1
|
Bassett J, Balasubramanian B, Clouse H, Trepakova E. High content imaging of relative ATP levels for mitochondrial toxicity prediction in human induced pluripotent stem cell derived cardiomyocytes. Toxicology 2025; 514:154088. [PMID: 39971086 DOI: 10.1016/j.tox.2025.154088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 02/05/2025] [Accepted: 02/13/2025] [Indexed: 02/21/2025]
Abstract
Human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) are increasingly being evaluated in assays aimed at better understanding potential cardiotoxic liability of newly developed therapeutic compounds. Disruption of mitochondria has been implicated in the mechanism of drug-induced cardiotoxicity of some compounds. Therefore, we have developed a high content imaging assay for the investigation of mitochondrial toxicity in hiPSC-CMs using ATP-Red, a fluorescent dye capable of detecting subcellular localization of relative ATP levels in living cells. We demonstrated time-dependent decreases in ATP-Red signal over 6 h treatment with known mitochondrial toxicants antimycin (0.03, 0.1 µM) or oligomycin (3, 10 µM). Concentration-dependent decreases in ATP-Red signal with antimycin (0.001-0.3 µM) and oligomycin (0.003-1 µM) were rescued by restoring glycolysis through glucose supplementation. Decreased ATP levels were also identified in a selection of clinically available drugs with reported association with mitochondrial toxicity but absent in compounds with no known association with mitochondrial dysfunction. ATP measurements using the ATP-Red imaging assay were consistent with orthogonal measurements of whole cell ATP levels in lysed hiPSC-CMs following compound treatment. Similar findings were also obtained with measurement of mitochondrial membrane potential, except amiodarone which had no change despite decreased ATP levels. The developed high throughput imaging assay, assessing subcellular ATP dynamics, could provide mechanistic insights for tested compounds.
Collapse
|
2
|
Yin J, Ding L, Yao S, Huang J, Xiao Y, Wang Y, Zhang B, Rehmutulla M, Gu L, Tong Q, Zhang Y. Y9, a Gboxin analog, displays anti-tumor effect in non-small cell lung cancer by inducing lysosomal dysfunction and apoptosis. Bioorg Chem 2024; 153:107820. [PMID: 39321714 DOI: 10.1016/j.bioorg.2024.107820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/27/2024]
Abstract
Non-small cell lung cancer (NSCLC) ranks among the most prevalent malignancies globally. Gboxin, a novel inhibitor of mitochondrial complex V that exerts unique anti-tumor effects via oxidative phosphorylation inhibition, but shows no efficacy against NSCLC in vivo. Through chemical structure optimization, we designed and synthesized Gboxin analog Y9, which demonstrates significantly enhanced potency over its predecessor. Specifically, Y9 inhibited NSCLC significantly more strongly than Gboxin and possessed the ability to inhibit cell cycle progression and induce oxidative stress similar to Gboxin. Further investigation revealed that unlike Gboxin, Y9 selectively acidifies lysosomes and induces lysosomal dysfunction. This leads to hyperactive autophagy with impaired substrate clearance, and ultimately resulting in apoptosis. Animal studies confirmed the efficacy of Y9 in suppressing tumor growth in a xenograft mouse model. Collectively, Y9 is a distinctive Gboxin analog that outperforms its prototype by inducing lysosomal dysfunction and apoptosis, and has the potential to be developed as a novel anti-NSCLC lead compound.
Collapse
Affiliation(s)
- Jie Yin
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Longjie Ding
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Si Yao
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jianzheng Huang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yang Xiao
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yue Wang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Biqiong Zhang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Mewlude Rehmutulla
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lianghu Gu
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Qingyi Tong
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Yonghui Zhang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
3
|
Corradi J, Thompson B, Fletcher PA, Bertram R, Sherman AS, Satin LS. K ATP channel activity and slow oscillations in pancreatic beta cells are regulated by mitochondrial ATP production. J Physiol 2023; 601:5655-5667. [PMID: 37983196 PMCID: PMC10842208 DOI: 10.1113/jp284982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 10/16/2023] [Indexed: 11/22/2023] Open
Abstract
Pancreatic beta cells secrete insulin in response to plasma glucose. The ATP-sensitive potassium channel (KATP ) links glucose metabolism to islet electrical activity in these cells by responding to increased cytosolic [ATP]/[ADP]. It was recently proposed that pyruvate kinase (PK) in close proximity to beta cell KATP locally produces the ATP that inhibits KATP activity. This proposal was largely based on the observation that applying phosphoenolpyruvate (PEP) and ADP to the cytoplasmic side of excised inside-out patches inhibited KATP . To test the relative contributions of local vs. mitochondrial ATP production, we recorded KATP activity using mouse beta cells and INS-1 832/13 cells. In contrast to prior reports, we could not replicate inhibition of KATP activity by PEP + ADP. However, when the pH of the PEP solutions was not corrected for the addition of PEP, strong channel inhibition was observed as a result of the well-known action of protons to inhibit KATP . In cell-attached recordings, perifusing either a PK activator or an inhibitor had little or no effect on KATP channel closure by glucose, further suggesting that PK is not an important regulator of KATP . In contrast, addition of mitochondrial inhibitors robustly increased KATP activity. Finally, by measuring the [ATP]/[ADP] responses to imposed calcium oscillations in mouse beta cells, we found that oxidative phosphorylation could raise [ATP]/[ADP] even when ADP was at its nadir during the burst silent phase, in agreement with our mathematical model. These results indicate that ATP produced by mitochondrial oxidative phosphorylation is the primary controller of KATP in pancreatic beta cells. KEY POINTS: Phosphoenolpyruvate (PEP) plus adenosine diphosphate does not inhibit KATP activity in excised patches. PEP solutions only inhibit KATP activity if the pH is unbalanced. Modulating pyruvate kinase has minimal effects on KATP activity. Mitochondrial inhibition, in contrast, robustly potentiates KATP activity in cell-attached patches. Although the ADP level falls during the silent phase of calcium oscillations, mitochondria can still produce enough ATP via oxidative phosphorylation to close KATP . Mitochondrial oxidative phosphorylation is therefore the main source of the ATP that inhibits the KATP activity of pancreatic beta cells.
Collapse
Affiliation(s)
- Jeremías Corradi
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Brehm Diabetes Research Center, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Benjamin Thompson
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Brehm Diabetes Research Center, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Patrick A. Fletcher
- Laboratory of Biological Modeling, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Richard Bertram
- Department of Mathematics and Programs in Neuroscience and Molecular Biophysics, Florida State University, Tallahassee, Florida, USA
| | - Arthur S. Sherman
- Laboratory of Biological Modeling, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Leslie S. Satin
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Brehm Diabetes Research Center, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
4
|
Wu S, Daston G, Rose J, Blackburn K, Fisher J, Reis A, Selman B, Naciff J. Identifying chemicals based on receptor binding/bioactivation/mechanistic explanation associated with potential to elicit hepatotoxicity and to support structure activity relationship-based read-across. Curr Res Toxicol 2023; 5:100108. [PMID: 37363741 PMCID: PMC10285556 DOI: 10.1016/j.crtox.2023.100108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 06/28/2023] Open
Abstract
The liver is the most common target organ in toxicology studies. The development of chemical structural alerts for identifying hepatotoxicity will play an important role in in silico model prediction and help strengthen the identification of analogs used in structure activity relationship (SAR)- based read-across. The aim of the current study is development of an SAR-based expert-system decision tree for screening of hepatotoxicants across a wide range of chemistry space and proposed modes of action for clustering of chemicals using defined core chemical categories based on receptor-binding or bioactivation. The decision tree is based on ∼ 1180 different chemicals that were reviewed for hepatotoxicity information. Knowledge of chemical receptor binding, metabolism and mechanistic information were used to group these chemicals into 16 different categories and 102 subcategories: four categories describe binders to 9 different receptors, 11 categories are associated with possible reactive metabolites (RMs) and there is one miscellaneous category. Each chemical subcategory has been associated with possible modes of action (MOAs) or similar key structural features. This decision tree can help to screen potential liver toxicants associated with core structural alerts of receptor binding and/or RMs and be used as a component of weight of evidence decisions based on SAR read-across, and to fill data gaps.
Collapse
|
5
|
Lee M, Ahn C, Kim K, Jeung EB. Mitochondrial Toxic Effects of Antiepileptic Drug Valproic Acid on Mouse Kidney Stem Cells. TOXICS 2023; 11:toxics11050471. [PMID: 37235285 DOI: 10.3390/toxics11050471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 05/13/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023]
Abstract
Valproic acid (VPA) is a histone deacetylase inhibitor that is used mainly as an antiepileptic and anticonvulsant drug. The side effects of VPA usually appears as hepatic injury and various metabolic disorders. On the other hand, it is rarely reported to cause kidney injury. Despite the many studies on the influence of VPA exposure on the kidneys, the specific mechanism remains unclear. This study examined the changes after VPA treatment to the mouse kidney stem cells (mKSCs). VPA triggers an increase in mitochondrial ROS, but there was no change in either mitochondrial membrane potential or the mitochondrial DNA copy number in mKSCs. The VPA treatment increased the mitochondrial complex III but decreased complex V significantly compared to the DMSO treatment as a control. The inflammatory marker (IL-6) and the expression of the apoptosis markers (Caspase 3) and were increased by VPA. In particular, the expression of the podocyte injury markers (CD2AP) was increased significantly. In conclusion, VPA exposure has adverse effects on mouse kidney stem cells.
Collapse
Affiliation(s)
- Minsu Lee
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Changhwan Ahn
- Laboratory of Veterinary Physiology, College of Veterinary Medicine, Jeju National University, Jeju 63243, Republic of Korea
- Veterinary Medical Research Institute, Jeju National University, Jeju 63243, Republic of Korea
| | - KangMin Kim
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Eui-Bae Jeung
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| |
Collapse
|
6
|
Tiwari AP, Tristan LJC, Albin B, Yang IH. Fluocinolone Acetonide Enhances Anterograde Mitochondria Trafficking and Promotes Neuroprotection against Paclitaxel-Induced Peripheral Neuropathy. ACS Chem Neurosci 2023. [PMID: 37167105 DOI: 10.1021/acschemneuro.3c00218] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023] Open
Abstract
Paclitaxel (PTX)-induced peripheral neuropathy (PIPN) is a debilitating health condition which is a result of degeneration of peripheral nerves found in extremities. Currently, there are no established treatment methods that can prevent or protect from PIPN. Fluocinolone acetonide (FA) has been recently identified as a potential candidate for protection from PIPN. However, the fundamental mechanism of action is still unknown. In this study, we showed that enhanced anterograde mitochondrial movement in dorsal root ganglion (DRG) cells has a major role in FA-mediated neuroprotection in PIPN. In this study, cells were treated with PTX or FA along with their combination followed by mitochondrial fluorescence staining. Somal (proximal) and axonal (distal) mitochondria were selectively stained using a microfluidic compartmentalized chamber with different MitoTrackers blue and red, respectively, which we termed, the two-color staining approach. Results revealed that axons were protected from degeneration by the PTX effect when treated along with FA. PTX exposure alone resulted in low mitochondrial mobility in DRG cells. However, cotreatment with PTX and FA showed significant enhancement of anterograde trafficking of somal (proximal) mitochondria to distal axons. Similarly, cotreatment with FA restored mitochondrial mobility significantly. Overall, this study affirms that increasing mitochondrial recruitment into the axon by cotreatment with FA can be a worthwhile strategy to protect or prevent PIPN. The proposed two-color staining approach can be extended to study trafficking for other neuron-specific subcellular organelles.
Collapse
Affiliation(s)
- Arjun Prasad Tiwari
- Center for Biomedical Engineering and Science, Department of Mechanical Engineering and Engineering Science, University of North Carolina at Charlotte, Charlotte, North Carolina 28223, United States
| | - Lee Ji Chao Tristan
- Department of Biomedical Engineering, National University of Singapore, Singapore 119077, Singapore
- School of Medicine, University of Western Australia, Perth, Western Australia 6009, Australia
| | - Bayne Albin
- Center for Biomedical Engineering and Science, Department of Mechanical Engineering and Engineering Science, University of North Carolina at Charlotte, Charlotte, North Carolina 28223, United States
| | - In Hong Yang
- Center for Biomedical Engineering and Science, Department of Mechanical Engineering and Engineering Science, University of North Carolina at Charlotte, Charlotte, North Carolina 28223, United States
| |
Collapse
|
7
|
Tavares-Negrete JA, Pedroza-González SC, Frías-Sánchez AI, Salas-Ramírez ML, de Santiago-Miramontes MDLÁ, Luna-Aguirre CM, Alvarez MM, Trujillo-de Santiago G. Supplementation of GelMA with Minimally Processed Tissue Promotes the Formation of Densely Packed Skeletal-Muscle-Like Tissues. ACS Biomater Sci Eng 2023. [PMID: 37126642 DOI: 10.1021/acsbiomaterials.2c01521] [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] [Indexed: 05/03/2023]
Abstract
We present a simple and cost-effective strategy for developing gelatin methacryloyl (GelMA) hydrogels supplemented with minimally processed tissue (MPT) to fabricate densely packed skeletal-muscle-like tissues. MPT powder was prepared from skeletal muscle by freeze-drying, grinding, and sieving. Cell-culture experiments showed that the incorporation of 0.5-2.0% (w/v) MPT into GelMA hydrogels enhances the proliferation of murine myoblasts (C2C12 cells) compared to proliferation in pristine GelMA hydrogels and GelMA supplemented with decellularized skeletal-muscle tissues (DCTs). MPT-supplemented constructs also preserved their three-dimensional (3D) integrity for 28 days. By contrast, analogous pristine GelMA constructs only maintained their structure for 14 days or less. C2C12 cells embedded in MPT-supplemented constructs exhibited a higher degree of cell alignment and reached a significantly higher density than cells loaded in pristine GelMA constructs. Our results suggest that the addition of MPT incorporates a rich source of biochemical and topological cues, such as growth factors, glycosaminoglycans (GAGs), and structurally preserved proteins (e.g., collagen). In addition, GelMA supplemented with MPT showed suitable rheological properties for use as bioinks for extrusion bioprinting. We envision that this simple and cost-effective strategy of hydrogel supplementation will evolve into an exciting spectrum of applications for tissue engineers, primarily in the biofabrication of relevant microtissues for in vitro models and cultured meat and ultimately for the biofabrication of transplant materials using autologous MPT.
