1
|
Shafik MS, El-Tanbouly DM, Bishr A, Soubh AA, Attia AS, Muhammad RN. Activation of Sig-1R by afobazole attenuates Tollip/HMGB1-mediaded CCN2 autophagic degradation and NETs formation in sunitinib-induced cardiotoxicity in mice: Involvement of IRE 1α/ASK1/JNK/ AP-1 trajectory. Toxicol Appl Pharmacol 2025:117423. [PMID: 40449751 DOI: 10.1016/j.taap.2025.117423] [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: 03/28/2025] [Revised: 05/25/2025] [Accepted: 05/28/2025] [Indexed: 06/03/2025]
Abstract
Although the contribution of sigma1 receptor (Sig-1R) to afobazole's cardioprotection has been meticulously investigated, Sig-1R-mediated cardioprotective effect of afobazole against sunitinib cardiotoxicity has not been studied yet. Hence, we aimed at studying the potential modulatory impact of afobazole on Sig-1R to combat sunitinib-induced endoplasmic reticulum (ER) stress, maladaptive autophagy, and hyperactivation of neutrophils that ends up with neutrophil extracellular traps (NETs) formation. Pre-treatment with afobazole attenuated sunitinib-induced cardiotoxicity and enhanced cardiac function via significant reduction of TNNT2 and CK-MB, and restoration of nearly normal hemodynamic measurements. Afobazole-mediated Sig-1R activation mitigated the ER stress sensor, IRE1α activation and its downstream (ASK/JNK/AP-1) pathways along with caspase-3 and FK18. Subsequently, afobazole hindered NETs formation by prohibiting ER stress-induced activation of caspase-2 and pro-inflammatory cytokines; IL-1β and TNFα, as indicated by the significant reduction of NETs' specific components, namely, PAD4, NE, and MPO, along with the NETs' specific marker Cit H3. Afobazole also downregulated sunitinib-induced maladaptive autophagy, as reflected by reducing the expressions of autophagy-regulating proteins (ATG5 and ATG7) and microtubule-associated protein light chain 3 (LC3-II/I) ratio as well p62 upregulation. Furthermore, afobazole exhibited a cardioprotective effect by restoring nearly normal CCN2 level that was degraded by Tollip and HMGB1. The above-mentioned outcomes triggered by afobazole were clearly negated upon administration of the Sig-1R antagonist (BD1047), confirming that Sig-1R activation predominantly mediates the observed cardioprotective effects. Afobazole demonstrated efficacy in mitigating sunitinib-induced cardiotoxicity, as evidenced through the enhancements in hemodynamic stability, reduction of ER stress, amelioration of maladaptive autophagy, and inhibition of NETs formation.
Collapse
Affiliation(s)
- Marihan S Shafik
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Ahram Canadian University, Egypt.
| | - Dalia M El-Tanbouly
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Egypt
| | - Abeer Bishr
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Ahram Canadian University, Egypt
| | - Ayman A Soubh
- Pharmacology and Toxicology Department, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, New Administrative Capital, Cairo, Egypt
| | - Amina S Attia
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Egypt
| | - Radwa N Muhammad
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Egypt
| |
Collapse
|
2
|
Shi Q, Ren L, Papineau K, Yang X, Pang L, Oliphant JH, Schnackenberg L, Mattes W. Pexidartinib impairs liver mitochondrial functions causing cell death in primary human hepatocytes at clinically relevant concentrations. Biochem Biophys Res Commun 2025; 773:152075. [PMID: 40424848 DOI: 10.1016/j.bbrc.2025.152075] [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: 05/15/2025] [Revised: 05/21/2025] [Accepted: 05/22/2025] [Indexed: 05/29/2025]
Abstract
Pexidartinib is a regulatory agency approved small molecule kinase inhibitor (KI) with a boxed warning for hepatotoxicity, and FDA requires a Risk Evaluation and Mitigation Strategy (REMS) to mitigate such risk. The mechanism of pexidartinib hepatotoxicity is poorly understood. As mitochondrial injury and hepatocyte toxicity have been proposed to be a shared mechanism for the hepatotoxicity induced by many KIs, here we examined pexidartinib for such liabilities. Freshly isolated rat liver mitochondria, submitochondrial fractions, and cryopreserved primary human hepatocytes (PHHs) - the gold standard in vitro model for drug hepatotoxicity - were treated with pexidartinib at clinically relevant concentrations, and mitochondrial functions and cytotoxicity were assessed. In isolated mitochondria, the state 3 oxygen consumption rates of glutamate/malate- and succinate-driven respiration were both decreased by pexidartinib, while the state 4 oxygen consumption rates were unaffected. In submitochondrial fractions, the activities of respiratory chain complex (RCC) I and V, but not II, III, IV, were significantly inhibited by pexidartinib. In PHHs, as measured by a Seahorse system, pexidartinib decreased basal, spare, maximal, and adenosine triphosphate (ATP)-linked respirations at 2 h in the absence of cell death. Pexidartinib also inhibited cellular ATP level, increased reactive oxygen species, and caused cell death after 24 h. However, activities of caspases were unaffected. Importantly, the detrimental effects noted above occurred at pexidartinib concentrations of 0.5- to 2.5-fold of the human peak blood concentration (Cmax) achieved with the recommended therapeutic dose. These data suggest that mitochondrial injury and hepatocyte toxicity are involved in the mechanism of pexidartinib-induced hepatotoxicity.
Collapse
Affiliation(s)
- Qiang Shi
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA.
| | - Lijun Ren
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA
| | - Katy Papineau
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA
| | - Xi Yang
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Li Pang
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA
| | - Jessica Hawes Oliphant
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA
| | - Laura Schnackenberg
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA
| | - William Mattes
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA
| |
Collapse
|
3
|
Wang X, Chen R, Liu J, Wang E, Luo H. Liver injury related to vascular endothelial growth factor tyrosine kinase inhibitors: a pharmacovigilance analysis of the USA FDA adverse event reporting system (FAERS) database. Expert Opin Drug Saf 2025:1-9. [PMID: 39881499 DOI: 10.1080/14740338.2025.2460449] [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: 08/06/2024] [Revised: 12/24/2024] [Accepted: 01/02/2025] [Indexed: 01/31/2025]
Abstract
BACKGROUND While vascular endothelial growth factor tyrosine kinase inhibitors (VEGFR-TKIs) are known to cause adverse events like cardiotoxicity and haematotoxicity, their impact on liver injury remains understudied. This study evaluates the association between VEGFR-TKIs and liver injury using data from the FDA Adverse Event Reporting System (FAERS) database from 2006 to 2024. RESEARCH DESIGN AND METHODS Nine VEGFR-TKIs (Axitinib, Vandetanib, Cabozantinib, Lenvatinib, Pazopanib, Ponatinib, Regorafenib, Sunitinib, Sorafenib) were analyzed. Disproportionality and Bayesian analyses identified cases of VEGFR-TKI-induced liver injury, assessing onset time, mortality, and hospitalization rates. RESULTS 8,619 cases of liver injury were identified. Pazopanib had the highest association with liver injury (reporting odds ratio 3.9). The median onset of liver injury was 21 days. Mortality was 28.5%, with Sorafenib linked to the highest mortality (48.6%). Lenvatinib had the highest hospitalization rate (56%). CONCLUSION VEGFR-TKIs are associated with liver injury. Close monitoring is required to mitigate the risks of hospitalization and early mortality during treatment.
Collapse
Affiliation(s)
- Xiang Wang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Rujie Chen
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jialin Liu
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - E Wang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hui Luo
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
4
|
López-Cánovas JL, Naranjo-Martínez B, Diaz-Ruiz A. Fasting in combination with the cocktail Sorafenib:Metformin blunts cellular plasticity and promotes liver cancer cell death via poly-metabolic exhaustion. Cell Oncol (Dordr) 2025; 48:161-182. [PMID: 38990489 PMCID: PMC11850423 DOI: 10.1007/s13402-024-00966-2] [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] [Accepted: 05/31/2024] [Indexed: 07/12/2024] Open
Abstract
PURPOSE Dual-Interventions targeting glucose and oxidative metabolism are receiving increasing attention in cancer therapy. Sorafenib (S) and Metformin (M), two gold-standards in liver cancer, are known for their mitochondrial inhibitory capacity. Fasting, a glucose-limiting strategy, is also emerging as chemotherapy adjuvant. Herein, we explore the anti-carcinogenic response of nutrient restriction in combination with sorafenib:metformin (NR-S:M). RESULTS Our data demonstrates that, independently of liver cancer aggressiveness, fasting synergistically boosts the anti-proliferative effects of S:M co-treatment. Metabolic and Cellular plasticity was determined by the examination of mitochondrial and glycolytic activity, cell cycle modulation, activation of cellular apoptosis, and regulation of key signaling and metabolic enzymes. Under NR-S:M conditions, early apoptotic events and the pro-apoptotic Bcl-xS/Bcl-xL ratio were found increased. NR-S:M induced the highest retention in cellular SubG1 phase, consistent with the presence of DNA fragments from cellular apoptosis. Mitochondrial functionality, Mitochondrial ATP-linked respiration, Maximal respiration and Spare respiratory capacity, were all found blunted under NR-S:M conditions. Basal Glycolysis, Glycolytic reserve, and glycolytic capacity, together with the expression of glycogenic (PKM), gluconeogenic (PCK1 and G6PC3), and glycogenolytic enzymes (PYGL, PGM1, and G6PC3), were also negatively impacted by NR-S:M. Lastly, a TMT-proteomic approach corroborated the synchronization of liver cancer metabolic reprogramming with the activation of molecular pathways to drive a quiescent-like status of energetic-collapse and cellular death. CONCLUSION Altogether, we show that the energy-based polytherapy NR-S:M blunts cellular, metabolic and molecular plasticity of liver cancer. Notwithstanding the in vitro design of this study, it holds a promising therapeutic tool worthy of exploration for this tumor pathology.
Collapse
Affiliation(s)
- Juan L López-Cánovas
- Laboratory of Cellular and Molecular Gerontology, Precision Nutrition and Aging Program, Institute IMDEA Food (CEI UAM+CSIC), Crta. de Canto Blanco nº 8, Madrid, E-28049, Spain
| | - Beatriz Naranjo-Martínez
- Laboratory of Cellular and Molecular Gerontology, Precision Nutrition and Aging Program, Institute IMDEA Food (CEI UAM+CSIC), Crta. de Canto Blanco nº 8, Madrid, E-28049, Spain
| | - Alberto Diaz-Ruiz
- Laboratory of Cellular and Molecular Gerontology, Precision Nutrition and Aging Program, Institute IMDEA Food (CEI UAM+CSIC), Crta. de Canto Blanco nº 8, Madrid, E-28049, Spain.
- CIBER Pathophysiology of Obesity and Nutrition (CIBERobn), Córdoba, Spain.
| |
Collapse
|
5
|
Xiang L, Qin Y, Li L, Xiang X, Zhang W, Jiao Q, Shao Y, Huang X, Wu M, Zhou T, Lin Y, Chen Y. Targeting hyperactive mitochondria in activated HSCs and inhibition of liver fibrogenesis in mice using sorafenib complex micelles. Int J Pharm 2025; 669:125058. [PMID: 39653289 DOI: 10.1016/j.ijpharm.2024.125058] [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: 10/09/2024] [Revised: 11/28/2024] [Accepted: 12/06/2024] [Indexed: 12/14/2024]
Abstract
Liver fibrosis is a pathological condition marked by the excessive buildup of extracellular matrix primarily resulting from the transformation of quiescent hepatic stellate cells (HSCs) to myofibroblastic (MF) phenotype and their resultant over-expansion. Activated HSCs completely rely on their hyperactive mitochondria to supply the energy and biomass for their rapid proliferation and collagen secretion, so an intervention targeting their mitochondria can effectively restrict their pathological amplification and contribution to liver fibrosis. Here we tried sorafenib, a drug that plays anticancer roles by inducing the disruption and loss of mitochondrial functions, to reach an antifibrotic goal. And a complex micellar system, VA-PEG-PCL/TPGS (VPP/TPGS), was specifically designed and fabricated to encapsulate and deliver sorafenib selectively to activated HSCs to overcome its application limitations in bioavailability, toxicity and intracellular stay, and eventually maximize its induction of mitochondrial dysfunction and therapeutically antifibrotic efficacy. The prepared sorafenib complex micelles not only exhibited a suitable particle size, uniform morphology, and nice stability, but also performed excellently in the biosafety and HSCs-targetability in vitro and in vivo. In human active HSC cell lines, they markedly attenuated mitochondrial hyperactivity, induced apoptosis, and downregulated fibrosis markers as expected; while in a CCl4-induced murine model of hepatic fibrosis, they effectively restricted the expansion of MF-HSCs, reduced collagen deposition, and promoted the healing of liver damage, showing a good potential in fibrosis curation. Collectively, our VPP/TPGS complex micelles provide an ideal drug delivery platform that has the potential to revolutionize the treatment of liver fibrosis via addressing its cellular and metabolic underpinnings and thus improve patient outcomes.
Collapse
Affiliation(s)
- Li Xiang
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China; Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China
| | - Yuting Qin
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Lei Li
- Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China
| | - Xianjing Xiang
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Wenhui Zhang
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Qiangqiang Jiao
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Yaru Shao
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Xinqiong Huang
- Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China
| | - Meichun Wu
- Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China
| | - Tianle Zhou
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Yukang Lin
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Yuping Chen
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China; Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China; MOE Key Laboratory of Rare Pediatric Diseases, Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China.
| |
Collapse
|
6
|
Shashni B, Tran HT, Vong LB, Chung RJ, Nagasaki Y. Sorafenib-Loaded Silica-Containing Redox Nanoparticle Decreases Tumorigenic Potential of Lewis Lung Carcinoma. Pharmaceutics 2025; 17:50. [PMID: 39861698 PMCID: PMC11768447 DOI: 10.3390/pharmaceutics17010050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/28/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
Background: Orally administered sorafenib has shown limited improvement in overall survival for non-small-cell lung cancer patients, likely due to poor pharmacokinetics and adverse effects, including gastrointestinal toxicity. To address these issues, we developed silica-containing antioxidant nanoparticles (siRNP) as a carrier to enhance the therapeutic efficacy of lipophilic sorafenib. Methods: Sorafenib was loaded into siRNP via dialysis (sora@siRNP). The therapeutic efficacy and safety of sora@siRNP (20 and 40 mg-sora/kg) were evaluated in a xenograft mouse model of Lewis lung carcinoma (subcutaneous tumors and experimental metastasis) following oral administration. Results: Crosslinking nanosilica in siRNP improved drug stability, enabling 8.9% sorafenib loading and pH resilience. Oral sora@siRNP exhibited dose-dependent tumor growth suppression by downregulating pMEK, outperforming free sorafenib, which showed inconsistent efficacy likely due to formulation variability. Intestinal damage, a major adverse effect of free sorafenib, was significantly reduced with sora@siRNP, attributed to siRNP's antioxidant property of mitigating oxidative damage. Survival rates in the experimental metastasis model were 66-74% for sorafenib but reached 100% for sora@siRNP, highlighting its superior efficacy and safety. Conclusions: These findings demonstrate that nanosilica-crosslinked antioxidant nanoparticles (siRNP) enhance the stability, delivery efficiency, and safety of lipophilic drugs like sorafenib for oral administration. This platform holds promise for improving therapeutic outcomes in lung cancer while minimizing adverse effects.
Collapse
Affiliation(s)
- Babita Shashni
- Department of Materials Science, Graduate School of Pure and Applied Sciences, University of Tsukuba, Tennoudai 1-1-1, Tsukuba 305-8573, Ibaraki, Japan; (B.S.); (H.T.T.)
| | - Hao Thi Tran
- Department of Materials Science, Graduate School of Pure and Applied Sciences, University of Tsukuba, Tennoudai 1-1-1, Tsukuba 305-8573, Ibaraki, Japan; (B.S.); (H.T.T.)
| | - Long Binh Vong
- School of Biomedical Engineering, International University, Ho Chi Minh City 700000, Vietnam;
- School of Biomedical Engineering, Vietnam National University, Ho Chi Minh City 700000, Vietnam
| | - Ren-Jei Chung
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei 10608, Taiwan;
- High-Value Biomaterials Research and Commercialization Center (HBRCC), National Taipei University of Technology, Taipei 10608, Taiwan
| | - Yukio Nagasaki
- Department of Materials Science, Graduate School of Pure and Applied Sciences, University of Tsukuba, Tennoudai 1-1-1, Tsukuba 305-8573, Ibaraki, Japan; (B.S.); (H.T.T.)
