1
|
Xue J, Lou X, Ning D, Yang Y, Shao R, Liu Y, Shi Z, Dai R, Wang F, Chen G. Ezetimibe Protects Against Alpha-Amanitin-Induced Hepatotoxicity by Targeting the NTCP Receptor: Mechanistic Insights from In Vitro and In Vivo Models. Toxicon 2025:108423. [PMID: 40449755 DOI: 10.1016/j.toxicon.2025.108423] [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: 02/17/2025] [Revised: 05/16/2025] [Accepted: 05/21/2025] [Indexed: 06/03/2025]
Abstract
BACKGROUND α-Amanitin, the primary lethal toxin of Amanita phalloides, induces irreversible hepatotoxicity by selectively inhibiting RNA polymerase II, leading to transcriptional arrest. Despite advancements in managing mushroom poisoning, a targeted antidote remains unavailable. The sodium taurocholate co-transporting polypeptide (NTCP), a hepatic bile acid transporter, facilitates α-amanitin entry into hepatocytes. Pharmacological blockade of NTCP represents a promising therapeutic strategy. OBJECTIVE To evaluate ezetimibe, an NTCP inhibitor, as a protective agent against α-amanitin-induced hepatotoxicity. METHODS Transcriptomic profiling of α-amanitin-exposed mouse liver tissues (NCBI accession: PRJNA809431) was conducted using DESeq2. Molecular docking simulations assessed interactions between NTCP, α-amanitin, and ezetimibe. Therapeutic efficacy was evaluated in vivo (mouse models) and in vitro (cultured hepatocytes). Key outcomes included survival rates, liver injury markers (ALT, AST), apoptosis (Bax/Bcl-2 ratio), and oxidative stress parameters. RESULTS NTCP expression was upregulated in α-amanitin-exposed livers. Molecular docking revealed α-amanitin binding at NTCP residue VAL-160, whereas ezetimibe interacted with LEU-14 and ASN-17. Ezetimibe (50 mg/kg) improved survival rates from 25% to 80% in α-amanitin-exposed mouse models (p < 0.01), reduced serum ALT (68 ± 5 U/L vs. 165 ± 12 U/L; p < 0.05) and AST (72 ± 6 U/L vs. 158 ± 10 U/L; p < 0.05), and attenuated apoptosis (60% decrease in Bax/Bcl-2; p < 0.05). In vitro, ezetimibe restored hepatocyte viability 2.1-fold (p < 0.05) and reduced oxidative stress (40% decrease in malondialdehyde; p < 0.05). Transcriptomic analysis linked α-amanitin toxicity to p53-mediated apoptosis. CONCLUSION Ezetimibe protects against α-amanitin hepatotoxicity by blocking NTCP-mediated uptake, supporting its potential clinical repurposing as a targeted antidote.
Collapse
Affiliation(s)
- Jinfang Xue
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Xiran Lou
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Deyuan Ning
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Yan Yang
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Ruifei Shao
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Yu Liu
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Zhuange Shi
- Medical School, Kunming University of Science and Technology, Kunming, China; Department of Emergency Medicine, The First People's Hospital of Yunnan Province, Kunming, China
| | - Ruanxian Dai
- Medical School, Kunming University of Science and Technology, Kunming, China; Department of Emergency Medicine, The First People's Hospital of Yunnan Province, Kunming, China
| | - Fuping Wang
- Department of Emergency Medicine, The First People's Hospital of Yunnan Province, Kunming, China
| | - Guobing Chen
- Department of Emergency Medicine, The First People's Hospital of Yunnan Province, Kunming, China.
| |
Collapse
|
2
|
Wang H, Zhang H, Miao L, Wang C, Teng H, Li X, Zhang X, Yang G, Wang S, Zeng X. α-amanitin induces hepatotoxicity via PPAR-γ inhibition and NLRP3 inflammasome activation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 290:117749. [PMID: 39862693 DOI: 10.1016/j.ecoenv.2025.117749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/07/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
Mushroom poisoning, predominantly caused by α-amanitin, is a critical food safety concern in worldwide, with severe cases leading to hepatotoxicity and fatalities. This study delves into the hepatotoxic effects of α-amanitin, focusing on the NLRP3 inflammasome and PPAR-γ's regulatory role in inflammation. In vitro studies with L-02 cells showed that α-amanitin reduces cell viability and triggers NLRP3 inflammasome activation, increasing NF-κB phosphorylation and pro-inflammatory cytokines IL-18 and IL-1β. The NLRP3 inhibitor MCC950 mitigated these effects without impacting NF-κB. Conversely, PPAR-γ knockdown intensified the inflammatory response. In vivo, α-amanitin induced dose-dependent liver injury in mice, evident by elevated serum ALT and AST, and histological liver damage. MCC950 pretreatment offered protection against hepatotoxicity, while PPAR-γ inhibition with GW9662 worsened the condition. The study highlights the interplay between α-amanitin, NLRP3, and PPAR-γ in hepatotoxicity, proposing potential therapeutic targets for mushroom poisoning-induced liver diseases.
Collapse
Affiliation(s)
- Haowei Wang
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Huijie Zhang
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Lin Miao
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Chan Wang
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Hanxin Teng
- Department of Pathogen Biology and Immunology, School of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Xiaodong Li
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Xiaoxing Zhang
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Genmeng Yang
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China.
| | - Shangwen Wang
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China.
| | - Xiaofeng Zeng
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China.
| |
Collapse
|
3
|
Zhao Z, Yi S, E H, Jiang L, Zhou C, Zhao X, Yang L. α-amanitin induce inflammatory response by activating ROS/NF-κB-NLRP3 signaling pathway in human hepatoma HepG2 cells. CHEMOSPHERE 2024; 364:143157. [PMID: 39178962 DOI: 10.1016/j.chemosphere.2024.143157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024]
Abstract
α-amanitin (AMA) is a hepatotoxic mushroom toxin responsible for over 90% of mushroom poisoning fatalities worldwide, seriously endangering human life and health. Few evidences have indicated that AMA leads to inflammatory responses and inflammatory infiltration in vitro and in vivo. However, the molecular mechanism remains unknown. In this study, human hepatocellular carcinomas cells (HepG2) were exposed to AMA at various concentrations for short period of times. Results revealed that AMA increased ROS production and elevated the releases of malondialdehyde (MDA) and lactate dehydrogenase (LDH), resulting in oxidative damage in HepG2 cells. Also, AMA exposure significantly increased the secreted levels of inflammatory cytokines and activated the NLRP3 inflammasome. The inflammatory responses were reversed by NLRP3 inhibitor MCC950 and NF-κB inhibitor Bay11-7082. Additionally, N-acetylcysteine (NAC) blocked the upregulation of the NF-κB/NLRP3 signaling pathway and remarkably alleviated the inflammatory response. These results demonstrated that AMA could induce inflammation through activating the NLRP3 inflammasome triggered by ROS/NF-κB signaling pathway. Our research provides new insights into the molecular mechanism of AMA-induced inflammation damage and may contribute to establish new prevention strategies for AMA hepatotoxicity.
