zhao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guobin Zheng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Zhihui Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Danping Zhuo
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Chuanrui Ma
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
16
|
Zhang SW, Wang H, Qiu YY, Huang RC, Dong ZC, Zhang L, Zhao LF, Xu HY, Sun WD. Photothermolysis Mediated by Gold Nanorods Conjugated with Epidermal Growth Factor Receptor (EGFR) Monoclonal Antibody Induces Apoptosis via the Mitochondrial Apoptosis Pathway in Laryngeal Squamous Cell Cancer. J Biomed Nanotechnol 2022; 18:754-762. [PMID: 35715914 DOI: 10.1166/jbn.2022.3272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Gold nanorods (AuNRs) have unique optical properties and biological affinity and can be used to treat tumors when conjugated with other protein molecules. Our previous studies have shown that EGFR monoclonal antibody (EGFRmAb)-modified AuNRs exert strong antitumor activity in vitro by inducing apoptosis. In this study, we tested the effects of EGFRmAb-modified AuNRs on laryngeal squamous cell cancer (LSCC) in vitro and in vivo. The in vitro results showed that EGFRmAb-modified AuNRs inhibited NP-69, BEAS-2B and Hep-2 cell growth and induced mitochondria-dependent apoptosis. The mitochondrial membrane potential was reduced, leading to the release of cytochrome C (Cyt C) and consequent activation of the intrinsic mitochondrial apoptosis pathway. Moreover, we observed that the occurrence of mitochondrial apoptosis is related to the destruction of the lysosome-mitochondria axis. To verify the effects in vivo, we also established a laryngeal tumor model in nude mice by subcutaneous transplantation. In model mice treated with EGFRmAb-modified AuNRs and irradiated with an NIR laser, tumor cell apoptosis and tumor growth were inhibited. These results suggest that EGFRmAb-modified AuNRs induced apoptosis through the intrinsic mitochondrial apoptotic pathway and are a potential candidate for cancer therapy.
Collapse
Affiliation(s)
- Shi-Wen Zhang
- First Department of Head and Neck Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
| | - Hao Wang
- First Department of Head and Neck Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
| | - You-Yu Qiu
- First Department of Head and Neck Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
| | - Ren-Chao Huang
- First Department of Head and Neck Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
| | - Zi-Chen Dong
- First Department of Head and Neck Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
| | - Lu Zhang
- First Department of Head and Neck Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
| | - Liu-Fang Zhao
- First Department of Head and Neck Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
| | - Hong-Yang Xu
- First Department of Head and Neck Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
| | - Wei-Di Sun
- First Department of Head and Neck Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
| |
Collapse
|
17
|
Lopez ME, Vacio AM, Cantu J, Holgado A. UNC-33L partially rescues life span and locomotion defects in unc-33 mutants but fails to rescue dauer formation defects. MICROPUBLICATION BIOLOGY 2022; 2022:10.17912/micropub.biology.000515. [PMID: 35088044 PMCID: PMC8787492 DOI: 10.17912/micropub.biology.000515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/12/2022] [Accepted: 01/15/2022] [Indexed: 11/29/2022]
Abstract
Herein, we tested the ability of UNC-33L to rescue dauer formation, lifespan, and locomotion defects of unc-33(mn407) mutants. Results show that the presence of UNC-33L does not rescue the defective dauer phenotype in unc-33(mn407) mutants. However, UNC-33L significantly rescued premature death and uncoordinated locomotion in young unc-33(mn407) adults. The degree of UNC-33L-mediated rescue was less noticeable as the nematodes aged, denoting that both age and the presence of UNC-33L interact in the production of the phenotypes.
Collapse
Affiliation(s)
- Melissa E Lopez
- Department of Biological Sciences, St. Edward's University, Austin, TX
| | - Arianna M Vacio
- Department of Biological Sciences, St. Edward's University, Austin, TX
| | - Jason Cantu
- Department of Biological Sciences, St. Edward's University, Austin, TX
| | - Andrea Holgado
- Department of Biological Sciences, St. Edward's University, Austin, TX,
Correspondence to: Andrea Holgado ()
| |
Collapse
|
18
|
Sirasanagandla SR, Al-Huseini I, Al Mushaiqri M, Al-Abri N, Al-Ghafri F. Maternal resveratrol supplementation ameliorates bisphenol A-induced atherosclerotic lesions formation in adult offspring ApoE -/- mice. 3 Biotech 2022; 12:36. [PMID: 35070626 PMCID: PMC8727657 DOI: 10.1007/s13205-021-03078-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 11/26/2021] [Indexed: 01/03/2023] Open
Abstract
Current evidence suggests that intrauterine bisphenol A (BPA) exposure increases the risk of developing cardiovascular diseases in later stages of life. The beneficial effect of resveratrol (Rsv) on developmental programming of atherosclerosis lesions formation in offspring is seldom reported. Hence, we sought to study the effect of maternal Rsv in ameliorating perinatal BPA exposure-induced atherosclerosis lesions formation in adult offspring using the apolipoprotein E-deficient (ApoE-/-) mice model. The pregnant ApoE-/- mice were allocated into three groups: control, BPA, BPA + resveratrol (BPA + Rsv). The BPA group mice received BPA in their drinking water (1 μg/ml). BPA + Rsv group mice received BPA in their drinking water (1 μg/ml) and were treated orally with Rsv (20 mg kg-1 day-1). All the treatments were continued throughout the gestation and lactation period. Quantitative analysis of Sudan IV-stained aorta revealed a significantly increased area of atherosclerotic lesions in both female (p < 0.01) and male adult offspring mice (p < 0.01) in the BPA group. Supplementation with Rsv significantly reduced the BPA-induced atherosclerotic lesion development in the female offspring mice (p < 0.05). Transmission electron microscopy revealed the presence of a significantly high incidence of autophagic endothelial, smooth muscle, and macrophage cells in the aorta of BPA-exposed mice. Rsv treatment reduced the incidence of autophagic cells in BPA-exposed mice. In conclusion, maternal Rsv supplementation significantly prevents the BPA-induced atherosclerotic lesions formation in a sex-dependent manner potentially by acting as an autophagy modulator. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1007/s13205-021-03078-y.
Collapse
Affiliation(s)
- Srinivasa Rao Sirasanagandla
- grid.412846.d0000 0001 0726 9430Department of Human and Clinical Anatomy, College of Medicine and Health Sciences, Sultan Qaboos University, Al-Khoudh, PO Box 35, Muscat, PC 123 Oman
| | - Isehaq Al-Huseini
- grid.412846.d0000 0001 0726 9430Department of Physiology, College of Medicine and Health Sciences, Sultan Qaboos University, Al-Khoudh, Muscat, 123 Oman
| | - Mohamed Al Mushaiqri
- grid.412846.d0000 0001 0726 9430Department of Human and Clinical Anatomy, College of Medicine and Health Sciences, Sultan Qaboos University, Al-Khoudh, PO Box 35, Muscat, PC 123 Oman
| | - Nadia Al-Abri
- grid.412846.d0000 0001 0726 9430Department of Pathology, College of Medicine and Health Sciences, Sultan Qaboos University, Al-Khoudh, Muscat, 123 Oman
| | - Fatma Al-Ghafri
- grid.412846.d0000 0001 0726 9430Department of Pathology, College of Medicine and Health Sciences, Sultan Qaboos University, Al-Khoudh, Muscat, 123 Oman
| |
Collapse
|
19
|
Tkachenko H, Kurhaluk N, Hetmański T, Włodarkiewicz A, Tomin V. Changes in energetic metabolism and lysosomal destruction in the skeletal muscle and cardiac tissues of pigeons (Columba livia f. urbana) from urban areas of the northern Pomeranian region (Poland). ECOTOXICOLOGY (LONDON, ENGLAND) 2021; 30:1170-1185. [PMID: 34076799 PMCID: PMC8295091 DOI: 10.1007/s10646-021-02423-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/15/2021] [Indexed: 05/05/2023]
Abstract
The aim of the present study was to evaluate the biochemical responses of the skeletal muscle and cardiac tissues of the urban pigeon as a bioindicator organism tested in diverse environments (Szpęgawa as a rural environment and Słupsk as an urban environment, Pomeranian Voivodeship, northern Poland), resulting in changes in the level of lipid peroxidation at the initial and final stages of this process and the activities of Krebs cycle enzymes (succinate dehydrogenase, pyruvate dehydrogenase, isocitrate dehydrogenase, and alfa-ketoglutarate dehydrogenase). Szpęgawa village was chosen due to the intensive use of the European motorway A1 with significant traffic and pollution levels. The concentration of Pb was higher in the soil and feathers of pigeons nesting in the polluted areas (Szpęgawa). Our studies have shown that the presence of lead in soil and feathers of the pigeons resulted in the activation of lipid peroxidation, destabilization and increased activity of lysosomal membranes, and activation of mitochondrial enzymes of the Krebs cycle with energy deficiency (reduction of ATP levels) in cardiac and skeletal muscle tissues simultaneously.
