101
|
Graham C, Stefanatos R, Yek AEH, Spriggs RV, Loh SHY, Uribe AH, Zhang T, Martins LM, Maddocks ODK, Scialo F, Sanz A. Mitochondrial ROS signalling requires uninterrupted electron flow and is lost during ageing in flies. GeroScience 2022; 44:1961-1974. [PMID: 35355221 PMCID: PMC9616974 DOI: 10.1007/s11357-022-00555-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 03/22/2022] [Indexed: 11/30/2022] Open
Abstract
Mitochondrial reactive oxygen species (mtROS) are cellular messengers essential for cellular homeostasis. In response to stress, reverse electron transport (RET) through respiratory complex I generates high levels of mtROS. Suppression of ROS production via RET (ROS-RET) reduces survival under stress, while activation of ROS-RET extends lifespan in basal conditions. Here, we demonstrate that ROS-RET signalling requires increased electron entry and uninterrupted electron flow through the electron transport chain (ETC). We find that in old fruit flies, ROS-RET is abolished when electron flux is decreased and that their mitochondria produce consistently high levels of mtROS. Finally, we demonstrate that in young flies, limiting electron exit, but not entry, from the ETC phenocopies mtROS generation observed in old individuals. Our results elucidate the mechanism by which ROS signalling is lost during ageing.
Collapse
Affiliation(s)
- Charlotte Graham
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, NE4 5PL, UK
| | - Rhoda Stefanatos
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, NE4 5PL, UK
- Faculty of Medical Sciences, Wellcome Centre for Mitochondrial Research, Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Angeline E H Yek
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, NE4 5PL, UK
| | - Ruth V Spriggs
- MRC Toxicology Unit, University of Cambridge, Cambridge, CB2 1QR, UK
- Hearing Sciences, School of Medicine, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Samantha H Y Loh
- MRC Toxicology Unit, University of Cambridge, Cambridge, CB2 1QR, UK
| | - Alejandro Huerta Uribe
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1QH, UK
| | - Tong Zhang
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1QH, UK
- Novartis Institutes for BioMedical Research, Shanghai, 201203, China
| | - L Miguel Martins
- MRC Toxicology Unit, University of Cambridge, Cambridge, CB2 1QR, UK
| | - Oliver D K Maddocks
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1QH, UK
| | - Filippo Scialo
- Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK.
- Dipartimento Di Scienze Mediche Traslazionali, Università Degli Studi Della Campania "Luigi Vanvitelli", 80131, Naples, Italy.
| | - Alberto Sanz
- Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK.
| |
Collapse
|
102
|
de Haan LR, Reiniers MJ, Reeskamp LF, Belkouz A, Ao L, Cheng S, Ding B, van Golen RF, Heger M. Experimental Conditions That Influence the Utility of 2′7′-Dichlorodihydrofluorescein Diacetate (DCFH2-DA) as a Fluorogenic Biosensor for Mitochondrial Redox Status. Antioxidants (Basel) 2022; 11:antiox11081424. [PMID: 35892626 PMCID: PMC9329753 DOI: 10.3390/antiox11081424] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/05/2022] [Accepted: 07/12/2022] [Indexed: 11/16/2022] Open
Abstract
Oxidative stress has been causally linked to various diseases. Electron transport chain (ETC) inhibitors such as rotenone and antimycin A are frequently used in model systems to study oxidative stress. Oxidative stress that is provoked by ETC inhibitors can be visualized using the fluorogenic probe 2′,7′-dichlorodihydrofluorescein-diacetate (DCFH2-DA). Non-fluorescent DCFH2-DA crosses the plasma membrane, is deacetylated to 2′,7′-dichlorodihydrofluorescein (DCFH2) by esterases, and is oxidized to its fluorescent form 2′,7′-dichlorofluorescein (DCF) by intracellular ROS. DCF fluorescence can, therefore, be used as a semi-quantitative measure of general oxidative stress. However, the use of DCFH2-DA is complicated by various protocol-related factors that mediate DCFH2-to-DCF conversion independently of the degree of oxidative stress. This study therefore analyzed the influence of ancillary factors on DCF formation in the context of ETC inhibitors. It was found that ETC inhibitors trigger DCF formation in cell-free experiments when they are co-dissolved with DCFH2-DA. Moreover, the extent of DCF formation depended on the type of culture medium that was used, the pH of the assay system, the presence of fetal calf serum, and the final DCFH2-DA solvent concentration. Conclusively, experiments with DCFH2-DA should not discount the influence of protocol-related factors such as medium and mitochondrial inhibitors (and possibly other compounds) on the DCFH2-DA-DCF reaction and proper controls should always be built into the assay protocol.
Collapse
Affiliation(s)
- Lianne R. de Haan
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, China; (L.R.d.H.); (M.J.R.); (L.A.); (B.D.)
- Laboratory for Experimental Oncology, Department of Pathology, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Megan J. Reiniers
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, China; (L.R.d.H.); (M.J.R.); (L.A.); (B.D.)
- Department of Surgery, Haaglanden Medisch Centrum, 2262 BA The Hague, The Netherlands
- Membrane Biochemistry and Biophysics, Department of Chemistry, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Laurens F. Reeskamp
- Department of Vascular Medicine, Amsterdam University Medical Centers, Location AMC, 1105 AZ Amsterdam, The Netherlands;
| | - Ali Belkouz
- Department of Medical Oncology, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Cancer Center Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| | - Lei Ao
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, China; (L.R.d.H.); (M.J.R.); (L.A.); (B.D.)
| | - Shuqun Cheng
- Department of Hepatic Surgery VI, The Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai 200438, China;
| | - Baoyue Ding
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, China; (L.R.d.H.); (M.J.R.); (L.A.); (B.D.)
| | - Rowan F. van Golen
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
| | - Michal Heger
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, China; (L.R.d.H.); (M.J.R.); (L.A.); (B.D.)
- Laboratory for Experimental Oncology, Department of Pathology, Erasmus MC, 3015 GD Rotterdam, The Netherlands
- Membrane Biochemistry and Biophysics, Department of Chemistry, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
- Correspondence: or ; Tel.: +31-6-2448-3083 or +31-30-2533-966
| |
Collapse
|
103
|
Gao H, Sun H, Yan N, Zhao P, Xu H, Zheng W, Zhang X, Wang T, Guo C, Zhong M. ATP13A2 Declines Zinc-Induced Accumulation of α-Synuclein in a Parkinson’s Disease Model. Int J Mol Sci 2022; 23:ijms23148035. [PMID: 35887392 PMCID: PMC9318580 DOI: 10.3390/ijms23148035] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/11/2022] [Accepted: 07/18/2022] [Indexed: 02/01/2023] Open
Abstract
Parkinson’s disease (PD) is characterized by the presence of Lewy bodies caused by α-synuclein. The imbalance of zinc homeostasis is a major cause of PD, promoting α-synuclein accumulation. ATP13A2, a transporter found in acidic vesicles, plays an important role in Zn2+ homeostasis and is highly expressed in Lewy bodies in PD-surviving neurons. ATP13A2 is involved in the transport of zinc ions in lysosomes and exosomes and inhibits the aggregation of α-synuclein. However, the potential mechanism underlying the regulation of zinc homeostasis and α-synuclein accumulation by ATP13A2 remains unexplored. We used α-synuclein-GFP transgenic mice and HEK293 α-synuclein-DsRed cell line as models. The spatial exploration behavior of mice was significantly reduced, and phosphorylation levels of α-synuclein increased upon high Zn2+ treatment. High Zn2+ also inhibited the autophagy pathway by reducing LAMP2a levels and changing the expression of LC3 and P62, by reducing mitochondrial membrane potential and increasing the expression of cytochrom C, and by activating the ERK/P38 apoptosis signaling pathway, ultimately leading to increased caspase 3 levels. These protein changes were reversed after ATP13A2 overexpression, whereas ATP13A2 knockout exacerbated α-synuclein phosphorylation levels. These results suggest that ATP13A2 may have a protective effect on Zn2+-induced abnormal aggregation of α-synuclein, lysosomal dysfunction, and apoptosis.
Collapse
Affiliation(s)
- Huiling Gao
- College of Life and Health Sciences, Northeastern University, Shenyang 110169, China; (H.G.); (H.S.); (P.Z.); (T.W.); (C.G.)
| | - Hehong Sun
- College of Life and Health Sciences, Northeastern University, Shenyang 110169, China; (H.G.); (H.S.); (P.Z.); (T.W.); (C.G.)
| | - Nan Yan
- School of Medical Applied Technology, Shenyang Medical College, Shenyang 110034, China;
| | - Pu Zhao
- College of Life and Health Sciences, Northeastern University, Shenyang 110169, China; (H.G.); (H.S.); (P.Z.); (T.W.); (C.G.)
| | - He Xu
- Department of Anatomy, Histology and Embryology, School of Medicine, Shenzhen University, Shenzhen 518060, China;
| | - Wei Zheng
- Department of Histology and Embryology, School of Basic Medical Sciences, China Medical University, Shenyang 110122, China;
| | - Xiaoyu Zhang
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China;
| | - Tao Wang
- College of Life and Health Sciences, Northeastern University, Shenyang 110169, China; (H.G.); (H.S.); (P.Z.); (T.W.); (C.G.)
| | - Chuang Guo
- College of Life and Health Sciences, Northeastern University, Shenyang 110169, China; (H.G.); (H.S.); (P.Z.); (T.W.); (C.G.)
| | - Manli Zhong
- College of Life and Health Sciences, Northeastern University, Shenyang 110169, China; (H.G.); (H.S.); (P.Z.); (T.W.); (C.G.)
- Correspondence:
| |
Collapse
|
104
|
Tsai CY, Wu JCC, Wu CJ, Chan SHH. Protective role of VEGF/VEGFR2 signaling against high fatality associated with hepatic encephalopathy via sustaining mitochondrial bioenergetics functions. J Biomed Sci 2022; 29:47. [PMID: 35786324 PMCID: PMC9251935 DOI: 10.1186/s12929-022-00831-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/27/2022] [Indexed: 11/30/2022] Open
Abstract
Background The lack of better understanding of the pathophysiology and cellular mechanisms associated with high mortality seen in hepatic encephalopathy (HE), a neurological complication arising from acute hepatic failure, remains a challenging medical issue. Clinical reports showed that the degree of baroreflex dysregulation is related to the severity of HE. Furthermore, mitochondrial dysfunction in the rostral ventrolateral medulla (RVLM), a key component of the baroreflex loop that maintains blood pressure and sympathetic vasomotor tone, is known to underpin impairment of baroreflex. Realizing that in addition to angiogenic and vasculogenic effects, by acting on its key receptor (VEGFR2), vascular endothelial growth factor (VEGF) elicits neuroprotection via maintenance of mitochondrial function, the guiding hypothesis of the present study is that the VEGF/VEGFR2 signaling plays a protective role against mitochondrial dysfunction in the RVLM to ameliorate baroreflex dysregulation that underpins the high fatality associated with HE. Methods Physiological, pharmacological and biochemical investigations were carried out in proof-of-concept experiments using an in vitro model of HE that involved incubation of cultured mouse hippocampal neurons with ammonium chloride. This was followed by corroboratory experiments employing a mouse model of HE, in which adult male C57BL/6 mice and VEGFR2 wild-type and heterozygous mice received an intraperitoneal injection of azoxymethane, a toxin used to induce acute hepatic failure. Results We demonstrated that VEGFR2 is present in cultured neurons, and observed that whereas recombinant VEGF protein maintained cell viability, gene-knockdown of vegfr2 enhanced the reduction of cell viability in our in vitro model of HE. In our in vivo model of HE, we found that VEGFR2 heterozygous mice exhibited shorter survival rate and time when compared to wild-type mice. In C57BL/6 mice, there was a progressive reduction in VEGFR2 mRNA and protein expression, mitochondrial membrane potential and ATP levels, alongside augmentation of apoptotic cell death in the RVLM, accompanied by a decrease in baroreflex-mediated sympathetic vasomotor tone and hypotension. Immunoneutralization of VEGF exacerbated all those biochemical and physiological events. Conclusions Our results suggest that, acting via VEGFR2, the endogenous VEGF plays a protective role against high fatality associated with HE by amelioration of the dysregulated baroreflex-mediated sympathetic vasomotor tone through sustaining mitochondrial bioenergetics functions and eliciting antiapoptotic action in the RVLM. Supplementary Information The online version contains supplementary material available at 10.1186/s12929-022-00831-0.
Collapse
Affiliation(s)
- Ching-Yi Tsai
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.
| | - Jacqueline C C Wu
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Chiung-Ju Wu
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Samuel H H Chan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| |
Collapse
|
105
|
Akande O, Chen Q, Cholyway R, Toldo S, Lesnefsky EJ, Quader M. Modulation of Mitochondrial Respiration During Early Reperfusion Reduces Cardiac Injury in Donation After Circulatory Death Hearts. J Cardiovasc Pharmacol 2022; 80:148-157. [PMID: 35579563 PMCID: PMC10441174 DOI: 10.1097/fjc.0000000000001290] [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: 12/22/2021] [Accepted: 04/03/2022] [Indexed: 11/25/2022]
Abstract
ABSTRACT Donation after circulatory death (DCD) donors are a potential source for heart transplantation. The DCD process has unavoidable ischemia and reperfusion (I/R) injury, primarily mediated through mitochondria, which limits routine utilization of hearts for transplantation. Amobarbital (AMO), a transient inhibitor of the electron transport chain, is known to decrease cardiac injury following ex vivo I/R. We studied whether AMO treatment during reperfusion can decrease injury in DCD hearts. Sprague Dawley rat hearts subjected to 25 minutes of in vivo ischemia (DCD hearts), or control beating donor hearts, were treated with AMO or vehicle for the first 5 minutes of reperfusion, followed by Krebs-Henseleit buffer reperfusion for 55 minutes (for mitochondrial isolation) or 85 minutes (for infarct size determination). Compared with vehicle, AMO treatment led to decreased infarct size (25.2% ± 1.5% vs. 31.5% ± 1.5%; P ≤ 0.05) and troponin I release (4.5 ± 0.05 ng/mL vs. 9.3 ± 0.24 ng/mL, P ≤ 0.05). AMO treatment decreased H 2 O 2 generation with glutamate as complex I substrate in both subsarcolemmal mitochondria (SSM) (37 ± 3.7 pmol·mg -1 ·min -1 vs. 56.9 ± 4.1 pmol·mg -1 ·min -1 ; P ≤ 0.05), and interfibrillar mitochondria (IFM) (31.8 ± 2.8 pmol·mg -1 ·min -1 vs. 46 ± 4.8 pmol·mg -1 ·min -1 ; P ≤ 0.05) and improved calcium retention capacity in SSM (360 ±17.2 nmol/mg vs. 277 ± 13 nmol/mg; P ≤ 0.05), and IFM (483 ± 20 nmol/mg vs. 377± 19 nmol/mg; P ≤ 0.05) compared with vehicle treatment. SSM and IFM retained more cytochrome c with AMO treatment compared with vehicle. In conclusion, brief inhibition of mitochondrial respiration during reperfusion using amobarbital is a promising approach to decrease injury in DCD hearts.