Collapse
Affiliation(s)
- Zhiyong Zhao
- Institute for Agro-food Standards and Testing Technology, Laboratory of Quality & Safety Risk Assessment for Agro-products (Shanghai), Ministry of Agriculture and Rural Affairs, Shanghai Academy of Agricultural Sciences, No.1000 Jinqi Road, Shanghai, 201403, PR China; Shanghai Guosen Biotechnology Co., Ltd., Shanghai, 201400, PR China.
| | - Siliang Yi
- Institute for Agro-food Standards and Testing Technology, Laboratory of Quality & Safety Risk Assessment for Agro-products (Shanghai), Ministry of Agriculture and Rural Affairs, Shanghai Academy of Agricultural Sciences, No.1000 Jinqi Road, Shanghai, 201403, PR China; College of Veterinary Medicine, Hunan Agricultural University, No.1 Nongda Road, Changsha, 410128, PR China
| | - Hengchao E
- Institute for Agro-food Standards and Testing Technology, Laboratory of Quality & Safety Risk Assessment for Agro-products (Shanghai), Ministry of Agriculture and Rural Affairs, Shanghai Academy of Agricultural Sciences, No.1000 Jinqi Road, Shanghai, 201403, PR China
| | - Lihuang Jiang
- Institute for Agro-food Standards and Testing Technology, Laboratory of Quality & Safety Risk Assessment for Agro-products (Shanghai), Ministry of Agriculture and Rural Affairs, Shanghai Academy of Agricultural Sciences, No.1000 Jinqi Road, Shanghai, 201403, PR China; College of Veterinary Medicine, Hunan Agricultural University, No.1 Nongda Road, Changsha, 410128, PR China
| | - Changyan Zhou
- Institute for Agro-food Standards and Testing Technology, Laboratory of Quality & Safety Risk Assessment for Agro-products (Shanghai), Ministry of Agriculture and Rural Affairs, Shanghai Academy of Agricultural Sciences, No.1000 Jinqi Road, Shanghai, 201403, PR China
| | - Xiaoyan Zhao
- Institute for Agro-food Standards and Testing Technology, Laboratory of Quality & Safety Risk Assessment for Agro-products (Shanghai), Ministry of Agriculture and Rural Affairs, Shanghai Academy of Agricultural Sciences, No.1000 Jinqi Road, Shanghai, 201403, PR China.
| | - Lingchen Yang
- College of Veterinary Medicine, Hunan Agricultural University, No.1 Nongda Road, Changsha, 410128, PR China.
| |
Collapse
|
4
|
Wang B, Xu Y, Wan AH, Wan G, Wang QP. Integrating genome-wide CRISPR screens and in silico drug profiling for targeted antidote development. Nat Protoc 2024; 19:2739-2770. [PMID: 38816517 DOI: 10.1038/s41596-024-00995-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 02/29/2024] [Indexed: 06/01/2024]
Abstract
Numerous toxins threaten humans, but specific antidotes are unavailable for most of them. Although CRISPR screening has aided the discovery of the mechanisms of some toxins, developing targeted antidotes remains a significant challenge. Recently, we established a systematic framework to develop antidotes by combining the identification of novel drug targets by using a genome-wide CRISPR screen with a virtual screen of drugs approved by the US Food and Drug Administration. This approach allows for a comprehensive understanding of toxin mechanisms at the whole-genome level and facilitates the identification of promising antidote drugs targeting specific molecules. Here, we present step-by-step instructions for executing genome-scale CRISPR-Cas9 knockout screens of toxins in HAP1 cells. We also provide detailed guidance for conducting an in silico drug screen and an in vivo drug validation. By using this protocol, it takes ~4 weeks to perform the genome-scale screen, 4 weeks for sequencing and data analysis, 4 weeks to validate candidate genes, 1 week for the virtual screen and 2 weeks for in vitro drug validation. This framework has the potential to accelerate the development of antidotes for a wide range of toxins and can rapidly identify promising drug candidates that are already known to be safe and effective. This could lead to the development of new antidotes much more quickly than traditional methods, protecting lives from diverse toxins and advancing human health.
Collapse
Affiliation(s)
- Bei Wang
- Laboratory of Metabolism and Aging, School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, P. R. China
| | - Yu Xu
- Laboratory of Metabolism and Aging, School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, P. R. China
| | - Arabella H Wan
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China
| | - Guohui Wan
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, P. R. China.
| | - Qiao-Ping Wang
- Laboratory of Metabolism and Aging, School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, P. R. China.
- Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
5
|
Gong M, Li Z, Xu H, Ma B, Gao P, Wang L, Li J, Wu Q, Wu J, Xie J. Amanitin-induced variable cytotoxicity in various cell lines is mediated by the different expression levels of OATP1B3. Food Chem Toxicol 2024; 188:114665. [PMID: 38641045 DOI: 10.1016/j.fct.2024.114665] [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: 02/02/2024] [Revised: 03/02/2024] [Accepted: 04/11/2024] [Indexed: 04/21/2024]
Abstract
Amanita phalloides is one of the deadliest mushrooms worldwide, causing most fatal cases of mushroom poisoning. Among the poisonous substances of Amanita phalloides, amanitins are the most lethal toxins to humans. Currently, there are no specific antidotes available for managing amanitin poisoning and treatments are lack of efficacy. Amanitin mainly causes severe injuries to specific organs, such as the liver, stomach, and kidney, whereas the lung, heart, and brain are hardly affected. However, the molecular mechanism of this phenomenon remains not understood. To explore the possible mechanism of organ specificity of amanitin-induced toxicity, eight human cell lines derived from different organs were exposed to α, β, and γ-amanitin at concentrations ranging from 0.3 to 100 μM. We found that the cytotoxicity of amanitin differs greatly in various cell lines, among which liver-derived HepG2, stomach-derived BGC-823, and kidney-derived HEK-293 cells are most sensitive. Further mechanistic study revealed that the variable cytotoxicity is mainly dependent on the different expression levels of the organic anion transporting polypeptide 1B3 (OATP1B3), which facilitates the internalization of amanitin into cells. Besides, knockdown of OATP1B3 in HepG2 cells prevented α-amanitin-induced cytotoxicity. These results indicated that OATP1B3 may be a crucial therapeutic target against amanitin-induced organ failure.
Collapse
Affiliation(s)
- Mengqiang Gong
- School of Agriculture, Yangtze University, Jingzhou, 434025, China; Laboratory of Toxicant Analysis, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Zhi Li
- Laboratory of Toxicant Analysis, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Hua Xu
- Laboratory of Toxicant Analysis, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Bo Ma
- Laboratory of Toxicant Analysis, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Pengxia Gao
- Laboratory of Toxicant Analysis, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Lili Wang
- Laboratory of Toxicant Analysis, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Junkai Li
- School of Agriculture, Yangtze University, Jingzhou, 434025, China
| | - Qinglai Wu
- School of Agriculture, Yangtze University, Jingzhou, 434025, China.
| | - Jianfeng Wu
- Laboratory of Toxicant Analysis, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China.
| | - Jianwei Xie
- Laboratory of Toxicant Analysis, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China.
| |
Collapse
|
6
|
Gouvinhas I, Silva J, Alves MJ, Garcia J. The most dreadful mushroom toxins: a review of their toxicological mechanisms, chemical structural characteristics, and treatment. EXCLI JOURNAL 2024; 23:833-859. [PMID: 39165585 PMCID: PMC11333700 DOI: 10.17179/excli2024-7257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/26/2024] [Indexed: 08/22/2024]
Abstract
Mushroom consumption is a worldwide custom that continues to grow in popularity. On the other hand, foraging for wild mushrooms can lead to serious disease and even death if deadly mushrooms are accidentally consumed. Mushroom poisoning is difficult to diagnose and treat since the symptoms are similar to those of other disorders. In terms of chemistry, mushroom poisoning is associated with extraordinarily strong toxins, meaning that isolating and identifying toxins has substantial scientific relevance, especially in understanding the lethal components of toxic mushrooms. Most of these toxins exhibit exceptional physiological features that might help enhance chemistry, biochemistry, physiology, and pharmacology research. Despite the discovery of more than 100 poisons, several dangerous mushrooms remain unexplored. This review covers the chemistry (including chemical structures, complete synthesis, and biosynthesis), as well as the toxicology, namely the toxicokinetics, mechanisms of toxicology, and clinical toxicology of these poisons, in addition to the discussion of the development of their most effective diagnostic and therapeutic strategies with the hopes of spurring additional studies, focusing on individual classes of toxins found in poisonous mushrooms such as amatoxins, gyromitrin, orellanine, and phallatoxins. See also the graphical abstract(Fig. 1).
