1
|
Qin Z, Chen J, Liu F, Li B, Zhang C, Wang X, Liu L, Wang M, Wang T, Wang S, Yu F, Wang S, Yang J. Jellyfish stings-induced cardiac failure was ameliorated through AAG-mediated glycogen-driven ATP production. EXPLORATION (BEIJING, CHINA) 2025; 5:20230089. [PMID: 40040825 PMCID: PMC11875447 DOI: 10.1002/exp.20230089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 01/16/2024] [Indexed: 07/27/2024]
Abstract
Jellyfish stings have become a common injury among fishermen and divers. Severe jellyfish stings could worsen cardiac function and even cause cardiac complications, ultimately leading to cardiac failure (CF). Currently, there are no effective drugs available. Single cell sequencing revealed alpha-1 acid glycoprotein (AAG), an energy regulatory protein targeting to glycogen, was highly expressed in jellyfish stings-induced CF patients. However, the mechanism remains elusive. It is postulated that AAG could increase glycogen metabolism, protecting against jellyfish stings-induced CF. AAG deletion exacerbated CF, while exogenous and endogenous AAG ameliorated CF. AAG also rescued the decline triggered by the AAG knockout (KO). Intriguingly, AAG improved cardiac function and metabolic adaptation by glycogen-driven ATP production, shifting mitochondrial/glycolytic ATP production towards glycolysis. Sorted by single-cell RNA sequencing and spatial transcription technology, CC-chemokine receptor 5 (CCR5) and Peroxisome proliferator-activated receptor-gamma coactivator-1alpha (PGC-1α) were differentially expressed. Mechanistically, CCR5 inhibitor MVC abolished AAG's protective effect and PGC-1α overexpression. Collectively, jellyfish stings-induced CF was ameliorated through AAG-mediated glycogen-driven ATP production, promoting glycolytic/mitochondrial metabolic switches to rely energetically primarily on glycolysis, which might serve as a therapeutic target of CF.
Collapse
Affiliation(s)
- Zhen Qin
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
| | - Jinhong Chen
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
| | - Fang Liu
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
| | - Bingbing Li
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
| | - Chenchen Zhang
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
| | - Xiuxiu Wang
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
| | - Lin Liu
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
- School of Health Sciences and EngineeringUniversity of Shanghai for Science and TechnologyShanghaiChina
| | - Mingke Wang
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
| | | | - Su Wang
- Center for Translational NeuromedicineUniversity of Rochester Medical CenterRochesterNew YorkUSA
| | - Feifei Yu
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
| | - Shifeng Wang
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
| | - Jishun Yang
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
| |
Collapse
|
2
|
Luo X, Yang J, Zhao Y, Nagayasu T, Chen J, Hu P, He Z, Li Z, Wu J, Zhao Z, Duan G, Sun X, Zhao L, Pan Y, Wang X. Engineering spatially-confined conduits to tune nerve self-organization and allodynic responses via YAP-mediated mechanotransduction. Nat Commun 2025; 16:66. [PMID: 39746959 PMCID: PMC11695937 DOI: 10.1038/s41467-024-55118-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 12/02/2024] [Indexed: 01/04/2025] Open
Abstract
Chronic allodynia stemming from peripheral stump neuromas can persist for extended periods, significantly compromising patients' quality of life. Conventional managements for nerve stumps have demonstrated limited effectiveness in ensuring their orderly termination. In this study, we present a spatially confined conduit strategy, designed to enhance the self-organization of regenerating nerves after truncation. This innovative approach elegantly enables the autonomous slowing of axonal outgrowth in response to the gradually constricting space, concurrently suppressing neuroinflammation through YAP-mediated mechanotransduction activation. Meanwhile, the decelerating axons exhibit excellent alignment and remyelination, thereby helping to prevent failure modes in nerve self-organization, such as axonal twisting in congested regions and overgrowth beyond the conduit's capacity. Additionally, proteins associated with mechanical allodynia, including TRPA1 and CGRP, exhibit a gradual reduction in expression as spatial constraints tighten, a trend inversely validated by the administration of the YAP-targeted inhibitor Verteporfin. This spatially confined conduit strategy significantly alleviates allodynia, thus preventing autotomy behavior and reducing pain-induced gait alterations.
Collapse
Affiliation(s)
- Xiaobin Luo
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China
- Department of Orthopedics, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Jia Yang
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China
| | - Yonggang Zhao
- State Key Laboratory for Performance and Structure Safety of Petroleum Tubular Goods and Equipment Materials, Tubular Goods Research Institute of CNPC, Xi'an, 710077, China
| | - Toshitatsu Nagayasu
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China
| | - Junlin Chen
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China
| | - Peilun Hu
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China
- Department of Orthopedics, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Zhi He
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China
| | - Zifan Li
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China
| | - Jun Wu
- Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100094, China
| | - Zhe Zhao
- Department of Orthopedics, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Guman Duan
- Department of Orthopedics, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Xiaodan Sun
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China
| | - Lingyun Zhao
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China
| | - Yongwei Pan
- Department of Orthopedics, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China.
| | - Xiumei Wang
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
3
|
Shi WT, Yao CP, Liu WH, Cao WY, Shao W, Liao SQ, Yu T, Zhu QF, Chen Z, Zang YJ, Farooq M, Wei WK, Zhang XA. An fusaric acid-based CRISPR library screen identifies MDH2 as a broad-spectrum regulator of Fusarium toxin-induced cell death. JOURNAL OF HAZARDOUS MATERIALS 2024; 480:135937. [PMID: 39342847 DOI: 10.1016/j.jhazmat.2024.135937] [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: 07/13/2024] [Revised: 09/13/2024] [Accepted: 09/21/2024] [Indexed: 10/01/2024]
Abstract
Fusarium mycotoxins are of great concern because they are the most common food-borne mycotoxins and environmental contaminants worldwide. Fusaric acid (FA), Deoxynivalenol (DON), Zearalenone (ZEA), T-2 toxin (T-2), and Fumonisin B1 (FB1) are important Fusarium toxins contaminating feeds and food and can cause serious health problems. FA can synergize with some other Fusarium toxins to enhance overall toxicity. However, the underlying molecular mechanism remains poorly understood. In this study, our CRISPR screening revealed Malate dehydrogenase 2 (MDH2) and Pyruvate dehydrogenase E1 subunit beta (PDHB) are the key genes for FA-induced cell death. Pathways associated with mitochondrial function, notably the TCA cycle, play a significant role in FA cytotoxicity. We found that MDH2 and PDHB depletion reduced FA-induced cell death, ROS accumulation, and the expression of caspase-3 and HIF-1α. The cell viability assays and flow cytometry demonstrated that MDH2 knockout but not PDHB decreased DON, ZEA, T-2, and FB1-induced cytotoxicity, apoptosis, and ROS accumulation. MDH2 inhibitor LW6 also decreased DON, ZEA, T-2, and FB1-induced toxicity. This suggested that MDH2, but not PDHB, is a common regulator of broad-spectrum Fusarium toxin (FA, DON, ZEA, T-2, and FB1)-induced cell death. Our work provides new avenues for the treatment of Fusarium toxin toxicity.
Collapse
Affiliation(s)
- Wei-Tao Shi
- Agro-biological Gene Research Center of Guangdong Academy of Agricultural Sciences, State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, PR China; Xinjiang Agricultural University, College of Animal Science, Urumqi 830052, PR China
| | - Chun-Peng Yao
- Vegetable Research Institute of Guangdong Academy of Agricultural Sciences, Guangdong Key Laboratory for New Technology Research of Vegetables, Guangzhou 510640, PR China
| | - Wen-Hua Liu
- Agro-biological Gene Research Center of Guangdong Academy of Agricultural Sciences, State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, PR China
| | - Wan-Yi Cao
- Agro-biological Gene Research Center of Guangdong Academy of Agricultural Sciences, State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, PR China
| | - Wei Shao
- Xinjiang Agricultural University, College of Animal Science, Urumqi 830052, PR China
| | - Shen-Quan Liao
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, 510640, PR China
| | - Ting Yu
- Agro-biological Gene Research Center of Guangdong Academy of Agricultural Sciences, State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, PR China
| | - Qing-Feng Zhu
- Agro-biological Gene Research Center of Guangdong Academy of Agricultural Sciences, State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, PR China
| | - Zhuang Chen
- Agro-biological Gene Research Center of Guangdong Academy of Agricultural Sciences, State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, PR China
| | - Ying-Jie Zang
- Agro-biological Gene Research Center of Guangdong Academy of Agricultural Sciences, State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, PR China
| | - Muhammad Farooq
- Agro-biological Gene Research Center of Guangdong Academy of Agricultural Sciences, State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, PR China
| | - Wen-Kang Wei
- Agro-biological Gene Research Center of Guangdong Academy of Agricultural Sciences, State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, PR China.