Collapse
Affiliation(s)
- Oluwatoyin Akande
- Division of Cardio-Thoracic Surgery, Department of Surgery, Virginia Commonwealth University, Richmond, VA
| | - Qun Chen
- Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA
- Pauley Heart Center, Virginia Commonwealth University, Richmond VA
| | - Renee Cholyway
- Division of Cardio-Thoracic Surgery, Department of Surgery, Virginia Commonwealth University, Richmond, VA
| | - Stefano Toldo
- Division of Cardio-Thoracic Surgery, Department of Surgery, Virginia Commonwealth University, Richmond, VA
- Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA
- Pauley Heart Center, Virginia Commonwealth University, Richmond VA
| | - Edward J. Lesnefsky
- Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA
- Pauley Heart Center, Virginia Commonwealth University, Richmond VA
- McGuire Veterans Administration Medical Center, Richmond VA
| | - Mohammed Quader
- Division of Cardio-Thoracic Surgery, Department of Surgery, Virginia Commonwealth University, Richmond, VA
- Pauley Heart Center, Virginia Commonwealth University, Richmond VA
- McGuire Veterans Administration Medical Center, Richmond VA
| |
Collapse
|
106
|
Wawi MJ, Mahler C, Inguimbert N, Marder TB, Ribou AC. A new mitochondrial probe combining pyrene and a triphenylphosphonium salt for cellular oxygen and free radical detection via fluorescence lifetime measurements. Free Radic Res 2022; 56:258-272. [PMID: 35772434 DOI: 10.1080/10715762.2022.2077202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
To improve and diversify the quantification of reactive oxygen species (ROS) in mitochondria of single cells, we connected pyrene derivatives (PB) to a triphenylphosphonium salt (TPP+) as a mitochondrial vector forming PB-TPP+ probes. Two pyrene isomers with the n-butyltriphenylphosphonium moieties attached at their 1- or 2- positions were synthesized and characterized. Using the long fluorescence lifetime of pyrene, it was possible to monitor the variation of cellular free radicals and oxygen and to follow the reversibility of both quenchers in real-time. We compared the behavior of these new probes to the previously published pyrene-probes, functionalized by a mitochondrial-penetrating peptide, allowing their transfer to the mitochondria (Mito-PB) or to the cytosolic membrane for pyrene butyric acid (PBA). The high cellular uptake of the new probes allows cells to be loaded with an initial concentration 40 times lower than that for Mito-PB probes, without inducing perturbations in cell growth. The variation in free radicals and oxygen levels was monitored within cells under different stress conditions through the fluorescence lifetime of the new TPP+-based probes giving comparable results to those obtained for MPP-based probes. However, at a loading concentration as low as 25 nM, our technique allows the detection of increased production of free radicals in the mitochondria in the presence of the TPP+ vector, a warning to the user of this well-known vector.
Collapse
Affiliation(s)
- Mohamad Jamal Wawi
- Espace-Dev, Univ Montpellier, IRD, Univ Guyane, Univ la Réunion, Univ Antilles, Montpellier, France.,Laboratoire IMAGES-ESPACE-DEV, Univ. Perpignan Via Domitia, Perpignan, France
| | - Christoph Mahler
- Institut für Anorganische Chemie, and Institute for Sustainable Chemistry & Catalysis with Boron, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Nicolas Inguimbert
- PSL Université Paris: EPHE-UPVD-CNRS, USR 3278 CRIOBE, Université de Perpignan Via Domitia, Perpignan, France
| | - Todd B Marder
- Institut für Anorganische Chemie, and Institute for Sustainable Chemistry & Catalysis with Boron, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Anne-Cécile Ribou
- Espace-Dev, Univ Montpellier, IRD, Univ Guyane, Univ la Réunion, Univ Antilles, Montpellier, France.,Laboratoire IMAGES-ESPACE-DEV, Univ. Perpignan Via Domitia, Perpignan, France
| |
Collapse
|
107
|
Milliken AS, Nadtochiy SM, Brookes PS. Inhibiting Succinate Release Worsens Cardiac Reperfusion Injury by Enhancing Mitochondrial Reactive Oxygen Species Generation. J Am Heart Assoc 2022; 11:e026135. [PMID: 35766275 PMCID: PMC9333399 DOI: 10.1161/jaha.122.026135] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Background The metabolite succinate accumulates during cardiac ischemia. Within 5 minutes of reperfusion, succinate returns to baseline levels via both its release from cells and oxidation by mitochondrial complex II. The latter drives reactive oxygen species (ROS) generation and subsequent opening of the mitochondrial permeability transition (PT) pore, leading to cell death. Targeting succinate dynamics (accumulation/oxidation/release) may be therapeutically beneficial in cardiac ischemia–reperfusion (IR) injury. It has been proposed that blocking MCT1 (monocarboxylate transporter 1) may be beneficial in IR injury, by preventing succinate release and subsequent engagement of downstream inflammatory signaling pathways. In contrast, herein we hypothesized that blocking MCT1 would retain succinate in cells, exacerbating ROS generation and IR injury. Methods and Results Using the mitochondrial ROS probe mitoSOX and a custom‐built murine heart perfusion rig built into a spectrofluorometer, we measured ROS generation in situ during the first moments of reperfusion. We found that acute MCT1 inhibition enhanced mitochondrial ROS generation at reperfusion and worsened IR injury (recovery of function and infarct size). Both of these effects were abrogated by tandem inhibition of mitochondrial complex II, suggesting that succinate retention worsens IR because it drives more mitochondrial ROS generation. Furthermore, using the PT pore inhibitor cyclosporin A, along with monitoring of PT pore opening via the mitochondrial membrane potential indicator tetramethylrhodamine ethyl ester, we herein provide evidence that ROS generation during early reperfusion is upstream of the PT pore, not downstream as proposed by others. In addition, pore opening was exacerbated by MCT1 inhibition. Conclusions Together, these findings highlight the importance of succinate dynamics and mitochondrial ROS generation as key determinants of PT pore opening and IR injury outcomes.
Collapse
Affiliation(s)
- Alexander S Milliken
- Department of Pharmacology and Physiology University of Rochester Medical Center Rochester NY
| | - Sergiy M Nadtochiy
- Department of Anesthesiology and Perioperative Medicine University of Rochester Medical Center Rochester NY
| | - Paul S Brookes
- Department of Anesthesiology and Perioperative Medicine University of Rochester Medical Center Rochester NY
| |
Collapse
|
108
|
Vasconcellos AF, Melo RM, Mandacaru SC, de Oliveira LS, de Oliveira AS, Moraes ECDS, Trugilho MRDO, Ricart CAO, Báo SN, Resende RO, Charneau S. Aedes aegypti Aag-2 Cell Proteome Modulation in Response to Chikungunya Virus Infection. Front Cell Infect Microbiol 2022; 12:920425. [PMID: 35782121 PMCID: PMC9240781 DOI: 10.3389/fcimb.2022.920425] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/18/2022] [Indexed: 01/16/2023] Open
Abstract
Chikungunya virus (CHIKV) is a single-stranded positive RNA virus that belongs to the genus Alphavirus and is transmitted to humans by infected Aedes aegypti and Aedes albopictus bites. In humans, CHIKV usually causes painful symptoms during acute and chronic stages of infection. Conversely, virus–vector interaction does not disturb the mosquito’s fitness, allowing a persistent infection. Herein, we studied CHIKV infection of Ae. aegypti Aag-2 cells (multiplicity of infection (MOI) of 0.1) for 48 h through label-free quantitative proteomic analysis and transmission electron microscopy (TEM). TEM images showed a high load of intracellular viral cargo at 48 h postinfection (hpi), as well as an unusual elongated mitochondria morphology that might indicate a mitochondrial imbalance. Proteome analysis revealed 196 regulated protein groups upon infection, which are related to protein synthesis, energy metabolism, signaling pathways, and apoptosis. These Aag-2 proteins regulated during CHIKV infection might have roles in antiviral and/or proviral mechanisms and the balance between viral propagation and the survival of host cells, possibly leading to the persistent infection.
Collapse
Affiliation(s)
- Anna Fernanda Vasconcellos
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
- Laboratory of Virology, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
| | - Reynaldo Magalhães Melo
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
| | - Samuel Coelho Mandacaru
- Laboratory of Toxinology and Center for Technological Development in Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Lucas Silva de Oliveira
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
| | - Athos Silva de Oliveira
- Laboratory of Virology, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
| | | | | | - Carlos André Ornelas Ricart
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
| | - Sônia Nair Báo
- Laboratory of Microscopy and Microanalysis, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
| | - Renato Oliveira Resende
- Laboratory of Virology, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
- *Correspondence: Sébastien Charneau, ; Renato Oliveira Resende,
| | - Sébastien Charneau
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
- *Correspondence: Sébastien Charneau, ; Renato Oliveira Resende,
| |
Collapse
|
109
|
Ahmad I, Newell-Fugate AE. Androgen and androgen receptor control of mitochondrial function. Am J Physiol Cell Physiol 2022; 323:C835-C846. [PMID: 35704694 DOI: 10.1152/ajpcell.00205.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The effects of androgens have been extensively studied in a variety of organs and cell types with increasing focus on the sexually dimorphic role androgens play not only with respect to cellular functions but also in metabolism. Although the classical mechanism of androgen action is via ligand-dependent binding with the nuclear transcription factor, androgen receptor (AR), cytosolic AR can also activate second messenger signaling pathways. Given that cytosolic AR can signal in this manner, there has been increased interest in the mechanisms by which androgens may control cellular organelle function. This review highlights the effects that androgens have on mitochondrial structure and function with emphasis on biogenesis, fusion/fission, mitophagy, bioenergetics (oxidative phosphorylation), and reactive oxygen species production. There are a number of publications on the effects of androgens in these general areas of mitochondrial function. However, the precise mechanisms by which androgens cause these effects are not known. Additionally, given that the nucleus and mitochondria work in tandem to control mitochondrial function and the mitochondria has its own DNA, future research efforts should focus on the direct, mechanistic effects of androgens on mitochondrial function.
Collapse
Affiliation(s)
- Irshad Ahmad
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| | - Annie E Newell-Fugate
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| |
Collapse
|
110
|
Ford HC, Allen WJ, Pereira GC, Liu X, Dillingham MS, Collinson I. Towards a molecular mechanism underlying mitochondrial protein import through the TOM and TIM23 complexes. eLife 2022; 11:75426. [PMID: 35674314 PMCID: PMC9255969 DOI: 10.7554/elife.75426] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 06/07/2022] [Indexed: 12/27/2022] Open
Abstract
Nearly all mitochondrial proteins need to be targeted for import from the cytosol. For the majority, the first port of call is the translocase of the outer membrane (TOM complex), followed by a procession of alternative molecular machines, conducting transport to their final destination. The pre-sequence translocase of the inner membrane (TIM23-complex) imports proteins with cleavable pre-sequences. Progress in understanding these transport mechanisms has been hampered by the poor sensitivity and time resolution of import assays. However, with the development of an assay based on split NanoLuc luciferase, we can now explore this process in greater detail. Here, we apply this new methodology to understand how ∆ψ and ATP hydrolysis, the two main driving forces for import into the matrix, contribute to the transport of pre-sequence-containing precursors (PCPs) with varying properties. Notably, we found that two major rate-limiting steps define PCP import time: passage of PCP across the outer membrane and initiation of inner membrane transport by the pre-sequence - the rates of which are influenced by PCP size and net charge. The apparent distinction between transport through the two membranes (passage through TOM is substantially complete before PCP-TIM engagement) is in contrast with the current view that import occurs through TOM and TIM in a single continuous step. Our results also indicate that PCPs spend very little time in the TIM23 channel - presumably rapid success or failure of import is critical for maintenance of mitochondrial fitness.
Collapse
Affiliation(s)
- Holly C Ford
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | - William J Allen
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | - Gonçalo C Pereira
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | - Xia Liu
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | | | - Ian Collinson
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
111
|
Haslem L, Hays JM, Hays FA. p66Shc in Cardiovascular Pathology. Cells 2022; 11:cells11111855. [PMID: 35681549 PMCID: PMC9180016 DOI: 10.3390/cells11111855] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 02/06/2023] Open
Abstract
p66Shc is a widely expressed protein that governs a variety of cardiovascular pathologies by generating, and exacerbating, pro-apoptotic ROS signals. Here, we review p66Shc’s connections to reactive oxygen species, expression, localization, and discuss p66Shc signaling and mitochondrial functions. Emphasis is placed on recent p66Shc mitochondrial function discoveries including structure/function relationships, ROS identity and regulation, mechanistic insights, and how p66Shc-cyt c interactions can influence p66Shc mitochondrial function. Based on recent findings, a new p66Shc mitochondrial function model is also put forth wherein p66Shc acts as a rheostat that can promote or antagonize apoptosis. A discussion of how the revised p66Shc model fits previous findings in p66Shc-mediated cardiovascular pathology follows.
Collapse
Affiliation(s)
- Landon Haslem
- Biochemistry and Molecular Biology Department, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (L.H.); (J.M.H.)
| | - Jennifer M. Hays
- Biochemistry and Molecular Biology Department, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (L.H.); (J.M.H.)
| | - Franklin A. Hays
- Biochemistry and Molecular Biology Department, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (L.H.); (J.M.H.)
- Stephenson Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Correspondence:
| |
Collapse
|
112
|
Bomer N, Pavez-Giani MG, Grote Beverborg N, Cleland JGF, van Veldhuisen DJ, van der Meer P. Micronutrient deficiencies in heart failure: Mitochondrial dysfunction as a common pathophysiological mechanism? J Intern Med 2022; 291:713-731. [PMID: 35137472 PMCID: PMC9303299 DOI: 10.1111/joim.13456] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Heart failure is a devastating clinical syndrome, but current therapies are unable to abolish the disease burden. New strategies to treat or prevent heart failure are urgently needed. Over the past decades, a clear relationship has been established between poor cardiac performance and metabolic perturbations, including deficits in substrate uptake and utilization, reduction in mitochondrial oxidative phosphorylation and excessive reactive oxygen species production. Together, these perturbations result in progressive depletion of cardiac adenosine triphosphate (ATP) and cardiac energy deprivation. Increasing the delivery of energy substrates (e.g., fatty acids, glucose, ketones) to the mitochondria will be worthless if the mitochondria are unable to turn these energy substrates into fuel. Micronutrients (including coenzyme Q10, zinc, copper, selenium and iron) are required to efficiently convert macronutrients to ATP. However, up to 50% of patients with heart failure are deficient in one or more micronutrients in cross-sectional studies. Micronutrient deficiency has a high impact on mitochondrial energy production and should be considered an additional factor in the heart failure equation, moving our view of the failing myocardium away from an "an engine out of fuel" to "a defective engine on a path to self-destruction." This summary of evidence suggests that supplementation with micronutrients-preferably as a package rather than singly-might be a potential therapeutic strategy in the treatment of heart failure patients.