Collapse
Affiliation(s)
- Irene Gouvinhas
- CITAB - Centre for the Research and Technology of Agro-Environment and Biological Sciences/ Inov4Agro - Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production, University of Trás-os-Montes e Alto Douro, 5001- 801 Vila Real, Portugal
| | - Jani Silva
- AquaValor - Centro de Valorização e Transferência de Tecnologia da Água - Associação, Rua Dr. Júlio Martins n.º 1, 5400-342 Chaves, Portugal
| | - Maria José Alves
- AquaValor - Centro de Valorização e Transferência de Tecnologia da Água - Associação, Rua Dr. Júlio Martins n.º 1, 5400-342 Chaves, Portugal
| | - Juliana Garcia
- CITAB - Centre for the Research and Technology of Agro-Environment and Biological Sciences/ Inov4Agro - Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production, University of Trás-os-Montes e Alto Douro, 5001- 801 Vila Real, Portugal
- AquaValor - Centro de Valorização e Transferência de Tecnologia da Água - Associação, Rua Dr. Júlio Martins n.º 1, 5400-342 Chaves, Portugal
| |
Collapse
|
7
|
Popescu M, Bratu A, Agapie M, Borjog T, Jafal M, Sima RM, Orban C. The Use and Potential Benefits of N-Acetylcysteine in Non-Acetaminophen Acute Liver Failure: An Etiology-Based Review. Biomedicines 2024; 12:676. [PMID: 38540289 PMCID: PMC10967777 DOI: 10.3390/biomedicines12030676] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 03/04/2024] [Accepted: 03/15/2024] [Indexed: 08/13/2024] Open
Abstract
Acute liver failure represents a life-threatening organ dysfunction with high mortality rates and an urgent need for liver transplantation. The etiology of the disease varies widely depending on various socio-economic factors and is represented mainly by paracetamol overdose and other drug-induced forms of liver dysfunction in the developed world and by viral hepatitis and mushroom poisoning in less developed countries. Current medical care constitutes either specific antidotes or supportive measures to ensure spontaneous recovery. Although it has been proven to have beneficial effects in paracetamol-induced liver failure, N-acetylcysteine is widely used for all forms of acute liver failure. Despite this, few well-designed studies have been conducted on the assessment of the potential benefits, dose regimens, or route of administration of N-acetylcysteine in non-acetaminophen liver failure. This review aims to summarize the current evidence behind the use of this drug in different forms of liver failure.
Collapse
Affiliation(s)
- Mihai Popescu
- Department of Anaesthesia and Intensive Care, “Carol Davila” University of Medicine and Pharmacy, 37 Dionisie Lupu Street, 020021 Bucharest, Romania; (M.A.); (T.B.); (M.J.); (C.O.)
- Department of Anaesthesia and Intensive Care, Bucharest University Emergency Hospital, 169 Independentei Street, 050098 Bucharest, Romania;
| | - Angelica Bratu
- Department of Anaesthesia and Intensive Care, Bucharest University Emergency Hospital, 169 Independentei Street, 050098 Bucharest, Romania;
| | - Mihaela Agapie
- Department of Anaesthesia and Intensive Care, “Carol Davila” University of Medicine and Pharmacy, 37 Dionisie Lupu Street, 020021 Bucharest, Romania; (M.A.); (T.B.); (M.J.); (C.O.)
- Department of Anaesthesia and Intensive Care, Bucharest University Emergency Hospital, 169 Independentei Street, 050098 Bucharest, Romania;
| | - Tudor Borjog
- Department of Anaesthesia and Intensive Care, “Carol Davila” University of Medicine and Pharmacy, 37 Dionisie Lupu Street, 020021 Bucharest, Romania; (M.A.); (T.B.); (M.J.); (C.O.)
- Department of Anaesthesia and Intensive Care, Bucharest University Emergency Hospital, 169 Independentei Street, 050098 Bucharest, Romania;
| | - Mugurel Jafal
- Department of Anaesthesia and Intensive Care, “Carol Davila” University of Medicine and Pharmacy, 37 Dionisie Lupu Street, 020021 Bucharest, Romania; (M.A.); (T.B.); (M.J.); (C.O.)
- Department of Anaesthesia and Intensive Care, Bucharest University Emergency Hospital, 169 Independentei Street, 050098 Bucharest, Romania;
| | - Romina-Marina Sima
- Department of Obstetrics and Gynecology, “Carol Davila” University of Medicine and Pharmacy, 37 Dionisie Lupu Street, 020021 Bucharest, Romania;
| | - Carmen Orban
- Department of Anaesthesia and Intensive Care, “Carol Davila” University of Medicine and Pharmacy, 37 Dionisie Lupu Street, 020021 Bucharest, Romania; (M.A.); (T.B.); (M.J.); (C.O.)
- Department of Anaesthesia and Intensive Care, Bucharest University Emergency Hospital, 169 Independentei Street, 050098 Bucharest, Romania;
| |
Collapse
|
8
|
Li Z, Ma B, Xu H, Gong M, Gao P, Wang L, Xie J. Divinyl sulfone, an oxidative metabolite of sulfur mustard, induces caspase-independent pyroptosis in hepatocytes. Arch Toxicol 2024; 98:897-909. [PMID: 38172301 DOI: 10.1007/s00204-023-03662-6] [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: 10/10/2023] [Accepted: 12/07/2023] [Indexed: 01/05/2024]
Abstract
Sulfur mustard (SM) is a highly toxic blister agent which has been used many times in several wars and conflicts and caused heavy casualties. Ease of production and lack of effective therapies make SM a potential threat to public health. SM intoxication causes severe damage on various target organs, such as the skin, eyes, and lungs. In addition, SM exposure can also lead to hepatotoxicity and severe liver injuries. However, despite decades of research, the molecular mechanism underlying SM-induced liver damage remains obscure. SM can be converted into various products via complex hepatic metabolism in vivo. There are some pieces of evidence that one of the oxidation products of SM, divinyl sulfone (DVS), exhibits even more significant toxicity than SM. Nevertheless, the molecular toxicology of DVS is still hardly known. In the present study, we confirmed that DVS is even more toxic than SM in the human hepatocellular carcinoma cell line HepG2. Further mechanistic study revealed that DVS exposure (200 μM) promotes pyroptosis in HepG2 cells, while SM (400 μM) mainly induces apoptosis. DVS induces gasdermin D (GSDMD) mediated pyroptosis, which is independent of caspases activation but depends on the large amounts of reactive oxygen species (ROS) and severe oxidative stress produced during DVS exposure. Our findings may provide novel insights for understanding the mechanism of SM poisoning and may be helpful to discover promising therapeutic strategies for SM intoxication.
Collapse
Affiliation(s)
- Zhi Li
- Laboratory of Toxicant Analysis, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Bo Ma
- Laboratory of Toxicant Analysis, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Hua Xu
- Laboratory of Toxicant Analysis, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China.
| | - Mengqiang Gong
- Laboratory of Toxicant Analysis, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Pengxia Gao
- Laboratory of Toxicant Analysis, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Lili Wang
- Laboratory of Toxicant Analysis, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Jianwei Xie
- Laboratory of Toxicant Analysis, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China.
| |
Collapse
|
9
|
Visser M, Hof WFJ, Broek AM, van Hoek A, de Jong JJ, Touw DJ, Dekkers BGJ. Unexpected Amanita phalloides-Induced Hematotoxicity-Results from a Retrospective Study. Toxins (Basel) 2024; 16:67. [PMID: 38393145 PMCID: PMC10891511 DOI: 10.3390/toxins16020067] [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: 12/19/2023] [Revised: 01/17/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
INTRODUCTION Amanita phalloides poisoning is a serious health problem with a mortality rate of 10-40%. Poisonings are characterized by severe liver and kidney toxicity. The effect of Amanita phalloides poisonings on hematological parameters has not been systematically evaluated thus far. METHODS Patients with suspected Amanita phalloides poisonings were retrospectively selected from the hospital database of the University Medical Center Groningen (UMCG). Medical data-including demographics; liver, kidney, and blood parameters; treatment; and outcomes-were collected. The severity of the poisoning was scored using the poison severity score. RESULTS Twenty-eight patients were identified who were admitted to the UMCG with suspected Amanita phalloides poisoning between 1994 and 2022. A time-dependent decrease was observed for hemoglobin and hematocrit concentrations, leukocytes, and platelets. Six out of twenty-eight patients developed acute liver failure (ALF). Patients with ALF showed a higher increase in liver enzymes, international normalized ratios, and PSS compared to patients without ALF. Conversely, hemoglobin and platelet numbers were decreased even further in these patients. Three out of six patients with ALF died and one patient received a liver transplant. CONCLUSION Our study shows that Amanita phalloides poisonings may be associated with hematotoxicity in patients. The quantification of hematological parameters is of relevance in intoxicated patients, especially in those with ALF.