- High-Value Biomaterials Research and Commercialization Center (HBRCC), National Taipei University of Technology, Taipei 10608, Taiwan
- Master’s School of Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tennoudai 1-1-1, Tsukuba 305-8573, Ibaraki, Japan
- Center for Research in Isotopes and Environmental Dynamics (CRiED), University of Tsukuba, Tennoudai 1-1-1, Tsukuba 305-8573, Ibaraki, Japan
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
7
|
Arzuk E. Investigation of the role of NLRP3 inflammasome activation in new-generation BCR-ABL1 tyrosine kinase inhibitors-induced hepatotoxicity. Toxicol Lett 2024; 400:71-80. [PMID: 39134127 DOI: 10.1016/j.toxlet.2024.08.007] [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: 05/27/2024] [Revised: 07/24/2024] [Accepted: 08/09/2024] [Indexed: 08/18/2024]
Abstract
New generation BCR-ABL1 TKIs raised attention regarding their adverse effects, including hepatotoxicity. Indeed, bosutinib and nilotinib were associated with severe hepatotoxicity compared with imatinib. Moreover, ponatinib has a boxed warning due to its potential to cause inflammatory liver damage, even death. However, the underlying mechanisms remain unclear. This study aimed to investigate the role of NLRP3 inflammasome activation in the underlying mechanism of ponatinib and bosutinib-induced hepatotoxicity. Furthermore, we determined the initiating event of this adverse outcome pathway by measuring the levels of reactive oxygen species as well as mitochondrial membrane potential in AML12 cells. The results demonstrated that ponatinib or bosutinib markedly inhibited cell viability and caused cytosolic membrane damage in cells. Moreover, drugs (IC50) dramatically induced oxidative stress and mitochondrial membrane potential disruption, which led to upregulation in the expression levels of NLRP3 inflammasome-related genes and proteins, activation of NLRP3 inflammasomes, cleavage of gasdermin-D and caspase-1, secretion of IL-1β, and cytosolic membrane damage. Furthermore, MCC950, a well-known specific inhibitor of NLRP3 inflammasome, and antioxidant N-acetyl-l-cysteine reversed the effects of drugs on the NLRP3 signaling pathway and cytosolic membranes. In summary, NLRP3 inflammasome activation is involved in new-generation BCR-ABL1 TKIs-triggered hepatotoxicity. Mitochondrial damage and reactive oxygen species accumulation were significant upstream signaling events in this signaling pathway.
Collapse
Affiliation(s)
- Ege Arzuk
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Ege University, İzmir, Turkey.
| |
Collapse
|
8
|
Hushmandi K, Einollahi B, Aow R, Suhairi SB, Klionsky DJ, Aref AR, Reiter RJ, Makvandi P, Rabiee N, Xu Y, Nabavi N, Saadat SH, Farahani N, Kumar AP. Investigating the interplay between mitophagy and diabetic neuropathy: Uncovering the hidden secrets of the disease pathology. Pharmacol Res 2024; 208:107394. [PMID: 39233055 PMCID: PMC11934918 DOI: 10.1016/j.phrs.2024.107394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 08/18/2024] [Accepted: 08/30/2024] [Indexed: 09/06/2024]
Abstract
Mitophagy, the cellular process of selectively eliminating damaged mitochondria, plays a crucial role in maintaining metabolic balance and preventing insulin resistance, both key factors in type 2 diabetes mellitus (T2DM) development. When mitophagy malfunctions in diabetic neuropathy, it triggers a cascade of metabolic disruptions, including reduced energy production, increased oxidative stress, and cell death, ultimately leading to various complications. Thus, targeting mitophagy to enhance the process may have emerged as a promising therapeutic strategy for T2DM and its complications. Notably, plant-derived compounds with β-cell protective and mitophagy-stimulating properties offer potential as novel therapeutic agents. This review highlights the intricate mechanisms linking mitophagy dysfunction to T2DM and its complications, particularly neuropathy, elucidating potential therapeutic interventions for this debilitating disease.
Collapse
Affiliation(s)
- Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Behzad Einollahi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Rachel Aow
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Suhana Binte Suhairi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Daniel J Klionsky
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Amir Reza Aref
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, Long School of Medicine, San Antonio, TX, USA
| | - Pooyan Makvandi
- Department of Biomaterials, Saveetha Dental College and Hospitals, SIMATS, Saveetha University, Chennai 600077, India; University Centre for Research & Development, Chandigarh University, Mohali, Punjab 140413, India
| | - Navid Rabiee
- Department of Biomaterials, Saveetha Dental College and Hospitals, SIMATS, Saveetha University, Chennai 600077, India
| | - Yi Xu
- Department of Science & Technology, Department of Urology, NanoBioMed Group, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, China
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia V8V 1P7, Canada
| | - Seyed Hassan Saadat
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
9
|
Mattii L, Moscato S, Ippolito C, Polizzi E, Novo G, Zucchi R, De Caterina R, Ghelardoni S, Madonna R. Empagliflozin mitigates ponatinib-induced cardiotoxicity by restoring the connexin 43-autophagy pathway. Biomed Pharmacother 2024; 178:117278. [PMID: 39116784 DOI: 10.1016/j.biopha.2024.117278] [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: 05/22/2024] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Empagliflozin (EMPA), a selective sodium-glucose cotransporter type 2 (SGLT2) inhibitor, has been shown to reduce major adverse cardiovascular events in patients with heart failure of different etiologies, although the underlying mechanism still remains unclear. Ponatinib (PON) is a multi-tyrosine kinase inhibitor successfully used against myeloid leukemia and other human malignancies, but its cardiotoxicity remains worrisome. Cardiac connexins (Cxs) are both substrates and regulators of autophagy and responsible for proper heart function. Alteration in connexin expression and localization have been described in patients with heart failure. AIMS To assess whether EMPA can mitigate PON-induced cardiac dysfunction by restoring the connexin 43-autophagy pathway. METHODS AND RESULTS Male C57BL/6 mice, randomized into four treatment groups (CNTRL, PON, EMPA, PON+EMPA) for 28 days, showed increased autophagy, decreased Cx43 expression as well as Cx43 lateralization, and attenuated systo-diastolic cardiac dysfunction after treatment with EMPA and PON compared with PON alone. Compared with CNTRL (DMSO), cardiomyocyte-differentiated H9c2 (dH9c2) cells treated with PON showed significantly reduced cell viability to approximately 20 %, decreased autophagy, increased cell senescence and reduced DNA binding activity of serum response factor (SRF) to serum response elements (SRE), which were paralleled by reduction in cardiac actin expression. Moreover, PON induced a significant increase of Cx43 protein and its S368-phosphorylated form (pS368-Cx43), as well as their displacement from the plasma membrane to the perinuclear and nuclear cellular region. All these effects were reverted by EMPA. CONCLUSION EMPA attenuates PON-induced cardiotoxicity by reducing senescence, enhancing the SRE-SRF binding and restoring the connexin 43-autophagy pathway. This effect may pave the way to use of SGLT2 inhibitors in attenuating tyrosine-kinase inhibitor cardiotoxicity.
Collapse
Affiliation(s)
- Letizia Mattii
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Stefania Moscato
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Chiara Ippolito
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Enza Polizzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Giuseppina Novo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Riccardo Zucchi
- Department of Pathology, Laboratory of Biochemistry, University of Pisa, Italy
| | | | - Sandra Ghelardoni
- Department of Pathology, Laboratory of Biochemistry, University of Pisa, Italy
| | - Rosalinda Madonna
- Department of Pathology, Cardiology Division, University of Pisa, Pisa, Italy.
| |
Collapse
|
10
|
Cheng M, Tao X, Wang F, Shen N, Xu Z, Hu Y, Huang P, Luo P, He Q, Zhang Y, Yan F. Underlying mechanisms and management strategies for regorafenib-induced toxicity in hepatocellular carcinoma. Expert Opin Drug Metab Toxicol 2024; 20:907-922. [PMID: 39225462 DOI: 10.1080/17425255.2024.2398628] [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: 04/22/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION Hepatocellular carcinoma (HCC) accounts for 85% of liver cancer cases and is the third leading cause of cancer death. Regorafenib is a multi-target inhibitor that dramatically prolongs progression-free survival in HCC patients who have failed sorafenib therapy. However, one of the primary factors limiting regorafenib's clinical utilization is toxicity. Using Clinical Trials.gov and PubMed, we gathered clinical data on regorafenib and conducted a extensive analysis of the medication's adverse reactions and mechanisms. Next, we suggested suitable management techniques to improve regorafenib's effectiveness. AREAS COVERED We have reviewed the mechanisms by which regorafenib-induced toxicity occurs and general management strategies through clinical trials of regorafenib. Furthermore, by examining the literature on regorafenib and other tyrosine kinase inhibition, we summarized the mechanics of the onset of regorafenib toxicity and mechanism-based intervention strategies by reviewing the literature related to regorafenib and other tyrosine kinase inhibition. EXPERT OPINION One of the primary factors restricting regorafenib's clinical utilization and combination therapy is its toxicity reactions. To optimize regorafenib treatment regimens, it is especially important to further understand the specific toxicity mechanisms of regorafenib as a multi-kinase inhibitor.
Collapse
Affiliation(s)
- Mengting Cheng
- College of Pharmaceutical Sciences, Center for Drug Safety Evaluation and Research of Zhejiang University, Hangzhou, China
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Xinyu Tao
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Fei Wang
- Outpatient Pharmacy, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Nonger Shen
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Zhifei Xu
- College of Pharmaceutical Sciences, Center for Drug Safety Evaluation and Research of Zhejiang University, Hangzhou, China
| | - Yuhuai Hu
- Department of Pharmacology and Toxicology, Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, China
| | - Ping Huang
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for malignant tumor, Hangzhou, Zhejiang, People's Republic of China
| | - Peihua Luo
- College of Pharmaceutical Sciences, Center for Drug Safety Evaluation and Research of Zhejiang University, Hangzhou, China
- Department of Pharmacology and Toxicology, Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, China
| | - Qiaojun He
- College of Pharmaceutical Sciences, Center for Drug Safety Evaluation and Research of Zhejiang University, Hangzhou, China
- Department of Pharmacology and Toxicology, Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, China
| | - Yiwen Zhang
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for malignant tumor, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
| | - Fangjie Yan
- Department of Pharmacology and Toxicology, Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, China
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
11
|
Wang Z, Jiang L, Lv X, Yin H, Wang Z, Li W, Liu Y. Higher risk of hepatotoxicity associated with cabozantinib in cancer patients. Crit Rev Oncol Hematol 2024; 196:104298. [PMID: 38364886 DOI: 10.1016/j.critrevonc.2024.104298] [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: 10/10/2023] [Revised: 02/05/2024] [Accepted: 02/12/2024] [Indexed: 02/18/2024] Open
Abstract
BACKGROUND The efficacy of cabozantinib has attracted interest in various solid tumors. The primary aim of this study was to evaluate the risk of hepatotoxicity associated with cabozantinib in the patients with cancer. METHODS PubMed, Cochrane, and EMBASE databases were searched for published randomized controlled trials (RCTs) from inception to September 9, 2023. The mainly outcomes were all-grade and grade ≥3 elevation of aspartate aminotransferase (AST) and alanine aminotransferase (ALT), expressed as relative risk (RR) and 95% confidence interval (CI). All data were pooled using fixed-effect or random-effects models according to the heterogeneity of the included RCTs. RESULTS Among the 922 records identified, 8 RCTs incorporating 2613 patients with cancer were included. For patients receiving cabozantinib, the relative risks of all-grade AST elevation (RR, 2.63; 95% CI, 2.16-3.20, P < 0.001), all-grade ALT elevation (RR, 2.89; 95% CI, 2.31-3.60, P < 0.001), grade ≥3 AST elevation (RR, 2.26; 95% CI, 1.34-3.83, P = 0.002), and grade ≥3 ALT elevation (RR, 3.40; 95% CI, 1.65-7.01, P < 0.001) were higher than those of patients who did not receive cabozantinib group. Further subgroup analysis showed that the relative risk of hepatotoxicity associated with cabozantinib was higher than that in the other TKIs (erlotinib, sunitinib, and sorafenib) and the non-TKI drug groups (everolimus, prednisone, mitoxantrone, and paclitaxel). CONCLUSIONS Compared with other solid tumor drugs, such as everolimus, sorafenib, sunitinib, paclitaxel, mitoxantrone-prednisone et al., cabozantinib has a higher risk of hepatotoxicity.
Collapse
Affiliation(s)
- Zhen Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Lili Jiang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Xin Lv
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Hang Yin
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Zhe Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Wenli Li
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Yong Liu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China.
| |
Collapse
|
12
|
Leung PY, Chen W, Sari AN, Sitaram P, Wu PK, Tsai S, Park JI. Erlotinib combination with a mitochondria-targeted ubiquinone effectively suppresses pancreatic cancer cell survival. World J Gastroenterol 2024; 30:714-727. [PMID: 38515951 PMCID: PMC10950623 DOI: 10.3748/wjg.v30.i7.714] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/13/2023] [Accepted: 01/17/2024] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND Pancreatic cancer is a leading cause of cancer-related deaths. Increased activity of the epidermal growth factor receptor (EGFR) is often observed in pancreatic cancer, and the small molecule EGFR inhibitor erlotinib has been approved for pancreatic cancer therapy by the food and drug administration. Nevertheless, erlotinib alone is ineffective and should be combined with other drugs to improve therapeutic outcomes. We previously showed that certain receptor tyrosine kinase inhibitors can increase mitochondrial membrane potential (Δψm), facilitate tumor cell uptake of Δψm-sensitive agents, disrupt mitochondrial homeostasis, and subsequently trigger tumor cell death. Erlotinib has not been tested for this effect. AIM To determine whether erlotinib can elevate Δψm and increase tumor cell uptake of Δψm-sensitive agents, subsequently triggering tumor cell death. METHODS Δψm-sensitive fluorescent dye was used to determine how erlotinib affects Δψm in pancreatic adenocarcinoma (PDAC) cell lines. The viability of conventional and patient-derived primary PDAC cell lines in 2D- and 3D cultures was measured after treating cells sequentially with erlotinib and mitochondria-targeted ubiquinone (MitoQ), a Δψm-sensitive MitoQ. The synergy between erlotinib and MitoQ was then analyzed using SynergyFinder 2.0. The preclinical efficacy of the two-drug combination was determined using immune-compromised nude mice bearing PDAC cell line xenografts. RESULTS Erlotinib elevated Δψm in PDAC cells, facilitating tumor cell uptake and mitochondrial enrichment of Δψm-sensitive agents. MitoQ triggered caspase-dependent apoptosis in PDAC cells in culture if used at high doses, while erlotinib pretreatment potentiated low doses of MitoQ. SynergyFinder suggested that these drugs synergistically induced tumor cell lethality. Consistent with in vitro data, erlotinib and MitoQ combination suppressed human PDAC cell line xenografts in mice more effectively than single treatments of each agent. CONCLUSION Our findings suggest that a combination of erlotinib and MitoQ has the potential to suppress pancreatic tumor cell viability effectively.
Collapse
Affiliation(s)
- Pui-Yin Leung
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Wenjing Chen
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Anissa N Sari
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Poojitha Sitaram
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Pui-Kei Wu
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Susan Tsai
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Jong-In Park
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| |
Collapse
|
13
|
Egidi MJ, Krug S, Haybaeck J, Michl P, Griesmann H. Anti-angiogenic therapy using the multi-tyrosine kinase inhibitor Regorafenib enhances tumor progression in a transgenic mouse model of ß-cell carcinogenesis. Br J Cancer 2023; 129:1225-1237. [PMID: 37620408 PMCID: PMC10575939 DOI: 10.1038/s41416-023-02389-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 07/12/2023] [Accepted: 07/31/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND Pancreatic neuroendocrine tumors (PNETs) represent a distinct hypervascularized tumor entity, often diagnosed at metastatic stage. Therapeutic efficacy of anti-angiogenic multi-kinase inhibitors is frequently limited by primary or acquired resistance in vivo. This study aimed to characterize the molecular mode of action as well as resistance mechanisms to the anti-angiogenic multi-tyrosine kinase inhibitor (TKI) Regorafenib in vitro and in vivo. METHODS In vitro, human and murine pancreatic neuroendocrine cell lines were comparatively treated with Regorafenib and other TKIs clinically used in PNETs. Effects on cell viability and proliferation were analyzed. In vivo, transgenic RIP1Tag2 mice were treated with Regorafenib at two different time periods during carcinogenesis and its impact on angiogenesis and tumor progression was evaluated. RESULTS Compared to the established TKI therapies with Sunitinib and Everolimus, Regorafenib showed the strongest effects on cell viability and proliferation in vitro, but was unable to induce apoptosis. Unexpectedly and in contrast to these in vitro findings, Regorafenib enhanced proliferation during early tumor development in RIP1Tag2 mice and had no significant effect in late tumor progression. In addition, invasiveness was increased at both time points. Mechanistically, we could identify an upregulation of the pro-survival protein Bcl-2, the induction of the COX2-PGE2-pathway as well as the infiltration of CSF1R positive immune cells into the tumors as potential resistance mechanisms following Regorafenib treatment. DISCUSSION Our data identify important tumor cell-autonomous and stroma-dependent mechanisms of resistance to antiangiogenic therapies.
Collapse
Affiliation(s)
- Maren Juliane Egidi
- Clinic for Internal Medicine I, Martin-Luther University Halle/Wittenberg, Ernst-Grube-Straße 40, D 06120, Halle, Germany
| | - Sebastian Krug
- Clinic for Internal Medicine I, Martin-Luther University Halle/Wittenberg, Ernst-Grube-Straße 40, D 06120, Halle, Germany
- Department of Internal Medicine IV, Heidelberg University Hospital, Heidelberg, Germany
| | - Johannes Haybaeck
- Department of Pathology, Neuropathology, and Molecular Pathology, Medical University of Innsbruck, Innsbruck, Austria
- Diagnostic & Research Center for Molecular Biomedicine, Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Patrick Michl
- Clinic for Internal Medicine I, Martin-Luther University Halle/Wittenberg, Ernst-Grube-Straße 40, D 06120, Halle, Germany.