Collapse
Affiliation(s)
- Nils Bomer
- Department of Cardiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Mario G Pavez-Giani
- Department of Cardiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Niels Grote Beverborg
- Department of Cardiology, University Medical Center Groningen, Groningen, The Netherlands
| | - John G F Cleland
- Robertson Centre for Biostatistics and Clinical Trials, University of Glasgow, Glasgow, UK.,National Heart & Lung Institute, Royal Brompton and Harefield Hospitals, Imperial College, London, UK
| | - Dirk J van Veldhuisen
- Department of Cardiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Peter van der Meer
- Department of Cardiology, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
113
|
Norton CE, Shaw RL, Mittler R, Segal SS. Endothelial cells promote smooth muscle cell resilience to H 2 O 2 -induced cell death in mouse cerebral arteries. Acta Physiol (Oxf) 2022; 235:e13819. [PMID: 35380737 DOI: 10.1111/apha.13819] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 03/28/2022] [Accepted: 03/31/2022] [Indexed: 12/01/2022]
Abstract
AIM Brain injury produces reactive oxygen species (ROS). However, little is known of how acute oxidative stress affects cell survival in the cerebral vascular supply. We hypothesized that endothelial cells (ECs) are more resilient to H2 O2 and protect vascular smooth muscle cells (SMCs) during acute oxidative stress. METHODS Mouse posterior cerebral arteries (PCAs; diameter, ~80 µm) were exposed to H2 O2 (200 µM, 50 min, 37°C). Nuclear staining identified dead and live cells of intact and endothelium-disrupted vessels. SMC [Ca2+ ]i was assessed with Fura-2 fluorescence, and superoxide production was assessed by dihydroethidium and MitoSOX fluorescence. RESULTS In response to H2 O2 : SMC death (21%) exceeded EC death (5%) and increased following endothelial disruption (to 48%) with a corresponding increase in SMC Ca2+ entry through transient receptor potential (TRP) channels. Whereas pharmacological inhibition of TRPV4 channels prevented SMC death and reduced Ca2+ entry for intact vessels, both remained elevated following endothelial disruption. In contrast, pharmacological inhibition or genetic deletion of TRPC3 prevented SMC death and attenuated Ca2+ entry for both intact and endothelium-disrupted vessels. Inhibiting gap junctions increased EC death (to 22%) while SMC death and [Ca2+ ]i responses were attenuated by inhibiting nitric oxide synthesis or scavenging superoxide/peroxynitrite. Inhibiting NADPH oxidases also prevented SMC Ca2+ entry and death. H2 O2 increased mitochondrial ROS production while scavenging mitochondria-derived superoxide prevented SMC death but not Ca2+ entry. CONCLUSIONS During acute exposure of cerebral arteries to acute oxidative stress, ECs are more resilient than SMCs and the endothelium may protect SMCs by reducing Ca2+ entry through TRPC3 channels.
Collapse
Affiliation(s)
- Charles E. Norton
- Department of Medical Pharmacology and Physiology University of Missouri Columbia Missouri USA
| | - Rebecca L. Shaw
- Department of Medical Pharmacology and Physiology University of Missouri Columbia Missouri USA
| | - Ron Mittler
- Department of Surgery University of Missouri Columbia Missouri USA
| | - Steven S. Segal
- Department of Medical Pharmacology and Physiology University of Missouri Columbia Missouri USA
- Dalton Cardiovascular Research Center Columbia Missouri USA
| |
Collapse
|
114
|
Eberhardt DR, Lee SH, Yin X, Balynas AM, Rekate EC, Kraiss JN, Lang MJ, Walsh MA, Streiff ME, Corbin AC, Li Y, Funai K, Sachse FB, Chaudhuri D. EFHD1 ablation inhibits cardiac mitoflash activation and protects cardiomyocytes from ischemia. J Mol Cell Cardiol 2022; 167:1-14. [PMID: 35304170 PMCID: PMC9107497 DOI: 10.1016/j.yjmcc.2022.03.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 02/23/2022] [Accepted: 03/11/2022] [Indexed: 12/07/2022]
Abstract
Altered levels of intracellular calcium (Ca2+) are a highly prevalent feature in different forms of cardiac injury, producing changes in contractility, arrhythmias, and mitochondrial dysfunction. In cardiac ischemia-reperfusion injury, mitochondrial Ca2+ overload leads to pathological production of reactive oxygen species (ROS), activates the permeability transition, and cardiomyocyte death. Here we investigated the cardiac phenotype caused by deletion of EF-hand domain-containing protein D1 (Efhd1-/-), a Ca2+-binding mitochondrial protein whose function is poorly understood. Efhd1-/- mice are viable and have no adverse cardiac phenotypes. They feature reductions in basal ROS levels and mitoflash events, both important precursors for mitochondrial injury, though cardiac mitochondria have normal susceptibility to Ca2+ overload. Notably, we also find that Efhd1-/- mice and their cardiomyocytes are resistant to hypoxic injury.
Collapse
Affiliation(s)
- David R Eberhardt
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah, United States of America
| | - Sandra H Lee
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah, United States of America
| | - Xue Yin
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah, United States of America
| | - Anthony M Balynas
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah, United States of America
| | - Emma C Rekate
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah, United States of America
| | - Jackie N Kraiss
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah, United States of America
| | - Marisa J Lang
- Diabetes & Metabolism Research Center, University of Utah, United States of America
| | - Maureen A Walsh
- Diabetes & Metabolism Research Center, University of Utah, United States of America
| | - Molly E Streiff
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah, United States of America; Department of Biomedical Engineering, University of Utah, United States of America
| | - Andrea C Corbin
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah, United States of America; Department of Biomedical Engineering, University of Utah, United States of America
| | - Ying Li
- Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, United States of America
| | - Katsuhiko Funai
- Diabetes & Metabolism Research Center, University of Utah, United States of America
| | - Frank B Sachse
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah, United States of America; Department of Biomedical Engineering, University of Utah, United States of America
| | - Dipayan Chaudhuri
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah, United States of America; Department of Biomedical Engineering, University of Utah, United States of America; Department of Internal Medicine, Division of Cardiovascular Medicine, University of Utah, Salt Lake City, UT, United States of America.
| |
Collapse
|
115
|
Sono-ReCORMs for synergetic sonodynamic-gas therapy of hypoxic tumor. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.06.076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
116
|
Ali MY, Oliva CR, Flor S, Goswami PC, Griguer CE. Cytochrome c oxidase mediates labile iron level and radioresistance in glioblastoma. Free Radic Biol Med 2022; 185:25-35. [PMID: 35476930 DOI: 10.1016/j.freeradbiomed.2022.04.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 04/11/2022] [Accepted: 04/20/2022] [Indexed: 02/06/2023]
Abstract
Radiotherapy is an important treatment modality for glioblastoma (GBM), yet the initial effectiveness of radiotherapy is eventually lost due to the development of adaptive radioresistance during fractionated radiation therapy. Defining the molecular mechanism(s) responsible for the adaptive radioresistance in GBM is necessary for the development of effective treatment options. The cellular labile iron pool (LIP) is very important for determining the cellular response to radiation, as it contributes to radiation-induced production of reactive oxygen species (ROS) such as lipid radicals through Fenton reactions. Recently, cytochrome c oxidase (CcO), a mitochondrial heme-containing enzyme also involved in regulating ROS production, was found to be involved in GBM chemoresistance. However, the role of LIP and CcO in GBM radioresistance is not known. Herein, we tested the hypothesis that CcO-mediated alterations in the level of labile iron contribute to adaptive radioresistance. Using an in vitro model of GBM adaptive radioresistance, we found an increase in CcO activity in radioresistant cells that associated with a decrease in the cellular LIP, decrease in lipid peroxidation, and a switch in the CcO subunit 4 (COX4) isoform expressed, from COX4-2 to COX4-1. Furthermore, knockdown of COX4-1 in radioresistant GBM cells decreased CcO activity and restored radiosensitivity, whereas overexpression of COX4-1 in radiosensitive cells increased CcO activity and rendered the cells radioresistant. Overexpression of COX4-1 in radiosensitive cells also significantly reduced the cellular LIP and lipid peroxidation. Pharmacological manipulation of the cellular labile iron level using iron chelators altered CcO activity and the radiation response. Overall, these results demonstrate a mechanistic link between CcO activity and LIP in GBM radioresistance and identify the CcO subunit isoform switch from COX4-2 to COX4-1 as a novel biochemical node for adaptive radioresistance of GBM. Manipulation of CcO and the LIP may restore the sensitivity to radiation in radioresistant GBM cells and thereby provide a strategy to improve therapeutic outcome in patients with GBM.
Collapse
Affiliation(s)
- Md Yousuf Ali
- Free Radical & Radiation Biology Program, Department of Radiation Oncology, The University of Iowa, Iowa City, IA, 52242, USA; Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, IA, 52242, USA
| | - Claudia R Oliva
- Free Radical & Radiation Biology Program, Department of Radiation Oncology, The University of Iowa, Iowa City, IA, 52242, USA
| | - Susanne Flor
- Free Radical & Radiation Biology Program, Department of Radiation Oncology, The University of Iowa, Iowa City, IA, 52242, USA
| | - Prabhat C Goswami
- Free Radical & Radiation Biology Program, Department of Radiation Oncology, The University of Iowa, Iowa City, IA, 52242, USA
| | - Corinne E Griguer
- Free Radical & Radiation Biology Program, Department of Radiation Oncology, The University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
117
|
Yin Z, Zheng H, Guo Z. Effect of Sodium-Glucose Co-transporter Protein 2 Inhibitors on Arrhythmia in Heart Failure Patients With or Without Type 2 Diabetes: A Meta-Analysis of Randomized Controlled Trials. Front Cardiovasc Med 2022; 9:902923. [PMID: 35665272 PMCID: PMC9157597 DOI: 10.3389/fcvm.2022.902923] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/04/2022] [Indexed: 01/10/2023] Open
Abstract
Aim Arrhythmic events such as atrial fibrillation (AF) are tightly associated with an increased risk of heart failure (HF). Previous studies have shown inconsistent results regarding the association between sodium-glucose co-transporter 2 inhibitors (SGLT2i) and the risk of arrhythmia. The purpose of this study was to investigate the association of SGLT2i treatment with arrhythmia outcomes in clinical trials of patients with HF. Methods We searched Embase, PubMed, Web of Science, Medline, The Cochrane Library, and JAMA databases to identify appropriate randomized controlled trials (RCTs) of SGLT2i interventions. Endpoint outcomes included AF, atrial flutter (AFL), AF/AFL, ventricular fibrillation (VF), ventricular tachycardia (VT), VF/VT, and bradycardia. A random-effects model was used for the meta-analysis of all outcomes. The risk of bias and quality of evidence was assessed by using the Cochrane tool and assessment framework. Results Out of 1,725 citations, 9 trials were included in this study, with follow-up from 4 weeks to 52 weeks for 10,344 participants (mean age 68.27 years; 69.62% of participants were men). Compared with placebo, SGLT2i reduced the incidence of AF by 37% [ratio risk (RR) 0.63; 95% confidence interval (CI) 0.45–0.87; p < 0.05] and AF/AFL by 34% (RR 0.66; 95% CI 0.49–0.90; p < 0.05). Conclusions SGLT2i can reduce the risk of cardiac arrhythmias, particularly the AF. Our study provides strong evidence for recommending the use of SGLT2i in patients with HF. Systematic Review Registration PROSPERO, identifier: CRD42022296696.
Collapse
Affiliation(s)
- Ziwei Yin
- Department of Cardiology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Huizhen Zheng
- Department of Cardiology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Zhihua Guo
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- *Correspondence: Zhihua Guo
| |
Collapse
|
118
|
Shiau JP, Chuang YT, Cheng YB, Tang JY, Hou MF, Yen CY, Chang HW. Impacts of Oxidative Stress and PI3K/AKT/mTOR on Metabolism and the Future Direction of Investigating Fucoidan-Modulated Metabolism. Antioxidants (Basel) 2022; 11:911. [PMID: 35624775 PMCID: PMC9137824 DOI: 10.3390/antiox11050911] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/05/2022] [Indexed: 12/22/2022] Open
Abstract
The critical factors for regulating cancer metabolism are oxidative stress and phosphoinositide-3-kinase/AKT serine-threonine kinase/mechanistic target of the rapamycin kinase (PI3K/AKT/mTOR). However, the metabolic impacts of oxidative stress and PI3K/AKT/mTOR on individual mechanisms such as glycolysis (Warburg effect), pentose phosphate pathway (PPP), fatty acid synthesis, tricarboxylic acid cycle (TCA) cycle, glutaminolysis, and oxidative phosphorylation (OXPHOS) are complicated. Therefore, this review summarizes the individual and interacting functions of oxidative stress and PI3K/AKT/mTOR on metabolism. Moreover, natural products providing oxidative stress and PI3K/AKT/mTOR modulating effects have anticancer potential. Using the example of brown algae-derived fucoidan, the roles of oxidative stress and PI3K/AKT/mTOR were summarized, although their potential functions within diverse metabolisms were rarely investigated. We propose a potential application that fucoidan may regulate oxidative stress and PI3K/AKT/mTOR signaling to modulate their associated metabolic regulations. This review sheds light on understanding the impacts of oxidative stress and PI3K/AKT/mTOR on metabolism and the future direction of metabolism-based cancer therapy of fucoidan.