Collapse
Affiliation(s)
- Halyna Tkachenko
- Department of Biology, Institute of Biology and Earth Sciences, Pomeranian University in Słupsk, Słupsk, Poland
| | - Natalia Kurhaluk
- Department of Biology, Institute of Biology and Earth Sciences, Pomeranian University in Słupsk, Słupsk, Poland.
| | - Tomasz Hetmański
- Department of Earth Sciences, Institute of Biology and Earth Sciences, Pomeranian University in Słupsk, Słupsk, Poland
| | - Agnieszka Włodarkiewicz
- Department of Physics, Institute of Science and Technology, Pomeranian University in Słupsk, Słupsk, Poland
| | - Vladimir Tomin
- Department of Physics, Institute of Science and Technology, Pomeranian University in Słupsk, Słupsk, Poland
| |
Collapse
|
20
|
Chen C, Guo Z, Ma G, Ma J, Zhang Z, Yu Q, Han L. Lysosomal Fe2+contributes to myofibrillar protein degradation through mitochondrial-dysfunction-induced apoptosis. Lebensm Wiss Technol 2021. [DOI: 10.1016/j.lwt.2021.111197] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
21
|
Marques ARA, Ramos C, Machado-Oliveira G, Vieira OV. Lysosome (Dys)function in Atherosclerosis-A Big Weight on the Shoulders of a Small Organelle. Front Cell Dev Biol 2021; 9:658995. [PMID: 33855029 PMCID: PMC8039146 DOI: 10.3389/fcell.2021.658995] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/11/2021] [Indexed: 12/15/2022] Open
Abstract
Atherosclerosis is a progressive insidious chronic disease that underlies most of the cardiovascular pathologies, including myocardial infarction and ischemic stroke. The malfunctioning of the lysosomal compartment has a central role in the etiology and pathogenesis of atherosclerosis. Lysosomes are the degradative organelles of mammalian cells and process endogenous and exogenous substrates in a very efficient manner. Dysfunction of these organelles and consequent inefficient degradation of modified low-density lipoproteins (LDL) and apoptotic cells in atherosclerotic lesions have, therefore, numerous deleterious consequences for cellular homeostasis and disease progression. Lysosome dysfunction has been mostly studied in the context of the inherited lysosomal storage disorders (LSDs). However, over the last years it has become increasingly evident that the consequences of this phenomenon are more far-reaching, also influencing the progression of multiple acquired human pathologies, such as neurodegenerative diseases, cancer, and cardiovascular diseases (CVDs). During the formation of atherosclerotic plaques, the lysosomal compartment of the various cells constituting the arterial wall is under severe stress, due to the tremendous amounts of lipoproteins being processed by these cells. The uncontrolled uptake of modified lipoproteins by arterial phagocytic cells, namely macrophages and vascular smooth muscle cells (VSMCs), is the initial step that triggers the pathogenic cascade culminating in the formation of atheroma. These cells become pathogenic "foam cells," which are characterized by dysfunctional lipid-laden lysosomes. Here, we summarize the current knowledge regarding the origin and impact of the malfunctioning of the lysosomal compartment in plaque cells. We further analyze how the field of LSD research may contribute with some insights to the study of CVDs, particularly how therapeutic approaches that target the lysosomes in LSDs could be applied to hamper atherosclerosis progression and associated mortality.
Collapse
Affiliation(s)
- André R A Marques
- iNOVA4Health, Chronic Diseases Research Center (CEDOC), NOVA Medical School (NMS), Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Cristiano Ramos
- iNOVA4Health, Chronic Diseases Research Center (CEDOC), NOVA Medical School (NMS), Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Gisela Machado-Oliveira
- iNOVA4Health, Chronic Diseases Research Center (CEDOC), NOVA Medical School (NMS), Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Otília V Vieira
- iNOVA4Health, Chronic Diseases Research Center (CEDOC), NOVA Medical School (NMS), Universidade NOVA de Lisboa, Lisbon, Portugal
| |
Collapse
|
22
|
Barbouti A, Lagopati N, Veroutis D, Goulas V, Evangelou K, Kanavaros P, Gorgoulis VG, Galaris D. Implication of Dietary Iron-Chelating Bioactive Compounds in Molecular Mechanisms of Oxidative Stress-Induced Cell Ageing. Antioxidants (Basel) 2021; 10:491. [PMID: 33800975 PMCID: PMC8003849 DOI: 10.3390/antiox10030491] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 02/07/2023] Open
Abstract
One of the prevailing perceptions regarding the ageing of cells and organisms is the intracellular gradual accumulation of oxidatively damaged macromolecules, leading to the decline of cell and organ function (free radical theory of ageing). This chemically undefined material known as "lipofuscin," "ceroid," or "age pigment" is mainly formed through unregulated and nonspecific oxidative modifications of cellular macromolecules that are induced by highly reactive free radicals. A necessary precondition for reactive free radical generation and lipofuscin formation is the intracellular availability of ferrous iron (Fe2+) ("labile iron"), catalyzing the conversion of weak oxidants such as peroxides, to extremely reactive ones like hydroxyl (HO•) or alcoxyl (RO•) radicals. If the oxidized materials remain unrepaired for extended periods of time, they can be further oxidized to generate ultimate over-oxidized products that are unable to be repaired, degraded, or exocytosed by the relevant cellular systems. Additionally, over-oxidized materials might inactivate cellular protection and repair mechanisms, thus allowing for futile cycles of increasingly rapid lipofuscin accumulation. In this review paper, we present evidence that the modulation of the labile iron pool distribution by nutritional or pharmacological means represents a hitherto unappreciated target for hampering lipofuscin accumulation and cellular ageing.
Collapse
Affiliation(s)
- Alexandra Barbouti
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece;
| | - Nefeli Lagopati
- Laboratory of Histology-Embryology, Molecular Carcinogenesis Group, Faculty of Medicine, School of Health Science, National and Kapodistrian University of Athens, 75, Mikras Asias Str., Goudi, 11527 Athens, Greece; (N.L.); (D.V.); (K.E.); (V.G.G.)
| | - Dimitris Veroutis
- Laboratory of Histology-Embryology, Molecular Carcinogenesis Group, Faculty of Medicine, School of Health Science, National and Kapodistrian University of Athens, 75, Mikras Asias Str., Goudi, 11527 Athens, Greece; (N.L.); (D.V.); (K.E.); (V.G.G.)
| | - Vlasios Goulas
- Department of Agricultural Sciences, Biotechnology and Food Science, Cyprus University of Technology, 3036 Lemesos, Cyprus;
| | - Konstantinos Evangelou
- Laboratory of Histology-Embryology, Molecular Carcinogenesis Group, Faculty of Medicine, School of Health Science, National and Kapodistrian University of Athens, 75, Mikras Asias Str., Goudi, 11527 Athens, Greece; (N.L.); (D.V.); (K.E.); (V.G.G.)
| | - Panagiotis Kanavaros
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece;
| | - Vassilis G. Gorgoulis
- Laboratory of Histology-Embryology, Molecular Carcinogenesis Group, Faculty of Medicine, School of Health Science, National and Kapodistrian University of Athens, 75, Mikras Asias Str., Goudi, 11527 Athens, Greece; (N.L.); (D.V.); (K.E.); (V.G.G.)
- Biomedical Research Foundation Academy of Athens, 11527 Athens, Greece
- Faculty of Biology, Medicine and Health Manchester Cancer Research Centre, Manchester Academic Health Sciences Centre, University of Manchester, Manchester M13 9PL, UK
- Center for New Biotechnologies and Precision Medicine, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Dimitrios Galaris
- Laboratory of Biological Chemistry, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece;
| |
Collapse
|
23
|
Carresi C, Mollace R, Macrì R, Scicchitano M, Bosco F, Scarano F, Coppoletta AR, Guarnieri L, Ruga S, Zito MC, Nucera S, Gliozzi M, Musolino V, Maiuolo J, Palma E, Mollace V. Oxidative Stress Triggers Defective Autophagy in Endothelial Cells: Role in Atherothrombosis Development. Antioxidants (Basel) 2021; 10:antiox10030387. [PMID: 33807637 PMCID: PMC8001288 DOI: 10.3390/antiox10030387] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 02/18/2021] [Accepted: 03/01/2021] [Indexed: 02/06/2023] Open
Abstract
Atherothrombosis, a multifactorial and multistep artery disorder, represents one of the main causes of morbidity and mortality worldwide. The development and progression of atherothrombosis is closely associated with age, gender and a complex relationship between unhealthy lifestyle habits and several genetic risk factors. The imbalance between oxidative stress and antioxidant defenses is the main biological event leading to the development of a pro-oxidant phenotype, triggering cellular and molecular mechanisms associated with the atherothrombotic process. The pathogenesis of atherosclerosis and its late thrombotic complications involve multiple cellular events such as inflammation, endothelial dysfunction, proliferation of vascular smooth muscle cells (SMCs), extracellular matrix (ECM) alterations, and platelet activation, contributing to chronic pathological remodeling of the vascular wall, atheromatous plague formation, vascular stenosis, and eventually, thrombus growth and propagation. Emerging studies suggest that clotting activation and endothelial cell (EC) dysfunction play key roles in the pathogenesis of atherothrombosis. Furthermore, a growing body of evidence indicates that defective autophagy is closely linked to the overproduction of reactive oxygen species (ROS) which, in turn, are involved in the development and progression of atherosclerotic disease. This topic represents a large field of study aimed at identifying new potential therapeutic targets. In this review, we focus on the major role played by the autophagic pathway induced by oxidative stress in the modulation of EC dysfunction as a background to understand its potential role in the development of atherothrombosis.