- Department of Internal Medicine IV, Heidelberg University Hospital, Heidelberg, Germany.
| | - Heidi Griesmann
- Clinic for Internal Medicine I, Martin-Luther University Halle/Wittenberg, Ernst-Grube-Straße 40, D 06120, Halle, Germany
| |
Collapse
|
14
|
Sun S, Qin J, Liao W, Gao X, Shang Z, Luo D, Xiong S. Mitochondrial Dysfunction in Cardiotoxicity Induced by BCR-ABL1 Tyrosine Kinase Inhibitors -Underlying Mechanisms, Detection, Potential Therapies. Cardiovasc Toxicol 2023; 23:233-254. [PMID: 37479951 DOI: 10.1007/s12012-023-09800-x] [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: 05/03/2023] [Accepted: 07/08/2023] [Indexed: 07/23/2023]
Abstract
The advent of BCR-ABL tyrosine kinase inhibitors (TKIs) targeted therapy revolutionized the treatment of chronic myeloid leukemia (CML) patients. Mitochondria are the key organelles for the maintenance of myocardial tissue homeostasis. However, cardiotoxicity associated with BCR-ABL1 TKIs can directly or indirectly cause mitochondrial damage and dysfunction, playing a pivotal role in cardiomyocytes homeostatic system and putting the cancer survivors at higher risk. In this review, we summarize the cardiotoxicity caused by BCR-ABL1 TKIs and the underlying mechanisms, which contribute dominantly to the damage of mitochondrial structure and dysfunction: endoplasmic reticulum (ER) stress, mitochondrial stress, damage of myocardial cell mitochondrial respiratory chain, increased production of mitochondrial reactive oxygen species (ROS), and other kinases and other potential mechanisms of cardiotoxicity induced by BCR-ABL1 TKIs. Furthermore, detection and management of BCR-ABL1 TKIs will promote our rational use, and cardioprotection strategies based on mitochondria will improve our understanding of the cardiotoxicity from a mitochondrial perspective. Ultimately, we hope shed light on clinical decision-making. By integrate and learn from both research and practice, we will endeavor to minimize the mitochondria-mediated cardiotoxicity and reduce the adverse sequelae associated with BCR-ABL1 TKIs.
Collapse
Affiliation(s)
- Sheng Sun
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Medical Oncology, Hospital of Chengdu University of Traditioanal Chinese Medicine, Chengdu, 610075, Sichuan Province, China
| | - Jiqiu Qin
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wenhao Liao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiang Gao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhoubiao Shang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dehua Luo
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shaoquan Xiong
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
- Department of Medical Oncology, Hospital of Chengdu University of Traditioanal Chinese Medicine, Chengdu, 610075, Sichuan Province, China.
| |
Collapse
|
15
|
Yan H, Wu W, Hu Y, Li J, Xu J, Chen X, Xu Z, Yang X, Yang B, He Q, Luo P. Regorafenib inhibits EphA2 phosphorylation and leads to liver damage via the ERK/MDM2/p53 axis. Nat Commun 2023; 14:2756. [PMID: 37179400 PMCID: PMC10182995 DOI: 10.1038/s41467-023-38430-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
The hepatotoxicity of regorafenib is one of the most noteworthy concerns for patients, however the mechanism is poorly understood. Hence, there is a lack of effective intervention strategies. Here, by comparing the target with sorafenib, we show that regorafenib-induced liver injury is mainly due to its nontherapeutic target Eph receptor A2 (EphA2). EphA2 deficiency attenuated liver damage and cell apoptosis under regorafenib treatment in male mice. Mechanistically, regorafenib inhibits EphA2 Ser897 phosphorylation and reduces ubiquitination of p53 by altering the intracellular localization of mouse double minute 2 (MDM2) by affecting the extracellular signal-regulated kinase (ERK)/MDM2 axis. Meanwhile, we found that schisandrin C, which can upregulate the phosphorylation of EphA2 at Ser897 also has protective effect against the toxicity in vivo. Collectively, our findings identify the inhibition of EphA2 Ser897 phosphorylation as a key cause of regorafenib-induced hepatotoxicity, and chemical activation of EphA2 Ser897 represents a potential therapeutic strategy to prevent regorafenib-induced hepatotoxicity.
Collapse
Affiliation(s)
- Hao Yan
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Wentong Wu
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yuhuai Hu
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, 310018, China
- Laboratory of Fruit Quality Biology/Zhejiang Provincial Key Laboratory of Horticultural Plant Integrative Biology/The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement, Zhejiang University, Hangzhou, 310058, China
| | - Jinjin Li
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jiangxin Xu
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xueqin Chen
- Department of Oncology, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou, 310002, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, China
| | - Zhifei Xu
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xiaochun Yang
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Bo Yang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Qiaojun He
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, 310018, China
| | - Peihua Luo
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- Department of Pharmacology and Toxicology, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310018, China.
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China.
| |
Collapse
|
16
|
Bolten JS, Mancuso RV, Roos NJ, Mayr A, Puligilla RD, Kraus L, Odermatt A, Fricker G, Huwyler J. Nephrotoxicity of iopamidol is associated with mitochondrial impairment in human cell and teleost models. Toxicol Appl Pharmacol 2023; 466:116493. [PMID: 36977437 DOI: 10.1016/j.taap.2023.116493] [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: 02/09/2023] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023]
Abstract
Iopamidol is a nonionic, low-osmolar iodinated contrast agent used for angiography. Its clinical use is associated with renal dysfunction. Patients suffering from preexisting kidney disease have an increased risk of renal failure upon iopamidol administration. Studies in animals confirmed renal toxicity, but the involved mechanisms remain unclear. Therefore, the aim of the present study was to use human embryonic kidney cells (HEK293T) as a general cell model of mitochondrial damage, as well as, zebrafish larvae, and isolated proximal tubules of killifish to investigate factors promoting renal tubular toxicity of iopamidol with a focus on mitochondrial damage. Results from in vitro HEK293T cell-based assays indicate that iopamidol affects mitochondrial function Treatment with iopamidol induces ATP depletion, reduces the mitochondrial membrane potential, and elevates mitochondrial superoxide and reactive oxygen species accumulation. Similar results were obtained with gentamicin sulfate and cadmium chloride, two well-known model compounds associated with renal tubular toxicity. Confocal microscopy confirms changes in mitochondrial morphology, such as mitochondrial fission. Importantly, these results were confirmed in proximal renal tubular epithelial cells using ex vivo and in vivo teleost models. In conclusion, this study provides evidence for iopamidol-induced mitochondrial damage in proximal renal epithelial cells. Teleost models allow studying proximal tubular toxicity with translational relevance for humans.
Collapse
Affiliation(s)
- Jan Stephan Bolten
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Technology, University of Basel, 4056, Switzerland
| | - Riccardo Vincenzo Mancuso
- Department of Pharmaceutical Sciences, Division of Clinical Pharmacology & Toxicology, University of Basel, 4031, Switzerland; Department of Pharmaceutical Sciences, Division of Molecular Pharmacy, University of Basel, 4056, Switzerland
| | - Noëmi Johanna Roos
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Technology, University of Basel, 4056, Switzerland
| | - Aline Mayr
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Technology, University of Basel, 4056, Switzerland
| | - Ramya Deepthi Puligilla
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Technology, University of Basel, 4056, Switzerland
| | - Lisa Kraus
- Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, 69120, Germany
| | - Alex Odermatt
- Department of Pharmaceutical Sciences, Division of Molecular and Systems Toxicology, University of Basel, 4056, Switzerland
| | - Gert Fricker
- Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, 69120, Germany; Mount Desert Island Biological Laboratory, Salsbury Cove, ME 04672, USA
| | - Jörg Huwyler
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Technology, University of Basel, 4056, Switzerland; Mount Desert Island Biological Laboratory, Salsbury Cove, ME 04672, USA.
| |
Collapse
|
17
|
Pearson A, Haenni D, Bouitbir J, Hunt M, Payne BAI, Sachdeva A, Hung RKY, Post FA, Connolly J, Nlandu-Khodo S, Jankovic N, Bugarski M, Hall AM. Integration of High-Throughput Imaging and Multiparametric Metabolic Profiling Reveals a Mitochondrial Mechanism of Tenofovir Toxicity. FUNCTION (OXFORD, ENGLAND) 2022; 4:zqac065. [PMID: 36654930 PMCID: PMC9840465 DOI: 10.1093/function/zqac065] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/14/2022] [Accepted: 12/20/2022] [Indexed: 12/26/2022]
Abstract
Nephrotoxicity is a major cause of kidney disease and failure in drug development, but understanding of cellular mechanisms is limited, highlighting the need for better experimental models and methodological approaches. Most nephrotoxins damage the proximal tubule (PT), causing functional impairment of solute reabsorption and systemic metabolic complications. The antiviral drug tenofovir disoproxil fumarate (TDF) is an archetypal nephrotoxin, inducing mitochondrial abnormalities and urinary solute wasting, for reasons that were previously unclear. Here, we developed an automated, high-throughput imaging pipeline to screen the effects of TDF on solute transport and mitochondrial morphology in human-derived RPTEC/TERT1 cells, and leveraged this to generate realistic models of functional toxicity. By applying multiparametric metabolic profiling-including oxygen consumption measurements, metabolomics, and transcriptomics-we elucidated a highly robust molecular fingerprint of TDF exposure. Crucially, we identified that the active metabolite inhibits complex V (ATP synthase), and that TDF treatment causes rapid, dose-dependent loss of complex V activity and expression. Moreover, we found evidence of complex V suppression in kidney biopsies from humans with TDF toxicity. Thus, we demonstrate an effective and convenient experimental approach to screen for disease relevant functional defects in kidney cells in vitro, and reveal a new paradigm for understanding the pathogenesis of a substantial cause of nephrotoxicity.
Collapse
Affiliation(s)
- Adam Pearson
- Institute of Anatomy, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Dominik Haenni
- Center for Microscopy and Image Analysis, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Jamal Bouitbir
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland
| | - Matthew Hunt
- Wellcome Centre for Mitochondrial Research, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Brendan A I Payne
- Wellcome Centre for Mitochondrial Research, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK,Department of Infection and Tropical Medicine, Royal Victoria Infirmary, Queen Victoria Road, Newcastle upon Tyne NE1 4LP, UK
| | - Ashwin Sachdeva
- Genito-Urinary Cancer Research Group, Division of Cancer Sciences, University of Manchester, Manchester, M20 4GJ, UK,Department of Surgery, The Christie Hospital NHS Foundation Trust, 550 Wilmslow Road, Manchester M20 4BX, UK
| | - Rachel K Y Hung
- King’s College Hospital and School of Immunology & Microbial Sciences, King’s College London, London, SE5 8AF, UK
| | - Frank A Post
- King’s College Hospital and School of Immunology & Microbial Sciences, King’s College London, London, SE5 8AF, UK
| | - John Connolly
- UCL Centre for Nephrology, Royal Free Hospital, Rowland Hill Street, London NW3 2PF, UK
| | - Stellor Nlandu-Khodo
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Nevena Jankovic
- Institute of Anatomy, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Milica Bugarski
- Institute of Anatomy, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | | |
Collapse
|
18
|
Liang F, Zhang K, Ma W, Zhan H, Sun Q, Xie L, Zhao Z. Impaired autophagy and mitochondrial dynamics are involved in Sorafenib-induced cardiomyocyte apoptosis. Toxicology 2022; 481:153348. [DOI: 10.1016/j.tox.2022.153348] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/22/2022] [Accepted: 10/04/2022] [Indexed: 02/04/2023]
|
19
|
Influence of Antibiotics on Functionality and Viability of Liver Cells In Vitro. Curr Issues Mol Biol 2022; 44:4639-4657. [PMID: 36286032 PMCID: PMC9600611 DOI: 10.3390/cimb44100317] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 09/17/2022] [Accepted: 09/22/2022] [Indexed: 11/22/2022] Open
Abstract
(1) Antibiotics are an important weapon in the fight against serious bacterial infections and are considered a common cause of drug-induced liver injury (DILI). The hepatotoxicity of many drugs, including antibiotics, is poorly analyzed in human in vitro models. (2) A standardized assay with a human hepatoma cell line was used to test the hepatotoxicity of various concentrations (Cmax, 5× Cmax, and 10× Cmax) of antibiotics. In an ICU, the most frequently prescribed antibiotics, ampicillin, cefepime, cefuroxime, levofloxacin, linezolid, meropenem, rifampicin, tigecycline, and vancomycin, were incubated with HepG2/C3A cells for 6 days. Cell viability (XTT assay, LDH release, and vitality), albumin synthesis, and cytochrome 1A2 activity were determined in cells. (3) In vitro, vancomycin, rifampicin, and tigecycline showed moderate hepatotoxic potential. The antibiotics ampicillin, cefepime, cefuroxime, levofloxacin, linezolid, and meropenem were associated with mild hepatotoxic reactions in test cells incubated with the testes Cmax concentration. Rifampicin and cefuroxime showed significantly negative effects on the viability of test cells. (4) Further in vitro studies and global pharmacovigilance reports should be conducted to reveal underlying mechanism of the hepatotoxic action of vancomycin, rifampicin, tigecycline, and cefuroxime, as well as the clinical relevance of these findings.
Collapse
|
20
|
Huang W, Liu WY, Chen LY, Ni L, Zou XX, Ye M, Zhang ZY, Zou SQ. Flavonoid and chromone-rich extract from Euscaphis Konishii Hayata leaf attenuated alcoholic liver injury in mice. JOURNAL OF ETHNOPHARMACOLOGY 2022; 295:115455. [PMID: 35697192 DOI: 10.1016/j.jep.2022.115455] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 06/06/2022] [Accepted: 06/08/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Euscaphis konishii Hayata is a traditional medicinal plant in China, and its leaves are usually used to make dishes for hepatic or gastrointestinal issues by Chinese She nationality. Pharmacological analysis showed that E. konishii leaves contain high levels of flavonoids and chromones with favorable anti-hepatoma effect. AIM OF THE STUDY The extract from E. konishii leaves was detected to evaluate its chemical composition, and the alcoholic liver injury mice model was adopted to elucidate its hepatoprotective effects. MATERIALS AND METHODS The total leaf extract from E. konishii was separated by polyamide column to get the flavonoid and chromone-rich extract (FCE). Single compounds from FCE was purified by gel and Sephadex LH-20 chromatography and analyzed by nuclear magnetic resonance (NMR). The chemical component of FCE was confirmed and quantified by HPLC-MS. The OH·, O2-, DPPH and ABTS + free radical assays were adopted to estimate the antioxidant activity of FCE in vitro. The alcohol-fed model mice were established to assess the hepatoprotective capacity of FCE in vivo, through biochemical determination, histopathological analysis, mitochondrial function measurement, quantitative Real-Time Polymerase Chain Reaction (qRT-PCR) detection and Western blot determination. RESULTS 8 flavonoids and 2 chromones were recognized in the FCEextract by both NMR and HPLC-MS. FCE represented strong free radicals scavenging activity in vitro. With oral administration, FCE (50, 100 and 200 mg/kg) dose-dependently decreased the serum levels of alanine aminotransferase (ALT), alkaline phosphatase (ALP) and aspartate aminotransferase (AST) in alcohol-fed mice. FCE gradually reduced the malondialdehyde (MDA) content, increased the activity of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) in the alcohol-treated liver tissues. FCE also alleviated the hepatic inflammation, inhibited the hepatocyte apoptosis and lessened the alcohol-induced histological alteration and lipid accumulation in the liver tissues. FCE administration inhibited the overexpression of endoplasmic reticulum (ER) chaperones signaling and unfolded protein response (UPR) pathways to defense the ER-induced apoptosis. Pretreatment with FCE also restored the mitochondrial membrane potentials andadenosine triphosphate (ATP) levels, which in turn suppressed the Cytochrome C release and mitochondria-induced apoptosis. CONCLUSIONS FCE conferred great protection against alcoholic liver injury, which might be associated with its viability through suppressing reactive oxygen species (ROS) stress and hepatocyte apoptosis.