| | - Xiao-Ai Zhang
- Agro-biological Gene Research Center of Guangdong Academy of Agricultural Sciences, State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, 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: 0] [Impact Index Per Article: 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
|
Pinheiro-Junior EL, Alirahimi E, Peigneur S, Isensee J, Schiffmann S, Erkoc P, Fürst R, Vilcinskas A, Sennoner T, Koludarov I, Hempel BF, Tytgat J, Hucho T, von Reumont BM. Diversely evolved xibalbin variants from remipede venom inhibit potassium channels and activate PKA-II and Erk1/2 signaling. BMC Biol 2024; 22:164. [PMID: 39075558 PMCID: PMC11288129 DOI: 10.1186/s12915-024-01955-5] [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: 02/17/2024] [Accepted: 07/09/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND The identification of novel toxins from overlooked and taxonomically exceptional species bears potential for various pharmacological applications. The remipede Xibalbanus tulumensis, an underwater cave-dwelling crustacean, is the only crustacean for which a venom system has been described. Its venom contains several xibalbin peptides that have an inhibitor cysteine knot (ICK) scaffold. RESULTS Our screenings revealed that all tested xibalbin variants particularly inhibit potassium channels. Xib1 and xib13 with their eight-cysteine domain similar to spider knottins also inhibit voltage-gated sodium channels. No activity was noted on calcium channels. Expanding the functional testing, we demonstrate that xib1 and xib13 increase PKA-II and Erk1/2 sensitization signaling in nociceptive neurons, which may initiate pain sensitization. Our phylogenetic analysis suggests that xib13 either originates from the common ancestor of pancrustaceans or earlier while xib1 is more restricted to remipedes. The ten-cysteine scaffolded xib2 emerged from xib1, a result that is supported by our phylogenetic and machine learning-based analyses. CONCLUSIONS Our functional characterization of synthesized variants of xib1, xib2, and xib13 elucidates their potential as inhibitors of potassium channels in mammalian systems. The specific interaction of xib2 with Kv1.6 channels, which are relevant to treating variants of epilepsy, shows potential for further studies. At higher concentrations, xib1 and xib13 activate the kinases PKA-II and ERK1/2 in mammalian sensory neurons, suggesting pain sensitization and potential applications related to pain research and therapy. While tested insect channels suggest that all probably act as neurotoxins, the biological function of xib1, xib2, and xib13 requires further elucidation. A novel finding on their evolutionary origin is the apparent emergence of X. tulumensis-specific xib2 from xib1. Our study is an important cornerstone for future studies to untangle the origin and function of these enigmatic proteins as important components of remipede but also other pancrustacean and arthropod venoms.
Collapse
Affiliation(s)
- Ernesto Lopes Pinheiro-Junior
- Toxicology and Pharmacology - Campus Gasthuisberg, University of Leuven (KU Leuven), Herestraat 49, PO Box 922, 3000, Louvain, Belgium
| | - Ehsan Alirahimi
- Department of Anesthesiology and Intensive Care Medicine, University Cologne, Translational Pain Research, University Hospital of Cologne, Cologne, Germany
| | - Steve Peigneur
- Toxicology and Pharmacology - Campus Gasthuisberg, University of Leuven (KU Leuven), Herestraat 49, PO Box 922, 3000, Louvain, Belgium
| | - Jörg Isensee
- Department of Anesthesiology and Intensive Care Medicine, University Cologne, Translational Pain Research, University Hospital of Cologne, Cologne, Germany
| | - Susanne Schiffmann
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596, Frankfurt Am Main, Germany
| | - Pelin Erkoc
- Institute of Pharmaceutical Biology, Goethe University Frankfurt, Max-Von-Laue-Str. 9, 60438, Frankfurt, Germany
- LOEWE Center for Translational Biodiversity Genomics (LOEWE-TBG), Senckenberganlage 25, 60325, Frankfurt, Germany
| | - Robert Fürst
- Institute of Pharmaceutical Biology, Goethe University Frankfurt, Max-Von-Laue-Str. 9, 60438, Frankfurt, Germany
- LOEWE Center for Translational Biodiversity Genomics (LOEWE-TBG), Senckenberganlage 25, 60325, Frankfurt, Germany
| | - Andreas Vilcinskas
- LOEWE Center for Translational Biodiversity Genomics (LOEWE-TBG), Senckenberganlage 25, 60325, Frankfurt, Germany
- Department of Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology (IME-BR), Ohlebergsweg 14, 35394, Giessen, Germany
| | - Tobias Sennoner
- Department of Informatics, Bioinformatics and Computational Biology, i12, Technical University of Munich, Boltzmannstr. 3, 85748, Garching, Munich, Germany
| | - Ivan Koludarov
- Department of Informatics, Bioinformatics and Computational Biology, i12, Technical University of Munich, Boltzmannstr. 3, 85748, Garching, Munich, Germany
| | - Benjamin-Florian Hempel
- Freie Unveristät Berlin, Veterinary Centre for Resistance Research (TZR), Robert-Von-Ostertag Str. 8, 14163, Berlin, Germany
| | - Jan Tytgat
- Toxicology and Pharmacology - Campus Gasthuisberg, University of Leuven (KU Leuven), Herestraat 49, PO Box 922, 3000, Louvain, Belgium
| | - Tim Hucho
- Department of Anesthesiology and Intensive Care Medicine, University Cologne, Translational Pain Research, University Hospital of Cologne, Cologne, Germany
| | - Björn M von Reumont
- LOEWE Center for Translational Biodiversity Genomics (LOEWE-TBG), Senckenberganlage 25, 60325, Frankfurt, Germany.
- Faculty of Biological Sciences, Institute of Cell Biology and Neuroscience, Goethe, Frankfurt, Max-Von-Laue-Str 13, 60438, Frankfurt, Germany.
| |
Collapse
|
6
|
Liu R, Wang Y, Kuai W, Li W, Wang Z, Xiao L, Wu J. Troxerutin suppress inflammation response and oxidative stress in jellyfish dermatitis by activating Nrf2/HO-1 signaling pathway. Front Immunol 2024; 15:1369849. [PMID: 38779681 PMCID: PMC11109374 DOI: 10.3389/fimmu.2024.1369849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 04/19/2024] [Indexed: 05/25/2024] Open
Abstract
Background Stomolophus meleagris envenomation causes severe cutaneous symptoms known as jellyfish dermatitis. The potential molecule mechanisms and treatment efficiency of dermatitis remain elusive because of the complicated venom components. The biological activity and molecular regulation mechanism of Troxerutin (TRX) was firstly examined as a potential treatment for jellyfish dermatitis. Methods We examined the inhibit effects of the TRX on tentacle extract (TE) obtained from S. meleagris in vivo and in vitro using the mice paw swelling models and corresponding assays for Enzyme-Linked Immunosorbent Assay (ELISA) Analysis, cell counting kit-8 assay, flow cytometry, respectively. The mechanism of TRX on HaCaT cells probed the altered activity of relevant signaling pathways by RNA sequencing and verified by RT-qPCR, Western blot to further confirm protective effects of TRX against the inflammation and oxidative damage caused by TE. Results TE significantly induced the mice paw skin toxicity and accumulation of inflammatory cytokines and reactive oxygen species in vivo and vitro. Moreover, a robust increase in the phosphorylation of mitogen-activated protein kinase (MAPKs) and nuclear factor-kappa B (NF-κB) signaling pathways was observed. While, the acute cutaneous inflammation and oxidative stress induced by TE were significantly ameliorated by TRX treatment. Notablly, TRX suppressed the phosphorylation of MAPK and NF-κB by initiating the nuclear factor erythroid 2-related factor 2 signaling pathway, which result in decreasing inflammatory cytokine release. Conclusion TRX inhibits the major signaling pathway responsible for inducing inflammatory and oxidative damage of jellyfish dermatitis, offering a novel therapy in clinical applications.
Collapse
Affiliation(s)
- Ran Liu
- Department of Dermatology, The First Affiliated Hospital of Naval Medical University, Navy Medical University, Shanghai, China
| | - Yulian Wang
- Department of Dermatology, The First Affiliated Hospital of Naval Medical University, Navy Medical University, Shanghai, China
| | - Wenhao Kuai
- Department of Dermatology, The First Affiliated Hospital of Naval Medical University, Navy Medical University, Shanghai, China
| | - Wenting Li
- Department of Dermatology, The First Affiliated Hospital of Naval Medical University, Navy Medical University, Shanghai, China
| | - Zengfa Wang
- Faculty of Naval Medicine, Naval Medical University, Shanghai, China
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun, China
| | - Liang Xiao
- Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Jianhua Wu
- Department of Dermatology, The First Affiliated Hospital of Naval Medical University, Navy Medical University, Shanghai, China
| |
Collapse
|
7
|
Geng H, Li R, Teng L, Yu C, Wang W, Gao K, Li A, Liu S, Xing R, Yu H, Li P. Exploring the Efficacy of Hydroxybenzoic Acid Derivatives in Mitigating Jellyfish Toxin-Induced Skin Damage: Insights into Protective and Reparative Mechanisms. Mar Drugs 2024; 22:205. [PMID: 38786596 PMCID: PMC11122885 DOI: 10.3390/md22050205] [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: 04/07/2024] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 05/25/2024] Open
Abstract
The escalation of jellyfish stings has drawn attention to severe skin reactions, underscoring the necessity for novel treatments. This investigation assesses the potential of hydroxybenzoic acid derivatives, specifically protocatechuic acid (PCA) and gentisic acid (DHB), for alleviating Nemopilema nomurai Nematocyst Venom (NnNV)-induced injuries. By employing an in vivo mouse model, the study delves into the therapeutic efficacy of these compounds. Through a combination of ELISA and Western blot analyses, histological examinations, and molecular assays, the study scrutinizes the inflammatory response, assesses skin damage and repair mechanisms, and investigates the compounds' ability to counteract venom effects. Our findings indicate that PCA and DHB significantly mitigate inflammation by modulating critical cytokines and pathways, altering collagen ratios through topical application, and enhancing VEGF and bFGF levels. Furthermore, both compounds demonstrate potential in neutralizing NnNV toxicity by inhibiting metalloproteinases and phospholipase-A2, showcasing the viability of small-molecule compounds in managing toxin-induced injuries.