Collapse
Affiliation(s)
- Cristina Carresi
- Research for Food Safety & Health IRC-FSH, Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (R.M.); (M.S.); (F.B.); (F.S.); (A.R.C.); (L.G.); (S.R.); (M.C.Z.); (S.N.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
- Correspondence: ; Tel.: +39-09613694128; Fax: +39-09613695737
| | - Rocco Mollace
- Research for Food Safety & Health IRC-FSH, Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (R.M.); (M.S.); (F.B.); (F.S.); (A.R.C.); (L.G.); (S.R.); (M.C.Z.); (S.N.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Roberta Macrì
- Research for Food Safety & Health IRC-FSH, Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (R.M.); (M.S.); (F.B.); (F.S.); (A.R.C.); (L.G.); (S.R.); (M.C.Z.); (S.N.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Miriam Scicchitano
- Research for Food Safety & Health IRC-FSH, Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (R.M.); (M.S.); (F.B.); (F.S.); (A.R.C.); (L.G.); (S.R.); (M.C.Z.); (S.N.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Francesca Bosco
- Research for Food Safety & Health IRC-FSH, Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (R.M.); (M.S.); (F.B.); (F.S.); (A.R.C.); (L.G.); (S.R.); (M.C.Z.); (S.N.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Federica Scarano
- Research for Food Safety & Health IRC-FSH, Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (R.M.); (M.S.); (F.B.); (F.S.); (A.R.C.); (L.G.); (S.R.); (M.C.Z.); (S.N.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Anna Rita Coppoletta
- Research for Food Safety & Health IRC-FSH, Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (R.M.); (M.S.); (F.B.); (F.S.); (A.R.C.); (L.G.); (S.R.); (M.C.Z.); (S.N.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Lorenza Guarnieri
- Research for Food Safety & Health IRC-FSH, Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (R.M.); (M.S.); (F.B.); (F.S.); (A.R.C.); (L.G.); (S.R.); (M.C.Z.); (S.N.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Stefano Ruga
- Research for Food Safety & Health IRC-FSH, Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (R.M.); (M.S.); (F.B.); (F.S.); (A.R.C.); (L.G.); (S.R.); (M.C.Z.); (S.N.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Maria Caterina Zito
- Research for Food Safety & Health IRC-FSH, Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (R.M.); (M.S.); (F.B.); (F.S.); (A.R.C.); (L.G.); (S.R.); (M.C.Z.); (S.N.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Saverio Nucera
- Research for Food Safety & Health IRC-FSH, Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (R.M.); (M.S.); (F.B.); (F.S.); (A.R.C.); (L.G.); (S.R.); (M.C.Z.); (S.N.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Micaela Gliozzi
- Research for Food Safety & Health IRC-FSH, Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (R.M.); (M.S.); (F.B.); (F.S.); (A.R.C.); (L.G.); (S.R.); (M.C.Z.); (S.N.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Vincenzo Musolino
- Research for Food Safety & Health IRC-FSH, Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (R.M.); (M.S.); (F.B.); (F.S.); (A.R.C.); (L.G.); (S.R.); (M.C.Z.); (S.N.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Jessica Maiuolo
- Research for Food Safety & Health IRC-FSH, Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (R.M.); (M.S.); (F.B.); (F.S.); (A.R.C.); (L.G.); (S.R.); (M.C.Z.); (S.N.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Ernesto Palma
- Research for Food Safety & Health IRC-FSH, Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (R.M.); (M.S.); (F.B.); (F.S.); (A.R.C.); (L.G.); (S.R.); (M.C.Z.); (S.N.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88100 Catanzaro, Italy
| | - Vincenzo Mollace
- Research for Food Safety & Health IRC-FSH, Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (R.M.); (M.S.); (F.B.); (F.S.); (A.R.C.); (L.G.); (S.R.); (M.C.Z.); (S.N.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88100 Catanzaro, Italy
| |
Collapse
|
24
|
Onyango IG, Bennett JP, Stokin GB. Regulation of neuronal bioenergetics as a therapeutic strategy in neurodegenerative diseases. Neural Regen Res 2021; 16:1467-1482. [PMID: 33433460 PMCID: PMC8323696 DOI: 10.4103/1673-5374.303007] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis are a heterogeneous group of debilitating disorders with multifactorial etiologies and pathogeneses that manifest distinct molecular mechanisms and clinical manifestations with abnormal protein dynamics and impaired bioenergetics. Mitochondrial dysfunction is emerging as an important feature in the etiopathogenesis of these age-related neurodegenerative diseases. The prevalence and incidence of these diseases is on the rise with the increasing global population and average lifespan. Although many therapeutic approaches have been tested, there are currently no effective treatment routes for the prevention or cure of these diseases. We present the current status of our knowledge and understanding of the involvement of mitochondrial dysfunction in these diseases and highlight recent advances in novel therapeutic strategies targeting neuronal bioenergetics as potential approach for treating these diseases.
Collapse
Affiliation(s)
- Isaac G Onyango
- Center for Translational Medicine, International Clinical Research Centre (ICRC), St. Anne's University Hospital, Brno, Czech Republic
| | - James P Bennett
- Neurodegeneration Therapeutics, 3050A Berkmar Drive, Charlottesville, VA, USA
| | - Gorazd B Stokin
- Center for Translational Medicine, International Clinical Research Centre (ICRC), St. Anne's University Hospital, Brno, Czech Republic
| |
Collapse
|
25
|
Forgotten partners and function regulators of inducible metallothioneins. Arh Hig Rada Toksikol 2020; 70:256-264. [PMID: 32623859 DOI: 10.2478/aiht-2019-70-3317] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 11/01/2019] [Indexed: 11/21/2022] Open
Abstract
Metallothioneins are peculiar cysteine rich, heat resistant, small cellular plasma proteins expressed through almost all life forms. The currently established biological functions of metallothioneins are the homeostasis of essential metals and protection against toxic transitional metals (TM) alongside defence from oxidative stress by direct scavenging of reactive oxygen and nitrogen species (ROS and RNS). In mammals, among the four main evolutionary conserved forms, only the ubiquitously expressed metallothionein 1 and 2 (here abbreviated as MT) are inducible by TM, oxidative stress, glucocorticoids and starvation among various other stimuli. However, more than sixty years after being discovered, metallothioneins still bear unresolved issues about their possible physiological function and regulation. The biological function of MTs has still not been associated with the in vitro-demonstrated capacity of MT interaction with cellular molecules glutathione (GSH) or adenosine triphosphate (ATP), or with the possibility of direct iron-MT binding in the reducing intracellular environment of some organelles, e.g. lysosomes. Iron as the most abundant cellular TM is also one of the main physiological sources of ROS. Moreover, iron exhibits strain, sex and age differences that reflected ROS generation and MT induction in (patho)physiology and toxicology studies. A recent study showed that iron sex differences follows expression of both ferritin and MT leading to wide implications from essential TM interconnectivity to aging. This review places emphasis on biochemically proven but physiologically ignored interactions of MT with iron to stimulate advanced research for establishing a wide frame of the biological roles of MTs important for health and longevity.
Collapse
|
26
|
(+)-Clausenamide protects against drug-induced liver injury by inhibiting hepatocyte ferroptosis. Cell Death Dis 2020; 11:781. [PMID: 32951003 PMCID: PMC7502081 DOI: 10.1038/s41419-020-02961-5] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 08/13/2020] [Accepted: 08/27/2020] [Indexed: 02/06/2023]
Abstract
Drug-induced liver injury is the major cause of acute liver failure. However, the underlying mechanisms seem to be multifaceted and remain poorly understood, resulting in few effective therapies. Here, we report a novel mechanism that contributes to acetaminophen-induced hepatotoxicity through the induction of ferroptosis, a distinctive form of programmed cell death. We subsequently identified therapies protective against acetaminophen-induced liver damage and found that (+)-clausenamide ((+)-CLA), an active alkaloid isolated from the leaves of Clausena lansium (Lour.) Skeels, inhibited acetaminophen-induced hepatocyte ferroptosis both in vivo and in vitro. Consistently, (+)-CLA significantly alleviated acetaminophen-induced or erastin-induced hepatic pathological damages, hepatic dysfunctions and excessive production of lipid peroxidation both in cultured hepatic cell lines and mouse liver. Furthermore, treatment with (+)-CLA reduced the mRNA level of prostaglandin endoperoxide synthase 2 while it increased the protein level of glutathione peroxidase 4 in hepatocytes and mouse liver, confirming that the inhibition of ferroptosis contributes to the protective effect of (+)-CLA on drug-induced liver damage. We further revealed that (+)-CLA specifically reacted with the Cys-151 residue of Keap1, which blocked Nrf2 ubiquitylation and resulted in an increased Nrf2 stability, thereby leading to the activation of the Keap1–Nrf2 pathway to prevent drug-induced hepatocyte ferroptosis. Our studies illustrate the innovative mechanisms of acetaminophen-induced liver damage and present a novel intervention strategy to treat drug overdose by using (+)-CLA.
Collapse
|
27
|
Li C, Hu W, Wang J, Song X, Xiong X, Liu Z. A highly specific probe for the imaging of inflammation-induced endogenous nitric oxide produced during the stroke process. Analyst 2020; 145:6125-6129. [PMID: 32851996 DOI: 10.1039/d0an00824a] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In this study, a turn-on two-photon fluorescent probe (Lyso-TP-NO) for nitric oxide (NO) was developed. It was synthesized using 4-ethylamino-1,8-naphthalimide as the two-photon fluorophore and N-methylaniline moiety as the reaction site. The probe and fluorophore were tested under one- and two-photon modes. The fluorescence intensity of the system was enhanced 23.1-fold after reacting with NO in the one-photon mode. However, the maximal two-photon action cross-section value of 200 GM was obtained under excitation at 840 nm. The probe exhibits high selectivity and sensitivity over other reactive oxygen species (ROS) and reactive nitrogen species (RNS), with a detection limit as low as 3.3 nM. The two-photon fluorescence imaging of living cells and mouse brain tissues can capture inflammation-induced endogenous NO production in lysosomes during stroke occurrence.
Collapse
Affiliation(s)
- Chenchen Li
- Hubei Key Laboratory of Biological Resources Protection and Utilization, Hubei Minzu University, Enshi 445000, China.
| | | | | | | | | | | |
Collapse
|
28
|
Latorraca LB, Feitosa WB, Mariano C, Moura MT, Fontes PK, Nogueira MFG, Paula-Lopes FF. Autophagy is a pro-survival adaptive response to heat shock in bovine cumulus-oocyte complexes. Sci Rep 2020; 10:13711. [PMID: 32792582 PMCID: PMC7426922 DOI: 10.1038/s41598-020-69939-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 06/04/2020] [Indexed: 12/11/2022] Open
Abstract
Autophagy is a physiological mechanism that can be activated under stress conditions. However, the role of autophagy during oocyte maturation has been poorly investigated. Therefore, this study characterized the role of autophagy on developmental competence and gene expression of bovine oocytes exposed to heat shock (HS). Cumulus-oocyte-complexes (COCs) were matured at Control (38.5 °C) and HS (41 °C) temperatures in the presence of 0 and 10 mM 3-methyladenine (3MA; autophagy inhibitor). Western blotting analysis revealed that HS increased autophagy marker LC3-II/LC3-I ratio in oocytes. However, there was no effect of temperature for oocytes matured with 3MA. On cumulus cells, 3MA reduced LC3-II/LC3-I ratio regardless of temperature. Inhibition of autophagy during IVM of heat-shocked oocytes (3MA-41 °C) reduced cleavage and blastocyst rates compared to standard in vitro matured heat-shocked oocytes (IVM-41 °C). Therefore, the magnitude of HS detrimental effects was greater in the presence of autophagy inhibitor. Oocyte maturation under 3MA-41 °C reduced mRNA abundance for genes related to energy metabolism (MTIF3), heat shock response (HSF1), and oocyte maturation (HAS2 and GREM1). In conclusion, autophagy is a stress response induced on heat shocked oocytes. Inhibition of autophagy modulated key functional processes rendering the oocyte more susceptible to the deleterious effects of heat shock.