Collapse
Affiliation(s)
- Wei Huang
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China; Engineering Research Institute of Conservation, Utilization of Natural Bioresources, Fujian Agriculture and Forestry University, Fuzhou, 350002, China; College of Agriculture, Fujian Agriculture and Forestry University, Fuzhou, 350002, China.
| | - Wan-Yi Liu
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Lu-Yao Chen
- Engineering Research Institute of Conservation, Utilization of Natural Bioresources, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Lin Ni
- Engineering Research Institute of Conservation, Utilization of Natural Bioresources, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Xiao-Xing Zou
- Engineering Research Institute of Conservation, Utilization of Natural Bioresources, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Min Ye
- Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350004, China
| | - Zhong-Yi Zhang
- College of Agriculture, Fujian Agriculture and Forestry University, Fuzhou, 350002, China.
| | - Shuang-Quan Zou
- Engineering Research Institute of Conservation, Utilization of Natural Bioresources, Fujian Agriculture and Forestry University, Fuzhou, 350002, China.
| |
Collapse
|
21
|
Biological Role of Pazopanib and Sunitinib Aldehyde Derivatives in Drug-Induced Liver Injury. Metabolites 2022; 12:metabo12090852. [PMID: 36144257 PMCID: PMC9505977 DOI: 10.3390/metabo12090852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/02/2022] [Accepted: 09/09/2022] [Indexed: 11/17/2022] Open
Abstract
Tyrosine kinase inhibitors pazopanib and sunitinib are both used to treat advanced renal cell carcinoma but expose patients to an increased risk of hepatotoxicity. We have previously identified two aldehyde derivatives for pazopanib and sunitinib (P-CHO and S-CHO, respectively) in liver microsomes. In this study, we aimed to decipher their role in hepatotoxicity by treating HepG2 and HepaRG hepatic cell lines with these derivatives and evaluating cell viability, mitochondrial dysfunction, and oxidative stress accumulation. Additionally, plasma concentrations of P-CHO were assessed in a cohort of patients treated with pazopanib. Results showed that S-CHO slightly decreased the viability of HepG2, but to a lesser extent than sunitinib, and affected the maximal respiratory capacity of the mitochondrial chain. P-CHO decreased viability and ATP production in HepG2. Traces of P-CHO were detected in the plasma of patients treated with pazopanib. Overall, these results showed that P-CHO and S-CHO affect hepatocyte integrity and could be involved in the pazopanib and sunitinib hepatotoxicity.
Collapse
|
22
|
Assessing Drug-Induced Mitochondrial Toxicity in Cardiomyocytes: Implications for Preclinical Cardiac Safety Evaluation. Pharmaceutics 2022; 14:pharmaceutics14071313. [PMID: 35890211 PMCID: PMC9319223 DOI: 10.3390/pharmaceutics14071313] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/09/2022] [Accepted: 06/14/2022] [Indexed: 02/07/2023] Open
Abstract
Drug-induced cardiotoxicity not only leads to the attrition of drugs during development, but also contributes to the high morbidity and mortality rates of cardiovascular diseases. Comprehensive testing for proarrhythmic risks of drugs has been applied in preclinical cardiac safety assessment for over 15 years. However, other mechanisms of cardiac toxicity have not received such attention. Of them, mitochondrial impairment is a common form of cardiotoxicity and is known to account for over half of cardiovascular adverse-event-related black box warnings imposed by the U.S. Food and Drug Administration. Although it has been studied in great depth, mitochondrial toxicity assessment has not yet been incorporated into routine safety tests for cardiotoxicity at the preclinical stage. This review discusses the main characteristics of mitochondria in cardiomyocytes, drug-induced mitochondrial toxicities, and high-throughput screening strategies for cardiomyocytes, as well as their proposed integration into preclinical safety pharmacology. We emphasize the advantages of using adult human primary cardiomyocytes for the evaluation of mitochondrial morphology and function, and the need for a novel cardiac safety testing platform integrating mitochondrial toxicity and proarrhythmic risk assessments in cardiac safety evaluation.
Collapse
|
23
|
Krstic J, Schindlmaier K, Prokesch A. Combination strategies to target metabolic flexibility in cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 373:159-197. [PMID: 36283766 DOI: 10.1016/bs.ircmb.2022.03.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Therapeutically interfering with metabolic pathways has great merit to curtail tumor growth because the demand for copious amounts of energy for growth-supporting biomass production is common to all cancer entities. A major impediment to a straight implementation of metabolic cancer therapy is the metabolic flexibility and plasticity of cancer cells (and their microenvironment) resulting in therapy resistance and evasion. Metabolic combination therapies, therefore, are promising as they are designed to target several energetic routes simultaneously and thereby diminish the availability of alternative substrates. Thus, dietary restrictions, specific nutrient limitations, and/or pharmacological interventions impinging on metabolic pathways can be combined to improve cancer treatment efficacy, to overcome therapy resistance, or even act as a preventive measure. Here, we review the most recent developments in metabolic combination therapies particularly highlighting in vivo reports of synergistic effects and available clinical data. We close with identifying the challenges of the field (metabolic tumor heterogeneity, immune cell interactions, inter-patient variabilities) and suggest a "metabo-typing" strategy to tailor evidence-based metabolic combination therapies to the energetic requirements of the tumors and the patient's nutritional habits and status.
Collapse
Affiliation(s)
- Jelena Krstic
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism & Aging, Medical University of Graz, Graz, Austria
| | - Katharina Schindlmaier
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Andreas Prokesch
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism & Aging, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
24
|
Zhang S, Wang Y, Cao Y, Wu J, Zhang Z, Ren H, Xu X, Kaznacheyeva E, Li Q, Wang G. Inhibition of the PINK1-Parkin Pathway Enhances the Lethality of Sorafenib and Regorafenib in Hepatocellular Carcinoma. Front Pharmacol 2022; 13:851832. [PMID: 35370635 PMCID: PMC8967359 DOI: 10.3389/fphar.2022.851832] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/28/2022] [Indexed: 12/30/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common fatal malignancies and the main cause of cancer-related deaths. The multitarget tyrosine kinase inhibitors (TKIs) sorafenib and regorafenib are systemic therapeutic drugs approved for the treatment of HCC. Here, we found that sorafenib and regorafenib injured mitochondria by inducing mitochondrial Ca2+ (mtCa2+) overload and mitochondrial permeability transition pore (mPTP) opening, resulting in mitochondria-mediated cell death, which was alleviated by cyclosporin A (CsA), an inhibitor of mPTP. Meanwhile, mPTP opening caused PINK1 accumulation on damaged mitochondria, which recruited Parkin to mitochondria to induce mitophagy. Inhibition of autophagy by the lysosomal inhibitor chloroquine (CQ) or inhibition of mitochondrial fission by mdivi-1 aggravated sorafenib- and regorafenib-induced cell death. Moreover, knockdown of PINK1 also promotes sorafenib- and regorafenib-induced cell death. An in vivo study showed that sorafenib and regorafenib inhibited HepG2 cell growth more effectively in PINK1 knockdown cells than in shNTC cells in null mice. Thus, our data demonstrate that PINK1-Parkin-mediated mitophagy alleviates sorafenib and regorafenib antitumor effects in vitro and in vivo.
Collapse
Affiliation(s)
- Shun Zhang
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Yixin Wang
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Yifan Cao
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Jin Wu
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Zubin Zhang
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Haigang Ren
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Xiaohui Xu
- Department of General Surgery, the First People's Hospital of Taicang, Taicang Affiliated Hospital of Soochow University, Suzhou, China
| | | | - Qing Li
- Department of Gastroenterology, the First People's Hospital of Taicang, Taicang Affiliated Hospital of Soochow University, Suzhou, China
| | - Guanghui Wang
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, China.,Center of Translational Medicine, the First People's Hospital of Taicang, Taicang Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
25
|
Tran HT, Vong LB, Nishikawa Y, Nagasaki Y. Sorafenib-loaded silica-containing redox nanoparticles for oral anti-liver fibrosis therapy. J Control Release 2022; 345:880-891. [PMID: 35395328 DOI: 10.1016/j.jconrel.2022.04.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 04/01/2022] [Accepted: 04/02/2022] [Indexed: 11/17/2022]
Abstract
Liver fibrosis is a chronic disease resulting from repetitive or prolonged liver injury with limited treatment options. Sorafenib has been reported to be a potential antifibrotic agent; however, its therapeutic effect is restricted because of its low bioavailability and severe adverse effects in the gastrointestinal (GI) tract. In this study, we developed sorafenib-loaded silica-containing redox nanoparticles (sora@siRNP) as an oral nanomedicine to treat liver fibrosis. The designed siRNP were prepared by self-assembly of amphiphilic block copolymers, which possess antioxidant nitroxide radicals as a side chain of the hydrophobic segment and porous silica particles in the nanoparticle core. The silica moieties in the core formed a crosslink between the self-assembling block copolymers to afford stable drug absorption, which could be useful in harsh GI conditions after oral drug administration. Based on in vitro evaluation, sora@siRNP exerted antiproliferative and antifibrotic effects against hepatic stellate cells (HSCs) and low toxicity against normal endothelial cells. A pharmacokinetic study showed that siRNP significantly improved the bioavailability and distribution of sorafenib in the liver. In an in vivo study using a mouse model of CCl4-induced liver fibrosis, oral administration of sora@siRNP significantly suppressed the fibrotic area in comparison to free sorafenib administration. In mice with CCl4-induced fibrosis, free sorafenib administration did not suppress the expression of α-smooth muscle actin; however, mice treated with sora@siRNP showed significantly suppressed expression of α-smooth muscle actin, indicating the inhibition of HSC activation, which was confirmed by in vitro experiments. Moreover, oral administration of free sorafenib induced severe intestinal damage and increased leakage into the gut, which can be attributed to the generation of reactive oxygen species (ROS). Our antioxidant nanocarriers, siRNP, reduced the adverse effects of local ROS scavenging in the GI tract. Our results suggest that sora@siRNP could serve as a promising oral nanomedicine for liver fibrosis.
Collapse
Affiliation(s)
- Hao Thi Tran
- Department of Materials Science, Graduate School of Pure and Applied Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8573, Japan
| | - Long Binh Vong
- Department of Materials Science, Graduate School of Pure and Applied Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8573, Japan; School of Biomedical Engineering, International University, Ho Chi Minh 703000, Viet Nam; Vietnam National University Ho Chi Minh City (VNU-HCM), Ho Chi Minh 703000, Viet Nam
| | - Yuji Nishikawa
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa 079-8501, Japan
| | - Yukio Nagasaki
- Department of Materials Science, Graduate School of Pure and Applied Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8573, Japan; Department of Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8577, Japan; Center for Research in Isotopes and Environmental Dynamics (CRiED), University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki, 305-8573, Japan.
| |
Collapse
|
26
|
Roos NJ, Mancuso RV, Sanvee GM, Bouitbir J, Krähenbühl S. Imatinib disturbs lysosomal function and morphology and impairs the activity of mTORC1 in human hepatocyte cell lines. Food Chem Toxicol 2022; 162:112869. [PMID: 35182693 DOI: 10.1016/j.fct.2022.112869] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 12/20/2021] [Accepted: 02/12/2022] [Indexed: 10/19/2022]
Abstract
The tyrosine kinase inhibitors (TKIs) imatinib and lapatinib are associated with severe hepatotoxicity, whose mechanisms are currently under investigation. As amphiphilic drugs, imatinib and lapatinib enrich in lysosomes. In the present study, we investigated their effects on lysosomal morphology and function in HepG2 and HuH-7 cells and explored possible links between lysosomal dysfunction and hepatotoxicity. Both TKIs increased the lysosomal volume time and concentration-dependently in HepG2 and HuH-7 cells. In HepG2 cells, lapatinib and imatinib raised the lysosomal pH and destabilized the lysosomal membrane, thereby impairing lysosomal proteolytic activity such as cathepsin B processing. Imatinib activated the transcription factor EB (TFEB), a regulator of lysosomal biogenesis and function, as demonstrated by nuclear TFEB accumulation and increased expression of TFEB-target genes. Because of lysosomal dysfunction, imatinib impaired mTORC1 activation, a protein complex activated on the lysosomal surface, which explained TFEB activation. HepG2 cells treated with imatinib showed increased levels of MAP1LC3A/B-II and of ATG13 (S318) phosphorylation, indicating induction of autophagy due to TFEB activation. Finally, imatinib induced apoptosis in HepG2 cells in a time and concentration-dependent manner, explained by lysosomal and mitochondrial toxicity. Our findings provide a new lysosome-centered mechanism for imatinib-induced hepatotoxicity that could be extended to other lysosomotropic drugs.
Collapse
Affiliation(s)
- Noëmi Johanna Roos
- Division of Clinical Pharmacology & Toxicology, University Hospital of Basel, Switzerland; Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Switzerland
| | - Riccardo Vincenzo Mancuso
- Division of Clinical Pharmacology & Toxicology, University Hospital of Basel, Switzerland; Division of Molecular Pharmacy, Department of Pharmaceutical Sciences, University of Basel, Switzerland
| | - Gerda Mawududzi Sanvee
- Division of Clinical Pharmacology & Toxicology, University Hospital of Basel, Switzerland; Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Switzerland
| | - Jamal Bouitbir
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Switzerland
| | - Stephan Krähenbühl
- Division of Clinical Pharmacology & Toxicology, University Hospital of Basel, Switzerland.
| |
Collapse
|
27
|
Milton CI, Selfe J, Aladowicz E, Man SYK, Bernauer C, Missiaglia E, Walters ZS, Gatz SA, Kelsey A, Generali M, Box G, Valenti M, de Haven‐Brandon A, Galiwango D, Hayes A, Clarke M, Izquierdo E, Gonzalez De Castro D, Raynaud FI, Kirkin V, Shipley JM. FGF7-FGFR2 autocrine signaling increases growth and chemoresistance of fusion-positive rhabdomyosarcomas. Mol Oncol 2022; 16:1272-1289. [PMID: 34850536 PMCID: PMC8936514 DOI: 10.1002/1878-0261.13145] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 09/30/2021] [Accepted: 11/29/2021] [Indexed: 11/09/2022] Open
Abstract
Rhabdomyosarcomas are aggressive pediatric soft-tissue sarcomas and include high-risk PAX3-FOXO1 fusion-gene-positive cases. Fibroblast growth factor receptor 4 (FGFR4) is known to contribute to rhabdomyosarcoma progression; here, we sought to investigate the involvement and potential for therapeutic targeting of other FGFRs in this disease. Cell-based screening of FGFR inhibitors with potential for clinical repurposing (NVP-BGJ398, nintedanib, dovitinib, and ponatinib) revealed greater sensitivity of fusion-gene-positive versus fusion-gene-negative rhabdomyosarcoma cell lines and was shown to be correlated with high expression of FGFR2 and its specific ligand, FGF7. Furthermore, patient samples exhibit higher mRNA levels of FGFR2 and FGF7 in fusion-gene-positive versus fusion-gene-negative rhabdomyosarcomas. Sustained intracellular mitogen-activated protein kinase (MAPK) activity and FGF7 secretion into culture media during serum starvation of PAX3-FOXO1 rhabdomyosarcoma cells together with decreased cell viability after genetic silencing of FGFR2 or FGF7 was in keeping with a novel FGF7-FGFR2 autocrine loop. FGFR inhibition with NVP-BGJ398 reduced viability and was synergistic with SN38, the active metabolite of irinotecan. In vivo, NVP-BGJ398 abrogated xenograft growth and warrants further investigation in combination with irinotecan as a therapeutic strategy for fusion-gene-positive rhabdomyosarcomas.