Collapse
Affiliation(s)
- Hao Geng
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (H.G.)
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), No. 1 Wenhai Road, Qingdao 266237, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Rongfeng Li
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (H.G.)
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), No. 1 Wenhai Road, Qingdao 266237, China
| | - Lichao Teng
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (H.G.)
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), No. 1 Wenhai Road, Qingdao 266237, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunlin Yu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (H.G.)
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), No. 1 Wenhai Road, Qingdao 266237, China
| | - Wenjie Wang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (H.G.)
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), No. 1 Wenhai Road, Qingdao 266237, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kun Gao
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (H.G.)
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), No. 1 Wenhai Road, Qingdao 266237, China
| | - Aoyu Li
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (H.G.)
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), No. 1 Wenhai Road, Qingdao 266237, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Song Liu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (H.G.)
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), No. 1 Wenhai Road, Qingdao 266237, China
| | - Ronge Xing
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (H.G.)
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), No. 1 Wenhai Road, Qingdao 266237, China
| | - Huahua Yu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (H.G.)
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), No. 1 Wenhai Road, Qingdao 266237, China
| | - Pengcheng Li
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (H.G.)
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), No. 1 Wenhai Road, Qingdao 266237, China
| |
Collapse
|
8
|
Silva RCMC, Ramos IB, Travassos LH, Mendez APG, Gomes FM. Evolution of innate immunity: lessons from mammalian models shaping our current view of insect immunity. J Comp Physiol B 2024; 194:105-119. [PMID: 38573502 DOI: 10.1007/s00360-024-01549-1] [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: 10/07/2023] [Revised: 01/23/2024] [Accepted: 03/09/2024] [Indexed: 04/05/2024]
Abstract
The innate immune system, a cornerstone for organismal resilience against environmental and microbial insults, is highly conserved across the evolutionary spectrum, underpinning its pivotal role in maintaining homeostasis and ensuring survival. This review explores the evolutionary parallels between mammalian and insect innate immune systems, illuminating how investigations into these disparate immune landscapes have been reciprocally enlightening. We further delve into how advancements in mammalian immunology have enriched our understanding of insect immune responses, highlighting the intertwined evolutionary narratives and the shared molecular lexicon of immunity across these organisms. Therefore, this review posits a holistic understanding of innate immune mechanisms, including immunometabolism, autophagy and cell death. The examination of how emerging insights into mammalian and vertebrate immunity inform our understanding of insect immune responses and their implications for vector-borne disease transmission showcases the imperative for a nuanced comprehension of innate immunity's evolutionary tale. This understanding is quintessential for harnessing innate immune mechanisms' potential in devising innovative disease mitigation strategies and promoting organismal health across the animal kingdom.
Collapse
Affiliation(s)
- Rafael Cardoso M C Silva
- Laboratory of Immunoreceptors and Signaling, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Isabela B Ramos
- Laboratório de Ovogênese Molecular de Vetores, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Entomologia Molecular, Rio de Janeiro, Brazil
| | - Leonardo H Travassos
- Laboratory of Immunoreceptors and Signaling, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Paula Guzman Mendez
- Laboratório de Ultraestrutura Celular Hertha Meyer, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fabio M Gomes
- Instituto Nacional de Entomologia Molecular, Rio de Janeiro, Brazil.
- Laboratório de Ultraestrutura Celular Hertha Meyer, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
9
|
Hughey SB, Kotler JA, Ozaki Y, Itani Y, Fukuzawa F, Yanagimoto T, Takamatsu K, Koito S, Suzuki H, Nishihira Y, Hughey AC, Nagata T. Marine Envenomation in Okinawa: Overview and Treatment Concept. Wilderness Environ Med 2024; 35:57-66. [PMID: 38379485 DOI: 10.1177/10806032231220401] [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] [Indexed: 02/22/2024]
Abstract
Okinawa prefecture is a popular tourist destination due to its beaches and reefs. The reefs host a large variety of animals, including a number of venomous species. Because of the popularity of the reefs and marine activities, people are frequently in close contact with dangerous venomous species and, thus, are exposed to potential envenomation. Commonly encountered venomous animals throughout Okinawa include the invertebrate cone snail, sea urchin, crown-of-thorns starfish, blue-ringed octopus, box jellyfish, and fire coral. The vertebrates include the stonefish, lionfish, sea snake, and moray eel. Treatment for marine envenomation can involve first aid, hot water immersion, antivenom, supportive care, regional anesthesia, and pharmaceutical administration. Information on venomous animals, their toxins, and treatment should be well understood by prehospital care providers and physicians practicing in the prefecture.
Collapse
Affiliation(s)
- Scott B Hughey
- Naval Hospital Okinawa, Okinawa, Japan
- Naval Biotechnology Group, Naval Medical Center Portsmouth, Portsmouth, VA
| | - Joshua A Kotler
- Naval Biotechnology Group, Naval Medical Center Portsmouth, Portsmouth, VA
- III Marine Expeditionary Force, Okinawa, Japan
| | | | | | | | | | | | - Shu Koito
- Naval Hospital Okinawa, Okinawa, Japan
| | | | | | | | - Takashi Nagata
- Department of Emergency Medicine, Japan Self-Defense Force Central Hospital, Tokyo, Japan
| |
Collapse
|
10
|
Yu C, Yin X, Li A, Li R, Yu H, Xing R, Liu S, Li P. Toxin metalloproteinases exert a dominant influence on pro-inflammatory response and anti-inflammatory regulation in jellyfish sting dermatitis. J Proteomics 2024; 292:105048. [PMID: 37981009 DOI: 10.1016/j.jprot.2023.105048] [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: 08/22/2023] [Revised: 11/02/2023] [Accepted: 11/10/2023] [Indexed: 11/21/2023]
Abstract
Toxin metalloproteinases are the primary components responsible for various toxicities in jellyfish venom, and there is still no effective specific therapy for jellyfish stings. The comprehension of the pathogenic mechanisms underlying toxin metalloproteinases necessitates further refinement. In this study, we conducted a differential analysis of a dermatitis mouse model induced by jellyfish Nemopilema nomurai venom (NnNV) samples with varying levels of metalloproteinase activity. Through skin tissue proteomics and serum metabolomics, the predominant influence of toxin metalloproteinase activity on inflammatory response was revealed, and the signal pathway involved in its regulation was identified. In skin tissues, many membrane proteins were significantly down-regulated, which might cause tissue damage. The expression of pro-inflammatory factors was mainly regulated by PI3K-Akt signaling pathway. In serum, many fatty acid metabolites were significantly down-regulated, which might be the anti-inflammation feedback regulated by NF-κB p65 signaling pathway. These results reveal the dermatitis mechanism of toxin metalloproteinases and provide new therapeutic targets for further studies. SIGNIFICANCE: Omics is an important method to analyze the pathological mechanism and discover the key markers, which can reveal the pathological characteristics of jellyfish stings. Our research first analyzed the impact of toxin metalloproteinases on jellyfish sting dermatitis by skin proteomics and serum metabolomics. The present results suggest that inhibition of toxin metalloproteinases may be an effective treatment strategy, and provide new references for further jellyfish sting studies.