Collapse
Affiliation(s)
- Miranda Visser
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen (UMCG), 9713 GZ Groningen, The Netherlands; (M.V.); (W.F.J.H.); (A.M.B.); (A.v.H.); (J.J.d.J.); (D.J.T.)
| | - Willemien F. J. Hof
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen (UMCG), 9713 GZ Groningen, The Netherlands; (M.V.); (W.F.J.H.); (A.M.B.); (A.v.H.); (J.J.d.J.); (D.J.T.)
| | - Astrid M. Broek
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen (UMCG), 9713 GZ Groningen, The Netherlands; (M.V.); (W.F.J.H.); (A.M.B.); (A.v.H.); (J.J.d.J.); (D.J.T.)
| | - Amanda van Hoek
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen (UMCG), 9713 GZ Groningen, The Netherlands; (M.V.); (W.F.J.H.); (A.M.B.); (A.v.H.); (J.J.d.J.); (D.J.T.)
| | - Joyce J. de Jong
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen (UMCG), 9713 GZ Groningen, The Netherlands; (M.V.); (W.F.J.H.); (A.M.B.); (A.v.H.); (J.J.d.J.); (D.J.T.)
| | - Daan J. Touw
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen (UMCG), 9713 GZ Groningen, The Netherlands; (M.V.); (W.F.J.H.); (A.M.B.); (A.v.H.); (J.J.d.J.); (D.J.T.)
- Department of Pharmaceutical Analysis, Groningen Research Institute of Pharmacy, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Bart G. J. Dekkers
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen (UMCG), 9713 GZ Groningen, The Netherlands; (M.V.); (W.F.J.H.); (A.M.B.); (A.v.H.); (J.J.d.J.); (D.J.T.)
| |
Collapse
|
10
|
Hof WFJ, Visser M, de Jong JJ, Rajasekar MN, Schuringa JJ, de Graaf IAM, Touw DJ, Dekkers BGJ. Unraveling Hematotoxicity of α-Amanitin in Cultured Hematopoietic Cells. Toxins (Basel) 2024; 16:61. [PMID: 38276537 PMCID: PMC10820516 DOI: 10.3390/toxins16010061] [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: 12/19/2023] [Revised: 01/13/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024] Open
Abstract
Amanita phalloides poisonings account for the majority of fatal mushroom poisonings. Recently, we identified hematotoxicity as a relevant aspect of Amanita poisonings. In this study, we investigated the effects of the main toxins of Amanita phalloides, α- and β-amanitin, on hematopoietic cell viability in vitro. Hematopoietic cell lines were exposed to α-amanitin or β-amanitin for up to 72 h with or without the pan-caspase inhibitor Z-VAD(OH)-FMK, antidotes N-acetylcysteine, silibinin, and benzylpenicillin, and organic anion-transporting polypeptide 1B3 (OATP1B3) inhibitors rifampicin and cyclosporin. Cell viability was established by trypan blue exclusion, annexin V staining, and a MTS assay. Caspase-3/7 activity was determined with Caspase-Glo assay, and cleaved caspase-3 was quantified by Western analysis. Cell number and colony-forming units were quantified after exposure to α-amanitin in primary CD34+ hematopoietic stem cells. In all cell lines, α-amanitin concentration-dependently decreased viability and mitochondrial activity. β-Amanitin was less toxic, but still significantly reduced viability. α-Amanitin increased caspase-3/7 activity by 2.8-fold and cleaved caspase-3 by 2.3-fold. Z-VAD(OH)-FMK significantly reduced α-amanitin-induced toxicity. In CD34+ stem cells, α-amanitin decreased the number of colonies and cells. The antidotes and OATP1B3 inhibitors did not reverse α-amanitin-induced toxicity. In conclusion, α-amanitin induces apoptosis in hematopoietic cells via a caspase-dependent mechanism.
Collapse
Affiliation(s)
- Willemien F. J. Hof
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen (UMCG), 9713 GZ Groningen, The Netherlands; (W.F.J.H.)
| | - Miranda Visser
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen (UMCG), 9713 GZ Groningen, The Netherlands; (W.F.J.H.)
| | - Joyce J. de Jong
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen (UMCG), 9713 GZ Groningen, The Netherlands; (W.F.J.H.)
| | - Marian N. Rajasekar
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen (UMCG), 9713 GZ Groningen, The Netherlands; (W.F.J.H.)
| | - Jan Jacob Schuringa
- Department of Experimental Hematology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Inge A. M. de Graaf
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen (UMCG), 9713 GZ Groningen, The Netherlands; (W.F.J.H.)
| | - Daan J. Touw
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen (UMCG), 9713 GZ Groningen, The Netherlands; (W.F.J.H.)
| | - Bart G. J. Dekkers
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen (UMCG), 9713 GZ Groningen, The Netherlands; (W.F.J.H.)
| |
Collapse
|
11
|
Kim D, Lee MS, Kim ND, Lee S, Lee HS. Identification of α-amanitin effector proteins in hepatocytes by limited proteolysis-coupled mass spectrometry. Chem Biol Interact 2023; 386:110778. [PMID: 37879594 DOI: 10.1016/j.cbi.2023.110778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/02/2023] [Accepted: 10/22/2023] [Indexed: 10/27/2023]
Abstract
The misuse of poisonous mushrooms containing amatoxins causes acute liver failure (ALF) in patients and is a cause of significant mortality. Although the toxic mechanisms of α-amanitin (α-AMA) and its interactions with RNA polymerase II (RNAP II) have been studied, α-AMA effector proteins that can interact with α-AMA in hepatocytes have not been systematically studied. Limited proteolysis-coupled mass spectrometry (LiP-MS) is an advanced technology that can quickly identify protein-ligand interactions based on global comparative proteomics. This study identified the α-AMA effector proteins found in human hepatocytes, following the detection of conformotypic peptides using LiP-MS coupled with tandem mass tag (TMT) technology. Proteins that are classified into protein processing in the endoplasmic reticulum and the ribosome during the KEGG pathway can be identified through affinity evaluation, according to α-AMA concentration-dependent LiP-MS and LiP-MS in hepatocytes derived from humans and mice, respectively. The possibility of interaction between α-AMA and proteins containing conformotypic peptides was evaluated through molecular docking studies. The results of this study suggest a novel path for α-AMA to induce hepatotoxicity through interactions with various proteins involved in protein synthesis, as well as with RNAP II.
