1
|
Kweon J, Lee H, Park J, Hong T, An G, Song G, Lim W, Jeong W. Developmental and organ toxicity of fenpropimorph in zebrafish: Involvement of apoptosis and inflammation. Chem Biol Interact 2025; 415:111512. [PMID: 40239885 DOI: 10.1016/j.cbi.2025.111512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 04/07/2025] [Accepted: 04/14/2025] [Indexed: 04/18/2025]
Abstract
Pesticides are increasingly the focus as a prominent factor in environmental pollution. Fenpropimorph, a widely utilized morpholine fungicide, is a significant water pollutant. Because of its extensive usage, fenpropimorph is readily detected in diverse aquatic ecosystems. Despite its well-known toxicity to aquatic organisms, its toxicity to zebrafish development and accompanying mechanics remain unexplored. To assess fenpropimorph's toxicity and potential mechanism, we employed the zebrafish model, a representative tool in toxicological studies. Our results showed that exposure to fenpropimorph reduced embryonic viability during the early stages of development and reduced head and body size. Moreover, fenpropimorph triggered apoptosis, DNA fragmentation, and inflammation. Aberrations in the vascular network were observed in the fli1:eGFP transgenic zebrafish model. Additionally, neurotoxic impacts were further assessed using transgenic olig2:dsRed zebrafish, accompanied by a reduction of liver size and fluorescence intensity of fabp10a:dsRed zebrafish. mRNA expression analysis related to corresponding organ development further supported our data. Overall, our research suggests that fenpropimorph may cause aberrations in aquatic organisms.
Collapse
Affiliation(s)
- Junhun Kweon
- Department of Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Hojun Lee
- Department of Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Junho Park
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Taeyeon Hong
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Garam An
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Gwonhwa Song
- Department of Biotechnology, Korea University, Seoul, 02841, Republic of Korea.
| | - Whasun Lim
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Wooyoung Jeong
- Department of Biomedical Sciences, Catholic Kwandong University, Gangneung, 25601, Republic of Korea.
| |
Collapse
|
2
|
Chan SJW, Xiao Z, Soh WWM, Zhu JY, Lopez-Garcia F, Deng H, Bazan GC. Membrane Intercalation of a Conjugated Oligoelectrolyte Photosensitizer Enables Efficient Anticancer Photodynamic Therapy. Adv Healthc Mater 2025:e2501300. [PMID: 40411856 DOI: 10.1002/adhm.202501300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 05/07/2025] [Indexed: 05/26/2025]
Abstract
Photodynamic therapy (PDT) complements traditional chemotherapeutic methods by leveraging an external optical stimulus to target and eradicate tumor cells. Photosensitizers with favorable photophysical properties are, however, often hampered by poor biodistribution and inadequate cellular uptake, highlighting the need for novel molecular design strategies. Conjugated oligoelectrolytes (COEs) are a class of optical reporters that readily incorporate within cells due to their lipid bilayer mimicking molecular topology. Herein, a COE-derived photosensitizer, COE-PP, which features a central porphyrin core flanked by ionic pendant groups is disclosed. COE-PP has a solubility in aqueous media of >20 mm, yet the membrane-mimicking molecular topology enables high cellular uptake and prolonged retention of the COE in vitro and in vivo of up to 21 days. COE-PP is an effective phototheranostic agent for simultaneous tumor imaging and eradication, with the generation of reactive oxygen species (ROS) upon illumination. The membrane localization of COE-PP promotes cell death during PDT by ROS and through downstream pathways involving lysosomal membrane permeabilization. These synergistic effects enable effective treatment of COE-containing tumors. From a broad design perspective, the molecular architecture of COE-PP demonstrates the potential of utilizing lipid bilayer-mimicking molecular topologies to design phototheranostic molecules that offer spatiotemporal control for therapeutic interventions.