Collapse
Affiliation(s)
- Chunlin Yu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China; College of Earth and Planetary Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiujing Yin
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China; College of Earth and Planetary Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Aoyu Li
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China; College of Earth and Planetary Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Rongfeng Li
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China
| | - Huahua Yu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China.
| | - Ronge Xing
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China
| | - Song Liu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China
| | - Pengcheng Li
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China
| |
Collapse
|
11
|
Kong X, Wang W, Zhong Y, Wang N, Bai K, Wu Y, Qi Q, Zhang Y, Liu X, Xie J. Recent advances in the exploration and discovery of SARS-CoV-2 inhibitory peptides from edible animal proteins. Front Nutr 2024; 11:1346510. [PMID: 38389797 PMCID: PMC10883054 DOI: 10.3389/fnut.2024.1346510] [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: 12/01/2023] [Accepted: 01/22/2024] [Indexed: 02/24/2024] Open
Abstract
The severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2), which causes the coronavirus disease 2019 (COVID-19), is spreading worldwide. Although the COVID-19 epidemic has passed its peak of transmission, the harm it has caused deserves our attention. Scientists are striving to develop medications that can effectively treat COVID-19 symptoms without causing any adverse reactions. SARS-CoV-2 inhibitory peptides derived from animal proteins have a wide range of functional activities in addition to safety. Identifying animal protein sources is crucial to obtaining SARS-CoV-2 inhibitory peptides from animal sources. This review aims to reveal the mechanisms of action of these peptides on SARS-CoV-2 and the possibility of animal proteins as a material source of SARS-CoV-2 inhibitory peptides. Also, it introduces the utilization of computer-aided design methods, phage display, and drug delivery strategies in the research on SARS-CoV-2 inhibitor peptides from animal proteins. In order to identify new antiviral peptides and boost their efficiency, we recommend investigating the interaction between SARS-CoV-2 inhibitory peptides from animal protein sources and non-structural proteins (Nsps) using a variety of technologies, including computer-aided drug approaches, phage display techniques, and drug delivery techniques. This article provides useful information for the development of novel anti-COVID-19 drugs.
Collapse
Affiliation(s)
- Xiaoyue Kong
- College of Food and Health, Zhejiang Agriculture and Forestry University, Hangzhou, China
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou, China
| | - Wei Wang
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou, China
| | - Yizhi Zhong
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Nan Wang
- College of Biology and Environmental Engineering, Zhejiang Shuren University, Hangzhou, China
| | - Kaiwen Bai
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou, China
| | - Yi Wu
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou, China
| | - Qianhui Qi
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou, China
| | - Yu Zhang
- Institute of Quality and Standard for Agriculture Products, Zhejiang Academy of Agricultural Science, Hangzhou, China
| | - Xingquan Liu
- College of Food and Health, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Junran Xie
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
12
|
Manion J, Musser MA, Kuziel GA, Liu M, Shepherd A, Wang S, Lee PG, Zhao L, Zhang J, Marreddy RKR, Goldsmith JD, Yuan K, Hurdle JG, Gerhard R, Jin R, Rakoff-Nahoum S, Rao M, Dong M. C. difficile intoxicates neurons and pericytes to drive neurogenic inflammation. Nature 2023; 622:611-618. [PMID: 37699522 PMCID: PMC11188852 DOI: 10.1038/s41586-023-06607-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 09/05/2023] [Indexed: 09/14/2023]
Abstract
Clostridioides difficile infection (CDI) is a major cause of healthcare-associated gastrointestinal infections1,2. The exaggerated colonic inflammation caused by C. difficile toxins such as toxin B (TcdB) damages tissues and promotes C. difficile colonization3-6, but how TcdB causes inflammation is unclear. Here we report that TcdB induces neurogenic inflammation by targeting gut-innervating afferent neurons and pericytes through receptors, including the Frizzled receptors (FZD1, FZD2 and FZD7) in neurons and chondroitin sulfate proteoglycan 4 (CSPG4) in pericytes. TcdB stimulates the secretion of the neuropeptides substance P (SP) and calcitonin gene-related peptide (CGRP) from neurons and pro-inflammatory cytokines from pericytes. Targeted delivery of the TcdB enzymatic domain, through fusion with a detoxified diphtheria toxin, into peptidergic sensory neurons that express exogeneous diphtheria toxin receptor (an approach we term toxogenetics) is sufficient to induce neurogenic inflammation and recapitulates major colonic histopathology associated with CDI. Conversely, mice lacking SP, CGRP or the SP receptor (neurokinin 1 receptor) show reduced pathology in both models of caecal TcdB injection and CDI. Blocking SP or CGRP signalling reduces tissue damage and C. difficile burden in mice infected with a standard C. difficile strain or with hypervirulent strains expressing the TcdB2 variant. Thus, targeting neurogenic inflammation provides a host-oriented therapeutic approach for treating CDI.
Collapse
Affiliation(s)
- John Manion
- Department of Urology, Boston Children's Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Melissa A Musser
- Division of Gastroenterology, Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Gavin A Kuziel
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
- Division of Infectious Diseases, Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Min Liu
- Department of Urology, Boston Children's Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Amy Shepherd
- Division of Gastroenterology, Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Siyu Wang
- Department of Urology, Boston Children's Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Pyung-Gang Lee
- Department of Urology, Boston Children's Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Leo Zhao
- Department of Urology, Boston Children's Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Jie Zhang
- Department of Urology, Boston Children's Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Ravi K R Marreddy
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX, USA
| | | | - Ke Yuan
- Division of Pulmonary Medicine, Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Julian G Hurdle
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX, USA
| | - Ralf Gerhard
- Institute of Toxicology, Hannover Medical School, Hannover, Germany
| | - Rongsheng Jin
- Department of Physiology and Biophysics, School of Medicine, University of California Irvine, Irvine, CA, USA
| | - Seth Rakoff-Nahoum
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Infectious Diseases, Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Meenakshi Rao
- Division of Gastroenterology, Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Min Dong
- Department of Urology, Boston Children's Hospital, Boston, MA, USA.
- Department of Surgery, Harvard Medical School, Boston, MA, USA.
- Department of Microbiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
13
|
Sidik S. Deadly mushroom poison might now have an antidote - with help from CRISPR. Nature 2023:10.1038/d41586-023-01630-9. [PMID: 37193870 DOI: 10.1038/d41586-023-01630-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
|
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
|
Jami S, Deuis JR, Klasfauseweh T, Cheng X, Kurdyukov S, Chung F, Okorokov AL, Li S, Zhang J, Cristofori-Armstrong B, Israel MR, Ju RJ, Robinson SD, Zhao P, Ragnarsson L, Andersson Å, Tran P, Schendel V, McMahon KL, Tran HNT, Chin YKY, Zhu Y, Liu J, Crawford T, Purushothamvasan S, Habib AM, Andersson DA, Rash LD, Wood JN, Zhao J, Stehbens SJ, Mobli M, Leffler A, Jiang D, Cox JJ, Waxman SG, Dib-Hajj SD, Neely GG, Durek T, Vetter I. Pain-causing stinging nettle toxins target TMEM233 to modulate Na V1.7 function. Nat Commun 2023; 14:2442. [PMID: 37117223 PMCID: PMC10147923 DOI: 10.1038/s41467-023-37963-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 04/08/2023] [Indexed: 04/30/2023] Open
Abstract
Voltage-gated sodium (NaV) channels are critical regulators of neuronal excitability and are targeted by many toxins that directly interact with the pore-forming α subunit, typically via extracellular loops of the voltage-sensing domains, or residues forming part of the pore domain. Excelsatoxin A (ExTxA), a pain-causing knottin peptide from the Australian stinging tree Dendrocnide excelsa, is the first reported plant-derived NaV channel modulating peptide toxin. Here we show that TMEM233, a member of the dispanin family of transmembrane proteins expressed in sensory neurons, is essential for pharmacological activity of ExTxA at NaV channels, and that co-expression of TMEM233 modulates the gating properties of NaV1.7. These findings identify TMEM233 as a previously unknown NaV1.7-interacting protein, position TMEM233 and the dispanins as accessory proteins that are indispensable for toxin-mediated effects on NaV channel gating, and provide important insights into the function of NaV channels in sensory neurons.
Collapse
Affiliation(s)
- Sina Jami
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Jennifer R Deuis
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Tabea Klasfauseweh
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Xiaoyang Cheng
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Sergey Kurdyukov
- Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre, Centenary Institute, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Felicity Chung
- Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre, Centenary Institute, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Andrei L Okorokov
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, Division of Medicine, University College London, Gower Street, London, WC1E 6BT, UK
| | - Shengnan Li
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, Division of Medicine, University College London, Gower Street, London, WC1E 6BT, UK
| | - Jiangtao Zhang
- Institute of Physics, Chinese Academy of Sciences, 100190, Beijing, P.R. China
| | - Ben Cristofori-Armstrong
- Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD, 4072, Australia
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Mathilde R Israel
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience, King's College London, SE1 1UL, London, UK
| | - Robert J Ju
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Samuel D Robinson
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Peng Zhao
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Lotten Ragnarsson
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Åsa Andersson
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Poanna Tran
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Vanessa Schendel
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Kirsten L McMahon
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Hue N T Tran
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Yanni K-Y Chin
- Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Yifei Zhu
- Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Junyu Liu
- Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Theo Crawford
- Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD, 4072, Australia
| | | | - Abdella M Habib
- College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - David A Andersson
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience, King's College London, SE1 1UL, London, UK
| | - Lachlan D Rash
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - John N Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, Division of Medicine, University College London, Gower Street, London, WC1E 6BT, UK
| | - Jing Zhao
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, Division of Medicine, University College London, Gower Street, London, WC1E 6BT, UK
| | - Samantha J Stehbens
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Mehdi Mobli
- Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Andreas Leffler
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover, 30625, Germany
| | - Daohua Jiang
- Institute of Physics, Chinese Academy of Sciences, 100190, Beijing, P.R. China
| | - James J Cox
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, Division of Medicine, University College London, Gower Street, London, WC1E 6BT, UK
| | - Stephen G Waxman
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Sulayman D Dib-Hajj
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - G Gregory Neely
- Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre, Centenary Institute, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Thomas Durek
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia.