Collapse
Affiliation(s)
- Jun-Ping Shiau
- Department of Surgery, Kaohsiung Municipal Siaogang Hospital, Kaohsiung 81267, Taiwan;
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Ya-Ting Chuang
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Yuan-Bin Cheng
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan;
| | - Jen-Yang Tang
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaoshiung Medical University, Kaohsiung 80708, Taiwan
| | - Ming-Feng Hou
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Ching-Yu Yen
- Department of Oral, Maxillofacial Surgery Chi-Mei Medical Center, Tainan 71004, Taiwan
- School of Dentistry, Taipei Medical University, Taipei 11031, Taiwan
| | - Hsueh-Wei Chang
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| |
Collapse
|
119
|
Walkowski B, Kleibert M, Majka M, Wojciechowska M. Insight into the Role of the PI3K/Akt Pathway in Ischemic Injury and Post-Infarct Left Ventricular Remodeling in Normal and Diabetic Heart. Cells 2022; 11:cells11091553. [PMID: 35563860 PMCID: PMC9105930 DOI: 10.3390/cells11091553] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/28/2022] [Accepted: 05/03/2022] [Indexed: 02/07/2023] Open
Abstract
Despite the significant decline in mortality, cardiovascular diseases are still the leading cause of death worldwide. Among them, myocardial infarction (MI) seems to be the most important. A further decline in the death rate may be achieved by the introduction of molecularly targeted drugs. It seems that the components of the PI3K/Akt signaling pathway are good candidates for this. The PI3K/Akt pathway plays a key role in the regulation of the growth and survival of cells, such as cardiomyocytes. In addition, it has been shown that the activation of the PI3K/Akt pathway results in the alleviation of the negative post-infarct changes in the myocardium and is impaired in the state of diabetes. In this article, the role of this pathway was described in each step of ischemia and subsequent left ventricular remodeling. In addition, we point out the most promising substances which need more investigation before introduction into clinical practice. Moreover, we present the impact of diabetes and widely used cardiac and antidiabetic drugs on the PI3K/Akt pathway and discuss the molecular mechanism of its effects on myocardial ischemia and left ventricular remodeling.
Collapse
Affiliation(s)
- Bartosz Walkowski
- Laboratory of Centre for Preclinical Research, Department of Experimental and Clinical Physiology, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland; (B.W.); (M.W.)
| | - Marcin Kleibert
- Laboratory of Centre for Preclinical Research, Department of Experimental and Clinical Physiology, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland; (B.W.); (M.W.)
- Correspondence: (M.K.); (M.M.)
| | - Miłosz Majka
- Laboratory of Centre for Preclinical Research, Department of Experimental and Clinical Physiology, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland; (B.W.); (M.W.)
- Correspondence: (M.K.); (M.M.)
| | - Małgorzata Wojciechowska
- Laboratory of Centre for Preclinical Research, Department of Experimental and Clinical Physiology, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland; (B.W.); (M.W.)
- Invasive Cardiology Unit, Independent Public Specialist Western Hospital John Paul II, Daleka 11, 05-825 Grodzisk Mazowiecki, Poland
| |
Collapse
|
120
|
A novel nitronyl nitroxide radical HPN-C6 attenuates brain damage in an acute hypobaric hypoxia mouse model through inhibition of the oxidative stress. Neurosci Lett 2022; 782:136650. [DOI: 10.1016/j.neulet.2022.136650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 04/16/2022] [Accepted: 04/19/2022] [Indexed: 11/22/2022]
|
121
|
Mooli RGR, Mukhi D, Ramakrishnan SK. Oxidative Stress and Redox Signaling in the Pathophysiology of Liver Diseases. Compr Physiol 2022; 12:3167-3192. [PMID: 35578969 PMCID: PMC10074426 DOI: 10.1002/cphy.c200021] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The increased production of derivatives of molecular oxygen and nitrogen in the form of reactive oxygen species (ROS) and reactive nitrogen species (RNS) lead to molecular damage called oxidative stress. Under normal physiological conditions, the ROS generation is tightly regulated in different cells and cellular compartments. Any disturbance in the balance between the cellular generation of ROS and antioxidant balance leads to oxidative stress. In this article, we discuss the sources of ROS (endogenous and exogenous) and antioxidant mechanisms. We also focus on the pathophysiological significance of oxidative stress in various cell types of the liver. Oxidative stress is implicated in the development and progression of various liver diseases. We narrate the master regulators of ROS-mediated signaling and their contribution to liver diseases. Nonalcoholic fatty liver diseases (NAFLD) are influenced by a "multiple parallel-hit model" in which oxidative stress plays a central role. We highlight the recent findings on the role of oxidative stress in the spectrum of NAFLD, including fibrosis and liver cancer. Finally, we provide a brief overview of oxidative stress biomarkers and their therapeutic applications in various liver-related disorders. Overall, the article sheds light on the significance of oxidative stress in the pathophysiology of the liver. © 2022 American Physiological Society. Compr Physiol 12:3167-3192, 2022.
Collapse
Affiliation(s)
- Raja Gopal Reddy Mooli
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Dhanunjay Mukhi
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Sadeesh K Ramakrishnan
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
122
|
Chen Q, Lesnefsky EJ. Time to Target Mitochondrial Reactive Oxygen Species Generation from Complex I. FUNCTION 2022; 3:zqac010. [PMID: 35359911 PMCID: PMC8951057 DOI: 10.1093/function/zqac010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/20/2022] [Accepted: 02/22/2022] [Indexed: 01/07/2023] Open
Affiliation(s)
- Qun Chen
- Department of Internal Medicine, Cardiology, Pauley Heart Center, Richmond, VA 23298, USA
| | - Edward J Lesnefsky
- Department of Internal Medicine, Cardiology, Pauley Heart Center, Richmond, VA 23298, USA
- Biochemistry, and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA
- Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA 23298, USA
- The Medical Service of the McGuire Veterans Affairs Medical Center in Richmond, VA 23249, USA
| |
Collapse
|
123
|
Karkucinska-Wieckowska A, Simoes ICM, Kalinowski P, Lebiedzinska-Arciszewska M, Zieniewicz K, Milkiewicz P, Górska-Ponikowska M, Pinton P, Malik AN, Krawczyk M, Oliveira PJ, Wieckowski MR. Mitochondria, oxidative stress and nonalcoholic fatty liver disease: A complex relationship. Eur J Clin Invest 2022; 52:e13622. [PMID: 34050922 DOI: 10.1111/eci.13622] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/19/2021] [Accepted: 05/22/2021] [Indexed: 02/06/2023]
Abstract
According to the 'multiple-hit' hypothesis, several factors can act simultaneously in nonalcoholic fatty liver disease (NAFLD) progression. Increased nitro-oxidative (nitroso-oxidative) stress may be considered one of the main contributors involved in the development and risk of NAFLD progression to nonalcoholic steatohepatitis (NASH) characterized by inflammation and fibrosis. Moreover, it has been repeatedly postulated that mitochondrial abnormalities are closely related to the development and progression of liver steatosis and NAFLD pathogenesis. However, it is difficult to determine with certainty whether mitochondrial dysfunction or oxidative stress are primary events or a simple consequence of NAFLD development. On the one hand, increasing lipid accumulation in hepatocytes could cause a wide range of effects from mild to severe mitochondrial damage with a negative impact on cell fate. This can start the cascade of events, including an increase of cellular reactive nitrogen species (RNS) and reactive oxygen species (ROS) production that promotes disease progression from simple steatosis to more severe NAFLD stages. On the other hand, progressing mitochondrial bioenergetic catastrophe and oxidative stress manifestation could be considered accompanying events in the vast spectrum of abnormalities observed during the transition from NAFL to NASH and cirrhosis. This review updates our current understanding of NAFLD pathogenesis and clarifies whether mitochondrial dysfunction and ROS/RNS are culprits or bystanders of NAFLD progression.
Collapse
Affiliation(s)
| | - Ines C M Simoes
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Piotr Kalinowski
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Magdalena Lebiedzinska-Arciszewska
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Krzysztof Zieniewicz
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Piotr Milkiewicz
- Liver and Internal Medicine Unit, Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland.,Translational Medicine Group, Pomeranian Medical University, Szczecin, Poland
| | | | - Paolo Pinton
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies, University of Ferrara, Ferrara, Italy
| | - Afshan N Malik
- Department of Diabetes, School of Life Course, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Marcin Krawczyk
- Laboratory of Metabolic Liver Diseases, Department of General, Transplant and Liver Surgery, Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland.,Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Paulo J Oliveira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, CIBB - Centre for Innovative Biomedicine and Biotechnology, Coimbra, Portugal
| | - Mariusz R Wieckowski
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
124
|
Mitochondrial dysfunction and oxidative stress contribute to cognitive and motor impairment in FOXP1 syndrome. Proc Natl Acad Sci U S A 2022; 119:2112852119. [PMID: 35165191 PMCID: PMC8872729 DOI: 10.1073/pnas.2112852119] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2021] [Indexed: 12/22/2022] Open
Abstract
FOXP1 haploinsufficiency underlies cognitive and motor impairments in individuals with FOXP1 syndrome. Here, we show that mice lacking one Foxp1 copy exhibit similar behavioral deficits, which may be caused by striatal dysfunction. Indeed, Foxp1+/− striatal medium spiny neurons display reduced neurite branching, and we show altered mitochondrial biogenesis and dynamics; increased mitophagy; reduced mitochondrial membrane potential, structure, and motility; and elevated oxygen species in the striatum of these animals. As FOXP1 is highly conserved, our data strongly suggest that mitochondrial dysfunction and excessive oxidative stress contribute to the motor and cognitive impairments seen in individuals with FOXP1 syndrome. Thus, mitochondrial homeostasis is critical for normal development and can explain deficits in neurodevelopmental disorders. FOXP1 syndrome caused by haploinsufficiency of the forkhead box protein P1 (FOXP1) gene is a neurodevelopmental disorder that manifests motor dysfunction, intellectual disability, autism, and language impairment. In this study, we used a Foxp1+/− mouse model to address whether cognitive and motor deficits in FOXP1 syndrome are associated with mitochondrial dysfunction and oxidative stress. Here, we show that genes with a role in mitochondrial biogenesis and dynamics (e.g., Foxo1, Pgc-1α, Tfam, Opa1, and Drp1) were dysregulated in the striatum of Foxp1+/− mice at different postnatal stages. Furthermore, these animals exhibit a reduced mitochondrial membrane potential and complex I activity, as well as decreased expression of the antioxidants superoxide dismutase 2 (Sod2) and glutathione (GSH), resulting in increased oxidative stress and lipid peroxidation. These features can explain the reduced neurite branching, learning and memory, endurance, and motor coordination that we observed in these animals. Taken together, we provide strong evidence of mitochondrial dysfunction in Foxp1+/− mice, suggesting that insufficient energy supply and excessive oxidative stress underlie the cognitive and motor impairment in FOXP1 deficiency.
Collapse
|
125
|
Marchini T, Magnani N, Garces M, Kelly J, Paz M, Caceres L, Calabro V, Lasagni Vitar R, Caltana L, Contin M, Reynoso S, Lago N, Vico T, Vanasco V, Wolf D, Tripodi V, Gonzalez Maglio D, Alvarez S, Buchholz B, Berra A, Gelpi R, Evelson P. Chronic exposure to polluted urban air aggravates myocardial infarction by impaired cardiac mitochondrial function and dynamics. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 295:118677. [PMID: 34906594 DOI: 10.1016/j.envpol.2021.118677] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 11/05/2021] [Accepted: 12/10/2021] [Indexed: 06/14/2023]
Abstract
Air pollution exposure positively correlates with increased cardiovascular morbidity and mortality rates, mainly due to myocardial infarction (MI). Herein, we aimed to study the metabolic mechanisms underlying this association, focusing on the evaluation of cardiac mitochondrial function and dynamics, together with its impact over MI progression. An initial time course study was performed in BALB/c mice breathing filtered air (FA) or urban air (UA) in whole-body exposure chambers located in Buenos Aires City downtown for up to 16 weeks (n = 8 per group and time point). After 12 weeks, lung inflammatory cell recruitment was evident in UA-exposed mice. Interestingly, impaired redox metabolism, characterized by decreased lung SOD activity and increased GSSG levels and NOX activity, precede local inflammation in this group. At this selected time point, additional mice were exposed to FA or UA (n = 12 per group) and alveolar macrophage PM uptake and nitric oxide (NO) production was observed in UA-exposed mice, together with increased pro-inflammatory cytokine levels (TNF-α and IL-6) in BAL and plasma. Consequently, impaired heart tissue oxygen metabolism and altered mitochondrial ultrastructure and function were observed in UA-exposed mice after 12 weeks, characterized by decreased active state respiration and ATP production rates, and enhanced mitochondrial H2O2 production. Moreover, disturbed cardiac mitochondrial dynamics was detected in this group. This scenario led to a significant increase in the area of infarcted tissue following myocardial ischemia reperfusion injury in vivo, from 43 ± 3% of the area at risk in mice breathing FA to 66 ± 4% in UA-exposed mice (n = 6 per group, p < 0.01), together with a sustained increase in LVEDP during myocardial reperfusion. Taken together, our data unravel cardiac mitochondrial mechanisms that contribute to the understanding of the adverse health effects of urban air pollution exposure, and ultimately highlight the importance of considering environmental factors in the development of cardiovascular diseases.
Collapse
Affiliation(s)
- Timoteo Marchini
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina; University Heart Center Freiburg-Bad Krozingen, Cardiology and Angiology I, Faculty of Medicine, University of Freiburg, Freiburg, 79106, Germany
| | - Natalia Magnani
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina
| | - Mariana Garces
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina
| | - Jazmin Kelly
- CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Departamento de Patología, Buenos Aires, C1113AAD, Argentina
| | - Mariela Paz
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Estudios de la Inmunidad Humoral (IDEHU), Buenos Aires, C1113AAD, Argentina
| | - Lourdes Caceres
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina
| | - Valeria Calabro
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina
| | - Romina Lasagni Vitar
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina
| | - Laura Caltana
- CONICET - Universidad de Buenos Aires, Instituto de Biología Celular y Neurociencias (IBCN), Buenos Aires, C1121ABG, Argentina
| | - Mario Contin
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Tecnología Farmacéutica, Buenos Aires, C1113AAD, Argentina
| | - Sofia Reynoso
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina
| | - Nestor Lago
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Patología, Buenos Aires, C1113AAD, Argentina
| | - Tamara Vico
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina
| | - Virginia Vanasco
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina
| | - Dennis Wolf
- University Heart Center Freiburg-Bad Krozingen, Cardiology and Angiology I, Faculty of Medicine, University of Freiburg, Freiburg, 79106, Germany
| | - Valeria Tripodi
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Tecnología Farmacéutica, Buenos Aires, C1113AAD, Argentina
| | - Daniel Gonzalez Maglio
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Estudios de la Inmunidad Humoral (IDEHU), Buenos Aires, C1113AAD, Argentina
| | - Silvia Alvarez
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina
| | - Bruno Buchholz
- CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Departamento de Patología, Buenos Aires, C1113AAD, Argentina
| | - Alejandro Berra
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Patología, Buenos Aires, C1113AAD, Argentina
| | - Ricardo Gelpi
- CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Departamento de Patología, Buenos Aires, C1113AAD, Argentina
| | - Pablo Evelson
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina.
| |
Collapse
|
126
|
Ebanks B, Chakrabarti L. Mitochondrial ATP Synthase is a Target of Oxidative Stress in Neurodegenerative Diseases. Front Mol Biosci 2022; 9:854321. [PMID: 35237666 PMCID: PMC8882969 DOI: 10.3389/fmolb.2022.854321] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 01/26/2022] [Indexed: 12/11/2022] Open
Abstract
The mitochondrial ATP synthase is responsible for the production of cellular ATP, and it does so by harnessing the membrane potential of the mitochondria that is produced by the sequential oxidation of select cellular metabolites. Since the structural features of ATP synthase were first resolved nearly three decades ago, significant progress has been made in understanding its role in health and disease. Mitochondrial dysfunction is common to neurodegeneration, with elevated oxidative stress a hallmark of this dysfunction. The patterns of this oxidative stress, including molecular targets and the form of oxidative modification, can vary widely. In this mini review we discuss the oxidative modifications of ATP synthase that have been observed in Alzheimer’s disease, Parkinson’s disease, and Huntington’s disease. Oxidative modifications of ATP synthase in Alzheimer’s disease are well-documented, and there is a growing body of knowledge on the subject in Parkinson’s disease. The consideration of ATP synthase as a pharmacological target in a variety of diseases underlines the importance of understanding these modifications, both as a potential target, and also as inhibitors of any pharmacological intervention.