Collapse
Affiliation(s)
- Lais B Latorraca
- Department of Pharmacology, Institute of Bioscience, São Paulo State University (UNESP), District of Rubião Junior S/N, Botucatu, São Paulo, 18618970, Brazil
| | - Weber B Feitosa
- Department of Biological Sciences, Federal University of São Paulo, Diadema, São Paulo, 09972270, Brazil
| | - Camila Mariano
- Department of Biological Sciences, Federal University of São Paulo, Diadema, São Paulo, 09972270, Brazil
| | - Marcelo T Moura
- Department of Biological Sciences, Federal University of São Paulo, Diadema, São Paulo, 09972270, Brazil
| | - Patrícia K Fontes
- Department of Pharmacology, Institute of Bioscience, São Paulo State University (UNESP), District of Rubião Junior S/N, Botucatu, São Paulo, 18618970, Brazil
| | - Marcelo F G Nogueira
- Department of Pharmacology, Institute of Bioscience, São Paulo State University (UNESP), District of Rubião Junior S/N, Botucatu, São Paulo, 18618970, Brazil
- Department of Biological Sciences, School of Sciences and Languages, UNESP, Assis, São Paulo, Brazil
| | - Fabíola F Paula-Lopes
- Department of Pharmacology, Institute of Bioscience, São Paulo State University (UNESP), District of Rubião Junior S/N, Botucatu, São Paulo, 18618970, Brazil.
- Department of Biological Sciences, Federal University of São Paulo, Diadema, São Paulo, 09972270, Brazil.
| |
Collapse
|
29
|
Zheng HJ, Zhang X, Guo J, Zhang W, Ai S, Zhang F, Wang Y, Liu WJ. Lysosomal dysfunction-induced autophagic stress in diabetic kidney disease. J Cell Mol Med 2020; 24:8276-8290. [PMID: 32583573 PMCID: PMC7412686 DOI: 10.1111/jcmm.15301] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/26/2020] [Accepted: 04/02/2020] [Indexed: 12/14/2022] Open
Abstract
The catabolic process that delivers cytoplasmic constituents to the lysosome for degradation, known as autophagy, is thought to act as a cytoprotective mechanism in response to stress or as a pathogenic process contributing towards cell death. Animal and human studies have shown that autophagy is substantially dysregulated in renal cells in diabetes, suggesting that activating autophagy could be a therapeutic intervention. However, under prolonged hyperglycaemia with impaired lysosome function, increased autophagy induction that exceeds the degradative capacity in cells could contribute toward autophagic stress or even the stagnation of autophagy, leading to renal cytotoxicity. Since lysosomal function is likely key to linking the dual cytoprotective and cytotoxic actions of autophagy, it is important to develop novel pharmacological agents that improve lysosomal function and restore autophagic flux. In this review, we first provide an overview of the autophagic-lysosomal pathway, particularly focusing on stages of lysosomal degradation during autophagy. Then, we discuss the role of adaptive autophagy and autophagic stress based on lysosomal function. More importantly, we focus on the role of autophagic stress induced by lysosomal dysfunction according to the pathogenic factors (including high glucose, advanced glycation end products (AGEs), urinary protein, excessive reactive oxygen species (ROS) and lipid overload) in diabetic kidney disease (DKD), respectively. Finally, therapeutic possibilities aimed at lysosomal restoration in DKD are introduced.
Collapse
Affiliation(s)
- Hui Juan Zheng
- Renal Research Institution of Beijing University of Chinese Medicine, Beijing, China.,Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Xueqin Zhang
- Renal Research Institution of Beijing University of Chinese Medicine, Beijing, China.,Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Jing Guo
- Renal Research Institution of Beijing University of Chinese Medicine, Beijing, China.,Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Wenting Zhang
- Renal Research Institution of Beijing University of Chinese Medicine, Beijing, China.,Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Sinan Ai
- Renal Research Institution of Beijing University of Chinese Medicine, Beijing, China.,Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Fan Zhang
- Renal Research Institution of Beijing University of Chinese Medicine, Beijing, China.,Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Yaoxian Wang
- Renal Research Institution of Beijing University of Chinese Medicine, Beijing, China.,Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Wei Jing Liu
- Renal Research Institution of Beijing University of Chinese Medicine, Beijing, China.,Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China.,Institute of Nephrology, and Zhanjiang Key Laboratory of Prevention and Management of Chronic Kidney Disease, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
30
|
Peters AE, Mihalas BP, Bromfield EG, Roman SD, Nixon B, Sutherland JM. Autophagy in Female Fertility: A Role in Oxidative Stress and Aging. Antioxid Redox Signal 2020; 32:550-568. [PMID: 31892284 DOI: 10.1089/ars.2019.7986] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Significance: The precipitous age-related decline in female fertility is intimately associated with a reduction in both the quantity and quality of the germline (oocytes). Although complex etiologies undoubtedly contribute to the deterioration of oocyte quality, increasing attention has focused on the pervasive impact of oxidative stress. Indeed, the prolonged lifespan of the meiotically arrested oocyte places this cell at heightened risk of oxidative lesions, which commonly manifest in dysregulation of protein homeostasis (proteostasis). Although oocytes are able to mitigate this threat via the mobilization of a sophisticated network of surveillance, repair, and proteolytic pathways, these defenses are themselves prone to age-related defects, reducing their capacity to eliminate oxidatively damaged proteins. Recent Advances: Here, we give consideration to the quality control mechanisms identified within the ovary that afford protection to the female germline. Our primary focus is to review recent advances in our understanding of the autophagy pathway and its contribution to promoting oocyte longevity and modulating pathophysiological responses to oxidative stress. In addition, we explore the therapeutic potential of emerging strategies to fortify autophagic activity. Critical Issues: The complex interplay of oxidative stress and autophagy has yet to be fully elucidated within the context of the aging oocyte and surrounding ovarian environment. Future Directions: Emerging evidence provides a strong impetus to resolve the causal link between autophagy and oxidative stress-driven pathologies in the aging oocyte. Such research may ultimately inform novel therapeutic strategies to combat the age-related loss of female fertility via fortification of intrinsic autophagic activity.
Collapse
Affiliation(s)
- Alexandra E Peters
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science and Pharmacy and Environmental and Life Sciences, University of Newcastle, Callaghan, Australia.,Hunter Medical Research Institute, New Lambton Heights, Australia
| | - Bettina P Mihalas
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science and Pharmacy and Environmental and Life Sciences, University of Newcastle, Callaghan, Australia.,Hunter Medical Research Institute, New Lambton Heights, Australia
| | - Elizabeth G Bromfield
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science and Pharmacy and Environmental and Life Sciences, University of Newcastle, Callaghan, Australia.,Hunter Medical Research Institute, New Lambton Heights, Australia.,Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Shaun D Roman
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science and Pharmacy and Environmental and Life Sciences, University of Newcastle, Callaghan, Australia.,Hunter Medical Research Institute, New Lambton Heights, Australia.,Priority Research Centre for Drug Development, University of Newcastle, Callaghan, Australia
| | - Brett Nixon
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science and Pharmacy and Environmental and Life Sciences, University of Newcastle, Callaghan, Australia.,Hunter Medical Research Institute, New Lambton Heights, Australia
| | - Jessie M Sutherland
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science and Pharmacy and Environmental and Life Sciences, University of Newcastle, Callaghan, Australia.,Hunter Medical Research Institute, New Lambton Heights, Australia
| |
Collapse
|
31
|
Hu J, Lemasters JJ. Suppression of iron mobilization from lysosomes to mitochondria attenuates liver injury after acetaminophen overdose in vivo in mice: Protection by minocycline. Toxicol Appl Pharmacol 2020; 392:114930. [PMID: 32109512 DOI: 10.1016/j.taap.2020.114930] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 02/19/2020] [Accepted: 02/24/2020] [Indexed: 12/20/2022]
Abstract
Acetaminophen (APAP) overdose causes hepatotoxicity involving mitochondrial dysfunction. Previous studies showed that translocation of Fe2+ from lysosomes into mitochondria by the mitochondrial Ca2+ uniporter (MCU) promotes the mitochondrial permeability transition (MPT) after APAP. Here, our Aim was to assess protection by iron chelation and MCU inhibition against APAP hepatotoxicity in mice. C57BL/6 mice and hepatocytes were administered toxic doses of APAP with and without starch-desferal (an iron chelator), minocycline (MCU inhibitor), or N-acetylcysteine (NAC). In mice, starch-desferal and minocycline pretreatment decreased ALT and liver necrosis after APAP by >60%. At 24 h after APAP, loss of fluorescence of mitochondrial rhodamine 123 occurred in pericentral hepatocytes often accompanied by propidium iodide labeling, indicating mitochondrial depolarization and cell death. Starch-desferal and minocycline pretreatment decreased mitochondrial depolarization and cell death by more than half. In cultured hepatocytes, cell killing at 10 h after APAP decreased from 83% to 49%, 35% and 27%, respectively, by 1 h posttreatment with minocycline, NAC, and minocycline plus NAC. With 4 h posttreatment in vivo, minocycline and minocycline plus NAC decreased ALT and necrosis by ~20% and ~50%, respectively, but NAC alone was not effective. In conclusion, minocycline and starch-desferal decrease mitochondrial dysfunction and severe liver injury after APAP overdose, suggesting that the MPT is likely triggered by iron uptake into mitochondria through MCU. In vivo, minocycline and minocycline plus NAC posttreatment after APAP protect at later time points than NAC alone, indicating that minocycline has a longer window of efficacy than NAC.
Collapse
Affiliation(s)
- Jiangting Hu
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC 29425, United States of America; Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, United States of America
| | - John J Lemasters
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC 29425, United States of America; Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, United States of America; Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, United States of America.
| |
Collapse
|
32
|
Cathepsin B pH-Dependent Activity Is Involved in Lysosomal Dysregulation in Atrophic Age-Related Macular Degeneration. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:5637075. [PMID: 31885803 PMCID: PMC6925809 DOI: 10.1155/2019/5637075] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 10/11/2019] [Accepted: 11/05/2019] [Indexed: 12/16/2022]
Abstract
Age-related macular degeneration (AMD) is characterized by retinal pigment epithelial (RPE) cell dysfunction beginning at early stages of the disease. The lack of an appropriate in vitro model is a major limitation in understanding the mechanisms leading to the occurrence of AMD. This study compared human-induced pluripotent stem cell- (hiPSC-) RPE cells derived from atrophic AMD patients (77 y/o ± 7) to hiPSC-RPE cells derived from healthy elderly individuals with no drusen or pigmentary alteration (62.5 y/o ± 17.5). Control and AMD hiPSC-RPE cell lines were characterized by immunofluorescence, flow cytometry, and electronic microscopy. The toxicity level of iron after Fe-NTA treatment was evaluated by an MTT test and by the detection of dichloro-dihydro-fluorescein diacetate. Twelve hiPSC-RPE cell lines (6 AMD and 6 controls) were used for the experiment. Under basal conditions, all hiPSC-RPE cells expressed a phenotypic profile of senescent cells with rounded mitochondria at passage 2. However, the treatment with Fe-NTA induced higher reactive oxygen species production and cell death in hiPSC-RPE AMD cells than in hiPSC-RPE Control cells. Interestingly, functional analysis showed differences in lysosomal activity between the two populations. Indeed, Cathepsin B activity was higher in hiPSC-RPE AMD cells compared to hiPSC-RPE Control cells in basal condition and link to a pH more acidic in this cell population. Moreover, oxidative stress exposure leads to an increase of Cathepsin D immature form levels in both populations, but in a higher proportion in hiPSC-RPE AMD cells. These findings could demonstrate that hiPSC-RPE AMD cells have a typical disease phenotype compared to hiPSC-RPE Control cells.