Collapse
Affiliation(s)
- Doeun Kim
- College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Min Seo Lee
- BK21 Four-sponsored Advanced Program for SmartPharma Leaders, College of Pharmacy, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - Nam Doo Kim
- Voronoibio Inc., Incheon, 21984, Republic of Korea
| | - Sangkyu Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Hye Suk Lee
- BK21 Four-sponsored Advanced Program for SmartPharma Leaders, College of Pharmacy, The Catholic University of Korea, Bucheon, 14662, Republic of Korea.
| |
Collapse
|
12
|
Zheng C, Lv S, Ye J, Zou L, Zhu K, Li H, Dong Y, Li L. Metabolomic Insights into the Mechanisms of Ganoderic Acid: Protection against α-Amanitin-Induced Liver Injury. Metabolites 2023; 13:1164. [PMID: 37999259 PMCID: PMC10672867 DOI: 10.3390/metabo13111164] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 11/16/2023] [Accepted: 11/18/2023] [Indexed: 11/25/2023] Open
Abstract
α-Amanitin is a representative toxin found in the Amanita genus of mushrooms, and the consumption of mushrooms containing α-Amanitin can lead to severe liver damage. In this study, we conduct toxicological experiments to validate the protective effects of Ganoderic acid A against α-amanitin-induced liver damage. By establishing animal models with different durations of Ganoderic acid A treatment and conducting a metabolomic analysis of the serum samples, we further confirmed the differences in serum metabolites between the AMA+GA and AMA groups. The analysis of differential serum metabolites after the Ganoderic acid A intervention suggests that Ganoderic acid A may intervene in α-amanitin-induced liver damage by participating in the regulation of retinol metabolism, tyrosine and tryptophan biosynthesis, fatty acid biosynthesis, sphingosine biosynthesis, spermidine and spermine biosynthesis, and branched-chain amino acid metabolism. This provides initial insights into the protective intervention mechanisms of GA against α-amanitin-induced liver damage and offers new avenues for the development of therapeutic drugs for α-Amanitin poisoning.
Collapse
Affiliation(s)
- Chong Zheng
- Guizhou Provincial Center for Disease Control and Prevention, Guiyang 550004, China; (C.Z.)
| | - Shaofang Lv
- School of Pharmacy, Guizhou Medical University, Guiyang 550025, China
| | - Jianfang Ye
- Guizhou Provincial Center for Disease Control and Prevention, Guiyang 550004, China; (C.Z.)
| | - Lu Zou
- Guizhou Provincial Center for Disease Control and Prevention, Guiyang 550004, China; (C.Z.)
| | - Kai Zhu
- Guizhou Provincial Center for Disease Control and Prevention, Guiyang 550004, China; (C.Z.)
| | - Haichang Li
- Guiyang Provincial Center for Disease Control and Prevention, Guiyang 550002, China
| | - Yongxi Dong
- School of Pharmacy, Guizhou Medical University, Guiyang 550025, China
| | - Lei Li
- Guizhou Provincial Center for Disease Control and Prevention, Guiyang 550004, China; (C.Z.)
| |
Collapse
|
13
|
Xu Y, Wang S, Leung CK, Chen H, Wang C, Zhang H, Zhang S, Tan Y, Wang H, Miao L, Li Y, Huang Y, Zhang X, Yang G, Zhang R, Zeng X. α-amanitin induces autophagy through AMPK-mTOR-ULK1 signaling pathway in hepatocytes. Toxicol Lett 2023:S0378-4274(23)00204-7. [PMID: 37329965 DOI: 10.1016/j.toxlet.2023.06.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 03/28/2023] [Accepted: 06/14/2023] [Indexed: 06/19/2023]
Abstract
Amanitin poisoning is one of the most life-threatening mushroom poisonings. α-Amanitin plays a key role in Amanita phalloides intoxication. α-Amanitin shows toxic effects on the liver. However, the mechanism by which α-amanitin induces liver injury has not been elucidated. Autophagy plays a crucial role in maintaining cellular homeostasis and is closely related to the occurrence of a variety of diseases. Studies have shown that autophagy may play an important role in the process of α-amanitin-induced liver injury. However, the mechanism of α-amanitin-induced autophagy remains unclear. Thus, this study aimed to explore the mechanisms of α-amanitin in inducing hepatotoxicity in Sprague Dawley (SD) rats and the normal human liver cell line L02 cells. The SD rats and L02 cells exposed to α-amanitin were observed to determine whether α-amanitin could induce the autophagy of rat liver and L02 cells. The regulatory relationship between autophagy and the AMPK-mTOR- ULK pathway by exposing the autophagy agonist (rapamycin (RAPA)), autophagy inhibitor (3-methylademine (3-MA)), and AMPK inhibitor (compound C) was also explored. Autophagy-related proteins and AMPK-mTOR-ULK pathway-related proteins were detected using Western blot. The results of the study indicated that exposure to different concentrations of α-amanitin led to morphological changes in liver cells and significantly elevated levels of ALT and AST in the serum of SD rats. Additionally, the expression levels of LC3-II, Beclin-1, ATG5, ATG7, AMPK, p-AMPK, mTOR, p-mTOR, and ULK1 were significantly increased in the rat liver. And we found that L02 cells exposed to 0.5μM α-amanitin for 6h significantly induced autophagy and activated the AMPK-mTOR-ULK1 pathway. Pretreated with RAPA, 3-MA, and compound C for 1h, the expression levels of autophagy-related proteins and AMPK-mTOR-ULK pathway-related proteins significantly changed. Our results indicates that autophagy and the AMPK-mTOR-ULK pathway are involved in the process of α-amanitin-induced liver injury. This study may foster the identification of actionable therapeutic targets for A. phalloides intoxication.
Collapse
Affiliation(s)
- Yue Xu
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Shangwen Wang
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Chi-Kwan Leung
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong; CUHK-SDU Joint Laboratory of Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Hao Chen
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Chan Wang
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Huijie Zhang
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Shuwei Zhang
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Yi Tan
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Haowei Wang
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Lin Miao
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Yi Li
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Yizhen Huang
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Xiaoxing Zhang
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Genmeng Yang
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China.
| | - Ruilin Zhang
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China.
| | - Xiaofeng Zeng
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China.
| |
Collapse
|
14
|
Wang B, Wan AH, Xu Y, Zhang RX, Zhao BC, Zhao XY, Shi YC, Zhang X, Xue Y, Luo Y, Deng Y, Neely GG, Wan G, Wang QP. Identification of indocyanine green as a STT3B inhibitor against mushroom α-amanitin cytotoxicity. Nat Commun 2023; 14:2241. [PMID: 37193694 PMCID: PMC10188588 DOI: 10.1038/s41467-023-37714-3] [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: 07/07/2022] [Accepted: 03/21/2023] [Indexed: 05/18/2023] Open
Abstract
The "death cap", Amanita phalloides, is the world's most poisonous mushroom, responsible for 90% of mushroom-related fatalities. The most fatal component of the death cap is α-amanitin. Despite its lethal effect, the exact mechanisms of how α-amanitin poisons humans remain unclear, leading to no specific antidote available for treatment. Here we show that STT3B is required for α-amanitin toxicity and its inhibitor, indocyanine green (ICG), can be used as a specific antidote. By combining a genome-wide CRISPR screen with an in silico drug screening and in vivo functional validation, we discover that N-glycan biosynthesis pathway and its key component, STT3B, play a crucial role in α-amanitin toxicity and that ICG is a STT3B inhibitor. Furthermore, we demonstrate that ICG is effective in blocking the toxic effect of α-amanitin in cells, liver organoids, and male mice, resulting in an overall increase in animal survival. Together, by combining a genome-wide CRISPR screen for α-amanitin toxicity with an in silico drug screen and functional validation in vivo, our study highlights ICG as a STT3B inhibitor against the mushroom toxin.