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, St Lucia, QLD, 4072, Australia.
| | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia.
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, 4102, Australia.
| |
Collapse
|
16
|
Lüddecke T, Paas A, Harris RJ, Talmann L, Kirchhoff KN, Billion A, Hardes K, Steinbrink A, Gerlach D, Fry BG, Vilcinskas A. Venom biotechnology: casting light on nature's deadliest weapons using synthetic biology. Front Bioeng Biotechnol 2023; 11:1166601. [PMID: 37207126 PMCID: PMC10188951 DOI: 10.3389/fbioe.2023.1166601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/18/2023] [Indexed: 05/21/2023] Open
Abstract
Venoms are complex chemical arsenals that have evolved independently many times in the animal kingdom. Venoms have attracted the interest of researchers because they are an important innovation that has contributed greatly to the evolutionary success of many animals, and their medical relevance offers significant potential for drug discovery. During the last decade, venom research has been revolutionized by the application of systems biology, giving rise to a novel field known as venomics. More recently, biotechnology has also made an increasing impact in this field. Its methods provide the means to disentangle and study venom systems across all levels of biological organization and, given their tremendous impact on the life sciences, these pivotal tools greatly facilitate the coherent understanding of venom system organization, development, biochemistry, and therapeutic activity. Even so, we lack a comprehensive overview of major advances achieved by applying biotechnology to venom systems. This review therefore considers the methods, insights, and potential future developments of biotechnological applications in the field of venom research. We follow the levels of biological organization and structure, starting with the methods used to study the genomic blueprint and genetic machinery of venoms, followed gene products and their functional phenotypes. We argue that biotechnology can answer some of the most urgent questions in venom research, particularly when multiple approaches are combined together, and with other venomics technologies.
Collapse
Affiliation(s)
- Tim Lüddecke
- Department of Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology, Giessen, Germany
- LOEWE Centre for Translational Biodiversity Genomics (LOEWE-TBG), Frankfurt am Main, Germany
- *Correspondence: Tim Lüddecke,
| | - Anne Paas
- Department of Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology, Giessen, Germany
- LOEWE Centre for Translational Biodiversity Genomics (LOEWE-TBG), Frankfurt am Main, Germany
| | - Richard J. Harris
- Venom Evolution Lab, School of Biological Sciences, The University of Queensland, Brisbane, QLD, Australia
- Institute for Molecular Biosciences (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Lea Talmann
- Syngenta Crop Protection, Stein, Switzerland
| | - Kim N. Kirchhoff
- Department of Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology, Giessen, Germany
| | - André Billion
- Department of Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology, Giessen, Germany
| | - Kornelia Hardes
- Department of Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology, Giessen, Germany
- LOEWE Centre for Translational Biodiversity Genomics (LOEWE-TBG), Frankfurt am Main, Germany
- BMBF Junior Research Group in Infection Research “ASCRIBE”, Giessen, Germany
| | - Antje Steinbrink
- LOEWE Centre for Translational Biodiversity Genomics (LOEWE-TBG), Frankfurt am Main, Germany
- Institute for Insect Biotechnology, Justus Liebig University of Giessen, Giessen, Germany
| | - Doreen Gerlach
- Department of Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology, Giessen, Germany
| | - Bryan G. Fry
- Venom Evolution Lab, School of Biological Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Andreas Vilcinskas
- Department of Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology, Giessen, Germany
- LOEWE Centre for Translational Biodiversity Genomics (LOEWE-TBG), Frankfurt am Main, Germany
- Institute for Insect Biotechnology, Justus Liebig University of Giessen, Giessen, Germany
| |
Collapse
|
17
|
Venom-derived pain-causing toxins: insights into sensory neuron function and pain mechanisms. Pain 2022; 163:S46-S56. [DOI: 10.1097/j.pain.0000000000002701] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 05/19/2022] [Indexed: 11/25/2022]
|
18
|
The application of genome-wide CRISPR-Cas9 screens to dissect the molecular mechanisms of toxins. Comput Struct Biotechnol J 2022; 20:5076-5084. [PMID: 36187925 PMCID: PMC9489804 DOI: 10.1016/j.csbj.2022.09.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 11/29/2022] Open
Abstract
Many toxins are life-threatening to both animals and humans. However, specific antidotes are not available for most of those toxins. The molecular mechanisms underlying the toxicology of well-known toxins are not yet fully characterized. Recently, the advance in CRISPR-Cas9 technologies has greatly accelerated the process of revealing the toxic mechanisms of some common toxins on hosts from a genome-wide perspective. The high-throughput CRISPR screen has made it feasible to untangle complicated interactions between a particular toxin and its corresponding targeting tissue(s). In this review, we present an overview of recent advances in molecular dissection of toxins’ cytotoxicity by using genome-wide CRISPR screens, summarize the components essential for toxin-specific CRISPR screens, and propose new strategies for future research.
Collapse
|
19
|
Anziani Vente A, Jaloux C, Morand JJ, LE Bel S, Bosdure E, Dubus JC, Morand A. Severe box jellyfish envenomation in 2 children. Travel Med Infect Dis 2021; 45:102217. [PMID: 34973452 DOI: 10.1016/j.tmaid.2021.102217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 07/11/2021] [Accepted: 11/23/2021] [Indexed: 10/19/2022]
Affiliation(s)
- Audrey Anziani Vente
- Department of Infant Medicine and Specialized Pediatrics, University Hospital of La Timone, AP-HM, Marseille, France
| | - Charlotte Jaloux
- Department of Pediatric Plastic Surgery, University Hospital of La Timone, AP-HM, Marseille, France
| | | | - Stéphane LE Bel
- Department of Neonatal and Pediatric Intensive Care Unit, University Hospital of La Timone, AP-HM, Marseille, France
| | - Emmanuelle Bosdure
- Department of Infant Medicine and Specialized Pediatrics, University Hospital of La Timone, AP-HM, Marseille, France
| | - Jean-Christophe Dubus
- Aix-Marseille University, Faculty of Medecine, Marseille, France; Department of Infant Medicine and Specialized Pediatrics, University Hospital of La Timone, AP-HM, Marseille, France
| | - Aurélie Morand
- Aix-Marseille University, Faculty of Medecine, Marseille, France; Department of Infant Medicine and Specialized Pediatrics, University Hospital of La Timone, AP-HM, Marseille, France.
| |
Collapse
|
20
|
Kokotović T, Langeslag M, Lenartowicz EM, Manion J, Fell CW, Alehabib E, Tafakhori A, Darvish H, Bellefroid EJ, Neely GG, Kress M, Penninger JM, Nagy V. PRDM12 Is Transcriptionally Active and Required for Nociceptor Function Throughout Life. Front Mol Neurosci 2021; 14:720973. [PMID: 34646120 PMCID: PMC8502974 DOI: 10.3389/fnmol.2021.720973] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 08/20/2021] [Indexed: 12/21/2022] Open
Abstract
PR domain-containing member 12 (PRDM12) is a key developmental transcription factor in sensory neuronal specification and survival. Patients with rare deleterious variants in PRDM12 are born with congenital insensitivity to pain (CIP) due to the complete absence of a subtype of peripheral neurons that detect pain. In this paper, we report two additional CIP cases with a novel homozygous PRDM12 variant. To elucidate the function of PRDM12 during mammalian development and adulthood, we generated temporal and spatial conditional mouse models. We find that PRDM12 is expressed throughout the adult nervous system. We observed that loss of PRDM12 during mid-sensory neurogenesis but not in the adult leads to reduced survival. Comparing cellular biophysical nociceptive properties in developmental and adult-onset PRDM12 deletion mouse models, we find that PRDM12 is necessary for proper nociceptive responses throughout life. However, we find that PRDM12 regulates distinct age-dependent transcriptional programs. Together, our results implicate PRDM12 as a viable therapeutic target for specific pain therapies even in adults.