Collapse
|
33
|
Dhakal S, Kushairi N, Phan CW, Adhikari B, Sabaratnam V, Macreadie I. Dietary Polyphenols: A Multifactorial Strategy to Target Alzheimer's Disease. Int J Mol Sci 2019; 20:E5090. [PMID: 31615073 PMCID: PMC6834216 DOI: 10.3390/ijms20205090] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/11/2019] [Accepted: 10/11/2019] [Indexed: 02/06/2023] Open
Abstract
Ageing is an inevitable fundamental process for people and is their greatest risk factor for neurodegenerative disease. The ageing processes bring changes in cells that can drive the organisms to experience loss of nutrient sensing, disrupted cellular functions, increased oxidative stress, loss of cellular homeostasis, genomic instability, accumulation of misfolded protein, impaired cellular defenses and telomere shortening. Perturbation of these vital cellular processes in neuronal cells can lead to life threatening neurological disorders like Alzheimer's Disease, Parkinson's Disease, Huntington's Disease, Lewy body dementia, etc. Alzheimer's Disease is the most frequent cause of deaths in the elderly population. Various therapeutic molecules have been designed to overcome the social, economic and health care burden caused by Alzheimer's Disease. Almost all the chemical compounds in clinical practice have been found to treat symptoms only limiting them to palliative care. The reason behind such imperfect drugs may result from the inefficiencies of the current drugs to target the cause of the disease. Here, we review the potential role of antioxidant polyphenolic compounds that could possibly be the most effective preventative strategy against Alzheimer's Disease.
Collapse
Affiliation(s)
- Sudip Dhakal
- School of Science, RMIT University, Bundoora, Victoria 3083, Australia.
| | - Naufal Kushairi
- Mushroom Research Centre, University of Malaya, 50603 Kuala Lumpur, Malaysia.
- Department of Anatomy, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| | - Chia Wei Phan
- Mushroom Research Centre, University of Malaya, 50603 Kuala Lumpur, Malaysia.
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| | - Benu Adhikari
- School of Science, RMIT University, Bundoora, Victoria 3083, Australia.
| | - Vikineswary Sabaratnam
- Mushroom Research Centre, University of Malaya, 50603 Kuala Lumpur, Malaysia.
- Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| | - Ian Macreadie
- School of Science, RMIT University, Bundoora, Victoria 3083, Australia.
| |
Collapse
|
34
|
Lv H, Shang P. The significance, trafficking and determination of labile iron in cytosol, mitochondria and lysosomes. Metallomics 2019; 10:899-916. [PMID: 29923582 DOI: 10.1039/c8mt00048d] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The labile iron pool (LIP) is a pool of chelatable and redox-active iron, not only essential for a wide variety of metabolic process, but also as a catalyst in the Fenton reaction, causing the release of hazardous reactive oxygen species (ROS) with potential for inducing oxidative stress and cell damage. The cellular LIP represents the entirety of every heterogenous sub-pool of iron, not only present in the cytosol, but also in mitochondria, lysosomes and the nucleus, which have all been detected and characterized by various fluorescent methods. Accumulated evidence indicates that alterations in the intracellular LIP can substantially contribute to a variety of injurious processes and initiate pathological development. Herein, we present our understanding of the role of the cellular LIP. To fully review the LIP, firstly, the significance of cellular labile iron in different subcellular compartments is presented. And then, the trafficking processes of cellular labile iron between/in cytosol, mitochondria and lysosomes are discussed in detail. Then, the recent progress in uncovering and assessing the cellular LIP by fluorescent methods have been noted. Overall, this summary may help to comprehensively envision the important physiological and pathological roles of the LIP and shed light on profiling the LIP in a real-time and nondestructive manner with fluorescent methods. Undoubtedly, with the advent and development of iron biology, a better understanding of iron, especially the LIP, may also enhance treatments for iron-related diseases.
Collapse
Affiliation(s)
- Huanhuan Lv
- School of Life Sciences, Northwestern Polytechnical University, Youyi Xilu 127, 7100072, Xi'an, Shaanxi, China
| | | |
Collapse
|
35
|
Zhao Z. Iron and oxidizing species in oxidative stress and Alzheimer's disease. Aging Med (Milton) 2019; 2:82-87. [PMID: 31942516 PMCID: PMC6880687 DOI: 10.1002/agm2.12074] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 05/30/2019] [Accepted: 05/31/2019] [Indexed: 12/22/2022] Open
Abstract
Iron species can participate in the Fenton or Fenton-like reaction to generate oxidizing species that can cause oxidative damages to biomolecules and induce oxidative stress in the body. Furthermore, iron accumulation and oxidative stress have been shown to associate with the pathological progression of neurodegenerative disorders, including Alzheimer's disease (AD) and Parkinson's disease (PD). In this review, the role of iron species in generating the most deleterious free radical species (ie, hydroxyl radical) and effects of this species in causing oxidative stress in vivo are described. The implications of oxidative stress and the recently recognized cell death pathway (ie, ferroptosis) to AD are addressed. Strategies to combat this neurodegenerative disease, such as iron chelation and antioxidant therapies, and future research directions on this aspect are also discussed.
Collapse
Affiliation(s)
- Zhongwei Zhao
- School of Pharmaceutical SciencesCapital Medical UniversityBeijingChina
| |
Collapse
|
36
|
Abstract
The entry of pathogens into nonphagocytic host cells has received much attention in the past three decades, revealing a vast array of strategies employed by bacteria and viruses. A method of internalization that has been extensively studied in the context of viral infections is the use of the clathrin-mediated pathway. More recently, a role for clathrin in the entry of some intracellular bacterial pathogens was discovered. Classically, clathrin-mediated endocytosis was thought to accommodate internalization only of particles smaller than 150 nm; however, this was challenged upon the discovery that Listeria monocytogenes requires clathrin to enter eukaryotic cells. Now, with discoveries that clathrin is required during other stages of some bacterial infections, another paradigm shift is occurring. There is a more diverse impact of clathrin during infection than previously thought. Much of the recent data describing clathrin utilization in processes such as bacterial attachment, cell-to-cell spread and intracellular growth may be due to newly discovered divergent roles of clathrin in the cell. Not only does clathrin act to facilitate endocytosis from the plasma membrane, but it also participates in budding from endosomes and the Golgi apparatus and in mitosis. Here, the manipulation of clathrin processes by bacterial pathogens, including its traditional role during invasion and alternative ways in which clathrin supports bacterial infection, is discussed. Researching clathrin in the context of bacterial infections will reveal new insights that inform our understanding of host-pathogen interactions and allow researchers to fully appreciate the diverse roles of clathrin in the eukaryotic cell.
Collapse
Affiliation(s)
- Eleanor A Latomanski
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Hayley J Newton
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| |
Collapse
|
37
|
Role of autophagy in atherosclerosis: foe or friend? JOURNAL OF INFLAMMATION-LONDON 2019; 16:8. [PMID: 31073280 PMCID: PMC6498679 DOI: 10.1186/s12950-019-0212-4] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 04/16/2019] [Indexed: 12/16/2022]
Abstract
Athrosclerosis is conceived as a chronic inflammatory status affecting cells from vascular walls. Different mechanisms and pathological features are evident at the onset of atherosclerotic changes via the engaging different cells from the vascular wall and circulatory cells. Attempts are currently focused on the detection of cell compensatory mechanisms against atherosclerotic changes to restore cell function and/or postpone severe vasculitis. Autophagy is an intracellular self-digesting process commonly protrudes exhausted organelles and injured cytoplasmic constituents via double-lipid bilayer membrane vesicles out the target cells. Recent investigations point to the critical and defensive role of autophagy in the vascular cells behavioral function such as endothelial cells and smooth muscle cells against different insults. Autophagy response and related effectors could be modulated in the favor to restore cell function and reduce pro-inflammatory status under pathological conditions. In this review, the recent findings were collected regarding the role of autophagy during atherosclerotic changes. We aimed to answer the question of how autophagy stimulation and/or inhibition could provide a promising effect on developing a sophisticated treatment for AS.
Collapse
|
38
|
Martinet W, Coornaert I, Puylaert P, De Meyer GRY. Macrophage Death as a Pharmacological Target in Atherosclerosis. Front Pharmacol 2019; 10:306. [PMID: 31019462 PMCID: PMC6458279 DOI: 10.3389/fphar.2019.00306] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 03/12/2019] [Indexed: 12/20/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disorder characterized by the gradual build-up of plaques within the vessel wall of middle-sized and large arteries. Over the past decades, treatment of atherosclerosis mainly focused on lowering lipid levels, which can be accomplished by the use of statins. However, some patients do not respond sufficiently to statin therapy and therefore still have a residual cardiovascular risk. This issue highlights the need for novel therapeutic strategies. As macrophages are implicated in all stages of atherosclerotic lesion development, they represent an important alternative drug target. A variety of anti-inflammatory strategies have recently emerged to treat or prevent atherosclerosis. Here, we review the canonical mechanisms of macrophage death and their impact on atherogenesis and plaque stability. Macrophage death is a prominent feature of advanced plaques and is a major contributor to necrotic core formation and plaque destabilization. Mechanisms of macrophage death in atherosclerosis include apoptosis, passive or accidental necrosis as well as secondary necrosis, a type of death that typically occurs when apoptotic cells are insufficiently cleared by neighboring cells via a phagocytic process termed efferocytosis. In addition, less-well characterized types of regulated necrosis in macrophages such as necroptosis, pyroptosis, ferroptosis, and parthanatos may occur in advanced plaques and are also discussed. Autophagy in plaque macrophages is an important survival pathway that protects against cell death, yet massive stimulation of autophagy promotes another type of death, usually referred to as autosis. Multiple lines of evidence indicate that a better insight into the different mechanisms of macrophage death, and how they mutually interact, will provide novel pharmacological strategies to resolve atherosclerosis and stabilize vulnerable, rupture-prone plaques.