Collapse
Affiliation(s)
- Bei Wang
- Laboratory of Metabolism and Aging, School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, PR China
| | - Arabella H Wan
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Yu Xu
- Laboratory of Metabolism and Aging, School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, PR China
| | - Ruo-Xin Zhang
- Laboratory of Metabolism and Aging, School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, PR China
| | - Ben-Chi Zhao
- Laboratory of Metabolism and Aging, School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, PR China
| | - Xin-Yuan Zhao
- Laboratory of Metabolism and Aging, School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, PR China
| | - Yan-Chuan Shi
- Obesity and Metabolic Disease Research Group, Diabetes and Metabolism Division, Garvan Institute of Medical Research, Darlinghurst, Sydney, Australia
| | - Xiaolei Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Yongbo Xue
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, PR China
| | - Yong Luo
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, PR China
| | - Yinyue Deng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, PR China
| | - G Gregory Neely
- Dr. John and Anne Chong Laboratory for Functional Genomics, Charles Perkins Centre and School of Life & Environmental Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Guohui Wan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, PR China.
| | - Qiao-Ping Wang
- Laboratory of Metabolism and Aging, School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, PR China.
| |
Collapse
|
15
|
Liu Y, Li S, Feng Y, Zhang Y, Ouyang J, Li S, Wang J, Tan L, Zou L. Serum metabolomic analyses reveal the potential metabolic biomarkers for prediction of amatoxin poisoning. Toxicon 2023; 230:107153. [PMID: 37178797 DOI: 10.1016/j.toxicon.2023.107153] [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: 03/06/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023]
Abstract
Amatoxin poisoning leads to over 90% of deaths in mushroom poisoning. The objective of present study was to identify the potential metabolic biomarkers for early diagnosis of amatoxin poisoning. Serum samples were collected from 61 patients with amatoxin poisoning and 61 healthy controls. An untargeted metabolomics analysis was performed using the ultra-high-performance liquid chromatography-quadrupole time-of-flight tandem mass spectrometry (UPLC-QTOF-MS/MS). Multivariate statistical analysis revealed that the patients with amatoxin poisoning could be clearly separated from healthy controls on the basis of their metabolic fingerprints. There were 33 differential metabolites including 15 metabolites up-regulated metabolites and 18 down-regulated metabolites in patients with amatoxin poisoning compared to healthy controls. These metabolites mainly enriched in the lipid metabolism and amino acid metabolism pathways, such as Glycerophospholipid metabolism, Sphingolipid metabolism, Phenylalanine tyrosine and typtophan biosynthesis, Tyrosine metabolism, Arginine and proline metabolism, which may serve important roles in the amatoxin poisoning. Among the differential metabolites, a total of 8 significant metabolic markers were identified for discriminating patients with amatoxin poisoning from healthy controls, including Glycochenodeoxycholate-3-sulfate (GCDCA-S), 11-Oxo-androsterone glucuronide, Neomenthol-glucuronide, Dehydroisoandrosterone 3-glucuronide, Glucose 6-phosphate (G6P), Lanthionine ketimine, Glycerophosphocholine (GPC) and Nicotinamide ribotide, which achieved satisfactory diagnostic accuracy (AUC>0.8) in both discovery and validation cohorts. Strikingly, the Pearson's correlation analysis indicated that 11-Oxo-androsterone glucuronide, G6P and GCDCA-S were positively correlated with the liver injury induced by amatoxin poisoning. The findings of the current study may provide insight into the pathological mechanism of amatoxin poisoning and screened out the reliable metabolic biomarkers to contribute the clinical early diagnosis of amatoxin poisoning.
Collapse
Affiliation(s)
- Yarong Liu
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan, 410013, PR China; Institute of Clinical Translational Medicine, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan, 410005, PR China; Key Laboratory of Molecular Epidemiology of Hunan Province, Hunan Normal University, No. 371 Tongzipo Road, Changsha, Hunan, 410013, PR China
| | - Shumei Li
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan, 410013, PR China; Institute of Clinical Translational Medicine, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan, 410005, PR China; Key Laboratory of Molecular Epidemiology of Hunan Province, Hunan Normal University, No. 371 Tongzipo Road, Changsha, Hunan, 410013, PR China
| | - Yang Feng
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan, 410013, PR China; Institute of Clinical Translational Medicine, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan, 410005, PR China; Key Laboratory of Molecular Epidemiology of Hunan Province, Hunan Normal University, No. 371 Tongzipo Road, Changsha, Hunan, 410013, PR China
| | - Yiyuan Zhang
- Institute of Clinical Translational Medicine, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan, 410005, PR China
| | - Jielin Ouyang
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan, 410013, PR China; Institute of Clinical Translational Medicine, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan, 410005, PR China; Key Laboratory of Molecular Epidemiology of Hunan Province, Hunan Normal University, No. 371 Tongzipo Road, Changsha, Hunan, 410013, PR China
| | - Shutong Li
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan, 410013, PR China; Institute of Clinical Translational Medicine, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan, 410005, PR China; Key Laboratory of Molecular Epidemiology of Hunan Province, Hunan Normal University, No. 371 Tongzipo Road, Changsha, Hunan, 410013, PR China
| | - Jia Wang
- Institute of Clinical Translational Medicine, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan, 410005, PR China.
| | - Lihong Tan
- Institute of Clinical Translational Medicine, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan, 410005, PR China.
| | - Lianhong Zou
- Institute of Clinical Translational Medicine, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan, 410005, PR China; Key Laboratory of Molecular Epidemiology of Hunan Province, Hunan Normal University, No. 371 Tongzipo Road, Changsha, Hunan, 410013, PR China.
| |
Collapse
|
16
|
Kim D, Lee MS, Sim H, Lee S, Lee HS. Characterization of complement C3 as a marker of alpha-amanitin toxicity by comparative secretome profiling. Toxicol Res 2023; 39:251-262. [PMID: 37008699 PMCID: PMC10050625 DOI: 10.1007/s43188-022-00163-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/23/2022] [Accepted: 11/28/2022] [Indexed: 12/24/2022] Open
Abstract
In the human body, proteins secreted into peripheral blood vessels are known as the secretome, and they represent the physiological or pathological status of cells. The unique response of cells to toxin exposure can be confirmed via secretome analysis, which can be used to discover toxic mechanisms or exposure markers. Alpha-amanitin (α-AMA) is the most widely studied amatoxin and inhibits transcription and protein synthesis by directly interacting with RNA polymerase II. However, secretory proteins released during hepatic failure caused by α-AMA have not been fully characterized. In this study, we analyzed the secretome of α-AMA-treated Huh-7 cells and mice using a comparative proteomics technique. Overall, 1440 and 208 proteins were quantified in cell media and mouse serum, respectively. Based on the bioinformatics results for the commonly downregulated proteins in cell media and mouse serum, we identified complement component 3 (C3) as a marker for α-AMA-induced hepatotoxicity. Through western blot in cell secretome and C3 ELISA assays in mouse serum, we validated α-AMA-induced downregulation of C3. In conclusion, using comparative proteomics and molecular biology techniques, we found that α-AMA-induced hepatotoxicity reduced C3 levels in the secretome. We expect that this study will aid in identifying new toxic mechanisms, therapeutic targets, and exposure markers of α-AMA-induced hepatotoxicity. Supplementary Information The online version contains supplementary material available at 10.1007/s43188-022-00163-z.
Collapse
Affiliation(s)
- Doeun Kim
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566 Republic of Korea
| | - Min Seo Lee
- BK21 Four-sponsored Advanced Program for SmartPharma Leaders, College of Pharmacy, The Catholic University of Korea, Bucheon, 14662 Republic of Korea
| | - Hyunchae Sim
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566 Republic of Korea
| | - Sangkyu Lee
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566 Republic of Korea
| | - Hye Suk Lee
- BK21 Four-sponsored Advanced Program for SmartPharma Leaders, College of Pharmacy, The Catholic University of Korea, Bucheon, 14662 Republic of Korea
| |
Collapse
|
17
|
Xue J, Lou X, Ning D, Shao R, Chen G. Mechanism and treatment of α-amanitin poisoning. Arch Toxicol 2023; 97:121-131. [PMID: 36271256 DOI: 10.1007/s00204-022-03396-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 10/10/2022] [Indexed: 12/31/2024]
Abstract
Amanita poisoning has a high mortality rate. The α-amanitin toxin in Amanita is the main lethal toxin. There is no specific detoxification drug for α-amanitin, and the clinical treatment mainly focuses on symptomatic and supportive therapy. The pathogenesis of α-amanitin mainly includes: α-amanitin can inhibit the activity of RNA polymeraseII in the nucleus, including the inhibition of the largest subunit of RNA polymeraseII, RNApb1, bridge helix, and trigger loop. In addition, α-amanitin acts in vivo through the enterohepatic circulation and transport system. α-Amanitin can cause the cell death. The existing mechanisms of cell damage mainly focus on apoptosis, oxidative stress, and autophagy. In addition to the pathogenic mechanism, α-amanitin also has a role in cancer treatment, which is the focus of current research. The mechanism of action of α-amanitin on the body is still being explored.