Collapse
Affiliation(s)
- Tomislav Kokotović
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria.,CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.,Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Michiel Langeslag
- Department of Physiology and Medical Physics, Institute of Physiology, Medical University of Innsbruck, Innsbruck, Austria.,Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria.,Department of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Ewelina M Lenartowicz
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria.,Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - John Manion
- Charles Perkins Centre, Dr. John and Anne Chong Lab for Functional Genomics, Centenary Institute, and School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW, Australia
| | - Christopher W Fell
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria.,CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.,Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Elham Alehabib
- Student Research Committee, Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Tafakhori
- Iranian Center of Neurological Research, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Darvish
- Neuroscience Research Center, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Eric J Bellefroid
- ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - G Gregory Neely
- Charles Perkins Centre, Dr. John and Anne Chong Lab for Functional Genomics, Centenary Institute, and School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW, Australia
| | - Michaela Kress
- Department of Physiology and Medical Physics, Institute of Physiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, VBC - Vienna BioCenter, Vienna, Austria.,Department of Medical Genetics, Life Science Institute, University of British Columbia, Vancouver, BC, Canada
| | - Vanja Nagy
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria.,CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.,Department of Neurology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
21
|
Trim CM, Byrne LJ, Trim SA. Utilisation of compounds from venoms in drug discovery. PROGRESS IN MEDICINAL CHEMISTRY 2021; 60:1-66. [PMID: 34147202 DOI: 10.1016/bs.pmch.2021.01.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Difficult drug targets are becoming the normal course of business in drug discovery, sometimes due to large interacting surfaces or only small differences in selectivity regions. For these, a different approach is merited: compounds lying somewhere between the small molecule and the large antibody in terms of many properties including stability, biodistribution and pharmacokinetics. Venoms have evolved over millions of years to be complex mixtures of stable molecules derived from other somatic molecules, the stability comes from the pressure to be ready for delivery at a moment's notice. Snakes, spiders, scorpions, jellyfish, wasps, fish and even mammals have evolved independent venom systems with complex mixtures in their chemical arsenal. These venom-derived molecules have been proven to be useful tools, such as for the development of antihypotensive angiotensin converting enzyme (ACE) inhibitors and have also made successful drugs such as Byetta® (Exenatide), Integrilin® (Eptifibatide) and Echistatin. Only a small percentage of the available chemical space from venoms has been investigated so far and this is growing. In a new era of biological therapeutics, venom peptides present opportunities for larger target engagement surface with greater stability than antibodies or human peptides. There are challenges for oral absorption and target engagement, but there are venom structures that overcome these and thus provide substrate for engineering novel molecules that combine all desired properties. Venom researchers are characterising new venoms, species, and functions all the time, these provide great substrate for solving the challenges presented by today's difficult targets.
Collapse
Affiliation(s)
- Carol M Trim
- Faculty of Science, Engineering and Social Sciences, Natural and Applied Sciences, School of Psychology and Life Sciences, Canterbury Christ Church University, Canterbury, Kent, United Kingdom
| | - Lee J Byrne
- Faculty of Science, Engineering and Social Sciences, Natural and Applied Sciences, School of Psychology and Life Sciences, Canterbury Christ Church University, Canterbury, Kent, United Kingdom
| | | |
Collapse
|
22
|
Hon KL, Hui WF, Leung AK. Antidotes for childhood toxidromes. Drugs Context 2021; 10:dic-2020-11-4. [PMID: 34122588 PMCID: PMC8177957 DOI: 10.7573/dic.2020-11-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 04/20/2021] [Indexed: 11/21/2022] Open
Abstract
Background Poisoning causes significant morbidity and sometimes mortality in children worldwide. The clinical skill of toxidrome recognition followed by the timely administration of an antidote specific for the poison is essential for the management of children with suspected poisoning. This is a narrative review on antidotes for toxidromes in paediatric practice. Methods A literature search was conducted on PubMed with the keywords “antidote”, “poisoning”, “intoxication”, “children” and “pediatric”. The search was customized by applying the appropriate filters (species: humans; age: birth to 18 years) to obtain the most relevant articles for this review article. Results Toxidrome recognition may offer a rapid guide to possible toxicology diagnosis such that the specific antidote can be administered in a timely manner. This article summarizes toxidromes and their respective antidotes in paediatric poisoning, with an emphasis on the symptomatology and source of exposure. The antidote and specific management for each toxidrome are discussed. Antidotes are only available for a limited number of poisons responsible for intoxication. Antidotes for common poisonings include N-acetyl cysteine for paracetamol and sodium thiosulphate for poisoning by cyanide. Conclusion Poisoning is a common cause of paediatric injury. Physicians should be familiar with the recognition of common toxidromes and promptly use specific antidotes for the management of childhood toxidromes.
Collapse
Affiliation(s)
- Kam Lun Hon
- Department of a Paediatrics and Adolescent Medicine, The Hong Kong Children's Hospital, Hong Kong
| | - Wun Fung Hui
- Department of a Paediatrics and Adolescent Medicine, The Hong Kong Children's Hospital, Hong Kong
| | - Alexander Kc Leung
- Department of Pediatrics, The University of Calgary and The Alberta Children's Hospital, Calgary, Alberta, Canada
| |
Collapse
|
23
|
|
24
|
Li R, Yu H, Li A, Yu C, Li P. Preparation and Neutralization Efficacy of Novel Jellyfish Antivenoms against Cyanea nozakii Toxins. Toxins (Basel) 2021; 13:toxins13020165. [PMID: 33670073 PMCID: PMC7926751 DOI: 10.3390/toxins13020165] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 01/22/2023] Open
Abstract
Jellyfish stings are a common issue globally, particularly in coastal areas in the summer. Victims can suffer pain, itching, swelling, shock, and even death. Usually, hot water, vinegar, or alumen is used to treat the normal symptoms of a jellyfish sting. However, a specific antivenom may be an effective treatment to deal with severe jellyfish stings. Cyanea nozakii often reach a diameter of 60 cm and are responsible for hundreds of thousands of stings per year in coastal Chinese waters. However, there has been no specific C. nozakii antivenom until now, and so the development of this antivenom is very important. Herein, we collected C. nozakii antisera from tentacle extract venom immunized rabbits and purified the immunoglobulin (IgG) fraction antivenom (AntiCnTXs). Subsequently, two complete procedures to produce a refined F(ab')2 type of antivenom (F(ab')2-AntiCnTXs) and Fab type of antivenom (Fab-AntiCnTXs) by multiple optimizations and purification were established. The neutralization efficacy of these three types of antivenoms was compared and analyzed in vitro and in vivo, and the results showed that all types of antibodies displayed some neutralization effect on the lethality of C. nozakii venom toxins, with the neutralization efficacy as follows: F(ab')2-AntiCnTXs ≥ AntiCnTXs > Fab-AntiCnTXs. This study describes the preparation of novel C. nozakii jellyfish antivenom preparations towards the goal of developing a new, effective treatment for jellyfish stings.
Collapse
Affiliation(s)
- Rongfeng Li
- Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (H.Y.); (A.L.); (C.Y.)
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), No. 1 Wenhai Road, Qingdao 266237, China
- Correspondence: (R.L.); (P.L.); Tel.: +86-532-8289-8512 (R.L.); +86-532-8289-8707 (P.L.)
| | - Huahua Yu
- Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (H.Y.); (A.L.); (C.Y.)
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), No. 1 Wenhai Road, Qingdao 266237, China
| | - Aoyu Li
- Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (H.Y.); (A.L.); (C.Y.)
| | - Chunlin Yu
- Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (H.Y.); (A.L.); (C.Y.)
- College of Earth and Planetary Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Pengcheng Li
- Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (H.Y.); (A.L.); (C.Y.)
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), No. 1 Wenhai Road, Qingdao 266237, China
- Correspondence: (R.L.); (P.L.); Tel.: +86-532-8289-8512 (R.L.); +86-532-8289-8707 (P.L.)
| |
Collapse
|
25
|
Görücü Yilmaz S. Genome editing technologies: CRISPR, LEAPER, RESTORE, ARCUT, SATI, and RESCUE. EXCLI JOURNAL 2021; 20:19-45. [PMID: 33510590 PMCID: PMC7838830 DOI: 10.17179/excli2020-3070] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/10/2020] [Indexed: 12/16/2022]
Abstract
Genome editing technologies include techniques used for desired genetic modifications and allow the insertion, modification or deletion of specific DNA fragments. Recent advances in genome biology offer unprecedented promise for interdisciplinary collaboration and applications in gene editing. New genome editing technologies enable specific and efficient genome modifications. The sources that inspire these modifications and already exist in the genome are DNA degradation enzymes and DNA repair pathways. Six of these recent technologies are the clustered regularly interspaced short palindromic repeats (CRISPR), leveraging endogenous ADAR for programmable editing of RNA (LEAPER), recruiting endogenous ADAR to specific transcripts for oligonucleotide-mediated RNA editing (RESTORE), chemistry-based artificial restriction DNA cutter (ARCUT), single homology arm donor mediated intron-targeting integration (SATI), RNA editing for specific C-to-U exchange (RESCUE). These technologies are widely used from various biomedical researches to clinics, agriculture, and allow you to rearrange genomic sequences, create cell lines and animal models to solve human diseases. This review emphasizes the characteristics, superiority, limitations, also whether each technology can be used in different biological systems and the potential application of these systems in the treatment of several human diseases.