Collapse
Affiliation(s)
- Jorge A Tavares-Negrete
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, 64849 Monterrey, México
- Departamento de Ingeniería Mecatrónica y Eléctrica, Tecnológico de Monterrey, 64849 Monterrey, México
| | - Sara Cristina Pedroza-González
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, 64849 Monterrey, México
- Departamento de Ingeniería Mecatrónica y Eléctrica, Tecnológico de Monterrey, 64849 Monterrey, México
| | - Ada I Frías-Sánchez
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, 64849 Monterrey, México
- Departamento de Ingeniería Mecatrónica y Eléctrica, Tecnológico de Monterrey, 64849 Monterrey, México
| | - Miriam L Salas-Ramírez
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, 64849 Monterrey, México
- Departamento de Ingeniería Mecatrónica y Eléctrica, Tecnológico de Monterrey, 64849 Monterrey, México
| | | | - Claudia Maribel Luna-Aguirre
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, 64849 Monterrey, México
- Departamento de Ingeniería Mecatrónica y Eléctrica, Tecnológico de Monterrey, 64849 Monterrey, México
| | - Mario M Alvarez
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, 64849 Monterrey, México
- Departamento de Ingeniería Mecatrónica y Eléctrica, Tecnológico de Monterrey, 64849 Monterrey, México
| | - Grissel Trujillo-de Santiago
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, 64849 Monterrey, México
- Departamento de Ingeniería Mecatrónica y Eléctrica, Tecnológico de Monterrey, 64849 Monterrey, México
| |
Collapse
|
8
|
Hoogstraten CA, Lyon JJ, Smeitink JAM, Russel FGM, Schirris TJJ. Time to Change: A Systems Pharmacology Approach to Disentangle Mechanisms of Drug-Induced Mitochondrial Toxicity. Pharmacol Rev 2023; 75:463-486. [PMID: 36627212 DOI: 10.1124/pharmrev.122.000568] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 09/30/2022] [Accepted: 11/21/2022] [Indexed: 01/11/2023] Open
Abstract
An increasing number of commonly prescribed drugs are known to interfere with mitochondrial function, which is associated with almost half of all Food and Drug Administration black box warnings, a variety of drug withdrawals, and attrition of drug candidates. This can mainly be attributed to a historic lack of sensitive and specific assays to identify the mechanisms underlying mitochondrial toxicity during drug development. In the last decade, a better understanding of drug-induced mitochondrial dysfunction has been achieved by network-based and structure-based systems pharmacological approaches. Here, we propose the implementation of a tiered systems pharmacology approach to detect adverse mitochondrial drug effects during preclinical drug development, which is based on a toolset developed to study inherited mitochondrial disease. This includes phenotypic characterization, profiling of key metabolic alterations, mechanistic studies, and functional in vitro and in vivo studies. Combined with binding pocket similarity comparisons and bottom-up as well as top-down metabolic network modeling, this tiered approach enables identification of mechanisms underlying drug-induced mitochondrial dysfunction. After validation of these off-target mechanisms, drug candidates can be adjusted to minimize mitochondrial activity. Implementing such a tiered systems pharmacology approach could lead to a more efficient drug development trajectory due to lower drug attrition rates and ultimately contribute to the development of safer drugs. SIGNIFICANCE STATEMENT: Many commonly prescribed drugs adversely affect mitochondrial function, which can be detected using phenotypic assays. However, these methods provide only limited insight into the underlying mechanisms. In recent years, a better understanding of drug-induced mitochondrial dysfunction has been achieved by network-based and structure-based system pharmacological approaches. Their implementation in preclinical drug development could reduce the number of drug failures, contributing to safer drug design.
Collapse
Affiliation(s)
- Charlotte A Hoogstraten
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences (C.A.H., F.G.M.R., T.J.J.S.), Radboud Center for Mitochondrial Medicine (C.A.H., J.A.M.S., F.G.M.R., T.J.J.S.), and Department of Pediatrics (J.A.M.S.), Radboud University Medical Center, Nijmegen, The Netherlands; GlaxoSmithKline, Safety Assessment, Ware, Hertfordshire, United Kingdom (J.J.L.); and Khondrion BV, Nijmegen, The Netherlands (J.A.M.S.)
| | - Jonathan J Lyon
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences (C.A.H., F.G.M.R., T.J.J.S.), Radboud Center for Mitochondrial Medicine (C.A.H., J.A.M.S., F.G.M.R., T.J.J.S.), and Department of Pediatrics (J.A.M.S.), Radboud University Medical Center, Nijmegen, The Netherlands; GlaxoSmithKline, Safety Assessment, Ware, Hertfordshire, United Kingdom (J.J.L.); and Khondrion BV, Nijmegen, The Netherlands (J.A.M.S.)
| | - Jan A M Smeitink
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences (C.A.H., F.G.M.R., T.J.J.S.), Radboud Center for Mitochondrial Medicine (C.A.H., J.A.M.S., F.G.M.R., T.J.J.S.), and Department of Pediatrics (J.A.M.S.), Radboud University Medical Center, Nijmegen, The Netherlands; GlaxoSmithKline, Safety Assessment, Ware, Hertfordshire, United Kingdom (J.J.L.); and Khondrion BV, Nijmegen, The Netherlands (J.A.M.S.)
| | - Frans G M Russel
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences (C.A.H., F.G.M.R., T.J.J.S.), Radboud Center for Mitochondrial Medicine (C.A.H., J.A.M.S., F.G.M.R., T.J.J.S.), and Department of Pediatrics (J.A.M.S.), Radboud University Medical Center, Nijmegen, The Netherlands; GlaxoSmithKline, Safety Assessment, Ware, Hertfordshire, United Kingdom (J.J.L.); and Khondrion BV, Nijmegen, The Netherlands (J.A.M.S.)
| | - Tom J J Schirris
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences (C.A.H., F.G.M.R., T.J.J.S.), Radboud Center for Mitochondrial Medicine (C.A.H., J.A.M.S., F.G.M.R., T.J.J.S.), and Department of Pediatrics (J.A.M.S.), Radboud University Medical Center, Nijmegen, The Netherlands; GlaxoSmithKline, Safety Assessment, Ware, Hertfordshire, United Kingdom (J.J.L.); and Khondrion BV, Nijmegen, The Netherlands (J.A.M.S.)
| |
Collapse
|
9
|
Salama RM, Tayel SG. Silymarin attenuates escitalopram (cipralex) induced pancreatic injury in adult male albino rats: a biochemical, histological, and immunohistochemical approach. Anat Cell Biol 2023; 56:122-136. [PMID: 36624692 PMCID: PMC9989791 DOI: 10.5115/acb.22.204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/11/2022] [Accepted: 11/25/2022] [Indexed: 01/11/2023] Open
Abstract
Depression is a prevalent global problem since ages, predominately treated with SSRI. Cipralex, is an antidepressant of the SSRIs class used as a remedy for mood, depression and anxiety. Silymarin (SIL), a natural free radical scavenging, has an antioxidant and anti-inflammatory properties. This hypothesis evaluates, for the first time, the role of cipralex on the structure of the endocrine and exocrine components of the pancreas and assess the beneficial effects of SIL on these changes. Forty-five rats were divided into control, cipralex, and cipralex plus SIL groups. During sacrifice, all rats and pancreases were weighed and the ratio of pancreatic weight (PW) to rat weight (RW) was calculated, blood samples were collected to estimate fasting glucose, insulin and amylase levels, the specimens were prepared for histological, immunohistochemical (inducible nitric oxide synthase [iNOS], tumour necrosis factor-alpha [TNF-α], caspase 3, proliferating cell nuclear antigen [PCNA], and anti-insulin antibody), and morphometrical studies. Cipralex group exhibited marked destruction of the pancreatic architecture of the exocrine and endocrine parts, with a dense collagen fiber deposition. Also, there is highly significant decrease (P<0.001) of PW/RT ratio, insulin, and amylase levels, the number and diameter of islets of Langerhans, the number of PCNA positive immunoreactive cells, and the number of insulin positive β-cells. Furthermore, a highly significant increase of glucose level, iNOS, TNF-α, and caspase-3 positive immunoreactive cells in the islets of Langerhans and acinar cells were observed. SIL improves the pancreatic histological architecture, weight loss, biochemical, and immunohistochemical analyses. Administering SIL is advantageous in managing cipralex induced pancreatic injury via its anti-inflammatory, antioxidant, and anti-apoptotic qualities.
Collapse
Affiliation(s)
- Rasha Mamdouh Salama
- Department of Anatomy and Embryology, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| | - Sara Gamal Tayel
- Department of Anatomy and Embryology, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| |
Collapse
|
10
|
The potential role of environmental factors in modulating mitochondrial DNA epigenetic marks. VITAMINS AND HORMONES 2023; 122:107-145. [PMID: 36863791 DOI: 10.1016/bs.vh.2023.01.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Many studies implicate mitochondrial dysfunction in the development and progression of numerous chronic diseases. Mitochondria are responsible for most cellular energy production, and unlike other cytoplasmic organelles, mitochondria contain their own genome. Most research to date, through investigating mitochondrial DNA copy number, has focused on larger structural changes or alterations to the entire mitochondrial genome and their role in human disease. Using these methods, mitochondrial dysfunction has been linked to cancers, cardiovascular disease, and metabolic health. However, like the nuclear genome, the mitochondrial genome may experience epigenetic alterations, including DNA methylation that may partially explain some of the health effects of various exposures. Recently, there has been a movement to understand human health and disease within the context of the exposome, which aims to describe and quantify the entirety of all exposures people encounter throughout their lives. These include, among others, environmental pollutants, occupational exposures, heavy metals, and lifestyle and behavioral factors. In this chapter, we summarize the current research on mitochondria and human health, provide an overview of the current knowledge on mitochondrial epigenetics, and describe the experimental and epidemiologic studies that have investigated particular exposures and their relationships with mitochondrial epigenetic modifications. We conclude the chapter with suggestions for future directions in epidemiologic and experimental research that is needed to advance the growing field of mitochondrial epigenetics.
Collapse
|
11
|
Kuretu A, Arineitwe C, Mothibe M, Ngubane P, Khathi A, Sibiya N. Drug-induced mitochondrial toxicity: Risks of developing glucose handling impairments. Front Endocrinol (Lausanne) 2023; 14:1123928. [PMID: 36860368 PMCID: PMC9969099 DOI: 10.3389/fendo.2023.1123928] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/31/2023] [Indexed: 02/15/2023] Open
Abstract
Mitochondrial impairment has been associated with the development of insulin resistance, the hallmark of type 2 diabetes mellitus (T2DM). However, the relationship between mitochondrial impairment and insulin resistance is not fully elucidated due to insufficient evidence to support the hypothesis. Insulin resistance and insulin deficiency are both characterised by excessive production of reactive oxygen species and mitochondrial coupling. Compelling evidence states that improving the function of the mitochondria may provide a positive therapeutic tool for improving insulin sensitivity. There has been a rapid increase in reports of the toxic effects of drugs and pollutants on the mitochondria in recent decades, interestingly correlating with an increase in insulin resistance prevalence. A variety of drug classes have been reported to potentially induce toxicity in the mitochondria leading to skeletal muscle, liver, central nervous system, and kidney injury. With the increase in diabetes prevalence and mitochondrial toxicity, it is therefore imperative to understand how mitochondrial toxicological agents can potentially compromise insulin sensitivity. This review article aims to explore and summarise the correlation between potential mitochondrial dysfunction caused by selected pharmacological agents and its effect on insulin signalling and glucose handling. Additionally, this review highlights the necessity for further studies aimed to understand drug-induced mitochondrial toxicity and the development of insulin resistance.
Collapse
Affiliation(s)
- Auxiliare Kuretu
- Pharmacology Division, Faculty of Pharmacy, Rhodes University, Makhanda, South Africa
| | - Charles Arineitwe
- Pharmacology Division, Faculty of Pharmacy, Rhodes University, Makhanda, South Africa
| | - Mamosheledi Mothibe
- Pharmacology Division, Faculty of Pharmacy, Rhodes University, Makhanda, South Africa
| | - Phikelelani Ngubane
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Andile Khathi
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Ntethelelo Sibiya
- Pharmacology Division, Faculty of Pharmacy, Rhodes University, Makhanda, South Africa
- *Correspondence: Ntethelelo Sibiya,
| |
Collapse
|
12
|
Kinetic Mathematical Modeling of Oxidative Phosphorylation in Cardiomyocyte Mitochondria. Cells 2022; 11:cells11244020. [PMID: 36552784 PMCID: PMC9777548 DOI: 10.3390/cells11244020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022] Open
Abstract
Oxidative phosphorylation (OXPHOS) is an oxygen-dependent process that consumes catabolized nutrients to produce adenosine triphosphate (ATP) to drive energy-dependent biological processes such as excitation-contraction coupling in cardiomyocytes. In addition to in vivo and in vitro experiments, in silico models are valuable for investigating the underlying mechanisms of OXPHOS and predicting its consequences in both physiological and pathological conditions. Here, we compare several prominent kinetic models of OXPHOS in cardiomyocytes. We examine how their mathematical expressions were derived, how their parameters were obtained, the conditions of their experimental counterparts, and the predictions they generated. We aim to explore the general landscape of energy production mechanisms in cardiomyocytes for future in silico models.
Collapse
|
13
|
Bețiu AM, Noveanu L, Hâncu IM, Lascu A, Petrescu L, Maack C, Elmér E, Muntean DM. Mitochondrial Effects of Common Cardiovascular Medications: The Good, the Bad and the Mixed. Int J Mol Sci 2022; 23:13653. [PMID: 36362438 PMCID: PMC9656474 DOI: 10.3390/ijms232113653] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 10/20/2022] [Accepted: 10/28/2022] [Indexed: 07/25/2023] Open
Abstract
Mitochondria are central organelles in the homeostasis of the cardiovascular system via the integration of several physiological processes, such as ATP generation via oxidative phosphorylation, synthesis/exchange of metabolites, calcium sequestration, reactive oxygen species (ROS) production/buffering and control of cellular survival/death. Mitochondrial impairment has been widely recognized as a central pathomechanism of almost all cardiovascular diseases, rendering these organelles important therapeutic targets. Mitochondrial dysfunction has been reported to occur in the setting of drug-induced toxicity in several tissues and organs, including the heart. Members of the drug classes currently used in the therapeutics of cardiovascular pathologies have been reported to both support and undermine mitochondrial function. For the latter case, mitochondrial toxicity is the consequence of drug interference (direct or off-target effects) with mitochondrial respiration/energy conversion, DNA replication, ROS production and detoxification, cell death signaling and mitochondrial dynamics. The present narrative review aims to summarize the beneficial and deleterious mitochondrial effects of common cardiovascular medications as described in various experimental models and identify those for which evidence for both types of effects is available in the literature.