Collapse
Affiliation(s)
- Brad Ebanks
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
- *Correspondence: Brad Ebanks,
| | - Lisa Chakrabarti
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, Nottingham, United Kingdom
| |
Collapse
|
127
|
Network-Assisted Systems Biology Analysis of the Mitochondrial Proteome in a Pre-Clinical Model of Ischemia, Revascularization and Post-Conditioning. Int J Mol Sci 2022; 23:ijms23042087. [PMID: 35216205 PMCID: PMC8879554 DOI: 10.3390/ijms23042087] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/07/2022] [Accepted: 02/10/2022] [Indexed: 12/24/2022] Open
Abstract
Infarct size is the major risk predictor for developing heart failure after an acute myocardial infarction (AMI). The discovery of the conditioning phenomena (i.e., repetitive brief cycles of ischemia applied either before or after a prolonged ischemic insult) has highlighted the existence of endogenous protective mechanisms of the heart potentially limiting infarct size after revascularization. However, most cardioprotective strategies, aiming at infarct size reduction, have failed in clinical studies. Thus, cardioprotection is an unmet clinical need. In the present study, we took a network-assisted systems biology approach to explore the mitochondrial proteomic signature of the myocardium after ischemia, ischemia with direct revascularization, and ischemia with re-establishment of blood flow by post-conditioning in a swine model of AMI. Furthermore, network extension with the ENCODE project human regulatory data allowed the prediction of potential transcription factors at play in the response to post-conditioning of the myocardium. Collectively, our results identify cardiac metabolism as a driver of cardioprotection, highlighting a dual role for post-conditioning promoting metabolic reprogramming of the myocardium, and a protective response mediated by VDAC2 and DJ-1 in the mitochondria.
Collapse
|
128
|
Carr SN, Crites BR, Pate JL, Hughes CHK, Matthews JC, Bridges PJ. Form of Supplemental Selenium Affects the Expression of mRNA Transcripts Encoding Selenoproteins, and Proteins Regulating Cholesterol Uptake, in the Corpus Luteum of Grazing Beef Cows. Animals (Basel) 2022; 12:313. [PMID: 35158637 PMCID: PMC8833813 DOI: 10.3390/ani12030313] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/15/2021] [Accepted: 01/21/2022] [Indexed: 02/06/2023] Open
Abstract
Selenium (Se)-deficient soils necessitate supplementation of this mineral to the diet of forage-grazing cattle. Functionally, Se is incorporated into selenoproteins, some of which function as important antioxidants. We have previously shown that the source of supplemental Se; inorganic (sodium selenite or sodium selenate; ISe), organic (selenomethionine or selenocysteine; OSe) or 1:1 mix of ISe and OSe (MIX), provided to Angus-cross cows affects concentrations of progesterone (P4) during the early luteal phase of the estrous cycle. In this study, we sought to investigate (1) the effect of form of Se on the expression of mRNA encoding selenoproteins in the corpus luteum (CL), and (2) whether this previously reported MIX-induced increase in P4 is the result of increased luteal expression of key steroidogenic transcripts. Following a Se depletion and repletion regimen, 3-year-old, non-lactating, Angus- cross cows were supplemented with either ISe as the industry standard, or MIX for at least 90 days, with the CL then retrieved on Day 7 post-estrus. Half of each CL was used for analysis of targeted mRNA transcripts and the remainder was dissociated for culture with select agonists. The expression of three selenoprotein transcripts and one selenoprotein P receptor was increased (p < 0.05), with an additional five transcripts tending to be increased (p < 0.10), in cows supplemented with MIX versus ISe. In cultures of luteal cells, hCG-induced increases in P4 (p < 0.05) were observed in CL obtained from ISe-supplemented cows. The abundance of steroidogenic transcripts in the CL was not affected by the form of Se, however, the abundance of mRNA encoding 2 key transcripts regulating cholesterol availability (Ldlr and Hsl) was increased (p < 0.05) in MIX-supplemented cows. Overall, the form of Se provided to cows is reported to affect the expression of mRNA encoding several selenoproteins in the CL, and that the form of Se-induced effects on luteal production of P4 appears to be the result of changes in cholesterol availability rather than a direct effect on the expression of steroidogenic enzymes within the CL.
Collapse
Affiliation(s)
- Sarah N. Carr
- Department of Animal and Food Sciences, University of Kentucky, Lexington, KY 40546, USA; (S.N.C.); (B.R.C.); (J.C.M.)
| | - Benjamin R. Crites
- Department of Animal and Food Sciences, University of Kentucky, Lexington, KY 40546, USA; (S.N.C.); (B.R.C.); (J.C.M.)
| | - Joy L. Pate
- Department of Animal Sciences, Center for Reproductive Biology and Health, The Pennsylvania State University, University Park, PA 16802, USA; (J.L.P.); (C.H.K.H.)
| | - Camilla H. K. Hughes
- Department of Animal Sciences, Center for Reproductive Biology and Health, The Pennsylvania State University, University Park, PA 16802, USA; (J.L.P.); (C.H.K.H.)
| | - James C. Matthews
- Department of Animal and Food Sciences, University of Kentucky, Lexington, KY 40546, USA; (S.N.C.); (B.R.C.); (J.C.M.)
| | - Phillip J. Bridges
- Department of Animal and Food Sciences, University of Kentucky, Lexington, KY 40546, USA; (S.N.C.); (B.R.C.); (J.C.M.)
| |
Collapse
|
129
|
Is Mitochondrial Oxidative Stress a Viable Therapeutic Target in Preeclampsia? Antioxidants (Basel) 2022; 11:antiox11020210. [PMID: 35204094 PMCID: PMC8868187 DOI: 10.3390/antiox11020210] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/15/2022] [Accepted: 01/18/2022] [Indexed: 01/31/2023] Open
Abstract
Despite considerable research efforts over the past few decades, the pathology of preeclampsia (PE) remains poorly understood with no new FDA-approved treatments. There is a substantial amount of work being conducted by investigators around the world to identify targets to develop therapies for PE. Oxidative stress has been identified as one of the crucial players in pathogenesis of PE and has garnered a great deal of attention by several research groups including ours. While antioxidants have shown therapeutic benefit in preclinical models of PE, the clinical trials evaluating antioxidants (vitamin E and vitamin C) were found to be disappointing. Although the idea behind contribution of mitochondrial oxidative stress in PE is not new, recent years have seen an enormous interest in exploring mitochondrial oxidative stress as an important pathological mediator in PE. We and others using animals, cell models, and preeclamptic patient samples have shown the evidence for placental, renal, and endothelial cell mitochondrial oxidative stress, and its significance in PE. These studies offer promising results; however, the important and relevant question is can we translate these results into clinical efficacy in treating PE. Hence, the purpose of this review is to review the existing literature and offer our insights on the potential of mitochondrial antioxidants in treating PE.
Collapse
|
130
|
Wang Y, Pan ZN, Xing CH, Zhang HL, Sun SC. Nivalenol affects spindle formation and organelle functions during mouse oocyte maturation. Toxicol Appl Pharmacol 2022; 436:115882. [PMID: 35016910 DOI: 10.1016/j.taap.2022.115882] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 12/19/2021] [Accepted: 01/07/2022] [Indexed: 12/29/2022]
Abstract
Oocyte maturation is essential for fertilization and early embryo development, and proper organelle functions guarantee this process to maintain high-quality oocytes. The type B trichothecene nivalenol (NIV) is a mycotoxin produced by Fusarium oxysporum and is commonly found in contaminated food. NIV intake affect growth, the immune system, and the female reproductive system. Here, we investigated NIV toxicity on mouse oocyte quality. Transcriptome analysis results showed that NIV exposure altered the expression of multiple genes involved in spindle formation and organelle function in mouse oocytes, indicating its toxicity on mouse oocyte maturation. Further analysis indicated that NIV exposure disrupted spindle structure and chromosome alignment, possibly through tubulin acetylation. NIV exposure induced aberrant mitochondria distribution and reduced mitochondria number, mitochondria membrane potential (MMP), and ATP levels. In addition, NIV caused the abnormal distribution of the Golgi apparatus and altered the expression of the vesicle trafficking protein Rab11. ER distribution was also disturbed under NIV exposure, indicating the effects of NIV on protein modification and transport in oocytes. Thus, our results demonstrated that NIV exposure affected spindle structure and organelles function in mouse oocytes.
Collapse
Affiliation(s)
- Yue Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhen-Nan Pan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Chun-Hua Xing
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Hao-Lin Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Shao-Chen Sun
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
131
|
Li L, Thompson J, Hu Y, Lesnefsky EJ, Willard B, Chen Q. Calpain-mediated protein targets in cardiac mitochondria following ischemia-reperfusion. Sci Rep 2022; 12:138. [PMID: 34997008 PMCID: PMC8741987 DOI: 10.1038/s41598-021-03947-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 12/09/2021] [Indexed: 12/30/2022] Open
Abstract
Calpain 1 and 2 (CPN1/2) are calcium-dependent cysteine proteases that exist in cytosol and mitochondria. Pharmacologic inhibition of CPN1/2 decreases cardiac injury during ischemia (ISC)-reperfusion (REP) by improving mitochondrial function. However, the protein targets of CPN1/2 activation during ISC-REP are unclear. CPN1/2 include a large subunit and a small regulatory subunit 1 (CPNS1). Genetic deletion of CPNS1 eliminates the activities of both CPN1 and CPN2. Conditional cardiomyocyte specific CPNS1 deletion mice were used in the present study to clarify the role of CPN1/2 activation in mitochondrial damage during ISC-REP with an emphasis on identifying the potential protein targets of CPN1/2. Isolated hearts from wild type (WT) or CPNS1 deletion mice underwent 25 min in vitro global ISC and 30 min REP. Deletion of CPNS1 led to decreased cytosolic and mitochondrial calpain 1 activation compared to WT. Cardiac injury was decreased in CPNS1 deletion mice following ISC-REP as shown by the decreased infarct size compared to WT. Compared to WT, mitochondrial function was improved in CPNS1 deletion mice following ischemia-reperfusion as shown by the improved oxidative phosphorylation and decreased susceptibility to mitochondrial permeability transition pore opening. H2O2 generation was also decreased in mitochondria from deletion mice following ISC-REP compared to WT. Deletion of CPNS1 also resulted in less cytochrome c and truncated apoptosis inducing factor (tAIF) release from mitochondria. Proteomic analysis of the isolated mitochondria showed that deletion of CPNS1 increased the content of proteins functioning in regulation of mitochondrial calcium homeostasis (paraplegin and sarcalumenin) and complex III activity. These results suggest that activation of CPN1 increases cardiac injury during ischemia-reperfusion by impairing mitochondrial function and triggering cytochrome c and tAIF release from mitochondria into cytosol.
Collapse
Affiliation(s)
- Ling Li
- Proteomics Core, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Jeremy Thompson
- Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Ying Hu
- Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Edward J Lesnefsky
- Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, 23298, USA
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, 23298, USA
- McGuire Department of Veterans Affairs Medical Center, Richmond, VA, 23249, USA
| | - Belinda Willard
- Proteomics Core, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Qun Chen
- Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA.
| |
Collapse
|
132
|
The Oxidative Stress and Chronic Inflammatory Process in Chagas Disease: Role of Exosomes and Contributing Genetic Factors. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2021:4993452. [PMID: 34976301 PMCID: PMC8718323 DOI: 10.1155/2021/4993452] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/27/2021] [Accepted: 12/06/2021] [Indexed: 12/15/2022]
Abstract
Chagas disease is a neglected tropical disease caused by the flagellated protozoa Trypanosoma cruzi that affects several million people mainly in Latin American countries. Chagas disease has two phases, which are acute and chronic, both separated by an indeterminate time period in which the infected individual is relatively asymptomatic. The acute phase extends for 40-60 days with atypical and mild symptoms; however, about 30% of the infected patients will develop a symptomatic chronic phase, which is characterized by either cardiac, digestive, neurological, or endocrine problems. Cardiomyopathy is the most important and severe result of Chagas disease, which leads to left ventricular systolic dysfunction, heart failure, and sudden cardiac death. Most deaths are due to heart failure (70%) and sudden death (30%) resulting from cardiomyopathy. During the chronic phase, T. cruzi-infected macrophages respond with the production of proinflammatory cytokines and production of superoxide and nitric oxide by the NADPH oxidase 2 (NOX2) and inducible nitric oxide synthase (iNOS) enzymes, respectively. During the chronic phase, myocardial changes are produced as a result of chronic inflammation, oxidative stress, fibrosis, and cell death. The cellular inflammatory response is mainly the result of activation of the NF-κB-dependent pathway, which activates gene expression of inflammatory cytokines, leading to progressive tissue damage. The persisting production of reactive oxygen species (ROS) is the result of mitochondrial dysfunction in the cardiomyocytes. In this review, we will discuss inflammation and oxidative damage which is produced in the heart during the chronic phase of Chagas disease and recent evidence on the role of macrophages and the production of proinflammatory cytokines during the acute phase and the origin of macrophages/monocytes during the chronic phase of Chagas disease. We will also discuss the contributing factors and mechanisms leading to the chronic inflammation of the cardiac tissue during the chronic phase of the disease as well as the innate and adaptive host immune response. The contribution of genetic factors to the progression of the chronic inflammatory cardiomyopathy of chronic Chagas disease is also discussed. The secreted extracellular vesicles (exosomes) produced for both T. cruzi and infected host cells can play key roles in the host immune response, and those roles are described. Lastly, we describe potential treatments to attenuate the chronic inflammation of the cardiac tissue, designed to improve heart function in chagasic patients.