Collapse
Affiliation(s)
- Christopher I. Milton
- Sarcoma Molecular Pathology TeamDivisions of Molecular Pathology and Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
- Present address:
Signal Transduction and Molecular Pharmacology TeamCancer Research UK Cancer Therapeutics UnitThe Institute of Cancer ResearchSuttonUK
| | - Joanna Selfe
- Sarcoma Molecular Pathology TeamDivisions of Molecular Pathology and Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Ewa Aladowicz
- Sarcoma Molecular Pathology TeamDivisions of Molecular Pathology and Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Stella Y. K. Man
- Sarcoma Molecular Pathology TeamDivisions of Molecular Pathology and Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Carolina Bernauer
- Sarcoma Molecular Pathology TeamDivisions of Molecular Pathology and Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Edoardo Missiaglia
- Sarcoma Molecular Pathology TeamDivisions of Molecular Pathology and Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
- Present address:
Department of Molecular PathologyCentre Hospitalier Universitaire VaudoisLausanneSwitzerland
| | - Zoë S. Walters
- Sarcoma Molecular Pathology TeamDivisions of Molecular Pathology and Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
- Present address:
Translational Epigenomics TeamHuman Development and HealthFaculty of MedicineSouthampton General HospitalUK
| | - Susanne A. Gatz
- Sarcoma Molecular Pathology TeamDivisions of Molecular Pathology and Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
- Present address:
Institute of Cancer and Genomic SciencesUniversity of BirminghamUK
| | - Anna Kelsey
- Department of Paediatric HistopathologyManchester University NHS Foundation TrustRoyal Manchester Children’s HospitalUK
| | - Melanie Generali
- Sarcoma Molecular Pathology TeamDivisions of Molecular Pathology and Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
- Present address:
Center for Therapy Development and Good Manufacturing PracticeInstitute for Regenerative Medicine (IREM)University of ZurichSwitzerland
| | - Gary Box
- Cancer Pharmacology and Stress Response TeamDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Melanie Valenti
- Cancer Pharmacology and Stress Response TeamDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Alexis de Haven‐Brandon
- Cancer Pharmacology and Stress Response TeamDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - David Galiwango
- Drug Metabolism and Pharmacokinetics TeamDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Angela Hayes
- Drug Metabolism and Pharmacokinetics TeamDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Matthew Clarke
- Glioma TeamDivision of Molecular PathologyThe Institute of Cancer ResearchLondonUK
| | - Elisa Izquierdo
- Glioma TeamDivision of Molecular PathologyThe Institute of Cancer ResearchLondonUK
| | - David Gonzalez De Castro
- Molecular HaematologyDivision of Molecular PathologyThe Institute of Cancer ResearchLondonUK
- Present address:
School of MedicineDentistry and Biomedical sciencesQueens University BelfastUK
| | - Florence I. Raynaud
- Drug Metabolism and Pharmacokinetics TeamDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Vladimir Kirkin
- Cancer Pharmacology and Stress Response TeamDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Janet M. Shipley
- Sarcoma Molecular Pathology TeamDivisions of Molecular Pathology and Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| |
Collapse
|
28
|
Contreras L, Rodríguez-Gil A, Muntané J, de la Cruz J. Broad Transcriptomic Impact of Sorafenib and Its Relation to the Antitumoral Properties in Liver Cancer Cells. Cancers (Basel) 2022; 14:cancers14051204. [PMID: 35267509 PMCID: PMC8909169 DOI: 10.3390/cancers14051204] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 02/21/2022] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Hepatocellular carcinoma (HCC) is the fourth most frequent cause of cancer-related mortality worldwide. While ablation, resection and orthotopic liver transplantation are indicated at an early stage of the disease, Sorafenib (Sfb) is the current most administrated first-line treatment for advanced HCC, even though its therapeutic benefit is limited due to the appearance of resistance. Deep knowledge on the molecular consequences of Sfb-treatment is essentially required for optimizing novel therapeutic strategies to improve the outcomes for patients with advanced HCC. In this study, we analyzed differential gene expression changes in two well characterized liver cancer cell lines upon a Sfb-treatment, demonstrating that both lines responded similarly to the treatment. Our results provide valuable information on the molecular action of Sfb on diverse cellular fundamental processes such as DNA repair, translation and proteostasis and identify rationalization issues that could provide a different therapeutic perspective to Sfb. Abstract Hepatocellular carcinoma (HCC) is one of the most frequent and essentially incurable cancers in its advanced stages. The tyrosine kinase inhibitor Sorafenib (Sfb) remains the globally accepted treatment for advanced HCC. However, the extent of its therapeutic benefit is limited. Sfb exerts antitumor activity through its cytotoxic, anti-proliferative and pro-apoptotic roles in HCC cells. To better understand the molecular mechanisms underlying these effects, we used RNA sequencing to generate comprehensive transcriptome profiles of HepG2 and SNU423, hepatoblastoma- (HB) and HCC-derived cell lines, respectively, following a Sfb treatment at a pharmacological dose. This resulted in similar alterations of gene expression in both cell lines. Genes functionally related to membrane trafficking, stress-responsible and unfolded protein responses, circadian clock and activation of apoptosis were predominantly upregulated, while genes involved in cell growth and cycle, DNA replication and repair, ribosome biogenesis, translation initiation and proteostasis were downregulated. Our results suggest that Sfb causes primary effects on cellular stress that lead to upregulation of selective responses to compensate for its negative effect and restore homeostasis. No significant differences were found specifically affecting each cell line, indicating the robustness of the Sfb mechanism of action despite the heterogeneity of liver cancer. We discuss our results on terms of providing rationalization for possible strategies to improve Sfb clinical outcomes.
Collapse
Affiliation(s)
- Laura Contreras
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, E-41013 Seville, Spain; (L.C.); (A.R.-G.)
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, E-41012 Seville, Spain
| | - Alfonso Rodríguez-Gil
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, E-41013 Seville, Spain; (L.C.); (A.R.-G.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), E-28029 Madrid, Spain
- Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, E-41009 Sevilla, Spain
| | - Jordi Muntané
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, E-41013 Seville, Spain; (L.C.); (A.R.-G.)
- Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, E-41009 Sevilla, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), E-28029 Madrid, Spain
- Correspondence: (J.M.); (J.d.l.C.); Tel.: +34-955-923-122 (J.M.); +34-923-126 (J.d.l.C.)
| | - Jesús de la Cruz
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, E-41013 Seville, Spain; (L.C.); (A.R.-G.)
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, E-41012 Seville, Spain
- Correspondence: (J.M.); (J.d.l.C.); Tel.: +34-955-923-122 (J.M.); +34-923-126 (J.d.l.C.)
| |
Collapse
|
29
|
Bouitbir J, Panajatovic MV, Krähenbühl S. Mitochondrial Toxicity Associated with Imatinib and Sorafenib in Isolated Rat Heart Fibers and the Cardiomyoblast H9c2 Cell Line. Int J Mol Sci 2022; 23:ijms23042282. [PMID: 35216404 PMCID: PMC8878993 DOI: 10.3390/ijms23042282] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 02/01/2023] Open
Abstract
Tyrosine kinase inhibitors (TKIs) are associated with cardiac toxicity, which may be caused by mitochondrial toxicity. The underlying mechanisms are currently unclear and require further investigation. In the present study, we aimed to investigate in more detail the role of the enzyme complexes of the electron transfer system (ETS), mitochondrial oxidative stress, and mechanisms of cell death in cardiac toxicity associated with imatinib and sorafenib. Cardiac myoblast H9c2 cells were exposed to imatinib and sorafenib (1 to 100 µM) for 24 h. Permeabilized rat cardiac fibers were treated with both drugs for 15 min. H9c2 cells exposed to sorafenib for 24 h showed a higher membrane toxicity and ATP depletion in the presence of galactose (favoring mitochondrial metabolism) compared to glucose (favoring glycolysis) but not when exposed to imatinib. Both TKIs resulted in a higher dissipation of the mitochondrial membrane potential in galactose compared to glucose media. Imatinib inhibited Complex I (CI)- and CIII- linked respiration under both conditions. Sorafenib impaired CI-, CII-, and CIII-linked respiration in H9c2 cells cultured with glucose, whereas it inhibited all ETS complexes with galactose. In permeabilized rat cardiac myofibers, acute exposure to imatinib and sorafenib decreased CI- and CIV-linked respiration in the presence of the drugs. Electron microscopy showed enlarged mitochondria with disorganized cristae. In addition, both TKIs caused mitochondrial superoxide accumulation and decreased the cellular GSH pool. Both TKIs induced caspase 3/7 activation, suggesting apoptosis as a mechanism of cell death. Imatinib and sorafenib impaired the function of cardiac mitochondria in isolated rat cardiac fibers and in H9c2 cells at plasma concentrations reached in humans. Both imatinib and sorafenib impaired the function of enzyme complexes of the ETS, which was associated with mitochondrial ROS accumulation and cell death by apoptosis.
Collapse
Affiliation(s)
- Jamal Bouitbir
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
- Division of Clinical Pharmacology & Toxicology, University Hospital, Basel, Switzerland; (M.V.P.); (S.K.)
- Correspondence: ; Tel.: +41-61-207-6290
| | - Miljenko V. Panajatovic
- Division of Clinical Pharmacology & Toxicology, University Hospital, Basel, Switzerland; (M.V.P.); (S.K.)
| | - Stephan Krähenbühl
- Division of Clinical Pharmacology & Toxicology, University Hospital, Basel, Switzerland; (M.V.P.); (S.K.)
| |
Collapse
|
30
|
Krstic J, Reinisch I, Schindlmaier K, Galhuber M, Riahi Z, Berger N, Kupper N, Moyschewitz E, Auer M, Michenthaler H, Nössing C, Depaoli MR, Ramadani-Muja J, Usluer S, Stryeck S, Pichler M, Rinner B, Deutsch AJA, Reinisch A, Madl T, Chiozzi RZ, Heck AJR, Huch M, Malli R, Prokesch A. Fasting improves therapeutic response in hepatocellular carcinoma through p53-dependent metabolic synergism. SCIENCE ADVANCES 2022; 8:eabh2635. [PMID: 35061544 PMCID: PMC8782451 DOI: 10.1126/sciadv.abh2635] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 11/29/2021] [Indexed: 05/03/2023]
Abstract
Cancer cells voraciously consume nutrients to support their growth, exposing metabolic vulnerabilities that can be therapeutically exploited. Here, we show in hepatocellular carcinoma (HCC) cells, xenografts, and patient-derived organoids that fasting improves sorafenib efficacy and acts synergistically to sensitize sorafenib-resistant HCC. Mechanistically, sorafenib acts noncanonically as an inhibitor of mitochondrial respiration, causing resistant cells to depend on glycolysis for survival. Fasting, through reduction in glucose and impeded AKT/mTOR signaling, prevents this Warburg shift. Regulating glucose transporter and proapoptotic protein expression, p53 is necessary and sufficient for the sorafenib-sensitizing effect of fasting. p53 is also crucial for fasting-mediated improvement of sorafenib efficacy in an orthotopic HCC mouse model. Together, our data suggest fasting and sorafenib as rational combination therapy for HCC with intact p53 signaling. As HCC therapy is currently severely limited by resistance, these results should instigate clinical studies aimed at improving therapy response in advanced-stage HCC.
Collapse
Affiliation(s)
- Jelena Krstic
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, 8010 Graz, Austria
| | - Isabel Reinisch
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, 8010 Graz, Austria
| | - Katharina Schindlmaier
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, 8010 Graz, Austria
| | - Markus Galhuber
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, 8010 Graz, Austria
| | - Zina Riahi
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, 8010 Graz, Austria
| | - Natascha Berger
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, 8010 Graz, Austria
- Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria
| | - Nadja Kupper
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, 8010 Graz, Austria
| | - Elisabeth Moyschewitz
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, 8010 Graz, Austria
| | - Martina Auer
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, 8010 Graz, Austria
| | - Helene Michenthaler
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, 8010 Graz, Austria
| | - Christoph Nössing
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, 8010 Graz, Austria
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, UK
| | - Maria R. Depaoli
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
| | - Jeta Ramadani-Muja
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
| | - Sinem Usluer
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
| | - Sarah Stryeck
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
- Institute of Interactive Systems and Data Science, Graz University of Technology, 8010 Graz, Austria
- Know-Center GmbH, 8010 Graz, Austria
| | - Martin Pichler
- Division of Clinical Oncology, Department of Medicine, Comprehensive Cancer Center Graz, Medical University of Graz, 8036 Graz, Austria
| | - Beate Rinner
- Department for Biomedical Research, Medical University of Graz, Graz, Austria
| | - Alexander J. A. Deutsch
- Division of Hematology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Andreas Reinisch
- Division of Hematology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
- Division of Hematology, Department of Blood Group Serology and Transfusion Medicine Medical University of Graz, 8036 Graz, Austria
| | - Tobias Madl
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
- BioTechMed-Graz, 8010 Graz, Austria
| | - Riccardo Zenezini Chiozzi
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, 3584CH Utrecht, Netherlands
- Netherlands Proteomics Center, 3584CH Utrecht, Netherlands
| | - Albert J. R. Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, 3584CH Utrecht, Netherlands
- Netherlands Proteomics Center, 3584CH Utrecht, Netherlands
| | - Meritxell Huch
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Roland Malli
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
- BioTechMed-Graz, 8010 Graz, Austria
| | - Andreas Prokesch
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, 8010 Graz, Austria
- BioTechMed-Graz, 8010 Graz, Austria
| |
Collapse
|
31
|
Telerman A, Granot G, Leibovitch C, Yarchovsky-Dolberg O, Shacham-Abulafia A, Partouche S, Yeshurun M, Ellis MH, Raanani P, Wolach O. Neutrophil Extracellular Traps Are Increased in Chronic Myeloid Leukemia and Are Differentially Affected by Tyrosine Kinase Inhibitors. Cancers (Basel) 2021; 14:cancers14010119. [PMID: 35008283 PMCID: PMC8750902 DOI: 10.3390/cancers14010119] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/13/2021] [Accepted: 12/24/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Neutrophil extracellular traps (NETs) are a recently described form of neutrophil cellular death that has been associated with a thrombotic tendency in many diseases. We studied NET formation in neutrophils derived from patients with chronic myeloid leukemia (CML) and in CML neutrophil cell lines and demonstrated that NETs are increased in CML and that certain drugs used to treat CML (tyrosine kinase inhibitors—TKIs) increase NET formation. These findings may shed light on a novel mechanism linking CML, TKIs and vascular toxicity. Abstract Cardiovascular complications are increasingly reported with the use of certain tyrosine kinase inhibitors (TKIs) to treat chronic myeloid leukemia (CML). We studied neutrophil extracellular trap (NET) formation in CML and evaluated the effect of TKIs on NET formation. Neutrophils isolated from treatment-naïve patients with CML showed a significant increase in NET formation compared to matched controls at baseline and after stimulation with ionomycin (IO) and phorbol 12-myristate 13-acetate (PMA). Expression of citrullinated histone H3 (H3cit), peptidyl arginine deiminase 4 (PAD4) and reactive oxygen species (ROS) was significantly higher in CML samples compared to controls. Pre-treatment of neutrophils with TKIs was associated with a differential effect on NET formation, and ponatinib significantly augmented NET-associated elastase and ROS levels as compared to controls and other TKIs. BCR-ABL1 retroviral transduced HoxB8-immortalized mouse hematopoietic progenitors, which differentiate into neutrophils in-vitro, demonstrated increased H3cit & myeloperoxidase (MPO) expression consistent with excess NET formation. This was inhibited by Cl-amidine, a PAD4 inhibitor, but not by the NADPH inhibitor diphenyleneiodonium (DPI). Ponatinib pre-exposure significantly increased H3cit expression in HoxB8-BCR-ABL1 cells after stimulation with IO. In summary, CML is associated with increased NET formation, which is augmented by ponatinib, suggesting a possible role for NETs in promoting vascular toxicity in CML.
Collapse
Affiliation(s)
- Alona Telerman
- Felsenstein Medical Research Center, Rabin Medical Center, Beilinson Hospital, Petah-Tikva 4941492, Israel; (A.T.); (G.G.); (S.P.)
| | - Galit Granot
- Felsenstein Medical Research Center, Rabin Medical Center, Beilinson Hospital, Petah-Tikva 4941492, Israel; (A.T.); (G.G.); (S.P.)
| | - Chiya Leibovitch
- Sackler Faculty of Medicine, Tel Aviv University, Ramat-Aviv 39040, Israel; (C.L.); (O.Y.-D.); (A.S.-A.); (M.Y.); (M.H.E.); (P.R.)
- Davidoff Cancer Center, Rabin Medical Center, Institute of Hematology, Beilinson Hospital, Petah-Tikva 4941492, Israel
| | - Osnat Yarchovsky-Dolberg
- Sackler Faculty of Medicine, Tel Aviv University, Ramat-Aviv 39040, Israel; (C.L.); (O.Y.-D.); (A.S.-A.); (M.Y.); (M.H.E.); (P.R.)
- Meir Medical Center, Hematology Institute and Blood Bank, Kfar Saba 4428164, Israel
| | - Adi Shacham-Abulafia
- Sackler Faculty of Medicine, Tel Aviv University, Ramat-Aviv 39040, Israel; (C.L.); (O.Y.-D.); (A.S.-A.); (M.Y.); (M.H.E.); (P.R.)
- Davidoff Cancer Center, Rabin Medical Center, Institute of Hematology, Beilinson Hospital, Petah-Tikva 4941492, Israel
| | - Shirly Partouche
- Felsenstein Medical Research Center, Rabin Medical Center, Beilinson Hospital, Petah-Tikva 4941492, Israel; (A.T.); (G.G.); (S.P.)
| | - Moshe Yeshurun
- Sackler Faculty of Medicine, Tel Aviv University, Ramat-Aviv 39040, Israel; (C.L.); (O.Y.-D.); (A.S.-A.); (M.Y.); (M.H.E.); (P.R.)
- Davidoff Cancer Center, Rabin Medical Center, Institute of Hematology, Beilinson Hospital, Petah-Tikva 4941492, Israel
| | - Martin H. Ellis
- Sackler Faculty of Medicine, Tel Aviv University, Ramat-Aviv 39040, Israel; (C.L.); (O.Y.-D.); (A.S.-A.); (M.Y.); (M.H.E.); (P.R.)
- Meir Medical Center, Hematology Institute and Blood Bank, Kfar Saba 4428164, Israel
| | - Pia Raanani
- Sackler Faculty of Medicine, Tel Aviv University, Ramat-Aviv 39040, Israel; (C.L.); (O.Y.-D.); (A.S.-A.); (M.Y.); (M.H.E.); (P.R.)
- Davidoff Cancer Center, Rabin Medical Center, Institute of Hematology, Beilinson Hospital, Petah-Tikva 4941492, Israel
| | - Ofir Wolach
- Sackler Faculty of Medicine, Tel Aviv University, Ramat-Aviv 39040, Israel; (C.L.); (O.Y.-D.); (A.S.-A.); (M.Y.); (M.H.E.); (P.R.)