Collapse
Affiliation(s)
- Alina M. Bețiu
- Doctoral School Medicine-Pharmacy, “Victor Babeș” University of Medicine and Pharmacy from Timișoara, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
- Center for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy from Timișoara, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
| | - Lavinia Noveanu
- Department of Functional Sciences—Pathophysiology, “Victor Babeș” University of Medicine and Pharmacy from Timișoara, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
| | - Iasmina M. Hâncu
- Doctoral School Medicine-Pharmacy, “Victor Babeș” University of Medicine and Pharmacy from Timișoara, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
- Center for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy from Timișoara, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
| | - Ana Lascu
- Center for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy from Timișoara, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
- Department of Functional Sciences—Pathophysiology, “Victor Babeș” University of Medicine and Pharmacy from Timișoara, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
| | - Lucian Petrescu
- Doctoral School Medicine-Pharmacy, “Victor Babeș” University of Medicine and Pharmacy from Timișoara, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
- Center for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy from Timișoara, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
| | - Christoph Maack
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, 97078 Würzburg, Germany
- Department of Internal Medicine 1, University Clinic Würzburg, 97078 Würzburg, Germany
| | - Eskil Elmér
- Mitochondrial Medicine, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, BMC A13, 221 84 Lund, Sweden
- Abliva AB, Medicon Village, 223 81 Lund, Sweden
| | - Danina M. Muntean
- Center for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy from Timișoara, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
- Department of Functional Sciences—Pathophysiology, “Victor Babeș” University of Medicine and Pharmacy from Timișoara, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
| |
Collapse
|
14
|
Seal S, Carreras-Puigvert J, Trapotsi MA, Yang H, Spjuth O, Bender A. Integrating cell morphology with gene expression and chemical structure to aid mitochondrial toxicity detection. Commun Biol 2022; 5:858. [PMID: 35999457 PMCID: PMC9399120 DOI: 10.1038/s42003-022-03763-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 07/25/2022] [Indexed: 12/05/2022] Open
Abstract
Mitochondrial toxicity is an important safety endpoint in drug discovery. Models based solely on chemical structure for predicting mitochondrial toxicity are currently limited in accuracy and applicability domain to the chemical space of the training compounds. In this work, we aimed to utilize both -omics and chemical data to push beyond the state-of-the-art. We combined Cell Painting and Gene Expression data with chemical structural information from Morgan fingerprints for 382 chemical perturbants tested in the Tox21 mitochondrial membrane depolarization assay. We observed that mitochondrial toxicants differ from non-toxic compounds in morphological space and identified compound clusters having similar mechanisms of mitochondrial toxicity, thereby indicating that morphological space provides biological insights related to mechanisms of action of this endpoint. We further showed that models combining Cell Painting, Gene Expression features and Morgan fingerprints improved model performance on an external test set of 244 compounds by 60% (in terms of F1 score) and improved extrapolation to new chemical space. The performance of our combined models was comparable with dedicated in vitro assays for mitochondrial toxicity. Our results suggest that combining chemical descriptors with biological readouts enhances the detection of mitochondrial toxicants, with practical implications in drug discovery.
Collapse
Affiliation(s)
- Srijit Seal
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Rd, Cambridge, CB2 1EW, UK
| | - Jordi Carreras-Puigvert
- Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Box 591, SE-75124, Uppsala, Sweden
| | - Maria-Anna Trapotsi
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Rd, Cambridge, CB2 1EW, UK
| | - Hongbin Yang
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Rd, Cambridge, CB2 1EW, UK
| | - Ola Spjuth
- Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Box 591, SE-75124, Uppsala, Sweden.
| | - Andreas Bender
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Rd, Cambridge, CB2 1EW, UK.
| |
Collapse
|
15
|
Roth RA, Kana O, Filipovic D, Ganey PE. Pharmacokinetic and toxicodynamic concepts in idiosyncratic, drug-induced liver injury. Expert Opin Drug Metab Toxicol 2022; 18:469-481. [PMID: 36003040 PMCID: PMC9484408 DOI: 10.1080/17425255.2022.2113379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 08/11/2022] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Idiosyncratic drug-induced liver injury (IDILI) causes morbidity and mortality in patients and leads to curtailed use of efficacious pharmaceuticals. Unlike intrinsically toxic reactions, which depend on dose, IDILI occurs in a minority of patients at therapeutic doses. Much remains unknown about causal links among drug exposure, a mode of action, and liver injury. Consequently, numerous hypotheses about IDILI pathogenesis have arisen. AREAS COVERED Pharmacokinetic and toxicodynamic characteristics underlying current hypotheses of IDILI etiology are discussed and illustrated graphically. EXPERT OPINION Hypotheses to explain IDILI etiology all involve alterations in pharmacokinetics, which lead to plasma drug concentrations that rise above a threshold for toxicity, or in toxicodynamics, which result in a lowering of the toxicity threshold. Altered pharmacokinetics arise, for example, from changes in drug metabolism or from transporter polymorphisms. A lowered toxicity threshold can arise from drug-induced mitochondrial injury, accumulation of toxic endogenous factors or harmful immune responses. Newly developed, interactive freeware (DemoTox-PK; https://bit.ly/DemoTox-PK) allows the user to visualize how such alterations might lead to a toxic reaction. The illustrations presented provide a framework for conceptualizing idiosyncratic reactions and could serve as a stimulus for future discussion, education, and research into modes of action of IDILI.
Collapse
Affiliation(s)
- Robert A. Roth
- Department of Pharmacology and Toxicology and Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 49924
- ProbiTox LLC, Chapel Hill, NC 27514
| | - Omar Kana
- Department of Pharmacology and Toxicology and Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 49924
- Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI 48824
| | - David Filipovic
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824
- Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI 48824
- Department of Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, MI 48824
| | - Patricia E. Ganey
- Department of Pharmacology and Toxicology and Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 49924
- ProbiTox LLC, Chapel Hill, NC 27514
| |
Collapse
|
16
|
Yousefsani BS, Salimi A, Imani F, Ramezani M, Shirani K, Seydi E, Pourahmad J. Risperidone Toxicity on Human Blood Lymphocytes in Nano molar Concentrations. Drug Res (Stuttg) 2022; 72:343-349. [PMID: 35605969 DOI: 10.1055/a-1830-8701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2022]
Abstract
Risperidone is an atypical antipsychotic drug used for the pharmacotherapy of psychiatric disorders. Some reports indicate that risperidone is toxic to various systems of the body, including the immune system. This study evaluated the toxicity effect of risperidone on human blood lymphocytes. To achieve this aim, lymphocytes were isolated using Ficoll paque plus. The results showed that risperidone (12, 24 and 48 nM) causes toxicity in human blood lymphocytes by increasing the level of intracellular reactive oxygen species (ROS), damage to lysosomal membrane, the collapse of the mitochondrial membrane potential (MMP), and increased extracellular oxidized glutathione (GSSG). Also, exposure of human blood lymphocytes to risperidone is associated with a decrease in intracellular glutathione (GSH) levels. Finally, it could be concluded that oxidative stress is one of the mechanisms of risperidone-induced toxicity in human blood lymphocytes.
Collapse
Affiliation(s)
- Bahareh Sadat Yousefsani
- Research Institute for Islamic and Complementary Medicine, Iran University of Medical Sciences, Tehran, Iran.,School of Persian Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ahmad Salimi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Farnaz Imani
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maral Ramezani
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kobra Shirani
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Enayatollah Seydi
- Department of Occupational Health and Safety Engineering, School of Health, Alborz University of Medical Sciences, Karaj, Iran.,Research Center for Health, Safety and Environment, Alborz University of Medical Sciences, Karaj, Iran
| | - Jalal Pourahmad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Ahn D, Kim CW, Go RE, Choi KC. Evaluation of mitochondrial oxidative toxicity in mammalian cardiomyocytes by determining the highly reproducible and reliable increase in mitochondrial superoxides after exposure to therapeutic drugs. Toxicol In Vitro 2022; 83:105393. [PMID: 35618243 DOI: 10.1016/j.tiv.2022.105393] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 01/11/2023]
Abstract
Mitochondria are important cytoplasmic elements present in eukaryotic cells, and are involved in converting energy to ATP through oxidative phosphorylation. Mitochondria are vulnerable to reactive oxygen species (ROS), thereby making it imperative to evaluate the toxicity. However, existing methods that evaluate mitochondrial toxicity in cardiomyocytes are limited. In the current study, we aimed to determine a mitochondrial biomarker that measures the toxicity of mitochondria, and subsequently suggest an efficient evaluation system for evaluating mitochondrial-specific oxidative toxicity. To achieve this, AC16 human cardiomyocytes, H9C2 rat cardiomyocytes were exposed to acetaminophen (AP), amiodarone hydrochloride (AMD), doxorubicin hydrochloride (Dox), valproic acid sodium salt (Val), and (Z)-4-hydroxytamoxifen (4-OHT). Mitochondrial oxidative stress was determined by staining the drug-treated cells with MitoSOX™ red fluorescence dye, followed by imaging with a fluorescence microscope. All working concentrations of Dox showed increased levels of red fluorescence in AC16 and H9C2 cells, whereas exposure to Val did not alter the red fluorescence level of both cells. Considering our results, increased MitoSOX™ subsequent to drug exposure is a highly reproducible and reliable method to measure the mitochondrial-specific oxidative toxicity. These results indicate that a screening system using MitoSOX™ has the potential to be applied as a reliable biomarker for determining mitochondrial oxidative toxicity in new drug development.
Collapse
Affiliation(s)
- Dohee Ahn
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Cho-Won Kim
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Ryeo-Eun Go
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea.
| |
Collapse
|
18
|
Chiejina CO, Anih L, Okoye C, Aguzie IO, Ali D, Kumar G, Nwani CD. Haloperidol alters the behavioral, hematological and biochemical parameters of freshwater African catfish, Clarias gariepinus (Burchell 1822). Comp Biochem Physiol C Toxicol Pharmacol 2022; 254:109292. [PMID: 35114394 DOI: 10.1016/j.cbpc.2022.109292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 01/20/2022] [Accepted: 01/26/2022] [Indexed: 11/24/2022]
Abstract
The presence of drugs and their metabolites in surface waters and municipal effluents has been reported in several studies, but their impacts on aquatic organisms are not yet well studied. The present study investigated the effects of exposure to the antipsychotic drug, haloperidol on the behavioral, hematological and biochemical parameters in juvenile Clarias gariepinus. The fishes were exposed to 0.12, 0.24 and 0.48 mg/L haloperidol for 15 days and later withdrawn from the toxicant and allowed to recover for 5 days. Blood was sampled on days 1, 5, 10, 15, and after the 5-day recovery for hematological and biochemical analysis. The pack cell volume (PCV), red blood cells (RBC), hemoglobin (Hb), reticulocytes and lymphocyte counts were significantly reduced in the exposed fish. The neutrophil counts were increased while that of monocytes, basophils and eosinophils were not affected by the drug. The mean corpuscular volume (MCV), mean corpuscular hemoglobin (MCH) and mean corpuscular hemoglobin concentration (MCHC) were not different from the control on exposure to the drug. The activities of aspartate aminotransferase (AST), alanine aminotransferase (ALT), alkaline phosphatase (ALP), and acid phosphatase (ACP); and serum creatinine, bile acid and bilirubin were increased on 15-day exposure to the drug. The activity of the clotting factor fibrinogen was reduced compared to the control after exposure to the drug. Haloperidol at concentrations used on 15-day exposure were toxic to fish, but the effect appeared short-lived, as it dissipated on 5-day withdrawal from the drug. While further studies are needed to ascertain the impact of prolonged exposure to environmentally relevant concentrations, caution is advised to avoid eco-toxicological damage to aquatic organisms.
Collapse
Affiliation(s)
- Chike Obinna Chiejina
- Department of Zoology and Environmental Biology, University of Nigeria Nsukka, Enugu, Nigeria
| | - Lucy Anih
- Department of Zoology and Environmental Biology, University of Nigeria Nsukka, Enugu, Nigeria
| | - Charles Okoye
- Department of Zoology and Environmental Biology, University of Nigeria Nsukka, Enugu, Nigeria
| | - Ifeanyi Oscar Aguzie
- Department of Zoology and Environmental Biology, University of Nigeria Nsukka, Enugu, Nigeria
| | - Daoud Ali
- Department of Zoology, College of Science, King Saud University, PO Box 2455, Riyadh 11451, Saudi Arabia
| | - Gokhlesh Kumar
- Clinical Division of Fish Medicine, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | | |
Collapse
|
19
|
Abstract
Combination antiretroviral therapy (cART) dramatically changed the face of the HIV/AIDS pandemic, making it one of the most prominent medical breakthroughs of the past 3 decades. However, as the life span of persons living with HIV (PLWH) continues to approach that of the general population, the same cannot be said regarding their quality of life. PLWH are affected by comorbid conditions such as high blood pressure, diabetes, and neurocognitive impairment at a higher rate and increased severity than their age-matched counterparts. PLWH also have higher levels of inflammation, the drivers of which are not entirely clear. As cART treatment is lifelong, we assessed here the effects of cART, independent of HIV, on primary human monocyte-derived macrophages (MDMs). MDMs were unskewed or skewed to an alternative phenotype and treated with Atripla or Triumeq, two first-line cART treatments. We report that Triumeq skewed alternative MDMs toward an inflammatory nonsenescent phenotype. Both Atripla and Triumeq caused mitochondrial dysfunction, specifically efavirenz and abacavir. Additionally, transcriptome sequencing (RNA-seq) demonstrated that both Atripla and Triumeq caused differential regulation of genes involved in immune regulation and cell cycle and DNA repair. Collectively, our data demonstrate that cART, independent of HIV, alters the MDM phenotype. This suggests that cART may contribute to cell dysregulation in PLWH that subsequently results in increased susceptibility to comorbidities.