Collapse
Affiliation(s)
- Wim Martinet
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Isabelle Coornaert
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Pauline Puylaert
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
39
|
Koppenol WH, Hider RH. Iron and redox cycling. Do's and don'ts. Free Radic Biol Med 2019; 133:3-10. [PMID: 30236787 DOI: 10.1016/j.freeradbiomed.2018.09.022] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/01/2018] [Accepted: 09/14/2018] [Indexed: 12/23/2022]
Abstract
A major form of toxicity arises from the ability of iron to redox cycle, that is, to accept an electron from a reducing compound and to pass it on to H2O2 (the Fenton reaction). In order to do so, iron must be suitably complexed to avoid formation of Fe2O3. The ligands determine the electrode potential; this information should be known before experiments are carried out. Only one-electron transfer reactions are likely to be significant; thus two-electron potentials should not be used to determine whether an iron(III) complex can be reduced or oxidized. Ascorbate is the relevant reducing agent in blood serum, which means that iron toxicity in this compartment arises from the ascorbate-driven Fenton reaction. In the cytosol, an iron(II)-glutathione complex is likely to be the low-molecular weight iron complex involved in toxicity. When physiologically relevant concentrations are used the window of redox opportunity ranges from +0.1 V to +0.9 V. The electrode potential for non-transferrin-bound iron in the form of iron citrate is close to 0 V and the reduction of iron(III) citrate by ascorbate is slow. The clinically utilised chelators desferrioxamine, deferiprone and deferasirox in each case render iron complexes with large negative electrode potentials, thus being effective in preventing iron redox cycling and the associated toxicity resulting from such activity. There is still uncertainty about the product of the Fenton reaction, HO• or FeO2+.
Collapse
Affiliation(s)
- W H Koppenol
- Schwändibergstrasse 25, CH-8784 Braunwald, Switzerland; Emeritus, Department of Chemistry and Applied Biosciences, ETHZ, CH-8093 Zürich, Switzerland.
| | - R H Hider
- Department of Pharmacy, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| |
Collapse
|
40
|
Low density lipoprotein oxidation by ferritin at lysosomal pH. Chem Phys Lipids 2018; 217:51-57. [DOI: 10.1016/j.chemphyslip.2018.09.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 09/05/2018] [Accepted: 09/29/2018] [Indexed: 01/19/2023]
|
41
|
Meng Y, Wang L, Ling L. Changes of lysosomal membrane permeabilization and lipid metabolism in sidt2 deficient mice. Exp Ther Med 2018; 16:246-252. [PMID: 29896245 PMCID: PMC5995057 DOI: 10.3892/etm.2018.6187] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 03/29/2018] [Indexed: 01/13/2023] Open
Abstract
The SID1 transmembrane family member 2 (sidt2) deficient mouse model was used to investigate the function of sidt2 in lysosomal membrane permeabilization and lipid metabolism of liver tissue. The mouse model was established by Cre/LoxP technology. Enzymatic methods were used to analyze the sidt2−/− mouse serum lipids, aspartate transaminase, alanine transaminase and serum bilirubin, compared with sidt2+/+ mice. Defective lipid metabolism and damaged liver functions were observed in the sidt2−/− mice. By using hematoxylin and eosin and Oil Red O staining, changes of morphology were observed in sidt2−/− mice with optical microscopy. Transmission electron microscopy was also used. Hepatic steatosis and partial liver tissue apoptosis were observed. The tissue distribution of sidt2 protein and mRNA was measured in knockout mice. The results indicated that negligible sidt2 mRNA and protein expression were observed in sidt2−/− mice, and that sidt2−/− mice had abnormal liver functions. Transmission electron microscopy revealed membrane lipid droplets in the liver cell cytoplasm, and some apoptotic body formation. These results demonstrated that absence of the lysosomal membrane protein sidt2 led to changes in lysosomal membrane permeabilization and lipid metabolism.
Collapse
Affiliation(s)
- Yu Meng
- Anhui Province Key Laboratory of Biological Macro-molecules Research, Wannan Medical College, Wuhu, Anhui 241001, P.R. China.,Department of Biochemistry, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Lizhuo Wang
- Anhui Province Key Laboratory of Biological Macro-molecules Research, Wannan Medical College, Wuhu, Anhui 241001, P.R. China.,Department of Biochemistry, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Liefeng Ling
- Anhui Province Key Laboratory of Biological Macro-molecules Research, Wannan Medical College, Wuhu, Anhui 241001, P.R. China.,Department of Biochemistry, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| |
Collapse
|
42
|
Lysek-Gladysinska M, Wieczorek A, Walaszczyk A, Jelonek K, Jozwik A, Pietrowska M, Dörr W, Gabrys D, Widlak P. Long-term effects of low-dose mouse liver irradiation involve ultrastructural and biochemical changes in hepatocytes that depend on lipid metabolism. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2018; 57:123-132. [PMID: 29470638 DOI: 10.1007/s00411-018-0734-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 02/16/2018] [Indexed: 06/08/2023]
Abstract
The aim of the study was to investigate long-term effects of radiation on the (ultra)structure and function of the liver in mice. The experiments were conducted on wild-type C57BL/6J and apolipoprotein E knock-out (ApoE-/-) male mice which received a single dose (2 or 8 Gy) of X-rays to the heart with simultaneous exposure of liver to low doses (no more than 30 and 120 mGy, respectively). Livers were collected for analysis 60 weeks after irradiation and used for morphological, ultrastructural, and biochemical studies. The results show increased damage to mitochondrial ultrastructure and lipid deposition in hepatocytes of irradiated animals as compared to non-irradiated controls. Stronger radiation-related effects were noted in ApoE-/- mice than wild-type animals. In contrast, radiation-related changes in the activity of lysosomal hydrolases, including acid phosphatase, β-glucuronidase, N-acetyl-β-D-hexosaminidase, β-galactosidase, and α-glucosidase, were observed in wild type but not in ApoE-deficient mice, which together with ultrastructural picture suggests a higher activity of autophagy in ApoE-proficient animals. Irradiation caused a reduction of plasma markers of liver damage in wild-type mice, while an increased level of hepatic lipase was observed in plasma of ApoE-deficient mice, which collectively indicates a higher resistance of hepatocytes from ApoE-proficient animals to radiation-mediated damage. In conclusion, liver dysfunctions were observed as late effects of irradiation with an apparent association with malfunction of lipid metabolism.
Collapse
Affiliation(s)
- Malgorzata Lysek-Gladysinska
- Department of Cell Biology and Electron Microscopy, Institute of Biology, University of Jan Kochanowski, Swietokrzyska 15, 25-406, Kielce, Poland.
| | - Anna Wieczorek
- Department of Cell Biology and Electron Microscopy, Institute of Biology, University of Jan Kochanowski, Swietokrzyska 15, 25-406, Kielce, Poland
| | - Anna Walaszczyk
- Maria Sklodowska-Curie Institute, Oncology Center, Gliwice Branch Wybrzeze Armii Krajowej 15, 44-101, Gliwice, Poland
| | - Karol Jelonek
- Maria Sklodowska-Curie Institute, Oncology Center, Gliwice Branch Wybrzeze Armii Krajowej 15, 44-101, Gliwice, Poland
| | - Artur Jozwik
- Institute of Genetics and Animal Breeding, Polish Academy of Sciences, 05-552, Jastrzebiec, Poland
| | - Monika Pietrowska
- Maria Sklodowska-Curie Institute, Oncology Center, Gliwice Branch Wybrzeze Armii Krajowej 15, 44-101, Gliwice, Poland
| | - Wolfgang Dörr
- Department of Radiotherapy and Radiation Oncology, Medical Faculty Carl Gustav Carus, University of Technology, Dresden, Germany
- Department of Radiation Oncology, ATRAB, Applied and Translational Radiobiology, Medical University Vienna, Vienna, Austria
| | - Dorota Gabrys
- Maria Sklodowska-Curie Institute, Oncology Center, Gliwice Branch Wybrzeze Armii Krajowej 15, 44-101, Gliwice, Poland
| | - Piotr Widlak
- Maria Sklodowska-Curie Institute, Oncology Center, Gliwice Branch Wybrzeze Armii Krajowej 15, 44-101, Gliwice, Poland
| |
Collapse
|
43
|
Wieczorek A, Lysek-Gladysinska M, Walaszczyk A, Jelonek K, Smolarz M, Pietrowska M, Gabrys D, Kulik R, Widlak P. Changes in activity and structure of lysosomes from liver of mouse irradiated in vivo. Int J Radiat Biol 2018; 94:443-453. [PMID: 29611442 DOI: 10.1080/09553002.2018.1451005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
PURPOSE Lysosomes may have an important role in response to ionizing radiation. Moreover, radiation could affect autophagy, which process involves the activity of lysosomal enzymes. In the present study, the effect of ionizing radiation on the lysosomal compartment of mouse liver was investigated after in vivo exposure. MATERIALS AND METHODS Morphology and ultrastructure of hepatocytes were assessed by light and electron microscopy, and activities of selected lysosomal enzymes were assessed in 12, 36 and 120 h after exposure to the mean dose of 1 Gy. The levels of autophagy-related proteins LC3-II and p62 were compared by Western blotting between untreated and irradiated animals (120 h after exposure). RESULTS Increased number of autophagic vacuoles in hepatocytes from exposed animals was documented in the ultrastructural study; destroyed mitochondria were the dominant component of such vacuoles. Moreover, an increased activity of lysosomal hydrolases was observed after exposure. However, levels of autophagy substrates LC3-II and p62 were barely affected in exposed animals 120 h after irradiation when the accumulation of autophagic vacuoles was observed. CONCLUSION Effects of irradiation included an increased number of autophagic vacuoles, especially of autophagosomes, and increased activity of lysosomal enzymes. However, putative markers of autophagic flux were not observed, which suggested suppression of the completion of the radiation-mediated autophagy pathway.