- Davidoff Cancer Center, Rabin Medical Center, Institute of Hematology, Beilinson Hospital, Petah-Tikva 4941492, Israel
- Correspondence: ; Tel.: +972-50-406-5590
| |
Collapse
|
32
|
Receptor-Tyrosine Kinase Inhibitor Ponatinib Inhibits Meningioma Growth In Vitro and In Vivo. Cancers (Basel) 2021; 13:cancers13235898. [PMID: 34885009 PMCID: PMC8657092 DOI: 10.3390/cancers13235898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/04/2021] [Accepted: 11/17/2021] [Indexed: 11/17/2022] Open
Abstract
To date, there is no standard-of-care systemic therapy for the treatment of aggressive meningiomas. Receptor tyrosine kinases (RTK) are frequently expressed in aggressive meningiomas and are associated with poor survival. Ponatinib is a FDA- and EMA-approved RTK inhibitor and its efficacy in meningioma has not been studied so far. Therefore, we investigated ponatinib as a potential drug candidate against meningioma. Cell viability and cell proliferation of ponatinib-treated meningioma cells were assessed using crystal violet assay, manual counting and BrdU assay. Treated meningioma cell lines were subjected to flow cytometry to evaluate the effects on cell cycle and apoptosis. Meningioma-bearing mice were treated with ponatinib to examine antitumor effects in vivo. qPCR was performed to assess the mRNA levels of tyrosine kinase receptors after ponatinib treatment. Full-length cDNA sequencing was carried out to assess differential gene expression. IC50 values of ponatinib were between 171.2 and 341.9 nM in three meningioma cell lines. Ponatinib induced G0/G1 cell cycle arrest and subsequently led to an accumulation of cells in the subG1-phase. A significant induction of apoptosis was observed in vitro. In vivo, ponatinib inhibited meningioma growth by 72.6%. Mechanistically, this was associated with downregulation of PDGFRA/B and FLT3 mRNA levels, and mitochondrial dysfunction. Taken together, ponatinib is a promising candidate for targeted therapy in the treatment of aggressive meningioma.
Collapse
|
33
|
Hadrava Vanova K, Yang C, Meuter L, Neuzil J, Pacak K. Reactive Oxygen Species: A Promising Therapeutic Target for SDHx-Mutated Pheochromocytoma and Paraganglioma. Cancers (Basel) 2021; 13:cancers13153769. [PMID: 34359671 PMCID: PMC8345159 DOI: 10.3390/cancers13153769] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/21/2021] [Accepted: 07/24/2021] [Indexed: 11/26/2022] Open
Abstract
Simple Summary Pheochromocytoma and paraganglioma are rare neuroendocrine tumors that arise from chromaffin cells of the adrenal medulla or their neural crest progenitors located outside the adrenal gland, respectively. About 10–15% of patients develop metastatic disease for whom treatment options and availability are extremely limited. The risk of developing metastatic disease is increased for patients with mutations in succinate dehydrogenase subunit B, which leads to metabolic reprogramming and redox imbalance. From this perspective, we focus on redox imbalance caused by this mutation and explore potential opportunities to therapeutically target reactive oxygen species production in these rare tumors. Abstract Pheochromocytoma (PHEO) and paraganglioma (PGL) are rare neuroendocrine tumors derived from neural crest cells. Germline variants in approximately 20 PHEO/PGL susceptibility genes are found in about 40% of patients, half of which are found in the genes that encode succinate dehydrogenase (SDH). Patients with SDH subunit B (SDHB)-mutated PHEO/PGL exhibit a higher likelihood of developing metastatic disease, which can be partially explained by the metabolic cell reprogramming and redox imbalance caused by the mutation. Reactive oxygen species (ROS) are highly reactive molecules involved in a multitude of important signaling pathways. A moderate level of ROS production can help regulate cellular physiology; however, an excessive level of oxidative stress can lead to tumorigenic processes including stimulation of growth factor-dependent pathways and the induction of genetic instability. Tumor cells effectively exploit antioxidant enzymes in order to protect themselves against harmful intracellular ROS accumulation, which highlights the essential balance between ROS production and scavenging. Exploiting ROS accumulation can be used as a possible therapeutic strategy in ROS-scavenging tumor cells. Here, we focus on the role of ROS production in PHEO and PGL, predominantly in SDHB-mutated cases. We discuss potential strategies and approaches to anticancer therapies by enhancing ROS production in these difficult-to-treat tumors.
Collapse
Affiliation(s)
- Katerina Hadrava Vanova
- Section of Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA; (K.H.V.); (L.M.)
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Vestec, 252 50 Prague West, Czech Republic; or
| | - Chunzhang Yang
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Leah Meuter
- Section of Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA; (K.H.V.); (L.M.)
| | - Jiri Neuzil
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Vestec, 252 50 Prague West, Czech Republic; or
- School of Pharmacy and Medical Science, Griffith University, Southport, QLD 4222, Australia
| | - Karel Pacak
- Section of Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA; (K.H.V.); (L.M.)
- Correspondence: ; Tel.: +1-(301)-402-4594
| |
Collapse
|
34
|
Babaei Z, Panjehpour M, Parsian H, Aghaei M. SAR131675 Receptor Tyrosine Kinase Inhibitor Induces Apoptosis through Bcl-2/Bax/Cyto c Mitochondrial Pathway in Human Umbilical Vein Endothelial Cells. Anticancer Agents Med Chem 2021; 22:943-950. [PMID: 34238175 DOI: 10.2174/1871520621666210708102619] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 05/11/2021] [Accepted: 05/31/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Tyrosine kinase inhibitors (TKIs) can be used to inhibit cancer cell proliferation by targeting the vascular endothelial growth factor receptor (VEGFR) family. SAR131675 is a highly selective receptor tyrosine kinase inhibitor to VEGFR3 that reveals the inhibitory effect on proliferation in human lymphatic endothelial cells. However, the molecular mechanisms underlying this process are generally unclear. OBJECTIVE This study was performed to investigate the possible involvement of the Bcl-2/Bax/Cyto c apoptosis pathway in human umbilical vein endothelial cells (HUVECs). In addition, the role of reactive oxygen species (ROS) and mitochondrial membrane potential was evaluated. METHODS The effect of SAR131675 on HUVEC cell viability was evaluated by MTT assay. The activity of SAR131675 in inducing apoptosis was carried out through the detection of Annexin V-FITC/PI signal by flow cytometry. To determine the mechanisms underlying SAR131675 induced apoptosis, the mitochondrial membrane potential, ROS generation, the activity of caspase-3, and expression of apoptosis-related proteins such as Bcl-2, Bax, and cytochrome c were evaluated in HUVECs. RESULTS SAR131675 significantly inhibited cell viability and induced apoptosis in HUVECs in a dose-dependent manner. Moreover, SAR131675 induced mitochondrial dysfunction, ROS generation, Bcl-2 down-regulation, Bax up-regulation, cytochrome c release, and caspase-3 activation, which displays features of the mitochondria-dependent apoptosis signaling pathway. CONCLUSION Our present data demonstrated that SAR131675-induced cytotoxicity in HUVECs is associated with the mitochondria apoptotic pathway. These results suggest that further studies are required to fully elucidate the role of TKIs in these cellular processes.
Collapse
Affiliation(s)
- Zeinab Babaei
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mojtaba Panjehpour
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hadi Parsian
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Mahmoud Aghaei
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
35
|
Wang Z, Wang X, Wang Z, Feng Y, Jia Y, Jiang L, Xia Y, Cao J, Liu Y. Comparison of Hepatotoxicity Associated With New BCR-ABL Tyrosine Kinase Inhibitors vs Imatinib Among Patients With Chronic Myeloid Leukemia: A Systematic Review and Meta-analysis. JAMA Netw Open 2021; 4:e2120165. [PMID: 34292334 PMCID: PMC8299317 DOI: 10.1001/jamanetworkopen.2021.20165] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
IMPORTANCE Although BCR-ABL fusion oncoprotein tyrosine kinase inhibitors (BCR-ABL TKIs) can substantially improve the survival rate of chronic myeloid leukemia (CML), they are clinically accompanied by severe hepatotoxicity. OBJECTIVE To compare the relative risk (RR) of hepatotoxicity of new-generation BCR-ABL TKIs with that of imatinib, and to provide an overall assessment of the clinical benefit. DATA SOURCES PubMed, Embase, Cochrane library databases, and ClinicalTrials.gov were searched for clinical trials published between January 2000 and April 2020. STUDY SELECTION Study selection was conducted independently by 2 investigators according to the inclusion and exclusion criteria published previously in the protocol: only randomized phase 2 or phase 3 clinical trials that compared bosutinib, dasatinib, nilotinib, or ponatinib with imatinib were included. Among the 2666 records identified, 9 studies finally fulfilled the established criteria. DATA EXTRACTION AND SYNTHESIS Two investigators extracted study characteristics and data independently using a standardized data extraction form. Data were extracted according to Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) reporting guidelines. When substantial heterogeneity was observed, pooled estimates were calculated based on the random-effect model; otherwise, the fixed-effect model was used. MAIN OUTCOMES AND MEASURES Data extracted included study characteristics, baseline patient information, interventions and data on all-grade and high-grade (grades 3 and 4) elevation of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels, overall survival, and major molecular response (MMR). The RRs and 95% CIs were calculated using the inverse variance method. RESULTS Nine trials involving 3475 patients were analyzed; the median (range) age was 49 (18-91) years; 2059 (59.2%) were male patients. Increased risks were observed for each new-generation TKI except for dasatinib. Patients receiving new-generation TKIs were more likely to experience all grades of ALT elevation (pooled RR, 2.89; 95% CI, 1.78-4.69; P < .001) and grades 3 and 4 ALT elevation (pooled RR, 4.36; 95% CI, 2.00-9.50; P < .001) compared with those receiving imatinib. Patients receiving new-generation TKIs were also more likely to experience all grades of AST elevation (pooled RR, 2.20; 95% CI, 1.63-2.98; P < .001) and grades 3 and 4 AST elevation (pooled RR, 2.65; 95% CI, 1.59-4.42; P < .001) compared with those receiving imatinib. New-generation TKIs were associated with a significantly higher rate of MMR at 1 year compared with imatinib (pooled RR, 1.59; 95% CI, 1.44-1.75; P < .001). No statistical difference in overall survival at 1 year was found between new-generation TKIs and imatinib (pooled RR, 1.00; 95% CI, 1.00-1.01; P = .33). CONCLUSIONS AND RELEVANCE When compared to imatinib, bosutinib, nilotinib, and ponatinib had higher relative risks of hepatotoxicity. Treatment with new-generation TKIs was associated with a higher MMR rate at 1 year but not with 1-year overall survival.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Alanine Transaminase/blood
- Aniline Compounds/adverse effects
- Aspartate Aminotransferases/blood
- Chemical and Drug Induced Liver Injury/etiology
- Dasatinib/adverse effects
- Female
- Humans
- Imatinib Mesylate/adverse effects
- Imidazoles/adverse effects
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/blood
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Male
- Middle Aged
- Nitriles/adverse effects
- Oncogene Proteins v-abl/drug effects
- Protein Kinase Inhibitors/adverse effects
- Protein-Tyrosine Kinases/antagonists & inhibitors
- Proto-Oncogene Proteins c-bcr/drug effects
- Pyridazines/adverse effects
- Pyrimidines/adverse effects
- Quinolines/adverse effects
- Risk
- Young Adult
Collapse
Affiliation(s)
- Zhe Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Xiaoyu Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Zhen Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Yuyi Feng
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Yaqin Jia
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Lili Jiang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Yangliu Xia
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Jun Cao
- Department of Occupational and Environmental Health, Dalian Medical University, Dalian, China
| | - Yong Liu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| |
Collapse
|
36
|
Li S, Zhang J, Liu C, Wang Q, Yan J, Hui L, Jia Q, Shan H, Tao L, Zhang M. The Role of Mitophagy in Regulating Cell Death. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6617256. [PMID: 34113420 PMCID: PMC8154277 DOI: 10.1155/2021/6617256] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/27/2021] [Accepted: 04/24/2021] [Indexed: 02/06/2023]
Abstract
Mitochondria are multifaceted organelles that serve to power critical cellular functions, including act as power generators of the cell, buffer cytosolic calcium overload, production of reactive oxygen species, and modulating cell survival. The structure and the cellular location of mitochondria are critical for their function and depend on highly regulated activities such as mitochondrial quality control (MQC) mechanisms. The MQC is regulated by several sets of processes: mitochondrial biogenesis, mitochondrial fusion and fission, mitophagy, and other mitochondrial proteostasis mechanisms such as mitochondrial unfolded protein response (mtUPR) or mitochondrial-derived vesicles (MDVs). These processes are important for the maintenance of mitochondrial homeostasis, and alterations in the mitochondrial function and signaling are known to contribute to the dysregulation of cell death pathways. Recent studies have uncovered regulatory mechanisms that control the activity of the key components for mitophagy. In this review, we discuss how mitophagy is controlled and how mitophagy impinges on health and disease through regulating cell death.
Collapse
Affiliation(s)
- Sunao Li
- Department of Forensic Sciences, School of Basic Medicine and Biological Sciences, Affilated Guangji Hospital, Soochow University, Suzhou, China
| | - Jiaxin Zhang
- Department of Forensic Sciences, School of Basic Medicine and Biological Sciences, Affilated Guangji Hospital, Soochow University, Suzhou, China
| | - Chao Liu
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qianliang Wang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jun Yan
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Li Hui
- Department of Forensic Sciences, School of Basic Medicine and Biological Sciences, Affilated Guangji Hospital, Soochow University, Suzhou, China
| | - Qiufang Jia
- Department of Forensic Sciences, School of Basic Medicine and Biological Sciences, Affilated Guangji Hospital, Soochow University, Suzhou, China
| | - Haiyan Shan
- Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Luyang Tao
- Department of Forensic Sciences, School of Basic Medicine and Biological Sciences, Affilated Guangji Hospital, Soochow University, Suzhou, China
| | - Mingyang Zhang
- Department of Forensic Sciences, School of Basic Medicine and Biological Sciences, Affilated Guangji Hospital, Soochow University, Suzhou, China
| |
Collapse
|
37
|
Gunaydin Akyildiz A, Boran T, Jannuzzi AT, Alpertunga B. Mitochondrial dynamics imbalance and mitochondrial dysfunction contribute to the molecular cardiotoxic effects of lenvatinib. Toxicol Appl Pharmacol 2021; 423:115577. [PMID: 34019861 DOI: 10.1016/j.taap.2021.115577] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 12/19/2022]
Abstract
Lenvatinib is a tyrosine kinase inhibitor (TKI) approved for the treatment of resistant differentiated thyroid cancer, advanced renal cell carcinoma, unresectable hepatocellular carcinoma, and endometrial carcinoma. Although it is successful in cancer treatment, it can cause life-threatening side effects such as cardiotoxicity. The molecular mechanism of cardiotoxicity caused by lenvatinib is not fully known. In this study, the molecular mechanism of lenvatinib's cardiotoxicity was investigated focusing on mitochondrial toxicity in the H9c2 cardiomyoblastic cell line. Lenvatinib inhibited cell viability at 48 and 72 h exposure with three selected concentrations (1.25 μM, 5 μM and 10 μM); and inhibited intracellular ATP after 72 h exposure compared to the control group. Mitochondrial membrane potential was decreased after 48 h and did not show significant changes after 72 h exposure. Evaluated with real-time PCR, mitochondrial dynamics (Mfn1, Mfn2, OPA1, DRP1, Fis1) expression levels after lenvatinib treatment significantly changed. Lenvatinib triggered the tendency from fusion to fission in mitochondria after 48 h exposure, and increased both fusion and fission after 72 h. The mtDNA ratio increased after 48 h and decreased after 72 h. ASK1, JNK and AMPKα2 increased. UCP2 showed downregulation, SOD2 level showed upregulation and Cat levels decreased after drug treatment. Nrf1 and Nrf2 also changed concentration-dependently. Protein carbonyl levels increased significantly after lenvatinib treatments indicating oxidative stress. The protein levels of the electron transport chain complexes, LONP1, UCP2, and P21 showed significant differences after lenvatinib treatment. The outcome of our study is expected to be a contribution to the understanding of the molecular mechanisms of TKI-induced cardiotoxicity.
Collapse
Affiliation(s)
- Aysenur Gunaydin Akyildiz
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34116 Beyazit, Istanbul, Turkey; Bezmialem Vakif University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, Vatan Street, 34093 Fatih, Istanbul, Turkey
| | - Tugce Boran
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34116 Beyazit, Istanbul, Turkey
| | - Ayse Tarbin Jannuzzi
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34116 Beyazit, Istanbul, Turkey
| | - Buket Alpertunga
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34116 Beyazit, Istanbul, Turkey.
| |
Collapse
|
38
|
Xu S, Ling S, Shan Q, Ye Q, Zhan Q, Jiang G, Zhuo J, Pan B, Wen X, Feng T, Lu H, Wei X, Xie H, Zheng S, Xiang J, Shen Y, Xu X. Self-Activated Cascade-Responsive Sorafenib and USP22 shRNA Co-Delivery System for Synergetic Hepatocellular Carcinoma Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003042. [PMID: 33717848 PMCID: PMC7927615 DOI: 10.1002/advs.202003042] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/17/2020] [Indexed: 05/06/2023]
Abstract
Resistance to sorafenib severely hinders its effectiveness against hepatocellular carcinoma (HCC). Cancer stemness is closely connected with resistance to sorafenib. Methods for reversing the cancer stemness remains one of the largest concerns in research and the lack of such methods obstructs current HCC therapeutics. Ubiquitin-specific protease 22 (USP22) is reported to play a pivotal role in HCC stemness and multidrug resistance (MDR). Herein, a galactose-decorated lipopolyplex (Gal-SLP) is developed as an HCC-targeting self-activated cascade-responsive nanoplatform to co-delivery sorafenib and USP22 shRNA (shUSP22) for synergetic HCC therapy. Sorafenib, entrapped in the Gal-SLPs, induced a reactive oxygen species (ROS) cascade and triggered rapid shUSP22 release. Thus, Gal-SLPs dramatically suppressed the expression of USP22. The downregulation of USP22 suppresses multidrug resistance-associated protein 1 (MRP1) to induce intracellular sorafenib accumulation and hampers glycolysis of HCC cells. As a result, Gal-SLPs efficiently inhibit the viability, proliferation, and colony formation of HCC cells. A sorafenib-insensitive patient-derived xenograft (PDX) model is established and adopted to evaluate in vivo antitumor effect of Gal-SLPs. Gal-SLPs exhibit potent antitumor efficiency and biosafety. Therefore, Gal-SLPs are expected to have great potential in the clinical treatment of HCC.