Collapse
Affiliation(s)
- Samuel Jun Wei Chan
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
- Institute for Functional Intelligent Materials, National University of Singapore, Singapore, 117544, Singapore
| | - Zhaolin Xiao
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Targeted Tracer Research and Development Laboratory Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Wilson Wee Mia Soh
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
- Institute for Functional Intelligent Materials, National University of Singapore, Singapore, 117544, Singapore
| | - Ji-Yu Zhu
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
- Institute for Functional Intelligent Materials, National University of Singapore, Singapore, 117544, Singapore
- Institute for Digital Molecular Analytics and Science, Nanyang Technological University, Singapore, 636921, Singapore
| | - Fernando Lopez-Garcia
- Institute for Functional Intelligent Materials, National University of Singapore, Singapore, 117544, Singapore
| | - Hui Deng
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Targeted Tracer Research and Development Laboratory Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Guillermo Carlos Bazan
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
- Institute for Functional Intelligent Materials, National University of Singapore, Singapore, 117544, Singapore
- Institute for Digital Molecular Analytics and Science, Nanyang Technological University, Singapore, 636921, Singapore
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore, 117585, Singapore
| |
Collapse
|
3
|
Anand V, El-Dana F, Baran N, Borgman J, Yin Z, Zhao H, Wong ST, Andreeff M, Battula VL. GD3 synthase drives resistance to p53-induced apoptosis in breast cancer by modulating mitochondrial function. Oncogene 2025:10.1038/s41388-025-03432-x. [PMID: 40382494 DOI: 10.1038/s41388-025-03432-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 04/21/2025] [Accepted: 04/29/2025] [Indexed: 05/20/2025]
Abstract
TP53 mutations are common in breast cancer (BC) and are associated with poor prognosis. GD3 synthase (GD3S/ST8SIA1), a gene associated with breast cancer stem cells, is upregulated in tumors with p53 mutations. However, the functional relationship between GD3S and p53 is unknown. Here, we show that GD3S levels are highest in breast tumors with specific p53 mutations. Functional studies revealed that wild-type (WT) p53 inhibits GD3S expression, whereas mutation in p53 enhances GD3S expression by upregulating GD3S promoter activity. Moreover, we found that GD3S inhibits wild-type p53-induced apoptosis in BC cells, while BC cells harboring gain-of-function p53 mutations are dependent on GD3S for their growth. Mechanistic insights indicate that GD3S strengthens mitochondrial function by regulating their oxygen consumption rate and membrane polarity. Our findings demonstrate that specific GOF p53 mutations rely on GD3S to exert their tumor-promoting effects and that GD3S is a novel anti-apoptotic factor in BC cells. Stabilizing WT p53 and reducing mutant p53 levels downregulates GD3S expression, thereby augmenting apoptosis. GD3S overexpression counteracts the cell death triggered by WT p53 stabilization in BC cells, as well as that triggered by p53 knockdown in cells with specific GOF p53 mutations, which suggests that GD3S helps confer apoptosis resistance.
Collapse
Affiliation(s)
- Vivek Anand
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fouad El-Dana
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Natalia Baran
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Hematology and Central Hematological Laboratory, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Jenny Borgman
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zheng Yin
- Department of Systems Medicine and Bioengineering, Houston Methodist Neal Cancer Center, Weill Cornell Medicine, Houston, TX, USA
| | - Hong Zhao
- Department of Systems Medicine and Bioengineering, Houston Methodist Neal Cancer Center, Weill Cornell Medicine, Houston, TX, USA
| | - Stephen T Wong
- Department of Systems Medicine and Bioengineering, Houston Methodist Neal Cancer Center, Weill Cornell Medicine, Houston, TX, USA
| | - Michael Andreeff
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - V Lokesh Battula
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Internal Medicine, Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
4
|
Roman A, Motoc A, Marcovici I, Dehelean C, Nicolescu L, Boru C. Genistein Improves the Cytotoxic, Apoptotic, and Oxidative-Stress-Inducing Properties of Doxorubicin in SK-MEL-28 Cancer Cells. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:798. [PMID: 40428756 PMCID: PMC12113575 DOI: 10.3390/medicina61050798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 04/17/2025] [Accepted: 04/23/2025] [Indexed: 05/29/2025]
Abstract
Background and Objectives: Cutaneous melanoma (CM) poses a continuous challenge in oncology due to the developing resistance to available treatments. Doxorubicin (DOX) is noted as one of the most effective chemotherapeutics, although associated toxicity and resistance limit its use in CM treatment. Consequently, DOX has become a promising candidate for combination therapies targeting this neoplasm. Genistein (GEN) gathered significant attention due to its anti-neoplastic properties and ability to enhance the effects of DOX against several cancers, yet this association remains underexplored in CM. Therefore, this study investigated the combination therapy regimen comprising GEN and DOX in terms of anti-melanoma activity and safety profile. Materials and Methods: The in vitro experiments were performed on SK-MEL-28 and HaCaT cells. Cell viability was determined using MTT assay. Cell morphology and confluence were inspected microscopically. Nuclear and cytoskeletal aspects were assessed via immunofluorescence. Apoptosis and oxidative stress were quantified through caspase activity and intracellular reactive oxygen species (ROS) production, respectively. The irritant effect was evaluated on the chorioallantoic membrane. Results: The results revealed that the combination of GEN 10 µM with DOX (0.5 and 1 µM) provided augmented cytotoxic events (e.g., reduced cell viability, altered cell morphology and confluence, apoptotic-like impairments in nuclear shape and cytoskeletal network, increased caspases-3/7 and -9 activity, and elevated ROS) in SK-MEL-28 cells, compared to individual treatments, and exerted a strong synergistic interaction. Simultaneously, GEN 10 µM efficiently surpassed the toxic effects (e.g., viability and confluence loss, hypertrophy, and cytoskeletal condensation) of DOX (0.5 and 1 µM) in HaCaT cells. In ovo, GEN 10 µM + DOX 1 µM treatment was classified as non-irritant. Conclusions: These findings stand as one of the first contributions revealing the beneficial therapeutic interplay between GEN and DOX at physiologically achievable concentrations that resulted in elevated anti-tumor properties in CM cells and alleviated toxicity in keratinocytes.