Collapse
|
133
|
Lee Y, Lee J, Hyun SH, Lee GS, Lee E. In vitro maturation using αMEM containing reduced NaCl enhances maturation and developmental competence of pig oocytes after somatic cell nuclear transfer. J Vet Sci 2022; 23:e31. [PMID: 35363440 PMCID: PMC8977537 DOI: 10.4142/jvs.21279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/19/2021] [Accepted: 01/03/2022] [Indexed: 11/21/2022] Open
Abstract
Background Compared to medium containing 108 mM sodium chloride (NaCl), in vitro maturation (IVM) using a simple medium with reduced (61.6 mM) NaCl increases the cytoplasmic maturation and embryonic development of pig oocytes. Objectives This study determines the effect of a complex medium containing reduced NaCl on the IVM and embryonic development of pig oocytes. Methods Pig oocytes were matured in Minimum Essential Medium Eagle-alpha modification (αMEM) supplemented with 61.6 (61αMEM) or 108 (108αMEM) mM NaCl, and containing polyvinyl alcohol (PVA) (αMEMP) or pig follicular fluid (PFF) (αMEMF). Medium-199 (M199) served as the control for conventional IVM. Cumulus cell expansion, nuclear maturation, intra-oocyte glutathione (GSH) contents, size of perivitelline space (PVS), and embryonic development after parthenogenesis (PA) and somatic cell nuclear transfer (SCNT) were evaluated after IVM. Results Regardless of PVA or PFF supplementation, oocytes matured in 61αMEM showed increased intra-oocyte GSH contents and width of PVS (p < 0.05), as well as increased blastocyst formation (p < 0.05) after PA and SCNT, as compared to oocytes matured in 108αMEMP and M199. Under conditions of PFF-enriched αMEM, SCNT oocytes matured in 61αMEMF showed higher blastocyst formation (p < 0.05), compared to maturation in 108αMEMF and M199, whereas PA cultured oocytes showed no significant difference. Conclusions IVM in αMEM supplemented with reduced NaCl (61.6 mM) enhances the embryonic developmental competence subsequent to PA and SCNT, which attributes toward improved oocyte maturation.
Collapse
Affiliation(s)
- Yongjin Lee
- College of Veterinary Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Joohyeong Lee
- Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Korea
| | - Sang-Hwan Hyun
- Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Korea
| | - Geun-Shik Lee
- College of Veterinary Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Eunsong Lee
- College of Veterinary Medicine, Kangwon National University, Chuncheon 24341, Korea
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), College of Veterinary Medicine, Chungbuk National University and Institute of Stem Cell & Regenerative Medicine, Chungbuk National University, Cheongju 28644, Korea
| |
Collapse
|
134
|
Kamboj S, Singh R. Chromanone-A Prerogative Therapeutic Scaffold: An Overview. ARABIAN JOURNAL FOR SCIENCE AND ENGINEERING 2022; 47:75-111. [PMID: 34226859 PMCID: PMC8244469 DOI: 10.1007/s13369-021-05858-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 06/09/2021] [Indexed: 02/06/2023]
Abstract
Chromanone or Chroman-4-one is the most important and interesting heterobicyclic compound and acts as a building block in medicinal chemistry for isolation, designing and synthesis of novel lead compounds. Structurally, absence of a double bond in chromanone between C-2 and C-3 shows a minor difference from chromone but exhibits significant variations in biological activities. In the present review, various studies published on synthesis, pharmacological evaluation on chroman-4-one analogues are addressed to signify the importance of chromanone as a versatile scaffold exhibiting a wide range of pharmacological activities. But, due to poor yield in the case of chemical synthesis and expensive isolation procedure from natural compounds, more studies are required to provide the most effective and cost-effective methods to synthesize novel chromanone analogs to give leads to chemistry community. Considering the versatility of chromanone, this review is designed to impart comprehensive, critical and authoritative information about chromanone template in drug designing and development.
Collapse
Affiliation(s)
- Sonia Kamboj
- Maharishi Markandeshwar College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Haryana 133203 India ,Ch. Devi Lal College of Pharmacy, Jagadhri, Haryana 135003 India
| | - Randhir Singh
- Maharishi Markandeshwar College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Haryana 133203 India
| |
Collapse
|
135
|
Liu C, Ma N, Guo Z, Zhang Y, Zhang J, Yang F, Su X, Zhang G, Xiong X, Xing Y. Relevance of mitochondrial oxidative stress to arrhythmias: Innovative concepts to target treatments. Pharmacol Res 2021; 175:106027. [PMID: 34890774 DOI: 10.1016/j.phrs.2021.106027] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 11/26/2021] [Accepted: 12/05/2021] [Indexed: 12/13/2022]
Abstract
Cardiac arrhythmia occurs frequently worldwide, and in severe cases can be fatal. Mitochondria are the power plants of cardiomyocytes. In recent studies, mitochondria under certain stimuli produced excessive reactive oxygen species (ROS), which affect the normal function of cardiomyocytes through ion channels and related proteins. Mitochondrial oxidative stress (MOS) plays a key role in diseases with multifactorial etiopathogenesis, such as arrhythmia; MOS can lead to arrhythmias such as atrial fibrillation and ventricular tachycardia. This review discusses the mechanisms of arrhythmias caused by MOS, particularly of ROS produced by mitochondria. MOS can cause arrhythmias by affecting the activities of Ca2+-related proteins, the mitochondrial permeability transition pore protein, connexin 43, hyperpolarization-activated cyclic nucleotide-gated potassium channel 4, and ion channels. Based on these mechanisms, we discuss possible new treatments for arrhythmia. Targeted treatments focusing on mitochondria may reduce the progression of arrhythmias, as well as the occurrence of severe arrhythmias, and may be effective for personalized disease prevention.
Collapse
Affiliation(s)
- Can Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Ning Ma
- Dezhou Second People's Hospital, Dezhou 253000, China
| | - Ziru Guo
- Xingtai People's Hospital, Xingtai 054001, China
| | - Yijun Zhang
- The First Affiliated Hospital, Hebei North University, Zhangjiakou 075000, China
| | - Jianzhen Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Fan Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xin Su
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Guoxia Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xingjiang Xiong
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Yanwei Xing
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
136
|
Zhong Y, Luo R, Liu Q, Zhu J, Lei M, Liang X, Wang X, Peng X. Jujuboside A ameliorates high fat diet and streptozotocin induced diabetic nephropathy via suppressing oxidative stress, apoptosis, and enhancing autophagy. Food Chem Toxicol 2021; 159:112697. [PMID: 34826549 DOI: 10.1016/j.fct.2021.112697] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 11/19/2021] [Accepted: 11/21/2021] [Indexed: 12/13/2022]
Abstract
Jujuboside A (JuA) is a triterpenoid saponins isolated from the seed of jujube (semen Ziziphi spinosae) with anti-oxidant, anti-inflammation and anti-apoptosis properties. The present study aimed to investigate the reno-protective effects of JuA on type II diabetes. JuA (20 mg/kg) and Metformin (Met, 300 mg/kg) were administrated to diabetic Sprague Dawley rat for 8 weeks daily. Our results showed that JuA reduced blood glucose and kidney function markers including 24 h urinary protein, urinary β-NAG/urinary creatinine, serum urea nitrogen, serum uric acid and serum creatinine, and relieved renal pathological changes. In addition, JuA decreased O2- and H2O2 level, enhanced SOD, CAT and GPx activities, decreased NOX4 expression and improved mitochondrial respiratory chain function through regulating respiratory chain complex expression. Moreover, JuA downregulated the expressions of mitochondrial apoptosis proteins: Bax, CytC, Apaf-1 and caspase 9. Apoptosis mediated by ER stress also been inhibited by JuA via downregulating p-PERK, p-IRE1, XBP1s, ATF4, p-CHOP and caspase 12 expressions. JuA also enhanced autophagy and mitophagy via regulating CaMKK2-AMPK-p-mTOR and PINK1/Parkin pathways. Collectively, these results indicated that JuA protected against type II diabetic nephropathy through inhibiting oxidative stress and apoptosis mediated by mitochondria and ER stress. In addition, autophagy and mitophagy was enhanced by JuA.
Collapse
Affiliation(s)
- Yujie Zhong
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Ruilin Luo
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Qi Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Jiachang Zhu
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Min Lei
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Xiaofei Liang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Xin Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Xiaoli Peng
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| |
Collapse
|
137
|
MicroRNA-214 in Health and Disease. Cells 2021; 10:cells10123274. [PMID: 34943783 PMCID: PMC8699121 DOI: 10.3390/cells10123274] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/16/2021] [Accepted: 11/20/2021] [Indexed: 12/24/2022] Open
Abstract
MicroRNAs (miRNAs) are endogenously expressed, non-coding RNA molecules that mediate the post-transcriptional repression and degradation of mRNAs by targeting their 3′ untranslated region (3′-UTR). Thousands of miRNAs have been identified since their first discovery in 1993, and miR-214 was first reported to promote apoptosis in HeLa cells. Presently, miR-214 is implicated in an extensive range of conditions such as cardiovascular diseases, cancers, bone formation and cell differentiation. MiR-214 has shown pleiotropic roles in contributing to the progression of diseases such as gastric and lung cancers but may also confer cardioprotection against excessive fibrosis and oxidative damage. These contrasting functions are achieved through the diverse cast of miR-214 targets. Through silencing or overexpressing miR-214, the detrimental effects can be attenuated, and the beneficial effects promoted in order to improve health outcomes. Therefore, discovering novel miR-214 targets and understanding how miR-214 is dysregulated in human diseases may eventually lead to miRNA-based therapies. MiR-214 has also shown promise as a diagnostic biomarker in identifying breast cancer and coronary artery disease. This review provides an up-to-date discussion of miR-214 literature by describing relevant roles in health and disease, areas of disagreement, and the future direction of the field.
Collapse
|
138
|
van der Vlies AJ, Xu J, Ghasemi M, Bator C, Bell A, Rosoff-Verbit B, Liu B, Gomez ED, Hasegawa U. Thioether-Based Polymeric Micelles with Fine-Tuned Oxidation Sensitivities for Chemotherapeutic Drug Delivery. Biomacromolecules 2021; 23:77-88. [PMID: 34762396 DOI: 10.1021/acs.biomac.1c01010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Oxidation-sensitive drug delivery systems (DDSs) have attracted attention due to the potential to improve efficacy and safety of chemotherapeutics. These systems are designed to release the payload in response to oxidative stress conditions, which are associated with many types of cancer. Despite extensive research on the development of oxidation-sensitive DDS, the lack of selectivity toward cancer cells over healthy cells remains a challenge. Here, we report the design and characterization of polymeric micelles containing thioether groups with varying oxidation sensitivities within the micellar core, which become hydrophilic upon thioether oxidation, leading to destabilization of the micellar structure. We first used the thioether model compounds, 3-methylthiopropylamide (TPAM), thiomorpholine amide (TMAM), and 4-(methylthio)benzylamide (TPhAM) to investigate the effect of the chemical structures of the thioethers on the oxidation by hydrogen peroxide (H2O2). TPAM shows the fastest oxidation, followed by TMAM and TPhAM, showing that the oxidation reaction of thioethers can be modulated by changing the substituent groups bound to the sulfur atom. We next prepared micelles containing these different thioether groups within the core (TP, TM, and TPh micelles). The micelles containing the thioether groups with a higher oxidation sensitivity were destabilized by H2O2 at a lower concentration. Micelle destabilization was also tested in human liver cancer (HepG2) cells and human umbilical vein endothelial cells (HUVECs). The TP micelles having the highest oxidation sensitivity were destabilized in both HepG2 cells and HUVECs, while the TPh micelles, which showed the lowest reactivity toward H2O2, were stable in these cell lines. The TM micelles possessing a moderate oxidation sensitivity were destabilized in HepG2 cells but were stable in HUVECs. Furthermore, the micelles were loaded with doxorubicin (Dox) to evaluate their potential in drug delivery applications. Among the micelles, the TM micelles loaded with Dox showed the enhanced relative toxicity in HepG2 cells over HUVECs. Therefore, our approach to fine-tune the oxidation sensitivity of the micelles has potential for improving therapeutic efficacy and safety of drugs in cancer treatment.
Collapse
Affiliation(s)
- André J van der Vlies
- Department of Materials Science and Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Jiayi Xu
- Tim Taylor Department of Chemical Engineering, Kansas State University, Manhattan, Kansas 66506, United States
| | - Masoud Ghasemi
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Carol Bator
- Huck Life Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Amanda Bell
- Tim Taylor Department of Chemical Engineering, Kansas State University, Manhattan, Kansas 66506, United States
| | - Brett Rosoff-Verbit
- Department of Materials Science and Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Bin Liu
- Tim Taylor Department of Chemical Engineering, Kansas State University, Manhattan, Kansas 66506, United States
| | - Enrique D Gomez
- Department of Materials Science and Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States.,Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States.,Materials Research Institute, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Urara Hasegawa
- Department of Materials Science and Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States.,Tim Taylor Department of Chemical Engineering, Kansas State University, Manhattan, Kansas 66506, United States
| |
Collapse
|
139
|
Garcés M, Marchini T, Cáceres L, Calabró V, Mebert AM, Tuttolomondo MV, Vico T, Vanasco V, Tesan F, Salgueiro J, Zubillaga M, Desimone MF, Valacchi G, Alvarez S, Magnani ND, Evelson PA. Oxidative metabolism in the cardiorespiratory system after an acute exposure to nickel-doped nanoparticles in mice. Toxicology 2021; 464:153020. [PMID: 34740673 DOI: 10.1016/j.tox.2021.153020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/19/2021] [Accepted: 10/29/2021] [Indexed: 12/22/2022]
Abstract
There is an increasing concern over the harmful effects that metallic nanoparticles (NP) may produce on human health. Due to their redox properties, nickel (Ni) and Ni-containing NP are particularly relevant. Hence, the aim of this study was to establish the toxicological mechanisms in the cardiorespiratory oxidative metabolism initiated by an acute exposure to Ni-doped-NP. Mice were intranasally instilled with silica NP containing Ni (II) (Ni-NP) (1 mg Ni (II)/kg body weight) or empty NP as control, and 1 h after exposure lung, plasma, and heart samples were obtained to assess the redox metabolism. Results showed that, NP were mainly retained in the lungs triggering a significantly increased tissue O2 consumption rate, leading to Ni-NP-increased reactive oxygen species production by NOX activity, and mitochondrial H2O2 production rate. In addition, an oxidant redox status due to an altered antioxidant system showed by lung GSH/GSSG ratio decreased, and SOD activity increased, resulting in an increased phospholipid oxidation. Activation of circulating polymorphonuclear leukocytes, along with GSH/GSSG ratio decreased, and phospholipid oxidation were found in the Ni-NP-group plasma samples. Consequently, in distant organs such as heart, Ni-NP inhalation alters the tissue redox status. Our results showed that the O2 metabolism analysis is a critical area of study following Ni-NP inhalation. Therefore, this work provides novel data linking the redox metabolisms alterations elicited by exposure to Ni (II) adsorbed to NP and cardiorespiratory toxicity.