Collapse
|
20
|
Rodrigues D, Coyle L, Füzi B, Ferreira S, Jo H, Herpers B, Chung SW, Fisher C, Kleinjans JCS, Jennen D, de Kok TM. Unravelling Mechanisms of Doxorubicin-Induced Toxicity in 3D Human Intestinal Organoids. Int J Mol Sci 2022; 23:ijms23031286. [PMID: 35163210 PMCID: PMC8836276 DOI: 10.3390/ijms23031286] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 12/12/2022] Open
Abstract
Doxorubicin is widely used in the treatment of different cancers, and its side effects can be severe in many tissues, including the intestines. Symptoms such as diarrhoea and abdominal pain caused by intestinal inflammation lead to the interruption of chemotherapy. Nevertheless, the molecular mechanisms associated with doxorubicin intestinal toxicity have been poorly explored. This study aims to investigate such mechanisms by exposing 3D small intestine and colon organoids to doxorubicin and to evaluate transcriptomic responses in relation to viability and apoptosis as physiological endpoints. The in vitro concentrations and dosing regimens of doxorubicin were selected based on physiologically based pharmacokinetic model simulations of treatment regimens recommended for cancer patients. Cytotoxicity and cell morphology were evaluated as well as gene expression and biological pathways affected by doxorubicin. In both types of organoids, cell cycle, the p53 signalling pathway, and oxidative stress were the most affected pathways. However, significant differences between colon and SI organoids were evident, particularly in essential metabolic pathways. Short time-series expression miner was used to further explore temporal changes in gene profiles, which identified distinct tissue responses. Finally, in silico proteomics revealed important proteins involved in doxorubicin metabolism and cellular processes that were in line with the transcriptomic responses, including cell cycle and senescence, transport of molecules, and mitochondria impairment. This study provides new insight into doxorubicin-induced effects on the gene expression levels in the intestines. Currently, we are exploring the potential use of these data in establishing quantitative systems toxicology models for the prediction of drug-induced gastrointestinal toxicity.
Collapse
Affiliation(s)
- Daniela Rodrigues
- Department of Toxicogenomics, GROW School for Oncology and Developmental Biology, Maastricht University, 6229 ER Maastricht, The Netherlands; (J.C.S.K.); (D.J.); (T.M.d.K.)
- Correspondence:
| | - Luke Coyle
- Boehringer Ingelheim International GmbH, Pharmaceuticals Inc., Ridgefield, CT 06877, USA; (L.C.); (S.-W.C.)
| | - Barbara Füzi
- Department of Pharmaceutical Sciences, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria;
| | - Sofia Ferreira
- Certara UK Limited, Simcyp Division, Sheffield S1 2BJ, UK; (S.F.); (H.J.); (C.F.)
| | - Heeseung Jo
- Certara UK Limited, Simcyp Division, Sheffield S1 2BJ, UK; (S.F.); (H.J.); (C.F.)
| | - Bram Herpers
- Crown Bioscience Netherlands B.V., J.H. Oortweg 21, 2333 CH Leiden, The Netherlands;
| | - Seung-Wook Chung
- Boehringer Ingelheim International GmbH, Pharmaceuticals Inc., Ridgefield, CT 06877, USA; (L.C.); (S.-W.C.)
| | - Ciarán Fisher
- Certara UK Limited, Simcyp Division, Sheffield S1 2BJ, UK; (S.F.); (H.J.); (C.F.)
| | - Jos C. S. Kleinjans
- Department of Toxicogenomics, GROW School for Oncology and Developmental Biology, Maastricht University, 6229 ER Maastricht, The Netherlands; (J.C.S.K.); (D.J.); (T.M.d.K.)
| | - Danyel Jennen
- Department of Toxicogenomics, GROW School for Oncology and Developmental Biology, Maastricht University, 6229 ER Maastricht, The Netherlands; (J.C.S.K.); (D.J.); (T.M.d.K.)
| | - Theo M. de Kok
- Department of Toxicogenomics, GROW School for Oncology and Developmental Biology, Maastricht University, 6229 ER Maastricht, The Netherlands; (J.C.S.K.); (D.J.); (T.M.d.K.)
| |
Collapse
|
21
|
Mitochondria-affecting small molecules ameliorate proteostasis defects associated with neurodegenerative diseases. Sci Rep 2021; 11:17733. [PMID: 34489512 PMCID: PMC8421394 DOI: 10.1038/s41598-021-97148-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 08/12/2021] [Indexed: 02/06/2023] Open
Abstract
Macroautophagic recycling of dysfunctional mitochondria, known as mitophagy, is essential for mitochondrial homeostasis and cell viability. Accumulation of defective mitochondria and impaired mitophagy have been widely implicated in many neurodegenerative diseases, and loss-of-function mutations of PINK1 and Parkin, two key regulators of mitophagy, are amongst the most common causes of heritable parkinsonism. This has led to the hypothesis that pharmacological stimulation of mitophagy may be a feasible approach to combat neurodegeneration. Toward this end, we screened ~ 45,000 small molecules using a high-throughput, whole-organism, phenotypic screen that monitored accumulation of PINK-1 protein, a key event in mitophagic activation, in a Caenorhabditis elegans strain carrying a Ppink-1::PINK-1::GFP reporter. We obtained eight hits that increased mitochondrial fragmentation and autophagosome formation. Several of the compounds also reduced ATP production, oxygen consumption, mitochondrial mass, and/or mitochondrial membrane potential. Importantly, we found that treatment with two compounds, which we named PS83 and PS106 (more commonly known as sertraline) reduced neurodegenerative disease phenotypes, including delaying paralysis in a C. elegans β-amyloid aggregation model in a PINK-1-dependent manner. This report presents a promising step toward the identification of compounds that will stimulate mitochondrial turnover.
Collapse
|
22
|
Lin YT, Lin KH, Huang CJ, Wei AC. MitoTox: a comprehensive mitochondrial toxicity database. BMC Bioinformatics 2021; 22:369. [PMID: 34266386 PMCID: PMC8283953 DOI: 10.1186/s12859-021-04285-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 07/06/2021] [Indexed: 11/22/2022] Open
Abstract
Background Mitochondria play essential roles in regulating cellular functions. Some drug treatments and molecular interventions have been reported to have off-target effects damaging mitochondria and causing severe side effects. The development of a database for the management of mitochondrial toxicity-related molecules and their targets is important for further analyses. Results To correlate chemical, biological and mechanistic information on clinically relevant mitochondria-related toxicity, a comprehensive mitochondrial toxicity database (MitoTox) was developed. MitoTox is an electronic repository that integrates comprehensive information about mitochondria-related toxins and their targets. Information and data related to mitochondrial toxicity originate from various sources, including scientific journals and other electronic databases. These resources were manually verified and extracted into MitoTox. The database currently contains over 1400 small-molecule compounds, 870 mitochondrial targets, and more than 4100 mitochondrial toxin-target associations. Each MitoTox data record contains over 30 fields, including biochemical properties, therapeutic classification, target proteins, toxicological data, mechanistic information, clinical side effects, and references. Conclusions MitoTox provides a fully searchable database with links to references and other databases. Potential applications of MitoTox include toxicity classification, prediction, reference and education. MitoTox is available online at http://www.mitotox.org.
Collapse
Affiliation(s)
- Yu-Te Lin
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - Ko-Hong Lin
- Department of Electrical Engineering, National Taiwan University, Taipei, Taiwan
| | - Chi-Jung Huang
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - An-Chi Wei
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan. .,Department of Electrical Engineering, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
23
|
Mani S, Swargiary G, Tyagi S, Singh M, Jha NK, Singh KK. Nanotherapeutic approaches to target mitochondria in cancer. Life Sci 2021; 281:119773. [PMID: 34192595 DOI: 10.1016/j.lfs.2021.119773] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/18/2021] [Accepted: 06/22/2021] [Indexed: 01/18/2023]
Abstract
Treatment of cancer cells exemplifies a difficult test in the light of challenges associated with the nature of cancer cells and the severe side effects too. After making a large number of trials using both traditional and advanced therapies (immunotherapy and hormone therapy), approaches to design new therapies have reached a saturation level. However, nanotechnology-based approaches exhibit higher efficacy and great potential to bypass many of such therapeutic limitations. Because of their higher target specificity, the use of nanoparticles offers incredible potential in cancer therapeutics. Mitochondria, acting as a factory of energy production in cells, reveal an important role in the death as well as the survival of cells. Because of its significant involvement in the proliferation of cancer cells, it is being regarded as an important target for cancer therapeutics. Numerous studies reveal that nanotechnology-based approaches to directly target the mitochondria may help in improving the survival rate of cancer patients. In the current study, we have detailed the significance of mitochondria in the development of cancer phenotype, as well as indicated it as the potential targets for cancer therapy. Our study further highlights the importance of different nanoparticle-based approaches to target mitochondria of cancer cells and the associated outcomes of different studies. Though, nanotechnology-based approaches to target mitochondria of cancer cells demonstrate a potential and efficient way in cancer therapeutics. Yet, further study is needed to overcome the linked limitations.
Collapse
Affiliation(s)
- Shalini Mani
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector 62, Noida, UP 201301, India.
| | - Geeta Swargiary
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector 62, Noida, UP 201301, India
| | - Sakshi Tyagi
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector 62, Noida, UP 201301, India
| | - Manisha Singh
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector 62, Noida, UP 201301, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida, Uttar Pradesh 201310, India
| | - Keshav K Singh
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
24
|
Hinkovska-Galcheva V, Treadwell T, Shillingford JM, Lee A, Abe A, Tesmer JJG, Shayman JA. Inhibition of lysosomal phospholipase A2 predicts drug-induced phospholipidosis. J Lipid Res 2021; 62:100089. [PMID: 34087196 PMCID: PMC8243516 DOI: 10.1016/j.jlr.2021.100089] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 11/17/2022] Open
Abstract
Phospholipidosis, the excessive accumulation of phospholipids within lysosomes, is a pathological response observed following exposure to many drugs across multiple therapeutic groups. A clear mechanistic understanding of the causes and implications of this form of drug toxicity has remained elusive. We previously reported the discovery and characterization of a lysosome-specific phospholipase A2 (PLA2G15) and later reported that amiodarone, a known cause of drug-induced phospholipidosis, inhibits this enzyme. Here, we assayed a library of 163 drugs for inhibition of PLA2G15 to determine whether this phospholipase was the cellular target for therapeutics other than amiodarone that cause phospholipidosis. We observed that 144 compounds inhibited PLA2G15 activity. Thirty-six compounds not previously reported to cause phospholipidosis inhibited PLA2G15 with IC50 values less than 1 mM and were confirmed to cause phospholipidosis in an in vitro assay. Within this group, fosinopril was the most potent inhibitor (IC50 0.18 μM). Additional characterization of the inhibition of PLA2G15 by fosinopril was consistent with interference of PLA2G15 binding to liposomes. PLA2G15 inhibition was more accurate in predicting phospholipidosis compared with in silico models based on pKa and ClogP, measures of protonation, and transport-independent distribution in the lysosome, respectively. In summary, PLA2G15 is a primary target for cationic amphiphilic drugs that cause phospholipidosis, and PLA2G15 inhibition by cationic amphiphilic compounds provides a potentially robust screening platform for potential toxicity during drug development.
Collapse
Affiliation(s)
- Vania Hinkovska-Galcheva
- Department of Internal Medicine, University of Michigan Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Taylour Treadwell
- Department of Internal Medicine, University of Michigan Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Jonathan M Shillingford
- Department of Internal Medicine, University of Michigan Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Angela Lee
- Department of Internal Medicine, University of Michigan Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Akira Abe
- Department of Internal Medicine, University of Michigan Medical School, University of Michigan, Ann Arbor, MI, USA
| | - John J G Tesmer
- Departments of Biological Sciences and Medicinal Chemistry and Pharmacology, Purdue University, West Lafayette, IN, USA
| | - James A Shayman
- Department of Internal Medicine, University of Michigan Medical School, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
25
|
Mollazadeh H, Tavana E, Fanni G, Bo S, Banach M, Pirro M, von Haehling S, Jamialahmadi T, Sahebkar A. Effects of statins on mitochondrial pathways. J Cachexia Sarcopenia Muscle 2021; 12:237-251. [PMID: 33511728 PMCID: PMC8061391 DOI: 10.1002/jcsm.12654] [Citation(s) in RCA: 145] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/09/2020] [Accepted: 10/20/2020] [Indexed: 12/12/2022] Open
Abstract
Statins are a family of drugs that are used for treating hyperlipidaemia with a recognized capacity to prevent cardiovascular disease events. They inhibit β-hydroxy β-methylglutaryl-coenzyme A reductase, i.e. the rate-limiting enzyme in mevalonate pathway, reduce endogenous cholesterol synthesis, and increase low-density lipoprotein clearance by promoting low-density lipoprotein receptor expression mainly in the hepatocytes. Statins have pleiotropic effects including stabilization of atherosclerotic plaques, immunomodulation, anti-inflammatory properties, improvement of endothelial function, antioxidant, and anti-thrombotic action. Despite all beneficial effects, statins may elicit adverse reactions such as myopathy. Studies have shown that mitochondria play an important role in statin-induced myopathies. In this review, we aim to report the mechanisms of action of statins on mitochondrial function. Results have shown that statins have several effects on mitochondria including reduction of coenzyme Q10 level, inhibition of respiratory chain complexes, induction of mitochondrial apoptosis, dysregulation of Ca2+ metabolism, and carnitine palmitoyltransferase-2 expression. The use of statins has been associated with the onset of additional pathological conditions like diabetes and dementia as a result of interference with mitochondrial pathways by various mechanisms, such as reduction in mitochondrial oxidative phosphorylation, increase in oxidative stress, decrease in uncoupling protein 3 concentration, and interference in amyloid-β metabolism. Overall, data reported in this review suggest that statins may have major effects on mitochondrial function, and some of their adverse effects might be mediated through mitochondrial pathways.