Collapse
Affiliation(s)
- Anna Wieczorek
- a Department of Cell Biology and Electron Microscopy , Institute of Biology, University of Jan Kochanowski , Kielce , Poland
| | - Malgorzata Lysek-Gladysinska
- a Department of Cell Biology and Electron Microscopy , Institute of Biology, University of Jan Kochanowski , Kielce , Poland
| | - Anna Walaszczyk
- b Maria Sklodowska-Curie Institute - Oncology Center , Gliwice Branch , Gliwice , Poland
| | - Karol Jelonek
- b Maria Sklodowska-Curie Institute - Oncology Center , Gliwice Branch , Gliwice , Poland
| | - Mateusz Smolarz
- b Maria Sklodowska-Curie Institute - Oncology Center , Gliwice Branch , Gliwice , Poland
| | - Monika Pietrowska
- b Maria Sklodowska-Curie Institute - Oncology Center , Gliwice Branch , Gliwice , Poland
| | - Dorota Gabrys
- b Maria Sklodowska-Curie Institute - Oncology Center , Gliwice Branch , Gliwice , Poland
| | - Roland Kulik
- b Maria Sklodowska-Curie Institute - Oncology Center , Gliwice Branch , Gliwice , Poland
| | - Piotr Widlak
- b Maria Sklodowska-Curie Institute - Oncology Center , Gliwice Branch , Gliwice , Poland
| |
Collapse
|
44
|
Defective Autophagy in Atherosclerosis: To Die or to Senesce? OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:7687083. [PMID: 29682164 PMCID: PMC5846382 DOI: 10.1155/2018/7687083] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 12/29/2017] [Accepted: 01/18/2018] [Indexed: 11/17/2022]
Abstract
Autophagy is a subcellular process that plays an important role in the degradation of proteins and damaged organelles such as mitochondria (a process termed “mitophagy”) via lysosomes. It is crucial for regulating protein and mitochondrial quality control and maintaining cellular homeostasis, whereas dysregulation of autophagy has been implicated in a wide range of diseases including atherosclerosis. Recent evidence has shown that the autophagic process becomes dysfunctional during the progression of atherosclerosis, regardless of whether there are many autophagy-stimulating factors (e.g., reactive oxygen species, oxidized lipids, and cytokines) present within the atherosclerotic plaque. This review highlights the recent insights into the causes and consequences of defective autophagy in atherosclerosis, with a special focus on the role of autophagy and mitophagy in plaque macrophages, vascular smooth muscle cells (VSMCs), and endothelial cells (ECs). It has been shown that defective autophagy can promote apoptosis in macrophages but that it accelerates premature senescence in VSMCs. In the ECs, defective autophagy promotes both apoptosis and senescence. We will discuss the discrepancy between these three cell types in their response to autophagy deficiency and underline the cell type-dependent role of autophagy, which may have important implications for the efficacy of autophagy-targeted treatments for atherosclerosis.
Collapse
|
45
|
Jeong SJ, Kim S, Park JG, Jung IH, Lee MN, Jeon S, Kweon HY, Yu DY, Lee SH, Jang Y, Kang SW, Han KH, Miller YI, Park YM, Cheong C, Choi JH, Oh GT. Prdx1 (peroxiredoxin 1) deficiency reduces cholesterol efflux via impaired macrophage lipophagic flux. Autophagy 2017; 14:120-133. [PMID: 28605287 PMCID: PMC5846566 DOI: 10.1080/15548627.2017.1327942] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Oxidative stress activates macroautophagy/autophagy and contributes to atherogenesis via lipophagic flux, a form of lipid removal by autophagy. However, it is not known exactly how endogenous antioxidant enzymes are involved in lipophagic flux. Here, we demonstrate that the antioxidant PRDX1 (peroxiredoxin 1) has a crucial role in the maintenance of lipophagic flux in macrophages. PRDX1 is more highly expressed than other antioxidant enzymes in monocytes and macrophages. We determined that Prdx1 deficiency induced excessive oxidative stress and impaired maintenance of autophagic flux in macrophages. Prdx1-deficient macrophages had higher intracellular cholesterol mass and lower cholesterol efflux compared with wild type. This perturbation in cholesterol homeostasis was due to impaired lipophagic cholesterol hydrolysis caused by excessive oxidative stress, resulting in the inhibition of free cholesterol formation and the reduction of NR1H3 (nuclear receptor subfamily 1, group H, member 3) activity. Notably, impairment of both lipophagic flux and cholesterol efflux was restored by the 2-Cys PRDX-mimics ebselen and gliotoxin. Consistent with this observation, apoe −/− mice transplanted with bone marrow from prdx1−/−apoe−/− mice had increased plaque formation compared with apoe−/− BM-transplanted recipients. This study reveals that PRDX1 is crucial to regulating lipophagic flux and maintaining macrophage cholesterol homeostasis against oxidative stress. We suggest that PRDX1-dependent control of oxidative stress may provide a strategy for treating atherosclerosis and autophagy-related human diseases.
Collapse
Affiliation(s)
- Se-Jin Jeong
- a Immune and Vascular Cell Network Research Center, National Creative Initiatives , Department of Life Sciences , Ewha Womans University , Seoul , Korea.,b Cardiovascular Division , Department of Medicine , Washington University School of Medicine , St. Louis , MO , USA
| | - Sinai Kim
- a Immune and Vascular Cell Network Research Center, National Creative Initiatives , Department of Life Sciences , Ewha Womans University , Seoul , Korea
| | - Jong-Gil Park
- c Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience & Biotechnology , Daejeon , Korea
| | - In-Hyuk Jung
- b Cardiovascular Division , Department of Medicine , Washington University School of Medicine , St. Louis , MO , USA
| | - Mi-Ni Lee
- a Immune and Vascular Cell Network Research Center, National Creative Initiatives , Department of Life Sciences , Ewha Womans University , Seoul , Korea
| | - Sejin Jeon
- a Immune and Vascular Cell Network Research Center, National Creative Initiatives , Department of Life Sciences , Ewha Womans University , Seoul , Korea
| | - Hyae Yon Kweon
- a Immune and Vascular Cell Network Research Center, National Creative Initiatives , Department of Life Sciences , Ewha Womans University , Seoul , Korea
| | - Dae-Yeul Yu
- d Korea Aging Research Center, Korea Research Institute of Bioscience and Biotechnology , Daejeon , Korea
| | - Sang-Hak Lee
- e Division of Cardiology , Department of Internal Medicine , Yonsei University College of Medicine , Seoul , Korea
| | - Yangsoo Jang
- e Division of Cardiology , Department of Internal Medicine , Yonsei University College of Medicine , Seoul , Korea
| | - Sang Won Kang
- f Department of Life Science and Research Center for Cell Homeostasis , Ewha Womans University , Seoul , Korea ; Global Top5 Research program, Ewha Womans University , Seoul , Korea
| | - Ki-Hwan Han
- g Department of Anatomy , School of Medicine, Ewha Womans University , Seoul , Korea
| | - Yury I Miller
- h Department of Medicine , University of California, San Diego , San Diego , CA , USA
| | - Young Mi Park
- i Department of Molecular Medicine , Ewha Womans University School of Medicine , Seoul , Korea
| | - Cheolho Cheong
- j Department of Microbiology and Immunology , McGill Faculty of Medicine , Montréal , Canada
| | - Jae-Hoon Choi
- k Department of Life Science , College of Natural Sciences and Research Institute for Natural Sciences, Hanyang University , Seoul , Korea
| | - Goo Taeg Oh
- a Immune and Vascular Cell Network Research Center, National Creative Initiatives , Department of Life Sciences , Ewha Womans University , Seoul , Korea
| |
Collapse
|
46
|
Delbosc S, Bayles RG, Laschet J, Ollivier V, Ho-Tin-Noé B, Touat Z, Deschildre C, Morvan M, Louedec L, Gouya L, Guedj K, Nicoletti A, Michel JB. Erythrocyte Efferocytosis by the Arterial Wall Promotes Oxidation in Early-Stage Atheroma in Humans. Front Cardiovasc Med 2017; 4:43. [PMID: 28824922 PMCID: PMC5539175 DOI: 10.3389/fcvm.2017.00043] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 06/26/2017] [Indexed: 01/21/2023] Open
Abstract
Background Since red blood cells (RBCs) are the predominant cellular blood component interacting with the arterial wall, we explored the role of RBCs efferocytosis by vascular smooth muscle cells (vSMCs) in the initiation of human atheroma. Methods and results The comparison of human healthy aortas with aortic fatty streaks or fibroatheromas revealed that RBC angiophagy is implicated from the earliest stages of atherogenesis, as documented by the concomitant detection of redox-active iron, hemoglobin, glycophorin A, and ceroids. RBCs infiltration in the arterial wall was associated with local lipid and protein oxidation, as well as vascular response (expression of heme oxygenase-1 and of genes related to iron metabolism as well as those encoding for phagocytosis). These effects were recapitulated in vitro when vSMCs were co-cultured with phosphatidyl-exposing senescent (s) RBCs but not with fresh RBCs. VSMCs engulfing sRBC increased their intracellular iron content, accumulated hemoglobin, lipids, and activated their phagolysosomes. Strikingly, injections of sRBCs into rats promoted iron accumulation in the aortic wall. In rabbits, hypercholesterolemia increased circulating senescent RBCs and induced the subendothelial accumulation of iron-rich phagocytic foam cells. RBCs bring cholesterol and iron/heme into the vascular wall and interact with vSMCs that phagocytize them. Conclusion This study presents a previously unforeseen mechanism of plaque formation that implicates intimal RBC infiltration as one of the initial triggers for foam cell formation and intimal oxidation. Pathogenic effects exerted by several metabolic and hemodynamic factors may rely on their effect on RBC biology, thereby impacting how RBCs interact with the vascular wall.