Collapse
Affiliation(s)
- Mariana Garcés
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química General e Inorgánica, Argentina; Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Argentina.
| | - Timoteo Marchini
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química General e Inorgánica, Argentina; Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Argentina.
| | - Lourdes Cáceres
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química General e Inorgánica, Argentina; Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Argentina.
| | - Valeria Calabró
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química General e Inorgánica, Argentina; Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Argentina.
| | - Andrea M Mebert
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química Analítica Instrumental, Argentina; Universidad de Buenos Aires, CONICET, Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica, Argentina.
| | - María Victoria Tuttolomondo
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química Analítica Instrumental, Argentina; Universidad de Buenos Aires, CONICET, Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica, Argentina.
| | - Tamara Vico
- Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Argentina; Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Fisicoquímica, Argentina.
| | - Virginia Vanasco
- Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Argentina; Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Fisicoquímica, Argentina.
| | - Fiorella Tesan
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Fisicomatemática, Cátedra de Física, Argentina.
| | - Jimena Salgueiro
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Fisicomatemática, Cátedra de Física, Argentina.
| | - Marcela Zubillaga
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Fisicomatemática, Cátedra de Física, Argentina.
| | - Martín F Desimone
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química Analítica Instrumental, Argentina; Universidad de Buenos Aires, CONICET, Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica, Argentina.
| | - Giuseppe Valacchi
- NC State University, Plants for Human Health Institute, Animal Science Department, United States; Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy.
| | - Silvia Alvarez
- Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Argentina; Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Fisicoquímica, Argentina.
| | - Natalia D Magnani
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química General e Inorgánica, Argentina; Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Argentina.
| | - Pablo A Evelson
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química General e Inorgánica, Argentina; Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Argentina.
| |
Collapse
|
140
|
Liu Y, Yu S, Xing X, Qiao J, Yin Y, Wang J, Liu M, Zhang W. Ginsenoside Rh2 stimulates the production of mitochondrial reactive oxygen species and induces apoptosis of cervical cancer cells by inhibiting mitochondrial electron transfer chain complex. Mol Med Rep 2021; 24:873. [PMID: 34713297 PMCID: PMC8569524 DOI: 10.3892/mmr.2021.12513] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 10/07/2021] [Indexed: 11/29/2022] Open
Abstract
Ginsenoside Rh2 (G-Rh2) is a monomeric compound that extracted from ginseng and possesses anti-cancer activities both in vitro and in vivo. Previously, we reported that G-Rh2 induces apoptosis in HeLa cervical cancer cells and that the process was related to reactive oxygen species (ROS) accumulation and mitochondrial dysfunction. However, the upstream mechanisms of G-Rh2, along with its cellular targets, remain to be elucidated. In the present study, the Cell Counting Kit-8 assay, flow cytometry and Hoechst staining revealed that G-Rh2 significantly inhibited cell viability and induced apoptosis of cervical cancer cells. However, G-Rh2 was demonstrated to be non-toxic to End1/e6e7 cells. JC-1, rhodamine 123 staining, oxidative phosphorylation and glycolysis capacity assays demonstrated that G-Rh2 exposure caused an immediate decrease in mitochondrial transmembrane potential due to its inhibition of mitochondrial oxidative phosphorylation, as well as glycolysis, both of which reduced cellular ATP production. Western blotting and electron transport chain (ETC) activity assays revealed that G-Rh2 significantly inhibited the activity of ETC complexes I, III and V. Overexpression of ETC complex III partially significantly restored mitochondrial ROS and inhibited the apoptosis of cervical cancer cells induced by G-Rh2. The predicted results of binding energy in molecular docking, confirmed that G-Rh2 was highly likely to induce mitochondrial ROS production and promote cell apoptosis by targeting the ETC complex, especially for ETC complex III. Taken together, the present results revealed the potential anti-cervical cancer activity of G-Rh2 and provide direct evidence for the contribution of impaired ETC complex activity to cervical cancer cell death.
Collapse
Affiliation(s)
- Ying Liu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Shiting Yu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Xin Xing
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Juhui Qiao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Yiqiu Yin
- GeneScience Pharmaceuticals Co., Ltd., Changchun, Jilin 130012, P.R. China
| | - Jiawen Wang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Meichen Liu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Wei Zhang
- Scientific Research Department, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| |
Collapse
|
141
|
Galvan DL, Mise K, Danesh FR. Mitochondrial Regulation of Diabetic Kidney Disease. Front Med (Lausanne) 2021; 8:745279. [PMID: 34646847 PMCID: PMC8502854 DOI: 10.3389/fmed.2021.745279] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 08/30/2021] [Indexed: 12/14/2022] Open
Abstract
The role and nature of mitochondrial dysfunction in diabetic kidney disease (DKD) has been extensively studied. Yet, the molecular drivers of mitochondrial remodeling in DKD are poorly understood. Diabetic kidney cells exhibit a cascade of mitochondrial dysfunction ranging from changes in mitochondrial morphology to significant alterations in mitochondrial biogenesis, biosynthetic, bioenergetics and production of reactive oxygen species (ROS). How these changes individually or in aggregate contribute to progression of DKD remain to be fully elucidated. Nevertheless, because of the remarkable progress in our basic understanding of the role of mitochondrial biology and its dysfunction in DKD, there is great excitement on future targeted therapies based on improving mitochondrial function in DKD. This review will highlight the latest advances in understanding the nature of mitochondria dysfunction and its role in progression of DKD, and the development of mitochondrial targets that could be potentially used to prevent its progression.
Collapse
Affiliation(s)
- Daniel L Galvan
- Section of Nephrology, The University of Texas at MD Anderson Cancer Center, Houston, TX, United States
| | - Koki Mise
- Section of Nephrology, The University of Texas at MD Anderson Cancer Center, Houston, TX, United States.,Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Farhad R Danesh
- Section of Nephrology, The University of Texas at MD Anderson Cancer Center, Houston, TX, United States.,Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
142
|
Hosoi R, Fujii Y, Hiroyuki O, Shukuri M, Nishiyama S, Kanazawa M, Todoroki K, Arano Y, Sakai T, Tsukada H, Inoue O. Evaluation of intracellular processes in quinolinic acid-induced brain damage by imaging reactive oxygen species generation and mitochondrial complex I activity. EJNMMI Res 2021; 11:99. [PMID: 34628558 PMCID: PMC8502189 DOI: 10.1186/s13550-021-00841-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 09/17/2021] [Indexed: 11/10/2022] Open
Abstract
PURPOSE Our study aimed to elucidate the intracellular processes associated with quinolinic acid (QA)-induced brain injury by acquiring semiquantitative fluorescent images of reactive oxygen species (ROS) generation and positron emission tomography (PET) images of mitochondrial complex I (MC-I) activity. METHODS Ex vivo fluorescent imaging with dihydroethidium (DHE) and PET scans with 18F-BCPP-EF were conducted at 3 h and 24 h after QA injection into the rat striatum. Immunohistochemical studies were performed 24 h after QA injection into the rat brain using monoclonal antibodies against neuronal nuclei (NeuN) and CD11b. RESULTS A strong DHE-derived fluorescent signal was detected in a focal area within the QA-injected striatum 3 h after QA injection, and increased fluorescent signal spread throughout the striatum and parts of the cerebral cortex after 24 h. By contrast, 18F-BCPP-EF uptake in the QA-injected rat brain was unchanged after 3 h and markedly decreased after 24 h, not only in the striatum but also in the cerebral hemisphere. The fluorescent signal in the striatum 24 h after QA injection colocalised with microglial marker expression. CONCLUSIONS We successfully obtained functional images of focal ROS generation during the early period of excitotoxic injury, and microglial ROS generation and mitochondrial dysfunction were observed during the progression of the inflammatory response. Both ex vivo DHE imaging and in vivo 18F-BCPP-EF-PET were sufficiently sensitive to detect the respective processes of QA-induced brain damage. Our study contributes to the functional imaging of multiple events during the pathological process.
Collapse
Affiliation(s)
- Rie Hosoi
- Division of Health Sciences, Graduate School of Medicine, Osaka University, 1-7 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Yuka Fujii
- Division of Health Sciences, Graduate School of Medicine, Osaka University, 1-7 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Ohba Hiroyuki
- Central Research Laboratory, Hamamatsu Photonics K. K, 5000 Hirakuchi, Hamakita, Hamamatsu, Shizuoka, 434-8601, Japan
| | - Miho Shukuri
- Laboratory of Physical Chemistry, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo, 194-8543, Japan
| | - Shingo Nishiyama
- Central Research Laboratory, Hamamatsu Photonics K. K, 5000 Hirakuchi, Hamakita, Hamamatsu, Shizuoka, 434-8601, Japan
| | - Masakatsu Kanazawa
- Central Research Laboratory, Hamamatsu Photonics K. K, 5000 Hirakuchi, Hamakita, Hamamatsu, Shizuoka, 434-8601, Japan
| | - Kenichiro Todoroki
- Department of Analytical and Bio-Analytical Chemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka, 422-8526, Japan
| | - Yasushi Arano
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, Chiba, 260-8675, Japan
| | - Toshihiro Sakai
- Hanwa Intelligent Medical Center, Hanwa Daini Senboku Hospital, 3176 Fukaikitamachi, Naka-ku, Sakai, Osaka, 599-8271, Japan
| | - Hideo Tsukada
- Central Research Laboratory, Hamamatsu Photonics K. K, 5000 Hirakuchi, Hamakita, Hamamatsu, Shizuoka, 434-8601, Japan
| | - Osamu Inoue
- Hanwa Intelligent Medical Center, Hanwa Daini Senboku Hospital, 3176 Fukaikitamachi, Naka-ku, Sakai, Osaka, 599-8271, Japan
| |
Collapse
|
143
|
Federico M, Zavala M, Vico T, López S, Portiansky E, Alvarez S, Abrille MCV, Palomeque J. CaMKII activation in early diabetic hearts induces altered sarcoplasmic reticulum-mitochondria signaling. Sci Rep 2021; 11:20025. [PMID: 34625584 PMCID: PMC8501049 DOI: 10.1038/s41598-021-99118-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 07/20/2021] [Indexed: 01/01/2023] Open
Abstract
Prediabetic myocardium, induced by fructose-rich diet (FRD), is prone to increased sarcoplasmic reticulum (SR)-Ca2+ leak and arrhythmias due to increased activity of the Ca2+/calmodulin protein kinase II (CaMKII). However, little is known about the role of SR-mitochondria microdomains, mitochondrial structure, and mitochondrial metabolisms. To address this knowledge gap we measured SR-mitochondrial proximity, intracellular Ca2+, and mitochondrial metabolism in wild type (WT) and AC3-I transgenic mice, with myocardial-targeted CaMKII inhibition, fed with control diet (CD) or with FRD. Confocal images showed significantly increased spontaneous Ca2+ release events in FRD vs. CD WT cardiomyocytes. [3H]-Ryanodine binding assay revealed higher [3H]Ry binding in FRD than CD WT hearts. O2 consumption at State 4 and hydrogen peroxide (H2O2) production rate were increased, while respiratory control rate (RCR) and Ca2+ retention capacity (CRC) were decreased in FRD vs. CD WT isolated mitochondria. Transmission Electron Microscopy (TEM) images showed increased proximity at the SR-mitochondria microdomains, associated with increased tethering proteins, Mfn2, Grp75, and VDAC in FRD vs. CD WT. Mitochondria diameter was decrease and roundness and density were increased in FRD vs. CD WT specimens. The fission protein, Drp1 was significantly increased while the fusion protein, Opa1 was unchanged in FRD vs. CD WT hearts. These differences were prevented in AC3-I mice. We conclude that SR-mitochondria microdomains are subject to CaMKII-dependent remodeling, involving SR-Ca2+ leak and mitochondria fission, in prediabetic mice induced by FRD. We speculate that CaMKII hyperactivity induces SR-Ca2+ leak by RyR2 activation which in turn increases mitochondria Ca2+ content due to the enhanced SR-mitochondria tethering, decreasing CRC.
Collapse
Affiliation(s)
- Marilen Federico
- Centro de Investigaciones Cardiovasculares, UNLP-CONICET-CCT La Plata, Facultad de Ciencias Médicas, UNLP, 60 y 120 s/n, La Plata, CP 1900, Argentina
| | - Maite Zavala
- Centro de Investigaciones Cardiovasculares, UNLP-CONICET-CCT La Plata, Facultad de Ciencias Médicas, UNLP, 60 y 120 s/n, La Plata, CP 1900, Argentina
| | - Tamara Vico
- Instituto de Bioquímica y Medicina Molecular, UBA-CONICET, Facultad de Farmacia y Bioquímica, Buenos Aires, Argentina
| | - Sofía López
- Centro de Investigaciones Cardiovasculares, UNLP-CONICET-CCT La Plata, Facultad de Ciencias Médicas, UNLP, 60 y 120 s/n, La Plata, CP 1900, Argentina
| | - Enrique Portiansky
- Laboratorio de Análisis de Imágenes, UNLP, Facultad de Ciencias Veterinarias, La Plata, Argentina
| | - Silvia Alvarez
- Instituto de Bioquímica y Medicina Molecular, UBA-CONICET, Facultad de Farmacia y Bioquímica, Buenos Aires, Argentina
| | - Maria Celeste Villa Abrille
- Centro de Investigaciones Cardiovasculares, UNLP-CONICET-CCT La Plata, Facultad de Ciencias Médicas, UNLP, 60 y 120 s/n, La Plata, CP 1900, Argentina
| | - Julieta Palomeque
- Centro de Investigaciones Cardiovasculares, UNLP-CONICET-CCT La Plata, Facultad de Ciencias Médicas, UNLP, 60 y 120 s/n, La Plata, CP 1900, Argentina.
| |
Collapse
|
144
|
Mao W, Xiong G, Wu Y, Wang C, St. Clair D, Li JD, Xu R. RORα Suppresses Cancer-Associated Inflammation by Repressing Respiratory Complex I-Dependent ROS Generation. Int J Mol Sci 2021; 22:ijms221910665. [PMID: 34639006 PMCID: PMC8509002 DOI: 10.3390/ijms221910665] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/21/2021] [Accepted: 09/28/2021] [Indexed: 12/11/2022] Open
Abstract
Breast cancer development is associated with macrophage infiltration and differentiation in the tumor microenvironment. Our previous study highlights the crucial function of reactive oxygen species (ROS) in enhancing macrophage infiltration during the disruption of mammary tissue polarity. However, the regulation of ROS and ROS-associated macrophage infiltration in breast cancer has not been fully determined. Previous studies identified retinoid orphan nuclear receptor alpha (RORα) as a potential tumor suppressor in human breast cancer. In the present study, we showed that retinoid orphan nuclear receptor alpha (RORα) significantly decreased ROS levels and inhibited ROS-mediated cytokine expression in breast cancer cells. RORα expression in mammary epithelial cells inhibited macrophage infiltration by repressing ROS generation in the co-culture assay. Using gene co-expression and chromatin immunoprecipitation (ChIP) analyses, we identified complex I subunits NDUFS6 and NDUFA11 as RORα targets that mediated its function in suppressing superoxide generation in mitochondria. Notably, the expression of RORα in 4T1 cells significantly inhibited cancer metastasis, reduced macrophage accumulation, and enhanced M1-like macrophage differentiation in tumor tissue. In addition, reduced RORα expression in breast cancer tissue was associated with an increased incidence of cancer metastasis. These results provide additional insights into cancer-associated inflammation, and identify RORα as a potential target to suppress ROS-induced mammary tumor progression.