Collapse
Affiliation(s)
- Hamid Mollazadeh
- Department of Physiology and Pharmacology, Faculty of MedicineNorth Khorasan University of Medical SciencesBojnurdIran
- Natural Products and Medicinal Plants Research CenterNorth Khorasan University of Medical SciencesBojnurdIran
| | - Erfan Tavana
- Student Research Committee, School of MedicineNorth Khorasan University of Medical SciencesBojnurdIran
| | - Giovanni Fanni
- Department of Medical SciencesUniversity of TurinTurinItaly
| | - Simona Bo
- Department of Medical Sciences, AOU Città della Salute e della Scienza di TorinoUniversity of TurinTurinItaly
| | - Maciej Banach
- Department of HypertensionWAM University Hospital in LodzMedical University of Lodz, LodzPoland
- Polish Mother's Memorial Hospital Research Institute (PMMHRI), LodzPoland
| | - Matteo Pirro
- Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of MedicineUniversity of PerugiaPerugiaItaly
| | - Stephan von Haehling
- Department of Cardiology and PneumologyUniversity Medical Center GöttingenGöttingenGermany
- German Center for Cardiovascular Research (DZHK), partner site GöttingenGöttingenGermany
| | - Tannaz Jamialahmadi
- Department of Food Science and TechnologyIslamic Azad UniversityQuchanQuchanIran
- Department of Nutrition, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology InstituteMashhad University of Medical SciencesMashhadIran
- Neurogenic Inflammation Research CenterMashhad University of Medical SciencesMashhadIran
- Halal Research Center of IRIFDATehranIran
| |
Collapse
|
26
|
McCann MR, George De la Rosa MV, Rosania GR, Stringer KA. L-Carnitine and Acylcarnitines: Mitochondrial Biomarkers for Precision Medicine. Metabolites 2021; 11:51. [PMID: 33466750 PMCID: PMC7829830 DOI: 10.3390/metabo11010051] [Citation(s) in RCA: 179] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/08/2021] [Accepted: 01/11/2021] [Indexed: 12/11/2022] Open
Abstract
Biomarker discovery and implementation are at the forefront of the precision medicine movement. Modern advances in the field of metabolomics afford the opportunity to readily identify new metabolite biomarkers across a wide array of disciplines. Many of the metabolites are derived from or directly reflective of mitochondrial metabolism. L-carnitine and acylcarnitines are established mitochondrial biomarkers used to screen neonates for a series of genetic disorders affecting fatty acid oxidation, known as the inborn errors of metabolism. However, L-carnitine and acylcarnitines are not routinely measured beyond this screening, despite the growing evidence that shows their clinical utility outside of these disorders. Measurements of the carnitine pool have been used to identify the disease and prognosticate mortality among disorders such as diabetes, sepsis, cancer, and heart failure, as well as identify subjects experiencing adverse drug reactions from various medications like valproic acid, clofazimine, zidovudine, cisplatin, propofol, and cyclosporine. The aim of this review is to collect and interpret the literature evidence supporting the clinical biomarker application of L-carnitine and acylcarnitines. Further study of these metabolites could ultimately provide mechanistic insights that guide therapeutic decisions and elucidate new pharmacologic targets.
Collapse
Affiliation(s)
- Marc R. McCann
- The NMR Metabolomics Laboratory, Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Mery Vet George De la Rosa
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, MI 48109, USA; (M.V.G.); (G.R.R.)
| | - Gus R. Rosania
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, MI 48109, USA; (M.V.G.); (G.R.R.)
| | - Kathleen A. Stringer
- The NMR Metabolomics Laboratory, Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA;
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Michigan Center for Integrative Research in Critical Care, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
27
|
De Vries MC, Brown DA, Allen ME, Bindoff L, Gorman GS, Karaa A, Keshavan N, Lamperti C, McFarland R, Ng YS, O'Callaghan M, Pitceathly RDS, Rahman S, Russel FGM, Varhaug KN, Schirris TJJ, Mancuso M. Safety of drug use in patients with a primary mitochondrial disease: An international Delphi-based consensus. J Inherit Metab Dis 2020; 43:800-818. [PMID: 32030781 PMCID: PMC7383489 DOI: 10.1002/jimd.12196] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/18/2019] [Accepted: 11/19/2019] [Indexed: 12/29/2022]
Abstract
Clinical guidance is often sought when prescribing drugs for patients with primary mitochondrial disease. Theoretical considerations concerning drug safety in patients with mitochondrial disease may lead to unnecessary withholding of a drug in a situation of clinical need. The aim of this study was to develop consensus on safe medication use in patients with a primary mitochondrial disease. A panel of 16 experts in mitochondrial medicine, pharmacology, and basic science from six different countries was established. A modified Delphi technique was used to allow the panellists to consider draft recommendations anonymously in two Delphi rounds with predetermined levels of agreement. This process was supported by a review of the available literature and a consensus conference that included the panellists and representatives of patient advocacy groups. A high level of consensus was reached regarding the safety of all 46 reviewed drugs, with the knowledge that the risk of adverse events is influenced both by individual patient risk factors and choice of drug or drug class. This paper details the consensus guidelines of an expert panel and provides an important update of previously established guidelines in safe medication use in patients with primary mitochondrial disease. Specific drugs, drug groups, and clinical or genetic conditions are described separately as they require special attention. It is important to emphasise that consensus-based information is useful to provide guidance, but that decisions related to drug prescribing should always be tailored to the specific needs and risks of each individual patient. We aim to present what is current knowledge and plan to update this regularly both to include new drugs and to review those currently included.
Collapse
Affiliation(s)
- Maaike C. De Vries
- Radboudumc Amalia Children's HospitalRadboud Center for Mitochondrial MedicineNijmegenThe Netherlands
| | - David A. Brown
- Department of Human Nutrition, Foods, and Exercise and the Virginia Tech Center for Drug DiscoveryVirginia TechBlacksburgVirginia
| | - Mitchell E. Allen
- Department of Human Nutrition, Foods, and Exercise and the Virginia Tech Center for Drug DiscoveryVirginia TechBlacksburgVirginia
| | - Laurence Bindoff
- Department of Clinical MedicineUniversity of BergenBergenNorway
- Department of NeurologyHaukeland University HospitalBergenNorway
| | - Gráinne S. Gorman
- Wellcome Centre for Mitochondrial Research, Institute of NeuroscienceNewcastle UniversityNewcastle upon TyneUK
- The Newcastle upon Tyne Hospitals NHS Foundation TrustNewcastle upon TyneUK
| | - Amel Karaa
- Genetics Unit, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Nandaki Keshavan
- Mitochondrial Research GroupUCL Great Ormond Street Institute of Child HealthLondonUK
- Metabolic UnitGreat Ormond Street Hospital NHS Foundation TrustLondonUK
| | - Costanza Lamperti
- Unit of Medical Genetics and NeurogeneticsFondazione IRCCS Istituto Neurologico Carlo BestaMilanItaly
| | - Robert McFarland
- Wellcome Centre for Mitochondrial Research, Institute of NeuroscienceNewcastle UniversityNewcastle upon TyneUK
- The Newcastle upon Tyne Hospitals NHS Foundation TrustNewcastle upon TyneUK
| | - Yi Shiau Ng
- Wellcome Centre for Mitochondrial Research, Institute of NeuroscienceNewcastle UniversityNewcastle upon TyneUK
- The Newcastle upon Tyne Hospitals NHS Foundation TrustNewcastle upon TyneUK
| | - Mar O'Callaghan
- Department of Neurology, Metabolic UnitHospital Sant Joan de DéuBarcelonaSpain
- CIBERERInstituto de Salud Carlos IIIBarcelonaSpain
| | - Robert D. S. Pitceathly
- Department of Neuromuscular DiseasesUCL Queen Square Institute of Neurology and The National Hospital for Neurology and NeurosurgeryLondonUK
| | - Shamima Rahman
- Mitochondrial Research GroupUCL Great Ormond Street Institute of Child HealthLondonUK
- Metabolic UnitGreat Ormond Street Hospital NHS Foundation TrustLondonUK
| | - Frans G. M. Russel
- Department of Pharmacology and ToxicologyRadboud Institute for Molecular Life Sciences, Radboud Center for Mitochondrial Medicine, RadboudumcNijmegenThe Netherlands
| | - Kristin N. Varhaug
- Department of Clinical MedicineUniversity of BergenBergenNorway
- Department of NeurologyHaukeland University HospitalBergenNorway
| | - Tom J. J. Schirris
- Department of Pharmacology and ToxicologyRadboud Institute for Molecular Life Sciences, Radboud Center for Mitochondrial Medicine, RadboudumcNijmegenThe Netherlands
| | - Michelangelo Mancuso
- Department of Clinical and Experimental Medicine, Neurological InstituteUniversity of PisaPisaItaly
| |
Collapse
|
28
|
Brew DW, Black MC, Santos M, Rodgers J, Henderson WM. Metabolomic Investigations of the Temporal Effects of Exposure to Pharmaceuticals and Personal Care Products and Their Mixture in the Eastern Oyster (Crassostrea virginica). ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2020; 39:419-436. [PMID: 31661721 DOI: 10.1002/etc.4627] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 05/21/2019] [Accepted: 10/22/2019] [Indexed: 06/10/2023]
Abstract
The eastern oyster (Crassostrea virginica) supports a large aquaculture industry and is a keystone species along the Atlantic seaboard. Native oysters are routinely exposed to a complex mixture of contaminants that increasingly includes pharmaceuticals and personal care products (PPCPs). Unfortunately, the biological effects of chemical mixtures on oysters are poorly understood. Untargeted gas chromatography-mass spectrometry metabolomics was utilized to quantify the response of oysters exposed to fluoxetine, N,N-diethyl-meta-toluamide, 17α-ethynylestradiol, diphenhydramine, and their mixture. Oysters were exposed to 1 µg/L of each chemical or mixture for 10 d, followed by an 8-d depuration period. Adductor muscle (n = 14/treatment) was sampled at days 0, 1, 5, 10, and 18. Trajectory analysis illustrated that metabolic effects and class separation of the treatments varied at each time point and that, overall, the oysters were only able to partially recover from these exposures post-depuration. Altered metabolites were associated with cellular energetics (i.e., Krebs cycle intermediates), as well as amino acid metabolism and fatty acids. Exposure to these PPCPs also affected metabolic pathways associated with anaerobic metabolism, osmotic stress, and oxidative stress, in addition to the physiological effects of each chemical's postulated mechanism of action. Following depuration, fewer metabolites were altered, but none of the treatments returned them to their initial control values, indicating that metabolic disruptions were long-lasting. Interestingly, the mixture did not directly cluster with individual treatments in the scores plot from partial least squares discriminant analysis, and many of its affected metabolic pathways were not well predicted from the individual treatments. The present study highlights the utility of untargeted metabolomics in developing exposure biomarkers for compounds with different modes of action in bivalves. Environ Toxicol Chem 2020;39:419-436. © 2019 SETAC.
Collapse
Affiliation(s)
- David W Brew
- Department of Environmental Health Science, University of Georgia, Athens, Georgia, USA
| | - Marsha C Black
- Department of Environmental Health Science, University of Georgia, Athens, Georgia, USA
| | - Marina Santos
- Department of Environmental Health Science, University of Georgia, Athens, Georgia, USA
| | - Jackson Rodgers
- Department of Environmental Health Science, University of Georgia, Athens, Georgia, USA
| | - W Matthew Henderson
- National Exposure Research Laboratory, Office of Research and Development, US Environmental Protection Agency, Athens, Georgia
| |
Collapse
|
29
|
Moloudizargari M, Moradkhani F, Asghari N, Fallah M, Asghari MH, Moghadamnia AA, Abdollahi M. NLRP inflammasome as a key role player in the pathogenesis of environmental toxicants. Life Sci 2019; 231:116585. [PMID: 31226415 DOI: 10.1016/j.lfs.2019.116585] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 06/13/2019] [Accepted: 06/17/2019] [Indexed: 12/26/2022]
Abstract
Exposure to environmental toxicants (ET) results in specific organ damage and auto-immune diseases, mostly mediated by inflammatory responses. The NLRP3 inflammasome has been found to be the major initiator of the associated pathologic inflammation. It has been found that ETs can trigger all the signals required for an NLRP3-mediated response. The exaggerated activation of the NLRP3 inflammasome and its end product IL-1β, is responsible for the pathogenesis caused by many ETs including pesticides, organic pollutants, heavy metals, and crystalline compounds. Therefore, an extensive study of these chemicals and their mechanisms of inflammasome (INF) activation may provide the scientific evidence for possible targeting of this pathway by proposing possible protective agents that have been previously shown to affect INF compartments and its activation. Melatonin and polyunsaturated fatty acids (PUFA) are among the safest and the most studied of these agents, which affect a wide variety of cellular and physiological processes. These molecules have been shown to suppress the NLRP3 inflammasome mostly through the regulation of cellular redox status and the nuclear factor-κB (NF-κB) pathway, rendering them potential promising compounds to overcome ET-mediated organ damage. In the present review, we have made an effort to extensively review the ETs that exert their pathogenesis via the stimulation of inflammation, their precise mechanisms of action and the possible protective agents that could be potentially used to protect against such toxicants.
Collapse
Affiliation(s)
- Milad Moloudizargari
- Department of Immunology, School of Medicine, Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Moradkhani
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Narjes Asghari
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran, Islamic Republic of Iran
| | - Marjan Fallah
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Hossein Asghari
- Department of Pharmacology and Toxicology, School of Medicine, Babol University of Medical Sciences, Babol, Iran.
| | - Ali Akbar Moghadamnia
- Department of Pharmacology and Toxicology, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Mohammad Abdollahi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Toxicology and Diseases Group, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
30
|
The footprints of mitochondrial impairment and cellular energy crisis in the pathogenesis of xenobiotics-induced nephrotoxicity, serum electrolytes imbalance, and Fanconi's syndrome: A comprehensive review. Toxicology 2019; 423:1-31. [PMID: 31095988 DOI: 10.1016/j.tox.2019.05.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 04/29/2019] [Accepted: 05/09/2019] [Indexed: 12/19/2022]
Abstract
Fanconi's Syndrome (FS) is a disorder characterized by impaired renal proximal tubule function. FS is associated with a vast defect in the renal reabsorption of several chemicals. Inherited and/or acquired conditions seem to be connected with FS. Several xenobiotics including many pharmaceuticals are capable of inducing FS and nephrotoxicity. Although the pathological state of FS is well described, the exact underlying etiology and cellular mechanism(s) of xenobiotics-induced nephrotoxicity, serum electrolytes imbalance, and FS are not elucidated. Constant and high dependence of the renal reabsorption process to energy (ATP) makes mitochondrial dysfunction as a pivotal mechanism which could be involved in the pathogenesis of FS. The current review focuses on the footprints of mitochondrial impairment in the etiology of xenobiotics-induced FS. Moreover, the importance of mitochondria protecting agents and their preventive/therapeutic capability against FS is highlighted. The information collected in this review may provide significant clues to new therapeutic interventions aimed at minimizing xenobiotics-induced renal injury, serum electrolytes imbalance, and FS.