Collapse
Affiliation(s)
- Andrea Roman
- Faculty of Medicine, “Vasile Goldis” Western University of Arad, 94 Revolutiei Blvd., 310130 Arad, Romania
- Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Andrei Motoc
- Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Iasmina Marcovici
- Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Cristina Dehelean
- Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Laura Nicolescu
- Faculty of Medicine, “Vasile Goldis” Western University of Arad, 94 Revolutiei Blvd., 310130 Arad, Romania
| | - Casiana Boru
- Faculty of Medicine, “Vasile Goldis” Western University of Arad, 94 Revolutiei Blvd., 310130 Arad, Romania
| |
Collapse
|
5
|
Zhu C, Chen J, Yan Z, Wang F, Sun Z, Liu Z, Li Y, Chen X, Bao Z, Li Q, Chen Z. IL-22 Alleviates Sepsis-Induced Acute Lung Injury by Inhibiting Epithelial Cell Apoptosis Associated with STAT3 Signalling. J Inflamm Res 2025; 18:5383-5398. [PMID: 40291457 PMCID: PMC12024478 DOI: 10.2147/jir.s496387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 03/29/2025] [Indexed: 04/30/2025] Open
Abstract
Purpose Sepsis is a critical condition characterized by organ dysfunction due to an aberrant response to infection, which results in a life-threatening situation. The lung, which is the most vulnerable target organ, is often severely damaged during sepsis. Research has demonstrated that interleukin-22 (IL-22), which is secreted by various immunocytes, can mitigate inflammation-associated diseases. Nevertheless, the precise function of IL-22 in sepsis-induced acute lung injury (SALI) is still unclear. This study aimed to investigate the therapeutic efficacy of IL-22 in sepsis and explore the regulatory mechanisms involved. Methods A mouse caecal ligation and puncture (CLP) model of sepsis was established, and the effect of IL-22 was investigated as indicated. Immunohistochemistry, qRT‒PCR, ELISA, immunofluorescence, TUNEL, Western blotting, and flow cytometry assays were applied to investigate the protective efficacy and involved pathways. Additionally, an in vitro model of lipopolysaccharide (LPS)-induced bronchial epithelial cell (BEAS-2B) apoptosis was established, and these cells were treated with or without recombinant IL-22 (rIL-22) to further evaluate the effect of IL-22 and the underlying mechanism. Results The experimental results clearly confirmed that the levels of IL-22 were increased in the serum and lung tissue after CLP. The administration of rIL-22 was observed to increase the survival rate of septic mice. Notably, rIL-22 treatment resulted in decreased levels of proteins and a decreased cell number in the bronchoalveolar lavage fluid, as well as in a reduction in inflammatory cytokine release into the serum. Importantly, rIL-22 mitigated SALI by inhibiting lung cell apoptosis in septic mice. Furthermore, the results revealed that rIL-22 attenuated apoptosis of lung epithelial cells via the activation of the STAT3 signalling pathway. Conclusion The results of this study suggest that IL-22 alleviates lung epithelial cell apoptosis to protect mice against SALI in association with the STAT3 signalling pathway, highlighting the potential therapeutic value of IL-22 against sepsis.