Collapse
Affiliation(s)
- Jinfang Xue
- Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Xiran Lou
- Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Deyuan Ning
- Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Ruifei Shao
- Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Guobing Chen
- Department of Emergency Medicine, The First People's Hospital of Yunnan Province, No. 157 Jinbi Road, Xishan District, Kunming, 650032, People's Republic of China.
| |
Collapse
|
18
|
Garcia J, Carvalho A, das Neves RP, Malheiro R, Rodrigues DF, Figueiredo PR, Bovolini A, Duarte JA, Costa VM, Carvalho F. Antidotal effect of cyclosporine A against α-amanitin toxicity in CD-1 mice, at clinical relevant doses. Food Chem Toxicol 2022; 166:113198. [PMID: 35671903 DOI: 10.1016/j.fct.2022.113198] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 05/25/2022] [Accepted: 05/30/2022] [Indexed: 12/12/2022]
Abstract
Amanita phalloides is one of the most toxic mushrooms worldwide, being responsible for the majority of human fatal cases of mushroom intoxications. α-Amanitin, the most deleterious toxin of A. phalloides, inhibits RNA polymerase II (RNAP II), causing hepatic and renal failure. Herein, we used cyclosporine A after it showed potential to displace RNAP II α-amanitin in silico. That potential was not confirmed either by the incorporation of ethynyl-UTP or by the monitoring of fluorescent RNAP II levels. Nevertheless, concomitant incubation of cyclosporine A with α-amanitin, for a short period, provided significant protection against its toxicity in differentiated HepaRG cells. In mice, the concomitant administration of α-amanitin [0.45 mg/kg intraperitoneal (i.p.)] with cyclosporine A (10 mg/kg i.p. plus 2 × 10 mg/kg cyclosporine A i.p. at 8 and 12 h post α-amanitin) resulted in the full survival of α-amanitin-intoxicated mice, up to 30 days after the toxin's administration. Since α-amanitin is a substrate of the organic-anion-transporting polypeptide 1B3 and cyclosporine A inhibits this transporter and is a potent anti-inflammatory agent, we hypothesize that these mechanisms are responsible for the protection observed. These results indicate a potential antidotal effect of cyclosporine A, and its safety profile advocates for its use at an early stage of α-amanitin intoxications.
Collapse
Affiliation(s)
- Juliana Garcia
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal; Laboratório Associado i4HB - Instituto para a Saúde e a Bioeconomia, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, 4050-313, Porto, Portugal
| | - Alexandra Carvalho
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-517, Coimbra, Portugal; IIIUC-Institute of Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
| | - Ricardo Pires das Neves
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-517, Coimbra, Portugal
| | - Rui Malheiro
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal; Laboratório Associado i4HB - Instituto para a Saúde e a Bioeconomia, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, 4050-313, Porto, Portugal
| | - Daniela F Rodrigues
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal; Laboratório Associado i4HB - Instituto para a Saúde e a Bioeconomia, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, 4050-313, Porto, Portugal
| | - Pedro R Figueiredo
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-517, Coimbra, Portugal
| | | | - José Alberto Duarte
- CIAFEL, Faculty of Sport, University of Porto, Porto, Portugal; TOXRUN - Toxicology Research Unit, University Institute of Health Sciences, CESPU, CRL, 4585-116, Gandra, Portugal
| | - Vera Marisa Costa
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal; Laboratório Associado i4HB - Instituto para a Saúde e a Bioeconomia, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, 4050-313, Porto, Portugal.
| | - Félix Carvalho
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal; Laboratório Associado i4HB - Instituto para a Saúde e a Bioeconomia, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, 4050-313, Porto, Portugal.
| |
Collapse
|
19
|
Lin LY, Tong YL, Lu YQ. The characteristics of liver injury induced by Amanita and clinical value of α-amanitin detection. Hepatobiliary Pancreat Dis Int 2022; 21:257-266. [PMID: 35168873 DOI: 10.1016/j.hbpd.2022.01.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/17/2021] [Indexed: 02/05/2023]
Abstract
BACKGROUND Amanita poisoning as a foodborne disease has raised concerning mortality issues. Reducing the interval between mushroom ingestion and medical intervention could greatly influence the outcomes of Amanita poisoning patients, while treatment is highly dependent on a confirmed diagnosis. To this end, we developed an early detection-guided intervention strategy by optimizing diagnostic process with performing α-amanitin detection, and further explored whether this strategy influenced the progression of Amanita poisoning. METHODS This study was a retrospective analysis of 25 Amanita poisoning patients. Thirteen patients in the detection group were diagnosed mainly based on α-amanitin detection, and 12 patients were diagnosed essentially on the basis of mushroom consumption history, typical clinical patterns and mushroom identification (conventional group). Amanita poisoning patients received uniform therapy, in which plasmapheresis was executed once confirming the diagnosis of Amanita poisoning. We compared the demographic baseline, clinical and laboratory data, treatment and outcomes between the two groups, and further explored the predictive value of α-amanitin concentration in serum. RESULTS Liver injury induced by Amanita appeared worst at the fourth day and alanine aminotransferase (ALT) rose higher than aspartate aminotransferase (AST). The mortality rate was 7.7% (1/13) in the detection group and 50.0% (6/12) in the conventional group (P = 0.030), since patients in the detection group arrived hospital much earlier and received plasmapheresis at the early stage of disease. The early detection-guided intervention helped alleviate liver impairment caused by Amanita and decreased the peak AST as well as ALT. However, the predictive value of α-amanitin concentration in serum was still considered limited. CONCLUSIONS In the management of mushroom poisoning, consideration should be given to the rapid detection of α-amanitin in suspected Amanita poisoning patients and the immediate initiation of medical treatment upon a positive toxin screening result.
Collapse
Affiliation(s)
- Li-Ying Lin
- Department of Emergency Medicine, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Ya-Ling Tong
- Department of Emergency Medicine, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yuan-Qiang Lu
- Department of Emergency Medicine, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; The Key Laboratory for Diagnosis and Treatment of Aging and Physic-chemical Injury Diseases of Zhejiang Province, Hangzhou 310003, China.
| |
Collapse
|
20
|
Mao Z, Yu Y, Sun H, Cao Y, Jiang Q, Chu C, Sun Y, Huang S, Zhang J, Chen F. Rapid detection of α-amanitin and β-amanitin in rat plasma by ultra-performance liquid chromatography–tandem mass spectrometry and its application to the toxicokinetic study of Lepiota brunneoincarnata. Forensic Toxicol 2021; 40:111-118. [DOI: 10.1007/s11419-021-00607-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 11/19/2021] [Indexed: 11/29/2022]
|
21
|
Mao Z, Yu Y, Sun H, Cao Y, Jiang Q, Chu C, Sun Y, Huang S, Zhang J, Chen F. Development of a simple and reliable method for α-amanitin detection in rat plasma and its application to a toxicokinetic study. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2021; 35:e9184. [PMID: 34472672 DOI: 10.1002/rcm.9184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 06/13/2023]
Abstract
RATIONALE α-Amanitin is a highly toxic peptide widely found in species of poisonous mushrooms. The matrix effect has been a major obstacle for accurate determination of α-amanitin in plasma samples by liquid chromatography/tandem mass spectrometry (LC/MS/MS). In this study, the strategy to eliminate the matrix effect of α-amanitin with a one-step dilution approach after deproteinization was applied. METHODS Rat plasma samples were processed by protein precipitation with methanol followed by a nine-fold dilution with pure water. The matrix effect value of α-amanitin was 19.7%-22.2% by protein precipitation and then changed to 87.5%-88.7% after dilution. α-Amanitin and the internal standard (roxithromycin) were analyzed on an ACQUITY UPLC® BEH C18 (50 mm × 2.1 mm, 1.7 μm) column within 3.0 min by gradient elution. RESULTS The linear ranges were 0.90-600 ng/mL with a correlation coefficient r >0.9958. A lower limit of quantification (LLOQ) of 0.90 ng/mL was achieved using only 50 μL of rat plasma. The intra- and inter-day precisions for the analyte ranged from 3.2% to 7.5% and 3.1% to 7.1%, respectively, and the accuracy ranged from -5.3% to -8.0%. CONCLUSIONS The matrix effect of α-amanitin was reduced by sample dilution after plasma deproteinization. A reliable LC/MS/MS method for the determination of α-amanitin in rat plasma was developed. This method was successfully applied for a toxicokinetic study of rats after intravenous injection of α-amanitin with a subacute toxicity dose at 0.10 mg/kg.