Collapse
Affiliation(s)
- Sandrine Delbosc
- UMRS 1148, INSERM, Paris 7-Denis Diderot University, Hôpital Xavier Bichat, Paris, France.,Département Hospitalo-Universitaire DHU "FIRE", Paris, France
| | - Richard Graham Bayles
- UMRS 1148, INSERM, Paris 7-Denis Diderot University, Hôpital Xavier Bichat, Paris, France.,Département Hospitalo-Universitaire DHU "FIRE", Paris, France
| | - Jamila Laschet
- UMRS 1148, INSERM, Paris 7-Denis Diderot University, Hôpital Xavier Bichat, Paris, France.,Département Hospitalo-Universitaire DHU "FIRE", Paris, France
| | - Veronique Ollivier
- UMRS 1148, INSERM, Paris 7-Denis Diderot University, Hôpital Xavier Bichat, Paris, France.,Département Hospitalo-Universitaire DHU "FIRE", Paris, France
| | - Benoit Ho-Tin-Noé
- UMRS 1148, INSERM, Paris 7-Denis Diderot University, Hôpital Xavier Bichat, Paris, France.,Département Hospitalo-Universitaire DHU "FIRE", Paris, France
| | - Ziad Touat
- UMRS 1148, INSERM, Paris 7-Denis Diderot University, Hôpital Xavier Bichat, Paris, France.,Département Hospitalo-Universitaire DHU "FIRE", Paris, France
| | - Catherine Deschildre
- UMRS 1148, INSERM, Paris 7-Denis Diderot University, Hôpital Xavier Bichat, Paris, France.,Département Hospitalo-Universitaire DHU "FIRE", Paris, France
| | - Marion Morvan
- UMRS 1148, INSERM, Paris 7-Denis Diderot University, Hôpital Xavier Bichat, Paris, France.,Département Hospitalo-Universitaire DHU "FIRE", Paris, France
| | - Liliane Louedec
- UMRS 1148, INSERM, Paris 7-Denis Diderot University, Hôpital Xavier Bichat, Paris, France.,Département Hospitalo-Universitaire DHU "FIRE", Paris, France
| | - Laurent Gouya
- Département Hospitalo-Universitaire DHU "FIRE", Paris, France.,UMRS 1149, INSERM, Paris 7-Denis Diderot University, Hôpital Xavier Bichat, Paris, France
| | - Kevin Guedj
- UMRS 1148, INSERM, Paris 7-Denis Diderot University, Hôpital Xavier Bichat, Paris, France.,Département Hospitalo-Universitaire DHU "FIRE", Paris, France
| | - Antonino Nicoletti
- UMRS 1148, INSERM, Paris 7-Denis Diderot University, Hôpital Xavier Bichat, Paris, France.,Département Hospitalo-Universitaire DHU "FIRE", Paris, France
| | - Jean-Baptiste Michel
- UMRS 1148, INSERM, Paris 7-Denis Diderot University, Hôpital Xavier Bichat, Paris, France.,Département Hospitalo-Universitaire DHU "FIRE", Paris, France
| |
Collapse
|
47
|
Barardo DG, Newby D, Thornton D, Ghafourian T, de Magalhães JP, Freitas AA. Machine learning for predicting lifespan-extending chemical compounds. Aging (Albany NY) 2017; 9:1721-1737. [PMID: 28783712 PMCID: PMC5559171 DOI: 10.18632/aging.101264] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 07/12/2017] [Indexed: 12/12/2022]
Abstract
Increasing age is a risk factor for many diseases; therefore developing pharmacological interventions that slow down ageing and consequently postpone the onset of many age-related diseases is highly desirable. In this work we analyse data from the DrugAge database, which contains chemical compounds and their effect on the lifespan of model organisms. Predictive models were built using the machine learning method random forests to predict whether or not a chemical compound will increase Caenorhabditis elegans' lifespan, using as features Gene Ontology (GO) terms annotated for proteins targeted by the compounds and chemical descriptors calculated from each compound's chemical structure. The model with the best predictive accuracy used both biological and chemical features, achieving a prediction accuracy of 80%. The top 20 most important GO terms include those related to mitochondrial processes, to enzymatic and immunological processes, and terms related to metabolic and transport processes. We applied our best model to predict compounds which are more likely to increase C. elegans' lifespan in the DGIdb database, where the effect of the compounds on an organism's lifespan is unknown. The top hit compounds can be broadly divided into four groups: compounds affecting mitochondria, compounds for cancer treatment, anti-inflammatories, and compounds for gonadotropin-releasing hormone therapies.
Collapse
Affiliation(s)
- Diogo G. Barardo
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Danielle Newby
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
| | - Daniel Thornton
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | | | - João Pedro de Magalhães
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | | |
Collapse
|
48
|
Shi L, Ito F, Wang Y, Okazaki Y, Tanaka H, Mizuno M, Hori M, Hirayama T, Nagasawa H, Richardson DR, Toyokuni S. Non-thermal plasma induces a stress response in mesothelioma cells resulting in increased endocytosis, lysosome biogenesis and autophagy. Free Radic Biol Med 2017; 108:904-917. [PMID: 28465262 DOI: 10.1016/j.freeradbiomed.2017.04.368] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 04/19/2017] [Accepted: 04/28/2017] [Indexed: 12/31/2022]
Abstract
Non-thermal plasma (NTP) is a potential new therapeutic modality for cancer. However, its mechanism of action remains unclear. Herein, we studied the effect of NTP on mesothelioma cells and fibroblasts to understand its anti-proliferative efficacy. Interestingly, NTP demonstrated greater selective anti-proliferative activity against mesothelioma cells relative to fibroblasts than cisplatin, which is used for mesothelioma treatment. The anti-proliferative effect of NTP was enhanced by pre-incubation with the cellular iron donor, ferric ammonium citrate (FAC), and inhibited by iron chelation using desferrioxamine (DFO). Three oxidative stress probes (CM-H2DCFDA, MitoSOX and C11-BODIPY) demonstrated reactive oxygen species (ROS) generation by NTP, which was inhibited by DFO. Moreover, NTP decreased transferrin receptor-1 and increased ferritin-H and -L chain expression that was correlated with decreased iron-regulatory protein expression and RNA-binding activity. This regulation was potentially due to increased intracellular iron in lysosomes, which was demonstrated via the Fe(II)-selective probe, HMRhoNox-M, and was consistent with autophagic-induction. Immunofluorescence using LysoTracker and Pepstatin A probes demonstrated increased cellular lysosome content, which was confirmed by elevated LAMP1 expression. The enhanced lysosomal biogenesis after NTP could be due to the observed increase in fluid-phase endocytosis and early endosome formation. These results suggest NTP acts as a stressor, which results in increased endocytosis, lysosome content and autophagy. In fact, NTP rapidly increased autophagosome formation, as judged by increased LC3B-II expression, which co-localized with LAMP1, indicating autophagolysosome formation. Autophagic-induction by NTP was confirmed using electron microscopy. In summary, NTP acts as a cellular stressor to rapidly induce fluid-phase endocytosis, lysosome biogenesis and autophagy.
Collapse
Affiliation(s)
- Lei Shi
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Fumiya Ito
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Yue Wang
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Yasumasa Okazaki
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Hiromasa Tanaka
- Center for Advanced Medicine and Clinical Research, Nagoya University Hospital, Nagoya 466-8550, Japan
| | - Masaaki Mizuno
- Center for Advanced Medicine and Clinical Research, Nagoya University Hospital, Nagoya 466-8550, Japan
| | - Masaru Hori
- Plasma Nanotechnology Research Center, Nagoya University, Nagoya 464-8603, Japan
| | - Tasuku Hirayama
- The Laboratory of Pharmaceutical and Medicinal Chemistry, Gifu Pharmaceutical University, Gifu, Japan
| | - Hideko Nagasawa
- The Laboratory of Pharmaceutical and Medicinal Chemistry, Gifu Pharmaceutical University, Gifu, Japan
| | - Des R Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, The University of Sydney, Sydney, NSW 2006, Australia.
| | - Shinya Toyokuni
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
49
|
Fabris F, Magalhães JPD, Freitas AA. A review of supervised machine learning applied to ageing research. Biogerontology 2017; 18:171-188. [PMID: 28265788 PMCID: PMC5350215 DOI: 10.1007/s10522-017-9683-y] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 02/21/2017] [Indexed: 11/30/2022]
Abstract
Broadly speaking, supervised machine learning is the computational task of learning correlations between variables in annotated data (the training set), and using this information to create a predictive model capable of inferring annotations for new data, whose annotations are not known. Ageing is a complex process that affects nearly all animal species. This process can be studied at several levels of abstraction, in different organisms and with different objectives in mind. Not surprisingly, the diversity of the supervised machine learning algorithms applied to answer biological questions reflects the complexities of the underlying ageing processes being studied. Many works using supervised machine learning to study the ageing process have been recently published, so it is timely to review these works, to discuss their main findings and weaknesses. In summary, the main findings of the reviewed papers are: the link between specific types of DNA repair and ageing; ageing-related proteins tend to be highly connected and seem to play a central role in molecular pathways; ageing/longevity is linked with autophagy and apoptosis, nutrient receptor genes, and copper and iron ion transport. Additionally, several biomarkers of ageing were found by machine learning. Despite some interesting machine learning results, we also identified a weakness of current works on this topic: only one of the reviewed papers has corroborated the computational results of machine learning algorithms through wet-lab experiments. In conclusion, supervised machine learning has contributed to advance our knowledge and has provided novel insights on ageing, yet future work should have a greater emphasis in validating the predictions.
Collapse
Affiliation(s)
- Fabio Fabris
- School of Computing, University of Kent, Canterbury, Kent CT2 7NF UK
| | - João Pedro de Magalhães
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L7 8TX UK
| | - Alex A. Freitas
- School of Computing, University of Kent, Canterbury, Kent CT2 7NF UK
| |
Collapse
|
50
|
Kavčič N, Pegan K, Turk B. Lysosomes in programmed cell death pathways: from initiators to amplifiers. Biol Chem 2017; 398:289-301. [DOI: 10.1515/hsz-2016-0252] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 09/05/2016] [Indexed: 01/19/2023]
Abstract
Abstract
Lysosome is the central organelle for intracellular degradation of biological macromolecules and organelles. The material destined for degradation enters the lysosomes primarily via endocytosis, autophagy and phagocytosis, and is degraded through the concerted action of more than 50 lysosomal hydrolases. However, lysosomes are also linked with numerous other processes, including cell death, inflammasome activation and immune response, as well as with lysosomal secretion and cholesterol recycling. Among them programmed cell death pathways including apoptosis have received major attention. In most of these pathways, cell death was accompanied by lysosomal membrane permeabilization and release of lysosomal constituents with an involvement of lysosomal hydrolases, including the cathepsins. However, it is less clear, whether lysosomal membrane permeabilization is really critical for the initiation of cell death programme(s). Therefore, the role of lysosomal membrane permeabilization in various programmed cell death pathways is reviewed, as well as the mechanisms leading to it.
Collapse
|