Collapse
Affiliation(s)
- Chiying Zhu
- Shenzhen Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, 518116, People’s Republic of China
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, People’s Republic of China
| | - Jiabo Chen
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, People’s Republic of China
- Department of Anesthesiology, School of Medicine, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, 330006, People’s Republic of China
| | - Zhengzheng Yan
- Laboratory Animal Research Center, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People’s Hospital), Dongguan, 523000, People’s Republic of China
| | - Fei Wang
- The Tenth Affiliated Hospital, Southern Medical University (Dongguan People’s Hospital), Dongguan, 523059, People’s Republic of China
| | - Ziqi Sun
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, People’s Republic of China
- Department of Anesthesiology, School of Medicine, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, 330006, People’s Republic of China
| | - Zeyu Liu
- Shenzhen Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, 518116, People’s Republic of China
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, People’s Republic of China
| | - Ying Li
- Laboratory Animal Research Center, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People’s Hospital), Dongguan, 523000, People’s Republic of China
| | - Xiaona Chen
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, People’s Republic of China
- Department of Biology, School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, People’s Republic of China
| | - Ziwei Bao
- Shenzhen Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, 518116, People’s Republic of China
| | - Quan Li
- Shenzhen Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, 518116, People’s Republic of China
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, People’s Republic of China
- Department of Anesthesiology, School of Medicine, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, 330006, People’s Republic of China
- Department of Biology, School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, People’s Republic of China
| | - Zhixia Chen
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, People’s Republic of China
- Department of Anesthesiology, School of Medicine, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, 330006, People’s Republic of China
| |
Collapse
|
6
|
Alonaizan R, Purnama U, Malandraki-Miller S, Gunadasa-Rohling M, Lewis A, Smart N, Carr C. MicroRNA-210 Enhances Cell Survival and Paracrine Potential for Cardiac Cell Therapy While Targeting Mitophagy. J Funct Biomater 2025; 16:147. [PMID: 40278255 PMCID: PMC12028018 DOI: 10.3390/jfb16040147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 04/04/2025] [Accepted: 04/16/2025] [Indexed: 04/26/2025] Open
Abstract
The therapeutic potential of presumed cardiac progenitor cells (CPCs) in heart regeneration has garnered significant interest, yet clinical trials have revealed limited efficacy due to challenges in cell survival, retention, and expansion. Priming CPCs to survive the hostile hypoxic environment may be key to enhancing their regenerative capacity. We demonstrate that microRNA-210 (miR-210), known for its role in hypoxic adaptation, significantly improves CPC survival by inhibiting apoptosis through the downregulation of Casp8ap2, a ~40% reduction in caspase activity, and a ~90% decrease in DNA fragmentation. Contrary to the expected induction of Bnip3-dependent mitophagy by hypoxia, miR-210 did not upregulate Bnip3, indicating a distinct anti-apoptotic mechanism. Instead, miR-210 reduced markers of mitophagy and increased mitochondrial biogenesis and oxidative metabolism, suggesting a role in metabolic reprogramming. Furthermore, miR-210 enhanced the secretion of paracrine growth factors from CPCs, with a ~1.6-fold increase in the release of stem cell factor and of insulin growth factor 1, which promoted in vitro endothelial cell proliferation and cardiomyocyte survival. These findings elucidate the multifaceted role of miR-210 in CPC biology and its potential to enhance cell-based therapies for myocardial repair by promoting cell survival, metabolic adaptation, and paracrine signalling.
Collapse
Affiliation(s)
- Rita Alonaizan
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK (C.C.)
- King Faisal Specialist Hospital & Research Centre, Riyadh 12713, Saudi Arabia
| | - Ujang Purnama
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK (C.C.)
| | | | - Mala Gunadasa-Rohling
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK (C.C.)
| | - Andrew Lewis
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK (C.C.)
| | - Nicola Smart
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK (C.C.)
| | - Carolyn Carr
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK (C.C.)