Collapse
|
31
|
Burgos-Aceves MA, Cohen A, Paolella G, Lepretti M, Smith Y, Faggio C, Lionetti L. Modulation of mitochondrial functions by xenobiotic-induced microRNA: From environmental sentinel organisms to mammals. THE SCIENCE OF THE TOTAL ENVIRONMENT 2018; 645:79-88. [PMID: 30015121 DOI: 10.1016/j.scitotenv.2018.07.109] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 07/09/2018] [Accepted: 07/09/2018] [Indexed: 06/08/2023]
Abstract
Mitochondria play a crucial role in energetic metabolism, signaling pathways, and overall cell viability. They are in the first line in facing cellular energy requirements in stress conditions, such as in response to xenobiotic exposure. Recently, a novel regulatory key role of microRNAs (miRNAs) in important signaling pathways in mitochondria has been proposed. Consequently, alteration in miRNAs expression by xenobiotics could outcome into mitochondrial dysfunction, reactive oxygen species overexpression, and liberation of apoptosis or necrosis activating proteins. The aim of this review is to show the highlights about mitochondria-associated miRNAs in cellular processes exposed to xenobiotic stress in different cell types involved in detoxification processes or sensitive to environmental hazards in marine sentinel organisms and mammals.
Collapse
Affiliation(s)
- Mario Alberto Burgos-Aceves
- Department of Chemistry and Biology, University of Salerno, via Giovanni Paolo II, 132, 84084 Fisciano, SA, Italy
| | - Amit Cohen
- Genomic Data Analysis Unit, The Hebrew University of Jerusalem-Hadassah Medical School, P.O. Box 12272, Jerusalem 91120, Israel
| | - Gaetana Paolella
- Department of Chemistry and Biology, University of Salerno, via Giovanni Paolo II, 132, 84084 Fisciano, SA, Italy
| | - Marilena Lepretti
- Department of Chemistry and Biology, University of Salerno, via Giovanni Paolo II, 132, 84084 Fisciano, SA, Italy
| | - Yoav Smith
- Genomic Data Analysis Unit, The Hebrew University of Jerusalem-Hadassah Medical School, P.O. Box 12272, Jerusalem 91120, Israel
| | - Caterina Faggio
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres, 31, 98166 Messina, Italy.
| | - Lillà Lionetti
- Department of Chemistry and Biology, University of Salerno, via Giovanni Paolo II, 132, 84084 Fisciano, SA, Italy
| |
Collapse
|
32
|
Pyrazinyl ureas revisited: 1-(3-(Benzyloxy)pyrazin-2-yl)-3-(3,4-dichlorophenyl)urea, a new blocker of Aβ-induced mPTP opening for Alzheimer's disease. Eur J Med Chem 2018; 157:268-278. [DOI: 10.1016/j.ejmech.2018.07.068] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 07/27/2018] [Accepted: 07/29/2018] [Indexed: 12/22/2022]
|
33
|
Niknahad H, Heidari R, Mohammadzadeh R, Ommati MM, Khodaei F, Azarpira N, Abdoli N, Zarei M, Asadi B, Rasti M, Shirazi Yeganeh B, Taheri V, Saeedi A, Najibi A. Sulfasalazine induces mitochondrial dysfunction and renal injury. Ren Fail 2018; 39:745-753. [PMID: 29214868 PMCID: PMC6446160 DOI: 10.1080/0886022x.2017.1399908] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Sulfasalazine is a commonly used drug for the treatment of rheumatoid arthritis and inflammatory bowel disease. There are several cases of renal injury encompass sulfasalazine administration in humans. The mechanism of sulfasalazine adverse effects toward kidneys is obscure. Oxidative stress and its consequences seem to play a role in the sulfasalazine-induced renal injury. The current investigation was designed to investigate the effect of sulfasalazine on kidney mitochondria. Rats received sulfasalazine (400 and 600 mg/kg/day, oral) for 14 consecutive days. Afterward, kidney mitochondria were isolated and assessed. Sulfasalazine-induced renal injury was biochemically evident by the increase in serum blood urea nitrogen (BUN), gamma-glutamyl transferase (γ-GT), and creatinine (Cr). Histopathological presentations of the kidney in sulfasalazine-treated animals revealed by interstitial inflammation, tubular atrophy, and tissue necrosis. Markers of oxidative stress including an increase in reactive oxygen species (ROS) and lipid peroxidation (LPO), a defect in tissue antioxidant capacity, and glutathione (GSH) depletion were also detected in the kidney of sulfasalazine-treated groups. Decreased mitochondrial succinate dehydrogenase activity (SDA), mitochondrial depolarization, mitochondrial GSH depletion, increase in mitochondrial ROS, LPO, and mitochondrial swelling were also evident in sulfasalazine-treated groups. Current data suggested that oxidative stress and mitochondrial injury might be involved in the mechanism of sulfasalazine-induced renal injury.
Collapse
Affiliation(s)
- Hossein Niknahad
- a Pharmaceutical Sciences Research Center , Shiraz University of Medical Sciences , Shiraz , Iran.,b Department of Pharmacology and Toxicology, Faculty of Pharmacy , Shiraz University of Medical Sciences , Shiraz , Iran
| | - Reza Heidari
- a Pharmaceutical Sciences Research Center , Shiraz University of Medical Sciences , Shiraz , Iran
| | - Roya Mohammadzadeh
- b Department of Pharmacology and Toxicology, Faculty of Pharmacy , Shiraz University of Medical Sciences , Shiraz , Iran
| | - Mohammad Mehdi Ommati
- c Department of Animal Sciences, School of Agriculture , Shiraz University , Shiraz , Iran
| | - Forouzan Khodaei
- b Department of Pharmacology and Toxicology, Faculty of Pharmacy , Shiraz University of Medical Sciences , Shiraz , Iran
| | - Negar Azarpira
- d Transplant Research Center , Shiraz University of Medical Sciences , Shiraz , Iran
| | - Narges Abdoli
- e Food and Drug Organization, Ministry of Health , Tehran , Iran
| | - Mahdi Zarei
- b Department of Pharmacology and Toxicology, Faculty of Pharmacy , Shiraz University of Medical Sciences , Shiraz , Iran
| | - Behnam Asadi
- b Department of Pharmacology and Toxicology, Faculty of Pharmacy , Shiraz University of Medical Sciences , Shiraz , Iran
| | - Maryam Rasti
- b Department of Pharmacology and Toxicology, Faculty of Pharmacy , Shiraz University of Medical Sciences , Shiraz , Iran
| | - Babak Shirazi Yeganeh
- f Department of Pathology, School of Medicine , Shiraz University of Medical Sciences , Shiraz , Iran
| | - Vahid Taheri
- b Department of Pharmacology and Toxicology, Faculty of Pharmacy , Shiraz University of Medical Sciences , Shiraz , Iran
| | - Arastoo Saeedi
- b Department of Pharmacology and Toxicology, Faculty of Pharmacy , Shiraz University of Medical Sciences , Shiraz , Iran
| | - Asma Najibi
- b Department of Pharmacology and Toxicology, Faculty of Pharmacy , Shiraz University of Medical Sciences , Shiraz , Iran
| |
Collapse
|
34
|
Elmorsy E, Al-Ghafari A, Helaly ANM, Hisab AS, Oehrle B, Smith PA. Editor's Highlight: Therapeutic Concentrations of Antidepressants Inhibit Pancreatic Beta-Cell Function via Mitochondrial Complex Inhibition. Toxicol Sci 2018; 158:286-301. [PMID: 28482088 DOI: 10.1093/toxsci/kfx090] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Diabetes mellitus risk is increased by prolonged usage of antidepressants (ADs). Although various mechanisms are suggested for their diabetogenic potential, whether a direct effect of ADs on pancreatic β-cells is involved is unclear. We examined this idea for 3 ADs: paroxetine, clomipramine and, with particular emphasis, fluoxetine, on insulin secretion, mitochondrial function, cellular bioenergetics, KATP channel activity, and caspase activity in murine and human cell-line models of pancreatic β-cells. Metabolic assays showed that these ADs decreased the redox, oxidative respiration, and energetic potential of β-cells in a time and concentration dependent manner, even at a concentration of 100 nM, well within the therapeutic window. These effects were related to inhibition of mitochondrial complex I and III. Consistent with impaired mitochondrial function, lactate output was increased and insulin secretion decreased. Neither fluoxetine, antimycin nor rotenone could reactivate KATP channel activity blocked by glucose unlike the mitochondrial uncoupler, FCCP. Chronic, but not acute, AD increased oxidative stress and activated caspases, 3, 8, and 9. A close agreement was found for the rates of oxidative respiration, lactate output and modulation of KATP channel activity in MIN6 cells with those of primary murine cells; data that supports MIN6 as a valid model to study beta-cell bioenergetics. To conclude, paroxetine, clomipramine and fluoxetine were all cytotoxic at therapeutic concentrations on pancreatic beta-cells; an action suggested to arise by inhibition of mitochondrial bioenergetics, oxidative stress and induction of apoptosis. These actions help explain the diabetogenic potential of these ADs in humans.
Collapse
Affiliation(s)
- Ekramy Elmorsy
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ayat Al-Ghafari
- Biochemistry Department, Faculty of Science, King Abdulaziz University (KAU), Jeddah, Kingdom of Saudi Arabia
| | - Ahmed N M Helaly
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ahmed S Hisab
- University of Nottingham Medical School, University of Nottingham, Nottinghamshire, UK
| | - Bettina Oehrle
- University of Nottingham Medical School, University of Nottingham, Nottinghamshire, UK
| | - Paul A Smith
- University of Nottingham Medical School, University of Nottingham, Nottinghamshire, UK
| |
Collapse
|
35
|
Monzote L, Geroldinger G, Tonner M, Scull R, De Sarkar S, Bergmann S, Bacher M, Staniek K, Chatterjee M, Rosenau T, Gille L. Interaction of ascaridole, carvacrol, and caryophyllene oxide from essential oil of Chenopodium ambrosioides L. with mitochondria in Leishmania and other eukaryotes. Phytother Res 2018; 32:1729-1740. [PMID: 29672979 PMCID: PMC6208284 DOI: 10.1002/ptr.6097] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 03/07/2018] [Accepted: 03/27/2018] [Indexed: 11/22/2022]
Abstract
The antileishmanial activity of the essential oil (EO) from Chenopodium ambrosioides L. has been demonstrated in vitro and in animal models, attributed to the major components of the EO. This study focused on the effects of the three major EO compounds carvacrol, caryophyllene oxide (Caryo), and the antileishmanial endoperoxide ascaridole (Asc) on mitochondrial functions in Leishmania tarentolae promastigotes (LtP). EO and Caryo were able to partially inhibit the leishmanial electron transport chain, whereas other components failed to demonstrate a direct immediate effect. Caryo demonstrated inhibition of complex III activity in LtP and in isolated complex III from other species. The formation of superoxide radicals was studied in Leishmania by electron spin resonance spectroscopy in the presence of iron chelators wherein selected compounds failed to trigger a significant immediate additional superoxide production in LtP. However, upon prolonged incubation of Leishmania with Asc and especially in the absence of iron chelators (allowing the activation of Asc), an increased superoxide radical production and significant impairment of mitochondrial coupling in Leishmania was observed. Prolonged incubation with all EO components resulted in thiol depletion. Taken together, the major components of EO mediate their leishmanicidal activity via different mitochondrial targets and time profiles. Further studies are required to elucidate possible synergistic effects of carvacrol and Asc and the influence of minor compounds.
Collapse
Affiliation(s)
- Lianet Monzote
- Institute of Pharmacology and Toxicology, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Vienna, Austria.,Department of Parasitology, Institute of Tropical Medicine "Pedro Kourí", Havana, Cuba
| | - Gerald Geroldinger
- Institute of Pharmacology and Toxicology, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Matthias Tonner
- Institute of Pharmacology and Toxicology, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Ramón Scull
- Department of Chemistry, Institute of Pharmacy and Food, Havana University, Havana, Cuba
| | - Sritama De Sarkar
- Department of Pharmacology, Institute of Postgraduate Medical Education & Research, Kolkata, India
| | - Sophie Bergmann
- Institute of Pharmacology and Toxicology, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Markus Bacher
- Department of Chemistry, Division of Chemistry of Renewables, University of Natural Resources and Life Sciences, Tulln, Austria
| | - Katrin Staniek
- Institute of Pharmacology and Toxicology, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Mitali Chatterjee
- Department of Pharmacology, Institute of Postgraduate Medical Education & Research, Kolkata, India
| | - Thomas Rosenau
- Department of Chemistry, Division of Chemistry of Renewables, University of Natural Resources and Life Sciences, Tulln, Austria
| | - Lars Gille
- Institute of Pharmacology and Toxicology, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Vienna, Austria
| |
Collapse
|
36
|
Wolters JEJ, van Breda SGJ, Grossmann J, Fortes C, Caiment F, Kleinjans JCS. Integrated 'omics analysis reveals new drug-induced mitochondrial perturbations in human hepatocytes. Toxicol Lett 2018; 289:1-13. [PMID: 29501571 DOI: 10.1016/j.toxlet.2018.02.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 02/02/2018] [Accepted: 02/23/2018] [Indexed: 12/11/2022]
Abstract
We performed a multiple 'omics study by integrating data on epigenomic, transcriptomic, and proteomic perturbations associated with mitochondrial dysfunction in primary human hepatocytes caused by the liver toxicant valproic acid (VPA), to deeper understand downstream events following epigenetic alterations in the mitochondrial genome. Furthermore, we investigated persistence of cross-omics changes after terminating drug treatment. Upon transient methylation changes of mitochondrial genes during VPA-treatment, increasing complexities of gene-interaction networks across time were demonstrated, which normalized during washout. Furthermore, co-expression between genes and their corresponding proteins increased across time. Additionally, in relation to persistently decreased ATP production, we observed decreased expression of mitochondrial complex I and III-V genes. Persistent transcripts and proteins were related to citric acid cycle and β-oxidation. In particular, we identified a potential novel mitochondrial-nuclear signaling axis, MT-CO2-FN1-MYC-CPT1. In summary, this cross-omics study revealed dynamic responses of the mitochondrial epigenome to an impulse toxicant challenge resulting in persistent mitochondrial dysfunctioning. Moreover, this approach allowed for discriminating between the toxic effect of VPA and adaptation.