Collapse
Affiliation(s)
- Senay Görücü Yilmaz
- Department of Nutrition and Dietetics, Gaziantep University, Gaziantep, Turkey 27310
| |
Collapse
|
26
|
Jayathilake JMNJ, Gunathilake KVK. Cnidarian toxins: recent evidences for potential therapeutic uses. THE EUROPEAN ZOOLOGICAL JOURNAL 2020. [DOI: 10.1080/24750263.2020.1837268] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Affiliation(s)
- J. M. N. J. Jayathilake
- Department of Zoology, Faculty of Applied Sciences, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - K. V. K. Gunathilake
- Department of Zoology, Faculty of Applied Sciences, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| |
Collapse
|
27
|
Functional and Structural Variation among Sticholysins, Pore-Forming Proteins from the Sea Anemone Stichodactyla helianthus. Int J Mol Sci 2020; 21:ijms21238915. [PMID: 33255441 PMCID: PMC7727798 DOI: 10.3390/ijms21238915] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/15/2022] Open
Abstract
Venoms constitute complex mixtures of many different molecules arising from evolution in processes driven by continuous prey-predator interactions. One of the most common compounds in these venomous cocktails are pore-forming proteins, a family of toxins whose activity relies on the disruption of the plasmatic membranes by forming pores. The venom of sea anemones, belonging to the oldest lineage of venomous animals, contains a large amount of a characteristic group of pore-forming proteins known as actinoporins. They bind specifically to sphingomyelin-containing membranes and suffer a conformational metamorphosis that drives them to make pores. This event usually leads cells to death by osmotic shock. Sticholysins are the actinoporins produced by Stichodactyla helianthus. Three different isotoxins are known: Sticholysins I, II, and III. They share very similar amino acid sequence and three-dimensional structure but display different behavior in terms of lytic activity and ability to interact with cholesterol, an important lipid component of vertebrate membranes. In addition, sticholysins can act in synergy when exerting their toxin action. The subtle, but important, molecular nuances that explain their different behavior are described and discussed throughout the text. Improving our knowledge about sticholysins behavior is important for eventually developing them into biotechnological tools.
Collapse
|
28
|
Kim HJ, Noh JW, Amarsanaa K, Jeon SC, Yang YS, Hwang NH, Ko EA, Kang YJ, Jung SC. Peripheral Pain Modulation of Chrysaora pacifica Jellyfish Venom Requires Both Ca 2+ Influx and TRPA1 Channel Activation in Rats. Neurotox Res 2020; 38:900-913. [PMID: 32910305 DOI: 10.1007/s12640-020-00282-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 08/27/2020] [Accepted: 08/31/2020] [Indexed: 12/21/2022]
Abstract
The venom of jellyfish triggers severe dermal pain along with inflammation and tissue necrosis, and occasionally, induces internal organ dysfunction. However, the basic mechanisms underlying its cytotoxic effects are still unknown. Here, we report one of the mechanisms involved in peripheral pain modulation associated with inflammatory and neurotoxic oxidative signaling in rats using the venom of jellyfish, Chrysaora pacifica (CpV). This jellyfish is identified by brown tentacles carrying nematocysts filled with cytotoxic venom that induces severe pain, pruritus, tentacle marks, and blisters. The subcutaneous injection of CpV into rat forepaws in behavioral tests triggered nociceptive response with a decreased threshold for mechanical pain perception. These responses lasted up to 48 h and were completely blocked by verapamil and TTA-P2, T-type Ca2+ channel blockers, or HC030031, a transient receptor potential cation ankyrin 1 (TRPA1) channel blocker, while another Ca2+ channel blocker, nimodipine, was ineffective. Also, treatment with Ca2+ chelators (EGTA and BaptaAM) significantly alleviated the CpV-induced pain response. These results indicate that CpV-induced pain modulation may require both Ca2+ influx through the T-type Ca2+ channels and activation of TRPA1 channels. Furthermore, CpV induced Ca2+-mediated oxidative neurotoxicity in the dorsal root ganglion (DRG) and cortical neurons dissociated from rats, resulting in decreased neuronal viability and increased intracellular levels of ROS. Taken together, CpV may activate Ca2+-mediated oxidative signaling to produce excessive ROS acting as an endogenous agonist of TRPA1 channels in the peripheral terminals of the primary afferent neurons, resulting in persistent inflammatory pain. These findings provide strong evidence supporting the therapeutic effectiveness of blocking oxidative signaling against pain and cytotoxicity induced by jellyfish venom.
Collapse
Affiliation(s)
- Hye-Ji Kim
- Department of Physiology, School of Medicine, Jeju National University, Jeju, 63243, Republic of Korea
| | - Jin-Woo Noh
- Department of Physiology, School of Medicine, Jeju National University, Jeju, 63243, Republic of Korea
| | - Khulan Amarsanaa
- Department of Physiology, School of Medicine, Jeju National University, Jeju, 63243, Republic of Korea
| | - Sang-Chan Jeon
- Department of Physiology, School of Medicine, Jeju National University, Jeju, 63243, Republic of Korea
| | - Yoon-Sil Yang
- Department of Structure and Function of Neural Network, Korea Brain Research Institute, Daegu, 41068, Republic of Korea
| | - Na-Hye Hwang
- Department of Physiology, School of Medicine, Jeju National University, Jeju, 63243, Republic of Korea
| | - Eun-A Ko
- Department of Physiology, School of Medicine, Jeju National University, Jeju, 63243, Republic of Korea
| | - Young-Joon Kang
- Department of Emergency Medicine, School of Medicine, Jeju National University, Jeju, 63243, Republic of Korea.,Institute of Medical Science, Jeju National University, Jeju, 63243, Republic of Korea
| | - Sung-Cherl Jung
- Department of Physiology, School of Medicine, Jeju National University, Jeju, 63243, Republic of Korea. .,Institute of Medical Science, Jeju National University, Jeju, 63243, Republic of Korea. .,Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju, 63243, Republic of Korea.
| |
Collapse
|
29
|
The pathology of Chironex fleckeri venom and known biological mechanisms. Toxicon X 2020; 6:100026. [PMID: 32550582 PMCID: PMC7285912 DOI: 10.1016/j.toxcx.2020.100026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 01/28/2020] [Accepted: 02/04/2020] [Indexed: 12/20/2022] Open
Abstract
The large box jellyfish Chironex fleckeri is found in northern Australian waters. A sting from this cubozoan species can kill within minutes. From clinical and animal studies, symptoms comprise severe pain, welts, scarring, hypotension, vasospasms, cardiac irregularities and cardiac arrest. At present, there is no cure and opioids are used to manage pain. Antivenom is available but controversy exists over its effectiveness. Experimental and combination therapies performed in vitro and in vivo have shown varied efficacy. These inconsistent results are likely a consequence of the different methods used to extract venom. Recent omics analysis has shed light on the systems of C. fleckeri venom action, including new toxin classes that use pore formation, cell membrane collapse and ion channel modulation. This review covers what is known on C. fleckeri pathomechanisms and highlights current gaps in knowledge. A more complete understanding of the mechanisms of C. fleckeri venom-induced pathology may lead to novel treatments and possibly, the discovery of novel cell pathways, novel drug scaffolds and novel drug targets for human disease.
Collapse
|
30
|
Reinicke J, Kitatani R, Masoud SS, Galbraith KK, Yoshida W, Igarashi A, Nagasawa K, Berger G, Yanagihara A, Nagai H, Horgen FD. Isolation, Structure Determination, and Synthesis of Cyclic Tetraglutamic Acids from Box Jellyfish Species Alatina alata and Chironex yamaguchii. Molecules 2020; 25:molecules25040883. [PMID: 32079282 PMCID: PMC7070617 DOI: 10.3390/molecules25040883] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/08/2020] [Accepted: 02/10/2020] [Indexed: 11/16/2022] Open
Abstract
Cubozoan nematocyst venoms contain known cytolytic and hemolytic proteins, but small molecule components have not been previously reported from cubozoan venom. We screened nematocyst extracts of Alatina alata and Chironex yamaguchii by LC-MS for the presence of small molecule metabolites. Three isomeric compounds, cnidarins 4A (1), 4B (2), and 4C (3), were isolated from venom extracts and characterized by NMR and MS, which revealed their planar structure as cyclic γ-linked tetraglutamic acids. The full configurational assignments were established by syntheses of all six possible stereoisomers, comparison of spectral data and optical rotations, and stereochemical analysis of derivatized degradation products. Compounds 1-3 were subsequently detected by LC-MS in tissues of eight other cnidarian species. The most abundant of these compounds, cnidarin 4A (1), showed no mammalian cell toxicity or hemolytic activity, which may suggest a role for these cyclic tetraglutamates in nematocyst discharge.
Collapse
Affiliation(s)
- Justin Reinicke
- Department of Natural Sciences, Hawaii Pacific University, Kaneohe, HI 96744, USA; (J.R.); (K.K.G.)
- Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, HI 96720, USA
| | - Ryuju Kitatani
- Department of Marine Sciences, Tokyo University of Marine Science and Technology, Tokyo 108-8477, Japan; (R.K.); (A.I.)
| | - Shadi Sedghi Masoud
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Tokyo 184-8588, Japan; (S.S.M.); (K.N.)
| | - Kelly Kawabata Galbraith
- Department of Natural Sciences, Hawaii Pacific University, Kaneohe, HI 96744, USA; (J.R.); (K.K.G.)
- German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Str. 27, 53127 Bonn, Germany
| | - Wesley Yoshida
- Department of Chemistry, University of Hawaii at Manoa, Honolulu, HI 98622, USA;
| | - Ayako Igarashi
- Department of Marine Sciences, Tokyo University of Marine Science and Technology, Tokyo 108-8477, Japan; (R.K.); (A.I.)
| | - Kazuo Nagasawa
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Tokyo 184-8588, Japan; (S.S.M.); (K.N.)
| | - Gideon Berger
- Department of Natural Sciences, Hawaii Pacific University, Kaneohe, HI 96744, USA; (J.R.); (K.K.G.)