| |
Collapse
|
7
|
Xu F, Qiu J, Liu N, Wei H, Gao Y, Fei Y, Xi J, Yu Z, Fan X, Chen L, Xia Y, Dou X. Therapeutic Potential of Raspberry Extract in High-Fat Diet-Induced Liver Injury via Apoptosis and AMPK/PPARα Pathways. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:9408-9423. [PMID: 40168586 DOI: 10.1021/acs.jafc.4c09593] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
This study aimed to explore the efficacy and mechanisms of raspberry (Rubus idaeus L. fruit) aqueous extract (RE) in alleviating high-fat diet (HFD)-induced metabolic-associated fatty liver disease (MAFLD). The MAFLD mouse model was established to examine the effects of RE through liver transcriptome and metabolomics analysis. In this study, RE supplementation significantly alleviated HFD-induced liver injury, hepatosteatosis, inflammation, and insulin resistance. Liver transcriptome analysis demonstrated that RE supplementation favorably regulated signaling pathways involved in fatty acid metabolism and inflammation, including the AMPK signaling pathway, PPAR signaling pathway, apoptosis, etc. Furthermore, the injection of compound C, an antagonist of AMPK, notably reversed the hepatoprotective effects of RE, evidenced by increased lipid profile levels, accelerated fatty acid-related gene disorder, and increased positive tunnel staining area. Furthermore, liver metabolomics analysis demonstrated that RE treatment led to substantial enrichment of the liver tissue metabolite umbelliferone (UMB), which has the potential to ameliorate lipid accumulation and hepatocyte injury through the AMPK signaling pathway. In summary, RE intervention mitigated HFD-induced liver dysfunction in mice, with UMB likely being the primary component responsible for its therapeutic efficacy in the liver. In addition, this study provided new insights, suggesting that RE could be used as a promising therapeutic approach for modulating MAFLD via apoptosis and the AMPK/PPARα signaling pathway.
Collapse
Affiliation(s)
- Fangying Xu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Jiannan Qiu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Nian Liu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Huaxin Wei
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Yanyan Gao
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Yang Fei
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Jiale Xi
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Zhiling Yu
- Consun Chinese Medicines Research Centre for Renal Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 852, China
| | - Xiaohui Fan
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lin Chen
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Yongliang Xia
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Xiaobing Dou
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| |
Collapse
|
8
|
Chen W, Jiang Y, Zeng J, Liu D, Feng X, Cheng Y, Lu D, Sun Y, Zhu Q, Zhang X, Wang Q. FDX1 promotes elesclomol-induced PANoptosis in diffuse large B-cell lymphoma via activating IRF3/IFN-β signaling. Oncogene 2025:10.1038/s41388-025-03366-4. [PMID: 40240522 DOI: 10.1038/s41388-025-03366-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 03/02/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025]
Abstract
Diffuse large B-cell lymphoma (DLBCL) remains a major clinical challenge and requires the development of new therapeutic approaches. The identification of cuproptosis, a newly defined form of copper-induced cell death, has provided innovative insights for cancer therapy. Here, we report that loss of the mitochondrial matrix reductase FDX1 in DLBCL cells impairs the antitumor effect of elesclomol (ES), which performs its function by transporting excess copper into cells. Overexpressing (OE) FDX1 significantly sensitized DLBCL cells to ES-induced cell death in vitro and enhanced the anticancer activity of ES in vivo. Furthermore, treatment with ES in FDX1-high expression patient-derived xenograft (PDX) showed a significantly greater inhibitory effect than in FDX1-low expression PDX. Mechanistically, FDX1 promotes the induction of IFN-β-dependent PANoptosis by increasing IRF3 phosphorylation in DLBCL cells upon ES treatment. Consistent with this finding, patient cohort analysis revealed that FDX1 expression correlated positively with enhanced IRF3 phosphorylation. Together, our findings are the first to identify the central role of FDX1 in synergizing with ES to activate IFN-β signaling and induce PANoptosis. This study enables us to re-explore the clinical anticancer potential of ES as a novel therapeutic strategy for DLBCL.
Collapse
Affiliation(s)
- Weifeng Chen
- Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, 511518, China
- Affiliated Cancer Hospital/Institute and GMU-GIBH Joint School of Life Sciences of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, 511495, China
| | - Yuhang Jiang
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, China
| | - Jun Zeng
- Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, 511518, China
| | - Dandan Liu
- Affiliated Cancer Hospital/Institute and GMU-GIBH Joint School of Life Sciences of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, 511495, China
| | - Xiaoting Feng
- Affiliated Cancer Hospital/Institute and GMU-GIBH Joint School of Life Sciences of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, 511495, China
| | - Yiqiu Cheng
- Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, 511518, China
| | - Di Lu
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Yangbai Sun
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Qinyuan Zhu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, 311599, China.
| | - Xiaoren Zhang
- Affiliated Cancer Hospital/Institute and GMU-GIBH Joint School of Life Sciences of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, 511495, China.