Collapse
Affiliation(s)
- Shengjun Xu
- Department of Hepatobiliary and Pancreatic SurgeryAffiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhouZhejiang310006China
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalSchool of Medicine Zhejiang UniversityHangzhouZhejiang310003China
- NHC Key Lab of Combined Multi‐Organ TransplantationHangzhouZhejiang310003China
| | - Sunbin Ling
- Department of Hepatobiliary and Pancreatic SurgeryAffiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhouZhejiang310006China
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalSchool of Medicine Zhejiang UniversityHangzhouZhejiang310003China
- NHC Key Lab of Combined Multi‐Organ TransplantationHangzhouZhejiang310003China
| | - Qiaonan Shan
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalSchool of Medicine Zhejiang UniversityHangzhouZhejiang310003China
- NHC Key Lab of Combined Multi‐Organ TransplantationHangzhouZhejiang310003China
| | - Qianwei Ye
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalSchool of Medicine Zhejiang UniversityHangzhouZhejiang310003China
- NHC Key Lab of Combined Multi‐Organ TransplantationHangzhouZhejiang310003China
| | - Qifan Zhan
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalSchool of Medicine Zhejiang UniversityHangzhouZhejiang310003China
- NHC Key Lab of Combined Multi‐Organ TransplantationHangzhouZhejiang310003China
| | - Guangjiang Jiang
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalSchool of Medicine Zhejiang UniversityHangzhouZhejiang310003China
- NHC Key Lab of Combined Multi‐Organ TransplantationHangzhouZhejiang310003China
| | - Jianyong Zhuo
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalSchool of Medicine Zhejiang UniversityHangzhouZhejiang310003China
- NHC Key Lab of Combined Multi‐Organ TransplantationHangzhouZhejiang310003China
| | - Binhua Pan
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalSchool of Medicine Zhejiang UniversityHangzhouZhejiang310003China
- NHC Key Lab of Combined Multi‐Organ TransplantationHangzhouZhejiang310003China
| | - Xue Wen
- Department of Pathologythe First Affiliated HospitalSchool of Medicine Zhejiang UniversityHangzhouZhejiang310003China
| | - Tingting Feng
- Department of Abdominal Medical OncologyZhejiang Cancer HospitalHangzhouZhejiang310022China
| | - Haohao Lu
- Department of Polymer Science and EngineeringZhejiang UniversityHangzhouZhejiang310027China
| | - Xuyong Wei
- Department of Hepatobiliary and Pancreatic SurgeryAffiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhouZhejiang310006China
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalSchool of Medicine Zhejiang UniversityHangzhouZhejiang310003China
- NHC Key Lab of Combined Multi‐Organ TransplantationHangzhouZhejiang310003China
| | - Haiyang Xie
- NHC Key Lab of Combined Multi‐Organ TransplantationHangzhouZhejiang310003China
| | - Shusen Zheng
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalSchool of Medicine Zhejiang UniversityHangzhouZhejiang310003China
- NHC Key Lab of Combined Multi‐Organ TransplantationHangzhouZhejiang310003China
- Department of Hepatobiliary and Pancreatic SurgeryShulan (Hangzhou) HospitalHangzhouZhejiang310000China
| | - Jiajia Xiang
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of EducationCollege of Chemical and Biological EngineeringZhejiang UniversityHangzhouZhejiang310027China
| | - Youqing Shen
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of EducationCollege of Chemical and Biological EngineeringZhejiang UniversityHangzhouZhejiang310027China
| | - Xiao Xu
- Department of Hepatobiliary and Pancreatic SurgeryAffiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhouZhejiang310006China
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalSchool of Medicine Zhejiang UniversityHangzhouZhejiang310003China
- NHC Key Lab of Combined Multi‐Organ TransplantationHangzhouZhejiang310003China
| |
Collapse
|
39
|
Molecules and Mechanisms to Overcome Oxidative Stress Inducing Cardiovascular Disease in Cancer Patients. Life (Basel) 2021; 11:life11020105. [PMID: 33573162 PMCID: PMC7911715 DOI: 10.3390/life11020105] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/18/2021] [Accepted: 01/27/2021] [Indexed: 02/06/2023] Open
Abstract
Reactive oxygen species (ROS) are molecules involved in signal transduction pathways with both beneficial and detrimental effects on human cells. ROS are generated by many cellular processes including mitochondrial respiration, metabolism and enzymatic activities. In physiological conditions, ROS levels are well-balanced by antioxidative detoxification systems. In contrast, in pathological conditions such as cardiovascular, neurological and cancer diseases, ROS production exceeds the antioxidative detoxification capacity of cells, leading to cellular damages and death. In this review, we will first describe the biology and mechanisms of ROS mediated oxidative stress in cardiovascular disease. Second, we will review the role of oxidative stress mediated by oncological treatments in inducing cardiovascular disease. Lastly, we will discuss the strategies that potentially counteract the oxidative stress in order to fight the onset and progression of cardiovascular disease, including that induced by oncological treatments.
Collapse
|
40
|
Boran T, Akyildiz AG, Jannuzzi AT, Alpertunga B. Extended regorafenib treatment can be linked with mitochondrial damage leading to cardiotoxicity. Toxicol Lett 2020; 336:39-49. [PMID: 33166663 DOI: 10.1016/j.toxlet.2020.11.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/31/2020] [Accepted: 11/03/2020] [Indexed: 12/22/2022]
Abstract
Regorafenib (RGF) has a great success in the treatment of colorectal cancer, gastrointestinal stromal tumours and hepatocellular carcinoma by inhibiting angiogenic, stromal and oncogenic kinases. However, RGF can induce life-threatening cardiotoxicity including hypertension and cardiac ischemia/infarction. The molecular mechanism of the adverse effects has not been elucidated. Mitochondrial dysfunction is one of the major causes of cardiac diseases since cardiac cells highly need ATP for their contractility. Therefore, we aimed to investigate molecular mechanisms of RGF-induced cardiac adverse effects using H9c2 cell model by focusing on mitochondria. Cells were treated with 0-20 μM RGF for 48 and 72 h. According to our results, RGF inhibited cell proliferation and decreased the ATP content of the cells depending on the exposure time and concentration. Loss of mitochondrial membrane potential was also observed at high dose. Mitochondrial fusion/fission genes and antioxidant SOD2 (superoxide dismutase) gene expression levels increased at high doses in both treatments. Mitochondrial DNA content decreased as exposure time and concentration increased. Also, protein expression levels of mitochondrial complex I and V have reduced and stress protein HSP70 level has increased following RGF treatment. Structural abnormalities in mitochondria was seen with transmission electron microscopy at the applied higher doses. Our findings suggest that RGF-induced cardiotoxicity may be associated with mitochondrial damage in cardiac cells.
Collapse
Affiliation(s)
- Tugce Boran
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34116, Beyazıt, Istanbul, Turkey
| | - Aysenur Gunaydin Akyildiz
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34116, Beyazıt, Istanbul, Turkey; Bezmialem Vakif University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, Vatan Street, 34093, Fatih, Istanbul, Turkey
| | - Ayse Tarbin Jannuzzi
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34116, Beyazıt, Istanbul, Turkey
| | - Buket Alpertunga
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34116, Beyazıt, Istanbul, Turkey.
| |
Collapse
|
41
|
Inoue T, Iwazaki N, Araki T, Hitotsumachi H. Human-Induced Pluripotent Stem Cell-Derived Hepatocytes and their Culturing Methods to Maintain Liver Functions for Pharmacokinetics and Safety Evaluation of Pharmaceuticals. Curr Pharm Biotechnol 2020; 21:773-779. [PMID: 32003687 DOI: 10.2174/1389201021666200131123524] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 10/23/2019] [Accepted: 12/31/2019] [Indexed: 12/12/2022]
Abstract
Human hepatocytes are essential cell types for pharmacokinetics and the safety evaluation of pharmaceuticals. However, widely used primary hepatocytes with individual variations in liver function lose those functions rapidly in culture. Hepatic cell lines are convenient to use but have low liver functions. Human-Induced Pluripotent Stem (hiPS) cells can be expanded and potentially differentiated into any cell or tissue, including the liver. HiPS cell-derived Hepatocyte-Like Cells (hiPSHeps) are expected to be extensively used as consistent functional human hepatocytes. Many laboratories are investigating methods of using hiPS cells to differentiate hepatocytes, but the derived cells still have immature liver functions. In this paper, we describe the current uses and limitations of conventional hepatic cells, evaluating the suitability of hiPS-Heps to pharmacokinetics and the safety evaluation of pharmaceuticals, and discuss the potential future use of non-conventional non-monolayer culture methods to derive fully functional hiPS-Heps.
Collapse
Affiliation(s)
- Tomoaki Inoue
- Consortium for Safety Assessment using Human iPS Cells (CSAHi), Japan
| | - Norihiko Iwazaki
- Consortium for Safety Assessment using Human iPS Cells (CSAHi), Japan
| | - Tetsuro Araki
- Consortium for Safety Assessment using Human iPS Cells (CSAHi), Japan
| | | |
Collapse
|
42
|
Wang R, Liu Y, Mi X, Chen Q, Jiang P, Hou J, Lin Y, Li S, Ji B, Fang Y. Sirt3 promotes hepatocellular carcinoma cells sensitivity to regorafenib through the acceleration of mitochondrial dysfunction. Arch Biochem Biophys 2020; 689:108415. [PMID: 32562663 DOI: 10.1016/j.abb.2020.108415] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 04/15/2020] [Accepted: 05/16/2020] [Indexed: 01/01/2023]
Abstract
Regorafenib, a multiple kinase inhibitor, is recently approved for treatment of patients with advanced hepatocellular carcinoma (HCC). Previous studies demonstrated that regorafenib was a mitochondrial toxicant, which associated with the impairment of mitochondria. Sirt3 is involved in the regulation of mitochondrial function in cancers. This study aimed to investigate the mechanism of Sirt3 involved in the mitochondrial dysfunction which associated with regorafenib treatment in liver cancer cells. We found regorafenib inhibited Sirt3 and p-ERK expression in HCC cells in a dose-dependent manner. Bioinformatics analysis showed that Sirt3 expression was down-regulated in liver cancer tissues and its low expression was correlated with worse overall survival (OS) in liver cancer patients. After transfected with Sirt3 overexpression plasmid, we found that Sirt3 sensitized liver cancer cells to regorafenib and resulted in much more apoptosis with a significant increase of ROS level. However, exogenous antioxidant could not weaken the apoptosis. Mitochondrial membrane potential assay indicated that Sirt3 overexpression accelerated the mitochondrial depolarization process induced by regorafenib and aggravated mitochondrial injury. Cellular oxygen consumption assay showed that mitochondrial dysfunction was caused by the damage of the electron transport chain. The results demonstrated that Sirt3 overexpression promoted the increase of ROS and apoptosis induced by regorafenib through the acceleration of mitochondrial dysfunction by impairing function of the electron transport chain in liver cancer cells. Our studies verified the functional role of Sirt3 in regorafenib treatment and suggested that regorafenib accompanied with Sirt3 activator as a novel treatment strategy for HCC.
Collapse
Affiliation(s)
- Ruobing Wang
- Department of Hepatobiliary and Pancreatic Surgery, the First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Yahui Liu
- Department of Hepatobiliary and Pancreatic Surgery, the First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Xuguang Mi
- Tumor Biotherapy Center, Jilin Province People's Hospital, Changchun, Jilin, 130021, China; Laboratory Center, the First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Qingmin Chen
- Department of Hepatobiliary and Pancreatic Surgery, the First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Peiqiang Jiang
- Department of Hepatobiliary and Pancreatic Surgery, the First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Junjie Hou
- Tumor Biotherapy Center, Jilin Province People's Hospital, Changchun, Jilin, 130021, China
| | - Yifan Lin
- Tumor Biotherapy Center, Jilin Province People's Hospital, Changchun, Jilin, 130021, China
| | - Siqi Li
- School of Medical Technology, Beihua University, Jilin, Jilin, 132021, China
| | - Bai Ji
- Department of Hepatobiliary and Pancreatic Surgery, the First Hospital of Jilin University, Changchun, Jilin, 130021, China.
| | - Yanqiu Fang
- Tumor Biotherapy Center, Jilin Province People's Hospital, Changchun, Jilin, 130021, China.
| |
Collapse
|
43
|
Rodríguez-Hernández MA, de la Cruz-Ojeda P, Gallego P, Navarro-Villarán E, Staňková P, Del Campo JA, Kučera O, Elkalaf M, Maseko TE, Červinková Z, Muntané J. Dose-dependent regulation of mitochondrial function and cell death pathway by sorafenib in liver cancer cells. Biochem Pharmacol 2020; 176:113902. [PMID: 32156660 DOI: 10.1016/j.bcp.2020.113902] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 03/03/2020] [Indexed: 01/14/2023]
Abstract
Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer and the fourth most frequent cause of cancer-related death worldwide. Sorafenib is the first line recommended therapy for patients with locally advanced/metastatic HCC. The low response rate is attributed to intrinsic resistance of HCC cells to Sorafenib. The potential resistance to Sorafenib-induced cell death is multifactorial and involves all hallmarks of cancer. However, the presence of sub-therapeutic dose can negatively influence the antitumoral properties of the drug. In this sense, the present study showed that the sub-optimal Sorafenib concentration (10 nM) was associated with activation of caspase-9, AMP-activated protein kinase (AMPK), sustained autophagy, peroxisome proliferator-activated receptor-coactivator 1α (PGC-1α) and mitochondrial function in HepG2 cells. The increased mitochondrial respiration by Sorafenib (10 nM) was also observed in permeabilized HepG2 cells, but not in isolated rat mitochondria, which suggests the involvement of an upstream component in this regulatory mechanism. The basal glycolysis was dose dependently increased at early time point studied (6 h). Interestingly, Sorafenib increased nitric oxide (NO) generation that played an inhibitory role in mitochondrial respiration in sub-therapeutic dose of Sorafenib. The administration of sustained therapeutic dose of Sorafenib (10 µM, 24 h) induced mitochondrial dysfunction and dropped basal glycolysis derived acidification, as well as increased oxidative stress and apoptosis in HepG2. In conclusion, the accurate control of the administered dose of Sorafenib is relevant for the potential prosurvival or proapoptotic properties induced by the drug in liver cancer cells.
Collapse
Affiliation(s)
- María A Rodríguez-Hernández
- Institute of Biomedicine of Seville (IBIS), IBiS/Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain; CENTRO DE INVESTIGACIÓN BIOMÉDICA EN RED de Enfermedades Hepáticas y Digestivas (CIBERehd), Spain
| | - Patricia de la Cruz-Ojeda
- Institute of Biomedicine of Seville (IBIS), IBiS/Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
| | - Paloma Gallego
- Institute of Biomedicine of Seville (IBIS), IBiS/Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
| | - Elena Navarro-Villarán
- Institute of Biomedicine of Seville (IBIS), IBiS/Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain; CENTRO DE INVESTIGACIÓN BIOMÉDICA EN RED de Enfermedades Hepáticas y Digestivas (CIBERehd), Spain
| | - Pavla Staňková
- Department of Physiology, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic; COST-European Cooperation in Science & Technology, Mitoeagle Action number: CA15203, Brussels, Belgium
| | - José A Del Campo
- Institute of Biomedicine of Seville (IBIS), IBiS/Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain; CENTRO DE INVESTIGACIÓN BIOMÉDICA EN RED de Enfermedades Hepáticas y Digestivas (CIBERehd), Spain
| | - Otto Kučera
- Department of Physiology, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic; COST-European Cooperation in Science & Technology, Mitoeagle Action number: CA15203, Brussels, Belgium
| | - Moustafa Elkalaf
- Department of Physiology, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Tumisang E Maseko
- Department of Physiology, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Zuzana Červinková
- Department of Physiology, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic; COST-European Cooperation in Science & Technology, Mitoeagle Action number: CA15203, Brussels, Belgium
| | - Jordi Muntané
- Institute of Biomedicine of Seville (IBIS), IBiS/Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain; CENTRO DE INVESTIGACIÓN BIOMÉDICA EN RED de Enfermedades Hepáticas y Digestivas (CIBERehd), Spain; COST-European Cooperation in Science & Technology, Mitoeagle Action number: CA15203, Brussels, Belgium; Department of General Surgery, "Virgen del Rocío" University Hospital/IBiS/CSIC/University of Seville, Seville, Spain.
| |
Collapse
|
44
|
Rodríguez-Hernández MA, de la Cruz-Ojeda P, López-Grueso MJ, Navarro-Villarán E, Requejo-Aguilar R, Castejón-Vega B, Negrete M, Gallego P, Vega-Ochoa Á, Victor VM, Cordero MD, Del Campo JA, Bárcena JA, Padilla CA, Muntané J. Integrated molecular signaling involving mitochondrial dysfunction and alteration of cell metabolism induced by tyrosine kinase inhibitors in cancer. Redox Biol 2020; 36:101510. [PMID: 32593127 PMCID: PMC7322178 DOI: 10.1016/j.redox.2020.101510] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 03/16/2020] [Indexed: 12/21/2022] Open
Abstract
Cancer cells have unlimited replicative potential, insensitivity to growth-inhibitory signals, evasion of apoptosis, cellular stress, and sustained angiogenesis, invasiveness and metastatic potential. Cancer cells adequately adapt cell metabolism and integrate several intracellular and redox signaling to promote cell survival in an inflammatory and hypoxic microenvironment in order to maintain/expand tumor phenotype. The administration of tyrosine kinase inhibitor (TKI) constitutes the recommended therapeutic strategy in different malignancies at advanced stages. There are important interrelationships between cell stress, redox status, mitochondrial function, metabolism and cellular signaling pathways leading to cell survival/death. The induction of apoptosis and cell cycle arrest widely related to the antitumoral properties of TKIs result from tightly controlled events involving different cellular compartments and signaling pathways. The aim of the present review is to update the most relevant studies dealing with the impact of TKI treatment on cell function. The induction of endoplasmic reticulum (ER) stress and Ca2+ disturbances, leading to alteration of mitochondrial function, redox status and phosphatidylinositol 3-kinase (PI3K)-protein kinase B (Akt)-mammalian target of rapamycin (mTOR) and AMP-activated protein kinase (AMPK) signaling pathways that involve cell metabolism reprogramming in cancer cells will be covered. Emphasis will be given to studies that identify key components of the integrated molecular pattern including receptor tyrosine kinase (RTK) downstream signaling, cell death and mitochondria-related events that appear to be involved in the resistance of cancer cells to TKI treatments.