Collapse
Affiliation(s)
- Wei Mao
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; (W.M.); (G.X.); (Y.W.); (C.W.); (D.S.C.)
- Hunan International Scientific and Technological Cooperation Base of Animal Models for Human Disease, School of Life Sciences, Central South University, Changsha 410078, China;
| | - Gaofeng Xiong
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; (W.M.); (G.X.); (Y.W.); (C.W.); (D.S.C.)
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Yuanyuan Wu
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; (W.M.); (G.X.); (Y.W.); (C.W.); (D.S.C.)
| | - Chi Wang
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; (W.M.); (G.X.); (Y.W.); (C.W.); (D.S.C.)
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
| | - Daret St. Clair
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; (W.M.); (G.X.); (Y.W.); (C.W.); (D.S.C.)
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Jia-Da Li
- Hunan International Scientific and Technological Cooperation Base of Animal Models for Human Disease, School of Life Sciences, Central South University, Changsha 410078, China;
| | - Ren Xu
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; (W.M.); (G.X.); (Y.W.); (C.W.); (D.S.C.)
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
- Correspondence: ; Tel.: +1-859-323-7889
| |
Collapse
|
145
|
Herb M, Gluschko A, Schramm M. Reactive Oxygen Species: Not Omnipresent but Important in Many Locations. Front Cell Dev Biol 2021; 9:716406. [PMID: 34557488 PMCID: PMC8452931 DOI: 10.3389/fcell.2021.716406] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/19/2021] [Indexed: 01/08/2023] Open
Abstract
Reactive oxygen species (ROS), such as the superoxide anion or hydrogen peroxide, have been established over decades of research as, on the one hand, important and versatile molecules involved in a plethora of homeostatic processes and, on the other hand, as inducers of damage, pathologies and diseases. Which effects ROS induce, strongly depends on the cell type and the source, amount, duration and location of ROS production. Similar to cellular pH and calcium levels, which are both strictly regulated and only altered by the cell when necessary, the redox balance of the cell is also tightly regulated, not only on the level of the whole cell but in every cellular compartment. However, a still widespread view present in the scientific community is that the location of ROS production is of no major importance and that ROS randomly diffuse from their cellular source of production throughout the whole cell and hit their redox-sensitive targets when passing by. Yet, evidence is growing that cells regulate ROS production and therefore their redox balance by strictly controlling ROS source activation as well as localization, amount and duration of ROS production. Hopefully, future studies in the field of redox biology will consider these factors and analyze cellular ROS more specifically in order to revise the view of ROS as freely flowing through the cell.
Collapse
Affiliation(s)
- Marc Herb
- Institute for Medical Microbiology, Immunology and Hygiene, Cologne, Germany
| | - Alexander Gluschko
- Institute for Medical Microbiology, Immunology and Hygiene, Cologne, Germany
| | - Michael Schramm
- Institute for Medical Microbiology, Immunology and Hygiene, Cologne, Germany
| |
Collapse
|
146
|
Lindsay RT, Dieckmann S, Krzyzanska D, Manetta-Jones D, West JA, Castro C, Griffin JL, Murray AJ. β-hydroxybutyrate accumulates in the rat heart during low-flow ischaemia with implications for functional recovery. eLife 2021; 10:e71270. [PMID: 34491199 PMCID: PMC8423437 DOI: 10.7554/elife.71270] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/23/2021] [Indexed: 12/15/2022] Open
Abstract
Extrahepatic tissues which oxidise ketone bodies also have the capacity to accumulate them under particular conditions. We hypothesised that acetyl-coenzyme A (acetyl-CoA) accumulation and altered redox status during low-flow ischaemia would support ketone body production in the heart. Combining a Langendorff heart model of low-flow ischaemia/reperfusion with liquid chromatography coupled tandem mass spectrometry (LC-MS/MS), we show that β-hydroxybutyrate (β-OHB) accumulated in the ischaemic heart to 23.9 nmol/gww and was secreted into the coronary effluent. Sodium oxamate, a lactate dehydrogenase (LDH) inhibitor, increased ischaemic β-OHB levels 5.3-fold and slowed contractile recovery. Inhibition of β-hydroxy-β-methylglutaryl (HMG)-CoA synthase (HMGCS2) with hymeglusin lowered ischaemic β-OHB accumulation by 40%, despite increased flux through succinyl-CoA-3-oxaloacid CoA transferase (SCOT), resulting in greater contractile recovery. Hymeglusin also protected cardiac mitochondrial respiratory capacity during ischaemia/reperfusion. In conclusion, net ketone generation occurs in the heart under conditions of low-flow ischaemia. The process is driven by flux through both HMGCS2 and SCOT, and impacts on cardiac functional recovery from ischaemia/reperfusion.
Collapse
Affiliation(s)
- Ross T Lindsay
- Department of Physiology, Development and Neuroscience, University of CambridgeLondonUnited Kingdom
- Department of Biochemistry and Cambridge Systems Biology Centre, University of CambridgeLondonUnited Kingdom
| | - Sophie Dieckmann
- Department of Physiology, Development and Neuroscience, University of CambridgeLondonUnited Kingdom
| | - Dominika Krzyzanska
- Department of Physiology, Development and Neuroscience, University of CambridgeLondonUnited Kingdom
| | - Dominic Manetta-Jones
- Department of Physiology, Development and Neuroscience, University of CambridgeLondonUnited Kingdom
| | - James A West
- Department of Biochemistry and Cambridge Systems Biology Centre, University of CambridgeLondonUnited Kingdom
| | - Cecilia Castro
- Department of Biochemistry and Cambridge Systems Biology Centre, University of CambridgeLondonUnited Kingdom
| | - Julian L Griffin
- Department of Biochemistry and Cambridge Systems Biology Centre, University of CambridgeLondonUnited Kingdom
- Section of Biomolecular Medicine, Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College LondonLondonUnited Kingdom
| | - Andrew J Murray
- Department of Physiology, Development and Neuroscience, University of CambridgeLondonUnited Kingdom
| |
Collapse
|
147
|
Lucero García Rojas EY, Villanueva C, Bond RA. Hypoxia Inducible Factors as Central Players in the Pathogenesis and Pathophysiology of Cardiovascular Diseases. Front Cardiovasc Med 2021; 8:709509. [PMID: 34447792 PMCID: PMC8382733 DOI: 10.3389/fcvm.2021.709509] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 07/09/2021] [Indexed: 01/01/2023] Open
Abstract
Cardiovascular (CV) diseases are the major cause of death in industrialized countries. The main function of the CV system is to deliver nutrients and oxygen to all tissues. During most CV pathologies, oxygen and nutrient delivery is decreased or completely halted. Several mechanisms, including increased oxygen transport and delivery, as well as increased blood flow are triggered to compensate for the hypoxic state. If the compensatory mechanisms fail to sufficiently correct the hypoxia, irreversible damage can occur. Thus, hypoxia plays a central role in the pathogenesis and pathophysiology of CV diseases. Hypoxia inducible factors (HIFs) orchestrate the gene transcription for hundreds of proteins involved in erythropoiesis, glucose transport, angiogenesis, glycolytic metabolism, reactive oxygen species (ROS) handling, cell proliferation and survival, among others. The overall regulation of the expression of HIF-dependent genes depends on the severity, duration, and location of hypoxia. In the present review, common CV diseases were selected to illustrate that HIFs, and proteins derived directly or indirectly from their stabilization and activation, are related to the development and perpetuation of hypoxia in these pathologies. We further classify CV diseases into acute and chronic hypoxic states to better understand the temporal relevance of HIFs in the pathogenesis, disease progression and clinical outcomes of these diseases. We conclude that HIFs and their derived factors are fundamental in the genesis and progression of CV diseases. Understanding these mechanisms will lead to more effective treatment strategies leading to reduced morbidity and mortality.
Collapse
Affiliation(s)
| | - Cleva Villanueva
- Instituto Politecnico Nacional, Escuela Superior de Medicina, Mexico City, Mexico
| | - Richard A Bond
- Department of Pharmacology and Pharmaceutical Sciences, University of Houston, Houston, TX, United States
| |
Collapse
|
148
|
Tsai CY, Fang C, Wu JCC, Wu CJ, Dai KY, Chen SM. Neuroinflammation and Microglial Activation at Rostral Ventrolateral Medulla Underpin Cadmium-Induced Cardiovascular Dysregulation in Rats. J Inflamm Res 2021; 14:3863-3877. [PMID: 34408468 PMCID: PMC8364915 DOI: 10.2147/jir.s325528] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 07/28/2021] [Indexed: 12/19/2022] Open
Abstract
Purpose Cadmium is a heavy metal and environmental toxicant known to act on the central cardiovascular regulatory mechanisms, and one of its brain targets is the rostral ventrolateral medulla (RVLM), a brainstem site that maintains blood pressure and sympathetic vasomotor tone. The present study assessed the hypothesis that cadmium elicits cardiovascular dysregulation by inducing neuroinflammation and microglial activation, two potential cellular mechanisms, in RVLM. Methods Adult male Sprague-Dawley rats were used for measuring cardiovascular responses after intravenous administration of cadmium. We further conducted real-time PCR, immunofluorescence staining, in situ determination of mitochondrial superoxide, hematoxylin and eosin staining, and enzyme-linked immunosorbent assay (ELISA) to identify cytokine and chemokine mRNA expression, microglia activation, superoxide production, and necrotic and apoptotic cell death in RVLM. Results We found animals maintained under propofol anesthesia, intravenous administration of cadmium acetate (4 mg/kg) resulted in an increase, followed by a rebound and a secondary decrease in spontaneous baroreflex-mediated sympathetic vasomotor tone, a progressive reduction in mean arterial pressure and heart rate, alongside augmentation of pro-inflammatory cytokine and chemokine in RVLM. All those cardiovascular and neuroinflammatory events were reversed by pretreatment with an anti-inflammatory drug, pentoxifylline (50 mg/kg, i.p.). There were also concurrent microglial activation, reactive oxygen species production, hypoxia, reduced blood flow, and necrotic and apoptotic cell death in RVLM. Conclusion Based on these biochemical, pharmacological and morphological observations, we conclude that neuroinflammation and microglial activation at RVLM, and their downstream cellular mechanisms, causally underpin cadmium-induced cardiovascular dysregulation.
Collapse
Affiliation(s)
- Ching-Yi Tsai
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Chi Fang
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Jacqueline C C Wu
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Chiung-Ju Wu
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Kuang-Yu Dai
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Shu-Mi Chen
- Pharmacology and Toxicology, School of Medicine, Tzu Chi University, Hualien, Taiwan.,Department of Pharmacy, Lotung Poh-Ai Hospital, Yilan, Taiwan
| |
Collapse
|
149
|
The Role of Taurine in Mitochondria Health: More Than Just an Antioxidant. Molecules 2021; 26:molecules26164913. [PMID: 34443494 PMCID: PMC8400259 DOI: 10.3390/molecules26164913] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/08/2021] [Accepted: 08/10/2021] [Indexed: 12/21/2022] Open
Abstract
Taurine is a naturally occurring sulfur-containing amino acid that is found abundantly in excitatory tissues, such as the heart, brain, retina and skeletal muscles. Taurine was first isolated in the 1800s, but not much was known about this molecule until the 1990s. In 1985, taurine was first approved as the treatment among heart failure patients in Japan. Accumulating studies have shown that taurine supplementation also protects against pathologies associated with mitochondrial defects, such as aging, mitochondrial diseases, metabolic syndrome, cancer, cardiovascular diseases and neurological disorders. In this review, we will provide a general overview on the mitochondria biology and the consequence of mitochondrial defects in pathologies. Then, we will discuss the antioxidant action of taurine, particularly in relation to the maintenance of mitochondria function. We will also describe several reported studies on the current use of taurine supplementation in several mitochondria-associated pathologies in humans.
Collapse
|
150
|
The Role of Mitochondrial Dysfunction in Atrial Fibrillation: Translation to Druggable Target and Biomarker Discovery. Int J Mol Sci 2021; 22:ijms22168463. [PMID: 34445167 PMCID: PMC8395135 DOI: 10.3390/ijms22168463] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 01/02/2023] Open
Abstract
Atrial fibrillation (AF) is the most prevalent and progressive cardiac arrhythmia worldwide and is associated with serious complications such as heart failure and ischemic stroke. Current treatment modalities attenuate AF symptoms and are only moderately effective in halting the arrhythmia. Therefore, there is an urgent need to dissect molecular mechanisms that drive AF. As AF is characterized by a rapid atrial activation rate, which requires a high energy metabolism, a role of mitochondrial dysfunction in AF pathophysiology is plausible. It is well known that mitochondria play a central role in cardiomyocyte function, as they produce energy to support the mechanical and electrical function of the heart. Details on the molecular mechanisms underlying mitochondrial dysfunction are increasingly being uncovered as a contributing factor in the loss of cardiomyocyte function and AF. Considering the high prevalence of AF, investigating the role of mitochondrial impairment in AF may guide the path towards new therapeutic and diagnostic targets. In this review, the latest evidence on the role of mitochondria dysfunction in AF is presented. We highlight the key modulators of mitochondrial dysfunction that drive AF and discuss whether they represent potential targets for therapeutic interventions and diagnostics in clinical AF.
Collapse
|