Collapse
Affiliation(s)
- Jarno E J Wolters
- Department of Toxicogenomics, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. Box 616, 6200 MD Maastricht, The Netherlands.
| | - Simone G J van Breda
- Department of Toxicogenomics, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. Box 616, 6200 MD Maastricht, The Netherlands.
| | - Jonas Grossmann
- Functional Genomics Center Zurich, Functional Genomics Center Zurich, University Zurich/ETH Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland.
| | - Claudia Fortes
- Functional Genomics Center Zurich, Functional Genomics Center Zurich, University Zurich/ETH Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland.
| | - Florian Caiment
- Department of Toxicogenomics, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. Box 616, 6200 MD Maastricht, The Netherlands.
| | - Jos C S Kleinjans
- Department of Toxicogenomics, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. Box 616, 6200 MD Maastricht, The Netherlands.
| |
Collapse
|
37
|
Abstract
Recent decades have seen a rapid increase in reported toxic effects of drugs and pollutants on mitochondria. Researchers have also documented many genetic differences leading to mitochondrial diseases, currently reported to affect ∼1 person in 4,300, creating a large number of potential gene-environment interactions in mitochondrial toxicity. We briefly review this history, and then highlight cutting-edge areas of mitochondrial research including the role of mitochondrial reactive oxygen species in signaling; increased understanding of fundamental biological processes involved in mitochondrial homeostasis (DNA maintenance and mutagenesis, mitochondrial stress response pathways, fusion and fission, autophagy and biogenesis, and exocytosis); systemic effects resulting from mitochondrial stresses in specific cell types; mitochondrial involvement in immune function; the growing evidence of long-term effects of mitochondrial toxicity; mitochondrial-epigenetic cross-talk; and newer approaches to test chemicals for mitochondrial toxicity. We also discuss the potential importance of hormetic effects of mitochondrial stressors. Finally, we comment on future areas of research we consider critical for mitochondrial toxicology, including increased integration of clinical, experimental laboratory, and epidemiological (human and wildlife) studies; improved understanding of biomarkers in the human population; and incorporation of other factors that affect mitochondria, such as diet, exercise, age, and nonchemical stressors.
Collapse
Affiliation(s)
- Joel N Meyer
- Nicholas School of the Environment and Integrated Toxicology and Environmental Health Program, Duke University, Durham, North Carolina 27708-0328
| | - Jessica H Hartman
- Nicholas School of the Environment and Integrated Toxicology and Environmental Health Program, Duke University, Durham, North Carolina 27708-0328
| | - Danielle F Mello
- Nicholas School of the Environment and Integrated Toxicology and Environmental Health Program, Duke University, Durham, North Carolina 27708-0328
| |
Collapse
|
38
|
Delong W, Yongling W, Lanying W, Juntao F, Xing Z. Design, synthesis and evaluation of 3-arylidene azetidin-2-ones as potential antifungal agents against Alternaria solani Sorauer. Bioorg Med Chem 2017; 25:6661-6673. [PMID: 29137937 DOI: 10.1016/j.bmc.2017.11.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 10/26/2017] [Accepted: 11/02/2017] [Indexed: 12/29/2022]
Abstract
A new concise and facile method was explored to synthesize a collection of new 3-arylidene azetidin-2-ones, which could be regarded as the derivatives of the hybrid scaffold of bioactive natural cinnamamide and heterocycle azetidi-2-one. The structures of the synthesized compounds were characterized by 1H, 13C NMR, and MS; and their antifungal activity were evaluated against Alternaria solani Sorauer. These antifungal data were subjected to a quantitative structure-activity relationship (QSAR) analysis using Codessa software on the basis of the results from B3LYP/6-31G(d,p) quantum calculations. The best regressive model revealed that potentially more active compounds should have low dipole moments and QC-min (minimal net atomic charge for a C atom), and high QO-max (maximal net atomic charge for an O atom) and QN-min (minimal net atomic charge for an N atom). The most potent compound 7k could lead to intracellular accumulation of reactive oxygen species, dissipation of mitochondrial transmembrane potential, and an autophagy-like cell death process in A. solani Sorauer. Taken together, these results laid the foundation for further design of improved crop-protection agents based on this hybrid scaffold.
Collapse
Affiliation(s)
- Wang Delong
- Research & Development Center of Biorational Pesticide, Shaanxi Research Center of Biopesticide Engineering & Technology, Key Laboratory of Plant Protection Resources and Pest Management of Ministry of Education, Northwest A&F University, Yangling 712100, China
| | - Wu Yongling
- Research & Development Center of Biorational Pesticide, Shaanxi Research Center of Biopesticide Engineering & Technology, Key Laboratory of Plant Protection Resources and Pest Management of Ministry of Education, Northwest A&F University, Yangling 712100, China
| | - Wang Lanying
- Research & Development Center of Biorational Pesticide, Shaanxi Research Center of Biopesticide Engineering & Technology, Key Laboratory of Plant Protection Resources and Pest Management of Ministry of Education, Northwest A&F University, Yangling 712100, China; College of Environment and Plant Protection, Hainan University, Haikou, Hainan 570228, China
| | - Feng Juntao
- Research & Development Center of Biorational Pesticide, Shaanxi Research Center of Biopesticide Engineering & Technology, Key Laboratory of Plant Protection Resources and Pest Management of Ministry of Education, Northwest A&F University, Yangling 712100, China.
| | - Zhang Xing
- Research & Development Center of Biorational Pesticide, Shaanxi Research Center of Biopesticide Engineering & Technology, Key Laboratory of Plant Protection Resources and Pest Management of Ministry of Education, Northwest A&F University, Yangling 712100, China
| |
Collapse
|
39
|
Zhang H, Yu P, Ren JX, Li XB, Wang HL, Ding L, Kong WB. Development of novel prediction model for drug-induced mitochondrial toxicity by using naïve Bayes classifier method. Food Chem Toxicol 2017; 110:122-129. [PMID: 29042293 DOI: 10.1016/j.fct.2017.10.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 10/10/2017] [Accepted: 10/13/2017] [Indexed: 02/05/2023]
Abstract
Mitochondrial dysfunction has been considered as an important contributing factor in the etiology of drug-induced organ toxicity, and even plays an important role in the pathogenesis of some diseases. The objective of this investigation was to develop a novel prediction model of drug-induced mitochondrial toxicity by using a naïve Bayes classifier. For comparison, the recursive partitioning classifier prediction model was also constructed. Among these methods, the prediction performance of naïve Bayes classifier established here showed best, which yielded average overall prediction accuracies for the internal 5-fold cross validation of the training set and external test set were 95 ± 0.6% and 81 ± 1.1%, respectively. In addition, four important molecular descriptors and some representative substructures of toxicants produced by ECFP_6 fingerprints were identified. We hope the established naïve Bayes prediction model can be employed for the mitochondrial toxicity assessment, and these obtained important information of mitochondrial toxicants can provide guidance for medicinal chemists working in drug discovery and lead optimization.
Collapse
Affiliation(s)
- Hui Zhang
- College of Life Science, Northwest Normal University, Lanzhou, Gansu 730070, PR China; State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, PR China.
| | - Peng Yu
- College of Life Science, Northwest Normal University, Lanzhou, Gansu 730070, PR China
| | - Ji-Xia Ren
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, PR China; College of Life Science, Liaocheng University, Liaocheng, Shandong 252059, PR China
| | - Xi-Bo Li
- College of Life Science, Northwest Normal University, Lanzhou, Gansu 730070, PR China
| | - He-Li Wang
- College of Life Science, Northwest Normal University, Lanzhou, Gansu 730070, PR China
| | - Lan Ding
- College of Life Science, Northwest Normal University, Lanzhou, Gansu 730070, PR China.
| | - Wei-Bao Kong
- College of Life Science, Northwest Normal University, Lanzhou, Gansu 730070, PR China
| |
Collapse
|
40
|
Park JE, Elkamhawy A, Hassan AHE, Pae AN, Lee J, Paik S, Park BG, Roh EJ. Synthesis and evaluation of new pyridyl/pyrazinyl thiourea derivatives: Neuroprotection against amyloid-β-induced toxicity. Eur J Med Chem 2017; 141:322-334. [PMID: 29031076 DOI: 10.1016/j.ejmech.2017.09.043] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 09/20/2017] [Accepted: 09/21/2017] [Indexed: 12/14/2022]
Abstract
Herein, we report synthesis and evaluation of new twenty six small molecules against β amyloid (Aβ)-induced opening of mitochondrial permeability transition pore (mPTP) using JC-1 assay which measures the change of mitochondrial membrane potential (ΔΨm). The neuroprotective effect of seventeen compounds against Aβ-induced mPTP opening was superior to that of the standard Cyclosporin A (CsA). Fifteen derivatives eliciting increased green to red fluorescence percentage less than 40.0% were evaluated for their impact on ATP production, cell viability and neuroprotection against Aβ-induced neuronal cell death. Among evaluated compounds, derivatives 9w, 9r and 9k had safe profile regarding ATP production and cell viability. In addition, they exhibited significant neuroprotection (69.3, 51.8 and 48.2% respectively). Molecular modeling study using CDocker algorithm predicted plausible binding modes explaining the elicited mPTP blocking activity. Hence, this study suggests compounds 9w, 9r and 9k as leads for further development of novel therapy to Alzheimer's disease.
Collapse
Affiliation(s)
- Jung-Eun Park
- Chemical Kinomics Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Department of Chemistry, Sogang University, Seoul 04107, Republic of Korea
| | - Ahmed Elkamhawy
- Chemical Kinomics Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| | - Ahmed H E Hassan
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; Medicinal Chemistry Laboratory, Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Life and Nonopharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Ae Nim Pae
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| | - Jiyoun Lee
- Department of Global Medical Science, Sungshin Women's University, Seoul 142-732, Republic of Korea
| | - Sora Paik
- Chemical Kinomics Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Department of Fundamental Pharmaceutical Sciences, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Beoung-Geon Park
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Eun Joo Roh
- Chemical Kinomics Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea.
| |
Collapse
|
41
|
Wheat germ supplementation alleviates insulin resistance and cardiac mitochondrial dysfunction in an animal model of diet-induced obesity. Br J Nutr 2017; 118:241-249. [DOI: 10.1017/s0007114517002082] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
AbstractObesity is strongly associated with insulin resistance (IR), along with mitochondrial dysfunction to metabolically active tissues and increased production of reactive O2 species (ROS). Foods rich in antioxidants such as wheat germ (WG), protect tissues from damage due to ROS and modulate some negative effects of obesity. This study examined the effects of WG supplementation on markers of IR, mitochondrial substrate metabolism and innate antioxidant markers in two metabolically active tissues (i.e. liver and heart) of C57BL/6 mice fed a high-fat–high-sucrose (HFS) diet. Male C57BL/6 mice, 6-week-old, were randomised into four dietary treatment groups (n 12 mice/group): control (C, 10 % fat kcal), C+10 % WG, HFS (60 % fat kcal) or HFS+10 % WG (HFS+WG). After 12 weeks of treatment, HFS+WG mice had significantly less visceral fat (−16 %, P=0·006) compared with the HFS group. WG significantly reduced serum insulin (P=0·009), the insulinotropic hormone, gastric inhibitory peptide (P=0·0003), and the surrogate measure of IR, homoeostatic model assessment of IR (P=0·006). HFS diet significantly elevated (45 %, P=0·02) cardiac complex 2 mitochondrial VO2, suggesting increased metabolic stress, whereas WG stabilised this effect to the level of control. Consequently, genes which mediate antioxidant defense and mitochondrial biogenesis (superoxide dismutase 2 (Sod2) and PPARγ coactivator 1-α (Pgc1a), respectively) were significantly reduced (P<0·05) in the heart of the HFS group, whereas WG supplementation tended to up-regulate both genes. WG significantly increased hepatic gene expression of Sod2 (P=0·048) but not Pgc1a. Together, these results showed that WG supplementation in HFS diet, reduced IR and improved cardiac mitochondrial metabolic functions.
Collapse
|
42
|
Yeh A, Marcinek DJ, Meador JP, Gallagher EP. Effect of contaminants of emerging concern on liver mitochondrial function in Chinook salmon. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2017; 190:21-31. [PMID: 28668760 PMCID: PMC5590637 DOI: 10.1016/j.aquatox.2017.06.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 06/09/2017] [Accepted: 06/14/2017] [Indexed: 05/05/2023]
Abstract
We previously reported the bioaccumulation of contaminants of emerging concern (CECs), including pharmaceuticals and personal care products (PPCPs) and perfluorinated compounds, in field-collected juvenile Chinook salmon from urban estuaries of Puget Sound, WA (Meador et al., 2016). Although the toxicological impacts of CECs on salmon are poorly understood, several of the detected contaminants disrupt mitochondrial function in other species. Here, we sought to determine whether environmental exposures to CECs are associated with hepatic mitochondrial dysfunction in juvenile Chinook. Fish were exposed in the laboratory to a dietary mixture of 16 analytes representative of the predominant CECs detected in our field study. Liver mitochondrial content was reduced in fish exposed to CECs, which occurred concomitantly with a 24-32% reduction in expression of peroxisome proliferator-activated receptor (PPAR) Y coactivator-1a (pgc-1α), a positive transcriptional regulator of mitochondrial biogenesis. The laboratory exposures also caused a 40-70% elevation of state 4 respiration per unit mitochondria, which drove a 29-38% reduction of efficiency of oxidative phosphorylation relative to controls. The mixture-induced elevation of respiration was associated with increased oxidative injury as evidenced by increased mitochondrial protein carbonyls, elevated expression of glutathione (GSH) peroxidase 4 (gpx4), a mitochondrial-associated GSH peroxidase that protects against lipid peroxidation, and reduction of mitochondrial GSH. Juvenile Chinook sampled in a WWTP effluent-impacted estuary with demonstrated releases of CECs showed similar trends toward reduced liver mitochondrial content and elevated respiratory activity per mitochondria (including state 3 and uncoupled respiration). However, respiratory control ratios were greater in fish from the contaminated site relative to fish from a minimally-polluted reference site, which may have been due to differences in the timing of exposure to CECs under laboratory and field conditions. Our results indicate that exposure to CECs can affect both mitochondrial quality and content, and support the analysis of mitochondrial function as an indicator of the sublethal effects of CECs in wild fish.