Collapse
Affiliation(s)
- María A Rodríguez-Hernández
- Institute of Biomedicine of Seville (IBiS), IBiS/Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain; Centro de Investigación Biomédica en red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - P de la Cruz-Ojeda
- Institute of Biomedicine of Seville (IBiS), IBiS/Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
| | - Mª José López-Grueso
- Department of Biochemistry and Molecular Biology, University of Cordoba, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
| | - Elena Navarro-Villarán
- Institute of Biomedicine of Seville (IBiS), IBiS/Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain; Centro de Investigación Biomédica en red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Raquel Requejo-Aguilar
- Department of Biochemistry and Molecular Biology, University of Cordoba, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
| | - Beatriz Castejón-Vega
- Research Laboratory, Oral Medicine Department, University of Seville, Seville, Spain
| | - María Negrete
- Institute of Biomedicine of Seville (IBiS), IBiS/Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
| | - Paloma Gallego
- Unit for the Clinical Management of Digestive Diseases, Hospital University "Nuestra Señora de Valme", Sevilla, Spain
| | - Álvaro Vega-Ochoa
- Institute of Biomedicine of Seville (IBiS), IBiS/Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
| | - Victor M Victor
- Centro de Investigación Biomédica en red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain; Service of Endocrinology and Nutrition, Hospital University "Doctor Peset", Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain; Department of Physiology, University of Valencia, Valencia, Spain
| | - Mario D Cordero
- Research Laboratory, Oral Medicine Department, University of Seville, Seville, Spain; Department of Physiology, Institute of Nutrition and Food Technology "José Mataix", Biomedical Research Center (CIBM), University of Granada, Armilla, Spain
| | - José A Del Campo
- Unit for the Clinical Management of Digestive Diseases, Hospital University "Nuestra Señora de Valme", Sevilla, Spain
| | - J Antonio Bárcena
- Department of Biochemistry and Molecular Biology, University of Cordoba, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
| | - C Alicia Padilla
- Department of Biochemistry and Molecular Biology, University of Cordoba, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
| | - Jordi Muntané
- Institute of Biomedicine of Seville (IBiS), IBiS/Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain; Centro de Investigación Biomédica en red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain; Department of General Surgery, Hospital University "Virgen del Rocío"/IBiS/CSIC/University of Seville, Seville, Spain.
| |
Collapse
|
45
|
Elucidation of the Molecular Mechanisms Underlying Sorafenib-Induced Hepatotoxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7453406. [PMID: 32509153 PMCID: PMC7245685 DOI: 10.1155/2020/7453406] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/23/2020] [Accepted: 04/28/2020] [Indexed: 12/22/2022]
Abstract
Sorafenib is a small, orally-active multikinase inhibitor that is most frequently used for the management of renal cell carcinoma, hepatocellular carcinoma, and radioactive iodine-resistant thyroid carcinoma. However, recent reports have associated sorafenib with hepatotoxicity that can limit its clinical application, although the mechanism of hepatotoxicity is still to be elucidated. Thus, our study was designed to explore the molecular mechanisms underlying sorafenib-induced hepatotoxicity in an in vivo model. Twenty male adult Wistar rats were randomly placed into two groups; the first group received an oral dose of normal saline (vehicle), and the second received sorafenib (30 mg/kg) once daily for twenty-one consecutive days. After twenty-one days, liver tissues and blood samples were used for gene expression, protein expression, and biochemical analysis. Sorafenib treatment resulted in markedly increased levels of alanine aminotransferase and alkaline phosphatase, which indicate the presence of liver damage. Additionally, sorafenib administration induced the inflammatory and oxidative stress marker NF-κB-p65, while antioxidant enzymes were attenuated. Moreover, sorafenib caused upregulation of both gene and protein for the apoptotic markers cleaved Caspase-3, Bax, and Bid, and downregulation of the antiapoptotic protein Bcl-2. In conclusion, our findings suggest that sorafenib administration is associated with hepatotoxicity, which might be due to the activation of oxidative stress and apoptotic pathways.
Collapse
|
46
|
Shi Q, Yang X, Ren L, Mattes WB. Recent advances in understanding the hepatotoxicity associated with protein kinase inhibitors. Expert Opin Drug Metab Toxicol 2020; 16:217-226. [DOI: 10.1080/17425255.2020.1727886] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Qiang Shi
- Division of Systems Biology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, USA
| | - Xi Yang
- Division of Cardiovascular and Renal Products, Office of New Drugs I, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Lijun Ren
- Division of Systems Biology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, USA
| | - William B. Mattes
- Division of Systems Biology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, USA
| |
Collapse
|
47
|
Heslop KA, Rovini A, Hunt EG, Fang D, Morris ME, Christie CF, Gooz MB, DeHart DN, Dang Y, Lemasters JJ, Maldonado EN. JNK activation and translocation to mitochondria mediates mitochondrial dysfunction and cell death induced by VDAC opening and sorafenib in hepatocarcinoma cells. Biochem Pharmacol 2020; 171:113728. [PMID: 31759978 PMCID: PMC7309270 DOI: 10.1016/j.bcp.2019.113728] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 11/20/2019] [Indexed: 02/07/2023]
Abstract
The multikinase inhibitor sorafenib, and opening of voltage dependent anion channels (VDAC) by the erastin-like compound X1 promotes oxidative stress and mitochondrial dysfunction in hepatocarcinoma cells. Here, we hypothesized that X1 and sorafenib induce mitochondrial dysfunction by increasing reactive oxygen species (ROS) formation and activating c-Jun N-terminal kinases (JNKs), leading to translocation of activated JNK to mitochondria. Both X1 and sorafenib increased production of ROS and activated JNK. X1 and sorafenib caused a drop in mitochondrial membrane potential (ΔΨ), a readout of mitochondrial metabolism, after 60 min. Mitochondrial depolarization after X1 and sorafenib occurred in parallel with JNK activation, increased superoxide (O2•-) production, decreased basal and oligomycin sensitive respiration, and decreased maximal respiratory capacity. Increased production of O2•- after X1 or sorafenib was abrogated by JNK inhibition and antioxidants. S3QEL 2, a specific inhibitor of site IIIQo, at Complex III, prevented depolarization induced by X1. JNK inhibition by JNK inhibitors VIII and SP600125 also prevented mitochondrial depolarization. After X1, activated JNK translocated to mitochondria as assessed by proximity ligation assays. Tat-Sab KIM1, a peptide selectively preventing the binding of JNK to the outer mitochondrial membrane protein Sab, blocked the depolarization induced by X1 and sorafenib. X1 promoted cell death mostly by necroptosis that was partially prevented by JNK inhibition. These results indicate that JNK activation and translocation to mitochondria is a common mechanism of mitochondrial dysfunction induced by both VDAC opening and sorafenib.
Collapse
Affiliation(s)
- K A Heslop
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - A Rovini
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - E G Hunt
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - D Fang
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - M E Morris
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - C F Christie
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - M B Gooz
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - D N DeHart
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - Y Dang
- Department of Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - J J Lemasters
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States; Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC, United States; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - E N Maldonado
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States.
| |
Collapse
|
48
|
Klieser E, Mayr C, Kiesslich T, Wissniowski T, Fazio PD, Neureiter D, Ocker M. The Crosstalk of miRNA and Oxidative Stress in the Liver: From Physiology to Pathology and Clinical Implications. Int J Mol Sci 2019; 20:5266. [PMID: 31652839 PMCID: PMC6862076 DOI: 10.3390/ijms20215266] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/14/2019] [Accepted: 10/21/2019] [Indexed: 02/07/2023] Open
Abstract
The liver is the central metabolic organ of mammals. In humans, most diseases of the liver are primarily caused by an unhealthy lifestyle-high fat diet, drug and alcohol consumption- or due to infections and exposure to toxic substances like aflatoxin or other environmental factors. All these noxae cause changes in the metabolism of functional cells in the liver. In this literature review we focus on the changes at the miRNA level, the formation and impact of reactive oxygen species and the crosstalk between those factors. Both, miRNAs and oxidative stress are involved in the multifactorial development and progression of acute and chronic liver diseases, as well as in viral hepatitis and carcinogenesis, by influencing numerous signaling and metabolic pathways. Furthermore, expression patterns of miRNAs and antioxidants can be used for biomonitoring the course of disease and show potential to serve as possible therapeutic targets.
Collapse
Affiliation(s)
- Eckhard Klieser
- Institute of Pathology, Paracelsus Medical University/Salzburger Landeskliniken (SALK), 5020 Salzburg, Austria.
- Cancer Cluster Salzburg, 5020 Salzburg, Austria.
| | - Christian Mayr
- Department of Internal Medicine I, Paracelsus Medical University/Salzburger Landeskliniken (SALK), 5020 Salzburg, Austria.
- Institute of Physiology and Pathophysiology, Paracelsus Medical University/Salzburger Landeskliniken (SALK), 5020 Salzburg, Austria.
| | - Tobias Kiesslich
- Department of Internal Medicine I, Paracelsus Medical University/Salzburger Landeskliniken (SALK), 5020 Salzburg, Austria.
- Institute of Physiology and Pathophysiology, Paracelsus Medical University/Salzburger Landeskliniken (SALK), 5020 Salzburg, Austria.
| | - Till Wissniowski
- Department of Gastroenterology and Endocrinology, Philipps University Marburg, 35043 Marburg, Germany.
| | - Pietro Di Fazio
- Department of Visceral, Thoracic and Vascular Surgery, Philipps University Marburg, 35043 Marburg, Germany.
| | - Daniel Neureiter
- Institute of Pathology, Paracelsus Medical University/Salzburger Landeskliniken (SALK), 5020 Salzburg, Austria.
- Cancer Cluster Salzburg, 5020 Salzburg, Austria.
| | - Matthias Ocker
- Translational Medicine Oncology, Bayer AG, 13353 Berlin, Germany.
- Department of Gastroenterology CBF, Charité University Medicine Berlin, 12200 Berlin, Germany.
| |
Collapse
|
49
|
Garten A, Grohmann T, Kluckova K, Lavery GG, Kiess W, Penke M. Sorafenib-Induced Apoptosis in Hepatocellular Carcinoma Is Reversed by SIRT1. Int J Mol Sci 2019; 20:ijms20164048. [PMID: 31430957 PMCID: PMC6719220 DOI: 10.3390/ijms20164048] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 07/30/2019] [Accepted: 08/09/2019] [Indexed: 12/21/2022] Open
Abstract
Sorafenib is a multi-kinase inhibitor and one of the few systemic treatment options for patients with advanced hepatocellular carcinomas (HCCs). Resistance to sorafenib develops frequently and could be mediated by the nicotinamide adenine dinucleotide (NAD)-dependent deacetylase sirtuin (SIRT)1. We aimed to test whether sorafenib efficacy is influenced by cellular NAD levels and NAD-dependent SIRT1 function. We analyzed sorafenib effects on apoptosis induction, NAD salvage, mitochondrial function, and related signaling pathways in HCC cell lines (HepG2, Hep3B, und HUH7) overexpressing SIRT1 or supplemented with the NAD metabolite nicotinamide mononucleotide (NMN) compared to controls. Treatment of HCC cell lines with sorafenib dose-dependently induced apoptosis and a significant decrease in cellular NAD concentrations. The SIRT1 protein was downregulated in HUH7 cells but not in Hep3B cells. After sorafenib treatment, mitochondrial respiration in permeabilized cells was lower, citrate synthase activity was attenuated, and cellular adenosine triphosphate (ATP) levels were decreased. Concomitant to increased phosphorylation of adenosine monophosphate (AMP)-activated protein kinase (AMPK), sorafenib treatment led to decreased activity of the mechanistic target of rapamycin (mTOR), indicative of energy deprivation. Transient overexpression of SIRT1, as well as NAD repletion by NMN, decreased sorafenib-induced apoptosis. We can, therefore, conclude that sorafenib influences the NAD/SIRT1/AMPK axis. Overexpression of SIRT1 could be an underlying mechanism of resistance to sorafenib treatment in HCC.
Collapse
Affiliation(s)
- Antje Garten
- Center for Pediatric Research Leipzig (CPL), University Hospital for Children & Adolescents, Leipzig University, Liebigstr. 19, 04103 Leipzig, Germany.
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| | - Theresa Grohmann
- Center for Pediatric Research Leipzig (CPL), University Hospital for Children & Adolescents, Leipzig University, Liebigstr. 19, 04103 Leipzig, Germany
| | - Katarina Kluckova
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Gareth G Lavery
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Wieland Kiess
- Center for Pediatric Research Leipzig (CPL), University Hospital for Children & Adolescents, Leipzig University, Liebigstr. 19, 04103 Leipzig, Germany
| | - Melanie Penke
- Center for Pediatric Research Leipzig (CPL), University Hospital for Children & Adolescents, Leipzig University, Liebigstr. 19, 04103 Leipzig, Germany
| |
Collapse
|
50
|
Hurrell T, Segeritz CP, Vallier L, Lilley KS, Cromarty AD. Proteomic Comparison of Various Hepatic Cell Cultures for Preclinical Safety Pharmacology. Toxicol Sci 2019; 164:229-239. [PMID: 29635369 DOI: 10.1093/toxsci/kfy084] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Experimental drugs need to be screened for safety within time constraints. Hepatotoxicity is one concerning contributor to the failure of investigational new drugs and a major rationale for postmarketing withdrawal decisions. Ethical considerations in preclinical research force the requirement for highly predictive in vitro assays using human tissue which retains functionality reflective of primary tissue. Here, the proteome of cells commonly used to assess preclinical hepatotoxicity was compared. Primary human hepatocytes (PHHs), hepatocyte-like cells (HLCs) differentiated from human pluripotent stem cells, HepG2 cell monolayers and HepG2 cell 3D spheroids were cultured and collected as whole cell lysates. Over 6000 proteins were identified and quantified in terms of relative abundance in replicate proteomic experiments using isobaric tagging methods. Comparison of these quantitative data provides biological insight into the feasibility of using HLCs, HepG2 monolayers, and HepG2 3D spheroids for hepatotoxicity testing. Collectively these data reveal how HLCs differentiated for 35 days and HepG2 cells proteomes differ from one another and that of PHHs. HepG2 cells possess a strong cancer cell signature and do not adequately express key metabolic proteins which mark the hepatic phenotype, this was not substantially altered by culturing as 3D spheroids. These data suggest that while no single hepatic model reflects the diverse array of outcomes required to mimic the in vivo liver functions, that HLCs are the most suitable investigational avenue for replacing PHHs in vitro.
Collapse
Affiliation(s)
- Tracey Hurrell
- Department of Pharmacology, Faculty of Health Sciences, School of Medicine, University of Pretoria, Pretoria 0007, South Africa
| | - Charis-Patricia Segeritz
- Wellcome Trust-Medical Research Council Stem Cell Institute, Anne McLaren Laboratory for Regenerative Medicine, Department of Surgery, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Ludovic Vallier
- Wellcome Trust-Medical Research Council Stem Cell Institute, Anne McLaren Laboratory for Regenerative Medicine, Department of Surgery, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Kathryn S Lilley
- Department of Biochemistry, Cambridge Centre for Proteomics, University of Cambridge, Cambridge CB2 1QR, UK
| | - Allan Duncan Cromarty
- Department of Pharmacology, Faculty of Health Sciences, School of Medicine, University of Pretoria, Pretoria 0007, South Africa
| |
Collapse
|