- Correspondence: (G.B.); (A.Y.); (H.N.); (F.D.H.); Tel.: +1-808-236-3551 (G.B.); +1-808- 956-8328 (A.Y.); +81-3-5463-0454 (H.N.); +1-808-236-5864 (F.D.H.)
| | - Angel Yanagihara
- Békésy Laboratory of Neurobiology, Pacific Biosciences Research Center, School of Ocean and Earth Science and Technology, and Department of Tropical Medicine, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96822, USA
- Correspondence: (G.B.); (A.Y.); (H.N.); (F.D.H.); Tel.: +1-808-236-3551 (G.B.); +1-808- 956-8328 (A.Y.); +81-3-5463-0454 (H.N.); +1-808-236-5864 (F.D.H.)
| | - Hiroshi Nagai
- Department of Marine Sciences, Tokyo University of Marine Science and Technology, Tokyo 108-8477, Japan; (R.K.); (A.I.)
- Correspondence: (G.B.); (A.Y.); (H.N.); (F.D.H.); Tel.: +1-808-236-3551 (G.B.); +1-808- 956-8328 (A.Y.); +81-3-5463-0454 (H.N.); +1-808-236-5864 (F.D.H.)
| | - F. David Horgen
- Department of Natural Sciences, Hawaii Pacific University, Kaneohe, HI 96744, USA; (J.R.); (K.K.G.)
- Correspondence: (G.B.); (A.Y.); (H.N.); (F.D.H.); Tel.: +1-808-236-3551 (G.B.); +1-808- 956-8328 (A.Y.); +81-3-5463-0454 (H.N.); +1-808-236-5864 (F.D.H.)
| |
Collapse
|
31
|
Lau MT, Ghazanfar S, Parkin A, Chou A, Rouaen JR, Littleboy JB, Nessem D, Khuong TM, Nevoltris D, Schofield P, Langley D, Christ D, Yang J, Pajic M, Neely GG. Systematic functional identification of cancer multi-drug resistance genes. Genome Biol 2020; 21:27. [PMID: 32028983 PMCID: PMC7006212 DOI: 10.1186/s13059-020-1940-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 01/20/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Drug resistance is a major obstacle in cancer therapy. To elucidate the genetic factors that regulate sensitivity to anti-cancer drugs, we performed CRISPR-Cas9 knockout screens for resistance to a spectrum of drugs. RESULTS In addition to known drug targets and resistance mechanisms, this study revealed novel insights into drug mechanisms of action, including cellular transporters, drug target effectors, and genes involved in target-relevant pathways. Importantly, we identified ten multi-drug resistance genes, including an uncharacterized gene C1orf115, which we named Required for Drug-induced Death 1 (RDD1). Loss of RDD1 resulted in resistance to five anti-cancer drugs. Finally, targeting RDD1 leads to chemotherapy resistance in mice and low RDD1 expression is associated with poor prognosis in multiple cancers. CONCLUSIONS Together, we provide a functional landscape of resistance mechanisms to a broad range of chemotherapeutic drugs and highlight RDD1 as a new factor controlling multi-drug resistance. This information can guide personalized therapies or instruct rational drug combinations to minimize acquisition of resistance.
Collapse
Affiliation(s)
- Man-Tat Lau
- The Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre and School of Life & Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
- Genome Editing Initiative, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Shila Ghazanfar
- School of Mathematics and Statistics, The University of Sydney, Sydney, NSW, 2006, Australia
- The Judith and David Coffey Life Lab, Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Ashleigh Parkin
- The Kinghorn Cancer Centre, The Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW, 2010, Australia
| | - Angela Chou
- The Kinghorn Cancer Centre, The Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW, 2010, Australia
- The University of Sydney, Sydney, NSW, 2006, Australia
| | - Jourdin R Rouaen
- The Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre and School of Life & Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Jamie B Littleboy
- The Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre and School of Life & Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Danielle Nessem
- The Kinghorn Cancer Centre, The Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW, 2010, Australia
| | - Thang M Khuong
- The Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre and School of Life & Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Damien Nevoltris
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, 2010, Australia
| | - Peter Schofield
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, Sydney, NSW, 2010, Australia
| | - David Langley
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, 2010, Australia
| | - Daniel Christ
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, Sydney, NSW, 2010, Australia
| | - Jean Yang
- School of Mathematics and Statistics, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Marina Pajic
- The Kinghorn Cancer Centre, The Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW, 2010, Australia.
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, Sydney, NSW, 2010, Australia.
| | - G Gregory Neely
- The Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre and School of Life & Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia.
- Genome Editing Initiative, The University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
32
|
Manion J, Waller MA, Clark T, Massingham JN, Neely GG. Developing Modern Pain Therapies. Front Neurosci 2019; 13:1370. [PMID: 31920521 PMCID: PMC6933609 DOI: 10.3389/fnins.2019.01370] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 12/04/2019] [Indexed: 12/24/2022] Open
Abstract
Chronic pain afflicts as much as 50% of the population at any given time but our methods to address pain remain limited, ineffective and addictive. In order to develop new therapies an understanding of the mechanisms of painful sensitization is essential. We discuss here recent progress in the understanding of mechanisms underlying pain, and how these mechanisms are being targeted to produce modern, specific therapies for pain. Finally, we make recommendations for the next generation of targeted, effective, and safe pain therapies.
Collapse
Affiliation(s)
- John Manion
- The Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Matthew A. Waller
- The Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Teleri Clark
- The Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Joshua N. Massingham
- The Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, Australia
| | - G. Gregory Neely
- The Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, Australia
- Genome Editing Initiative, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
33
|
Merquiol L, Romano G, Ianora A, D'Ambra I. Biotechnological Applications of Scyphomedusae. Mar Drugs 2019; 17:E604. [PMID: 31653064 PMCID: PMC6891278 DOI: 10.3390/md17110604] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 10/21/2019] [Indexed: 12/31/2022] Open
Abstract
As people across the world live longer, chronic illness and diminished well-being are becoming major global public health challenges. Marine biotechnology may help overcome some of these challenges by developing new products and know-how derived from marine organisms. While some products from marine organisms such as microalgae, sponges, and fish have already found biotechnological applications, jellyfish have received little attention as a potential source of bioactive compounds. Nevertheless, recent studies have highlighted that scyphomedusae (Cnidaria, Scyphozoa) synthesise at least three main categories of compounds that may find biotechnological applications: collagen, fatty acids and components of crude venom. We review what is known about these compounds in scyphomedusae and their current biotechnological applications, which falls mainly into four categories of products: nutraceuticals, cosmeceuticals, biomedicals, and biomaterials. By defining the state of the art of biotechnological applications in scyphomedusae, we intend to promote the use of these bioactive compounds to increase the health and well-being of future societies.
Collapse
Affiliation(s)
- Louise Merquiol
- Integrative Marine Ecology Department, Stazione Zoologica Anton Dohrn, Villa Comunale, 80121 Napoli, Italy.
| | - Giovanna Romano
- Marine Biotechnology Department, Stazione Zoologica Anton Dohrn, Villa Comunale, 80121 Napoli, Italy.
| | - Adrianna Ianora
- Marine Biotechnology Department, Stazione Zoologica Anton Dohrn, Villa Comunale, 80121 Napoli, Italy.
| | - Isabella D'Ambra
- Integrative Marine Ecology Department, Stazione Zoologica Anton Dohrn, Villa Comunale, 80121 Napoli, Italy.
| |
Collapse
|
34
|
Pore-Forming Proteins from Cnidarians and Arachnids as Potential Biotechnological Tools. Toxins (Basel) 2019; 11:toxins11060370. [PMID: 31242582 PMCID: PMC6628452 DOI: 10.3390/toxins11060370] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 06/18/2019] [Accepted: 06/21/2019] [Indexed: 12/31/2022] Open
Abstract
Animal venoms are complex mixtures of highly specialized toxic molecules. Cnidarians and arachnids produce pore-forming proteins (PFPs) directed against the plasma membrane of their target cells. Among PFPs from cnidarians, actinoporins stand out for their small size and molecular simplicity. While native actinoporins require only sphingomyelin for membrane binding, engineered chimeras containing a recognition antibody-derived domain fused to an actinoporin isoform can nonetheless serve as highly specific immunotoxins. Examples of such constructs targeted against malignant cells have been already reported. However, PFPs from arachnid venoms are less well-studied from a structural and functional point of view. Spiders from the Latrodectus genus are professional insect hunters that, as part of their toxic arsenal, produce large PFPs known as latrotoxins. Interestingly, some latrotoxins have been identified as potent and highly-specific insecticides. Given the proteinaceous nature of these toxins, their promising future use as efficient bioinsecticides is discussed throughout this Perspective. Protein engineering and large-scale recombinant production are critical steps for the use of these PFPs as tools to control agriculturally important insect pests. In summary, both families of PFPs, from Cnidaria and Arachnida, appear to be molecules with promising biotechnological applications.
Collapse
|