Collapse
Affiliation(s)
- Andrew Yeh
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105-6099, United States
| | - David J Marcinek
- Department of Radiology, Pathology, and Bioengineering University of Washington Medical School, Seattle, WA 98195, United States
| | - James P Meador
- Environmental and Fisheries Sciences Division, Northwest Fisheries Science Center, National Marine Fisheries Service, National Oceanic and Atmospheric Administration, 2725 Montlake Blvd. East, Seattle, WA 98112, United States
| | - Evan P Gallagher
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105-6099, United States.
| |
Collapse
|
43
|
The use of high-throughput screening techniques to evaluate mitochondrial toxicity. Toxicology 2017; 391:34-41. [PMID: 28789971 DOI: 10.1016/j.tox.2017.07.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 07/28/2017] [Accepted: 07/31/2017] [Indexed: 01/30/2023]
Abstract
Toxicologists and chemical regulators depend on accurate and effective methods to evaluate and predict the toxicity of thousands of current and future compounds. Robust high-throughput screening (HTS) experiments have the potential to efficiently test large numbers of chemical compounds for effects on biological pathways. HTS assays can be utilized to examine chemical toxicity across multiple mechanisms of action, experimental models, concentrations, and lengths of exposure. Many agricultural, industrial, and pharmaceutical chemicals classified as harmful to human and environmental health exert their effects through the mechanism of mitochondrial toxicity. Mitochondrial toxicants are compounds that cause a decrease in the number of mitochondria within a cell, and/or decrease the ability of mitochondria to perform normal functions including producing adenosine triphosphate (ATP) and maintaining cellular homeostasis. Mitochondrial dysfunction can lead to apoptosis, necrosis, altered metabolism, muscle weakness, neurodegeneration, decreased organ function, and eventually disease or death of the whole organism. The development of HTS techniques to identify mitochondrial toxicants will provide extensive databases with essential connections between mechanistic mitochondrial toxicity and chemical structure. Computational and bioinformatics approaches can be used to evaluate compound databases for specific chemical structures associated with toxicity, with the goal of developing quantitative structure-activity relationship (QSAR) models and mitochondrial toxicophores. Ultimately these predictive models will facilitate the identification of mitochondrial liabilities in consumer products, industrial compounds, pharmaceuticals and environmental hazards.
Collapse
|
44
|
Jamshidzadeh A, Niknahad H, Heidari R, Azadbakht M, Khodaei F, Arabnezhad MR, Farshad O. Propylthiouracil-induced mitochondrial dysfunction in liver and its relevance to drug-induced hepatotoxicity. PHARMACEUTICAL SCIENCES 2017. [DOI: 10.15171/ps.2017.15] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
45
|
Djafarzadeh S, Vuda M, Jeger V, Takala J, Jakob SM. The Effects of Fentanyl on Hepatic Mitochondrial Function. Anesth Analg 2017; 123:311-25. [PMID: 27089001 DOI: 10.1213/ane.0000000000001280] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Remifentanil interferes with hepatic mitochondrial function. The aim of the present study was to evaluate whether hepatic mitochondrial function is affected by fentanyl, a more widely used opioid than remifentanil. METHODS Human hepatoma HepG2 cells were exposed to fentanyl or pretreated with naloxone (an opioid receptor antagonist) or 5-hydroxydecanoate (5-HD, an inhibitor of mitochondrial adenosine triphosphate (ATP)-sensitive potassium [mitoKATP] channels), followed by incubation with fentanyl. Mitochondrial function and metabolism were then analyzed. RESULTS Fentanyl marginally reduced maximal mitochondrial complex-specific respiration rates using exogenous substrates (decrease in medians: 11%-18%; P = 0.003-0.001) but did not affect basal cellular respiration rates (P = 0.834). The effect on stimulated respiration was prevented by preincubation with naloxone or 5-HD. Fentanyl reduced cellular ATP content in a dose-dependent manner (P < 0.001), an effect that was not significantly prevented by 5-HD and not explained by increased total ATPase concentration. However, in vitro ATPase activity of recombinant human permeability glycoprotein (an ATP-dependent drug efflux transporter) was significantly stimulated by fentanyl (P = 0.004). CONCLUSIONS Our data suggest that fentanyl reduces stimulated mitochondrial respiration of cultured human hepatocytes by a mechanism that is blocked by a mitoKATP channel antagonist. Increased energy requirements for fentanyl efflux transport may offer an explanation for the substantial decrease in cellular ATP concentration.
Collapse
Affiliation(s)
- Siamak Djafarzadeh
- From the *Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland; and †Department of Clinical Research, Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | | | | | | | | |
Collapse
|
46
|
Du H, Yang J, Bai J, Ming K, Shi J, Yao F, Zhang W, Yu Y, Chen Y, Xiong W, Wu Y, Wang D, Hu Y, Liu J. A flavone-polysaccharide based prescription attenuates the mitochondrial dysfunction induced by duck hepatitis A virus type 1. PLoS One 2017; 12:e0175495. [PMID: 28394931 PMCID: PMC5386289 DOI: 10.1371/journal.pone.0175495] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 03/27/2017] [Indexed: 12/17/2022] Open
Abstract
The principal target organ of duck hepatitis A virus type 1 (DHAV-1) is duckling liver, which is an energy-intensive organ and plays important roles in body’s energy metabolism and conversion. As the “power house” of the hepatocytes, mitochondria provide more than 90% of the energy. However, mitochondria are much vulnerable to the oxidative stress for their rich in polyunsaturated fatty acids. Although previous researches have demonstrated that DHAV-1 could induce the oxidative stress in the serum of the infected ducklings, no related study on the mitochondria during the pathological process of DVH has been reported by far. To address this issue, we examined the HE stained tissue pathological slices, detected the hepatic SOD, CAT and GPX activities and MDA contents and analyzed the ATP content, mitochondrial ultrastructure and the mitochondrial SOD, GPX activities and MDA content in the liver tissues. The results showed that the hepatic redox status was significantly disturbed so that causing the mitochondrial dysfunction, ATP depletion and mitochondrial oxidative stress during the process of the DHAV-1 infection, and a prescription formulated with Hypericum japonicum flavone, Radix Rehmanniae Recens polysaccharide and Salvia plebeia flavone (HRS), which had been demonstrated with good anti-oxidative activity in serum, could effectively alleviate the hepatic injury and the oxidative stress in liver tissue induced by DHAV-1 thus alleviating the mitochondrial injury and oxidative stress. In a word, this research discovers the oxidative stress induced mitochondrial dysfunction and oxidative stress during the DVH pathological process and demonstrates HRS exerts good anti-oxidative activity in liver tissue to protect mitochondria against reactive oxygen species (ROS).
Collapse
MESH Headings
- Adenosine Triphosphate/metabolism
- Animals
- Antiviral Agents/pharmacology
- Drugs, Chinese Herbal/pharmacology
- Ducks
- Flavones/pharmacology
- Glutathione Peroxidase/metabolism
- Hepatitis Virus, Duck
- Hepatitis, Viral, Animal/drug therapy
- Hepatitis, Viral, Animal/metabolism
- Hepatitis, Viral, Animal/mortality
- Hepatitis, Viral, Animal/pathology
- Lipid Peroxidation/drug effects
- Lipid Peroxidation/physiology
- Liver/drug effects
- Liver/metabolism
- Liver/pathology
- Malondialdehyde/metabolism
- Mitochondria/drug effects
- Mitochondria/metabolism
- Mitochondria/ultrastructure
- Oxidative Stress/drug effects
- Picornaviridae Infections/drug therapy
- Picornaviridae Infections/metabolism
- Picornaviridae Infections/mortality
- Picornaviridae Infections/pathology
- Polysaccharides/pharmacology
- Random Allocation
- Superoxide Dismutase/metabolism
- Treatment Outcome
Collapse
Affiliation(s)
- Hongxu Du
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, P R China
| | - Jingjing Yang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, P R China
| | - Jingying Bai
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, P R China
| | - Ke Ming
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, P R China
| | - Jintong Shi
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, P R China
| | - Fangke Yao
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, P R China
| | - Wei Zhang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, P R China
| | - Yang Yu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, P R China
| | - Yun Chen
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, P R China
| | - Wen Xiong
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, P R China
| | - Yi Wu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, P R China
| | - Deyun Wang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, P R China
| | - Yuanliang Hu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, P R China
| | - Jiaguo Liu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, P R China
- * E-mail:
| |
Collapse
|
47
|
New insights into the intracellular distribution pattern of cationic amphiphilic drugs. Sci Rep 2017; 7:44277. [PMID: 28281674 PMCID: PMC5345070 DOI: 10.1038/srep44277] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 02/06/2017] [Indexed: 12/28/2022] Open
Abstract
Cationic amphiphilic drugs (CADs) comprise a wide variety of different substance classes such as antidepressants, antipsychotics, and antiarrhythmics. It is well recognized that CADs accumulate in certain intracellular compartments leading to specific morphological changes of cells. So far, no adequate technique exists allowing for ultrastructural analysis of CAD in intact cells. Azidobupramine, a recently described multifunctional antidepressant analogue, allows for the first time to perform high-resolution studies of CADs on distribution pattern and morphological changes in intact cells. We showed here that the intracellular distribution pattern of azidobupramine strongly depends on drug concentration and exposure time. The mitochondrial compartment (mDsRed) and the late endo-lysosomal compartment (CD63-GFP) were the preferred localization sites at low to intermediate concentrations (i.e. 1 μM, 5 μM). In contrast, the autophagosomal compartment (LC3-GFP) can only be reached at high concentrations (10 μM) and long exposure times (72 hrs). At the morphological level, LC3-clustering became only prominent at high concentrations (10 μM), while changes in CD63 pattern already occurred at intermediate concentrations (5 μM). To our knowledge, this is the first study that establishes a link between intracellular CAD distribution pattern and morphological changes. Therewith, our results allow for gaining deeper understanding of intracellular effects of CADs.
Collapse
|
48
|
Liver Effects of Clinical Drugs Differentiated in Human Liver Slices. Int J Mol Sci 2017; 18:ijms18030574. [PMID: 28272341 PMCID: PMC5372590 DOI: 10.3390/ijms18030574] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 02/22/2017] [Accepted: 02/28/2017] [Indexed: 02/06/2023] Open
Abstract
Drugs with clinical adverse effects are compared in an ex vivo 3-dimensional multi-cellular human liver slice model. Functional markers of oxidative stress and mitochondrial function, glutathione GSH and ATP levels, were affected by acetaminophen (APAP, 1 mM), diclofenac (DCF, 1 mM) and etomoxir (ETM, 100 μM). Drugs targeting mitochondria more than GSH were dantrolene (DTL, 10 μM) and cyclosporin A (CSA, 10 μM), while GSH was affected more than ATP by methimazole (MMI, 500 μM), terbinafine (TBF, 100 μM), and carbamazepine (CBZ 100 μM). Oxidative stress genes were affected by TBF (18%), CBZ, APAP, and ETM (12%–11%), and mitochondrial genes were altered by CBZ, APAP, MMI, and ETM (8%–6%). Apoptosis genes were affected by DCF (14%), while apoptosis plus necrosis were altered by APAP and ETM (15%). Activation of oxidative stress, mitochondrial energy, heat shock, ER stress, apoptosis, necrosis, DNA damage, immune and inflammation genes ranked CSA (75%), ETM (66%), DCF, TBF, MMI (61%–60%), APAP, CBZ (57%–56%), and DTL (48%). Gene changes in fatty acid metabolism, cholestasis, immune and inflammation were affected by DTL (51%), CBZ and ETM (44%–43%), APAP and DCF (40%–38%), MMI, TBF and CSA (37%–35%). This model advances multiple dosing in a human ex vivo model, plus functional markers and gene profile markers of drug induced human liver side-effects.
Collapse
|
49
|
Niknahad H, Jamshidzadeh A, Heidari R, Abdoli N, Ommati MM, Jafari F, Zarei M, Asadi B. The Postulated Hepatotoxic Metabolite of Methimazole Causes Mitochondrial Dysfunction and Energy Metabolism Disturbances in Liver. PHARMACEUTICAL SCIENCES 2016. [DOI: 10.15171/ps.2016.35] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
50
|
Schultze N, Wanka H, Zwicker P, Lindequist U, Haertel B. Mitochondrial functions of THP-1 monocytes following the exposure to selected natural compounds. Toxicology 2016; 377:57-63. [PMID: 28013001 DOI: 10.1016/j.tox.2016.12.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 12/19/2016] [Accepted: 12/20/2016] [Indexed: 12/24/2022]
Abstract
The immune system is an important target of various xenobiotics, which may lead to severe adverse effects including immunosuppression or inappropriate immunostimulation. Mitochondrial toxicity is one possibility by which xenobiotics exert their toxic effects in cells or organs. In this study, we investigated the impact of three natural compounds, cyclosporine A (CsA), deoxynivalenol (DON) and cannabidiol (CBD) on mitochondrial functions in the THP-1 monocytic cell line. The cells were exposed for 24h to two different concentrations (IC10 and IC50 determined by MTT) of each compound. The cells showed concentration-dependent elevated intracellular reactive oxygen species (iROS) and induction of apoptosis (except DON) in response to the three test compounds. Mitochondrial functions were characterized by using bioenergetics profiling experiments. In THP-1 monocytes, the IC50 of CsA decreased basal and maximal respiration as well as ATP production with an impact on spare capacity indicating a mitochondrial dysfunction. Similar reaction patterns were observed following CBD exposure. The basal respiration level and ATP-production decreased in the THP-1 cells exposed to the IC50 of DON with no major impact on mitochondrial function. In conclusion, impaired mitochondrial function was accompanied by elevated iROS and apoptosis level in a monocytic cell line exposed to CsA and CBD. Mitochondrial dysfunction may be one explanation for the cytotoxicity of CBD and CsA also in other in immune cells.
Collapse
Affiliation(s)
- Nadin Schultze
- Institute of Pharmacy, Pharmaceutical Biology, Ernst-Moritz-Arndt University of Greifswald, D17489 Greifswald, Germany.
| | - Heike Wanka
- Institute of Physiology, University Medicine of Greifswald, D17495 Karlsburg, Germany
| | - Paula Zwicker
- Institute of Pharmacy, Pharmaceutical Biology, Ernst-Moritz-Arndt University of Greifswald, D17489 Greifswald, Germany
| | - Ulrike Lindequist
- Institute of Pharmacy, Pharmaceutical Biology, Ernst-Moritz-Arndt University of Greifswald, D17489 Greifswald, Germany
| | - Beate Haertel
- Institute of Pharmacy, Pharmaceutical Biology, Ernst-Moritz-Arndt University of Greifswald, D17489 Greifswald, Germany
| |
Collapse
|