Collapse
Affiliation(s)
- Zhengsheng Mao
- Department of Forensic Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Youjia Yu
- Department of Forensic Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hao Sun
- Department of Emergency, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yue Cao
- Department of Forensic Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qiaoyan Jiang
- Department of Forensic Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chunyan Chu
- Department of Forensic Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yang Sun
- Department of Forensic Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shuainan Huang
- Department of Forensic Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jinsong Zhang
- Department of Emergency, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Feng Chen
- Department of Forensic Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
22
|
Runft S, Mischke R, Hoppe S, Hewicker-Trautwein M. [Acute liver failure in a dog after mushroom intake, presumably of the genus Amanita]. TIERAERZTLICHE PRAXIS AUSGABE KLEINTIERE HEIMTIERE 2021; 49:382-389. [PMID: 34670314 DOI: 10.1055/a-1584-6098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
A 4-year-old, neutered male Husky-mix dog weighing 29.4 kg that reportedly ingested a mushroom most likely of the genus Amanita one day prior to presentation exhibited signs of diarrhea, vomitus, inappetence and progressively worsening lethargy. Clinical chemistry revealed hypoglycemia, hyperbilirubinemia, decreased prothrombin and thromboplastin time, as well as increased liver enzyme activities. Despite hospitalization and supportive therapy over a period of 3 days the dog's general condition worsened leading to euthanasia. The pathomorphological findings were characterized by hemorrhage in several organs, hemorrhagic ingesta, icterus, and marked hepatic cellular necrosis.
Collapse
Affiliation(s)
- Sandra Runft
- Institut für Pathologie, Stiftung Tierärztliche Hochschule Hannover
| | - Reinhard Mischke
- Klinik für Kleintiere, Stiftung Tierärztliche Hochschule Hannover
| | - Sonja Hoppe
- Klinik für Kleintiere, Stiftung Tierärztliche Hochschule Hannover
| | | |
Collapse
|
23
|
Abstract
Albumin is abundant in serum but is also excreted at mucosal surfaces and enters tissues when inflammation increases vascular permeability. Host-associated opportunistic pathogens encounter albumin during commensalism and when causing infections. Considering the ubiquitous presence of albumin, we investigated its role in the pathogenesis of infections with the model human fungal pathogen, Candida albicans. Albumin was introduced in various in vitro models that mimic different stages of systemic or mucosal candidiasis, where it reduced the ability of C. albicans to damage host cells. The amphipathic toxin candidalysin mediates necrotic host cell damage induced by C. albicans. Using cellular and biophysical assays, we determined that albumin functions by neutralizing candidalysin through hydrophobic interactions. We discovered that albumin, similarly, can neutralize a variety of fungal (α-amanitin), bacterial (streptolysin O and staurosporin), and insect (melittin) hydrophobic toxins. These data suggest albumin as a defense mechanism against toxins, which can play a role in the pathogenesis of microbial infections. IMPORTANCE Albumin is the most abundant serum protein in humans. During inflammation, serum albumin levels decrease drastically, and low albumin levels are associated with poor patient outcome. Thus, albumin may have specific functions during infection. Here, we describe the ability of albumin to neutralize hydrophobic microbial toxins. We show that albumin can protect against damage induced by the pathogenic yeast C. albicans by neutralizing its cytolytic toxin candidalysin. These findings suggest that albumin is a toxin-neutralizing protein that may play a role during infections with toxin-producing microorganisms.
Collapse
|
24
|
Li Y, Sun Y, Kulke M, Hechler T, Van der Jeught K, Dong T, He B, Miller KD, Radovich M, Schneider BP, Pahl A, Zhang X, Lu X. Targeted immunotherapy for HER2-low breast cancer with 17p loss. Sci Transl Med 2021; 13:eabc6894. [PMID: 33568521 PMCID: PMC8351376 DOI: 10.1126/scitranslmed.abc6894] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 01/13/2021] [Indexed: 12/21/2022]
Abstract
The clinical challenge for treating HER2 (human epidermal growth factor receptor 2)-low breast cancer is the paucity of actionable drug targets. HER2-targeted therapy often has poor clinical efficacy for this disease due to the low level of HER2 protein on the cancer cell surface. We analyzed breast cancer genomics in the search for potential drug targets. Heterozygous loss of chromosome 17p is one of the most frequent genomic events in breast cancer, and 17p loss involves a massive deletion of genes including the tumor suppressor TP53 Our analyses revealed that 17p loss leads to global gene expression changes and reduced tumor infiltration and cytotoxicity of T cells, resulting in immune evasion during breast tumor progression. The 17p deletion region also includes POLR2A, a gene encoding the catalytic subunit of RNA polymerase II that is essential for cell survival. Therefore, breast cancer cells with heterozygous loss of 17p are extremely sensitive to the inhibition of POLR2A via a specific small-molecule inhibitor, α-amanitin. Here, we demonstrate that α-amanitin-conjugated trastuzumab (T-Ama) potentiated the HER2-targeted therapy and exhibited superior efficacy in treating HER2-low breast cancer with 17p loss. Moreover, treatment with T-Ama induced immunogenic cell death in breast cancer cells and, thereby, delivered greater efficacy in combination with immune checkpoint blockade therapy in preclinical HER2-low breast cancer models. Collectively, 17p loss not only drives breast tumorigenesis but also confers therapeutic vulnerabilities that may be used to develop targeted precision immunotherapy.
Collapse
Affiliation(s)
- Yujing Li
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yifan Sun
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Michael Kulke
- Heidelberg Pharma Research GmbH, Ladenburg 68526, Germany
| | | | - Kevin Van der Jeught
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Tianhan Dong
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Bin He
- Departments of Surgery and Urology, Immunobiology and Transplant Science Center, Houston Methodist Cancer Center, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX 77030, USA
- Department of Medicine, Weill Cornell Medicine of Cornell University, New York, NY 10065, USA
| | - Kathy D Miller
- Indiana University Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Milan Radovich
- Indiana University Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Bryan P Schneider
- Indiana University Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Andreas Pahl
- Heidelberg Pharma Research GmbH, Ladenburg 68526, Germany
| | - Xinna Zhang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
- Indiana University Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Xiongbin Lu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
- Indiana University Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
25
|
Costa VM, Capela JP, Sousa JR, Eleutério RP, Rodrigues PRS, Dores-Sousa JL, Carvalho RA, Lourdes Bastos M, Duarte JA, Remião F, Almeida MG, Varner KJ, Carvalho F. Mitoxantrone impairs proteasome activity and prompts early energetic and proteomic changes in HL-1 cardiomyocytes at clinically relevant concentrations. Arch Toxicol 2020; 94:4067-4084. [DOI: 10.1007/s00204-020-02874-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 08/12/2020] [Indexed: 11/24/2022]
|