| | - Qi Wang
- Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, 511518, China.
| |
Collapse
|
9
|
Vidak E, Vizovišek M, Kavčič N, Biasizzo M, Fonović M, Turk B. Apoptotic Caspases-3 and -7 Cleave Extracellular Domains of Membrane-Bound Proteins from MDA-MB-231 Breast Cancer Cells. Int J Mol Sci 2025; 26:3466. [PMID: 40331965 PMCID: PMC12026882 DOI: 10.3390/ijms26083466] [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: 01/07/2025] [Revised: 04/01/2025] [Accepted: 04/03/2025] [Indexed: 05/08/2025] Open
Abstract
Apoptotic executioner caspases-3 and -7 are the main proteases responsible for the execution of apoptosis. Apoptosis is the main form of programmed cell death involved in organism development and maintenance of homeostasis and is commonly impaired in various pathologies. Predominately an immunologically silent form of cell death, it can become immunogenic upon loss of membrane integrity during progression to secondary necrosis, which mostly occurs when apoptotic bodies are not efficiently cleared by efferocytosis. In cancer, the efferocytic capacity can be overwhelmed following chemotherapeutic treatment, thereby providing an opportunity for the potential extracellular functions of executioner apoptotic caspases in the tumor microenvironment. By triggering apoptosis in Jurkat E6.1 acute T cell leukemia cells, we demonstrated that during progression to secondary necrosis, executioner caspases-3 and -7 can be found in the extracellular space. Furthermore, we showed that extracellularly active caspases-3 and -7 can cleave extracellular domains of membrane-bound proteins from MDA-MB-231 breast cancer cells, a function generally executed in the tumor microenvironment by several extracellular proteases from metalloprotease and cathepsin families. As such, this study provides the evidence for the potential involvement of apoptotic caspases-3 and -7 in extracellular proteolytic networks. Presented mass spectrometry data are available via ProteomeXchange with identifier PXD061399.
Collapse
Affiliation(s)
- Eva Vidak
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia; (E.V.); (M.V.); (N.K.); (M.F.)
- Jožef Stefan International Postgraduate School, Jamova cesta 39, SI-1000 Ljubljana, Slovenia
| | - Matej Vizovišek
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia; (E.V.); (M.V.); (N.K.); (M.F.)
| | - Nežka Kavčič
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia; (E.V.); (M.V.); (N.K.); (M.F.)
| | - Monika Biasizzo
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia; (E.V.); (M.V.); (N.K.); (M.F.)
| | - Marko Fonović
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia; (E.V.); (M.V.); (N.K.); (M.F.)
| | - Boris Turk
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia; (E.V.); (M.V.); (N.K.); (M.F.)
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Vecna pot 113, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
10
|
Xiao J, Wang L, Zhang B, Hou A. Cell death in acute lung injury: caspase-regulated apoptosis, pyroptosis, necroptosis, and PANoptosis. Front Pharmacol 2025; 16:1559659. [PMID: 40191423 PMCID: PMC11968751 DOI: 10.3389/fphar.2025.1559659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 03/03/2025] [Indexed: 04/09/2025] Open
Abstract
There has been abundant research on the variety of programmed cell death pathways. Apoptosis, pyroptosis, and necroptosis under the action of the caspase family are essential for the innate immune response. Caspases are classified into inflammatory caspase-1/4/5/11, apoptotic caspase-3/6/7, and caspase-2/8/9/10. Although necroptosis is not caspase-dependent to transmit cell death signals, it can cross-link with pyroptosis and apoptosis signals under the regulation of caspase-8. An increasing number of studies have reiterated the involvement of the caspase family in acute lung injuries caused by bacterial and viral infections, blood transfusion, and ventilation, which is influenced by noxious stimuli that activate or inhibit caspase engagement pathways, leading to subsequent lung injury. This article reviews the role of caspases implicated in diverse programmed cell death mechanisms in acute lung injury and the status of research on relevant inhibitors against essential target proteins of the described cell death mechanisms. The findings of this review may help in delineating novel therapeutic targets for acute lung injury.
Collapse
Affiliation(s)
| | | | | | - Ana Hou
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
11
|
Tang K, Zhou L, Tian X, Fang SY, Vandenbulcke E, Du A, Shen J, Cao H, Zhou J, Chen K, Kim HR, Luo Z, Xin S, Lin SH, Park D, Yang L, Zhang Y, Suzuki K, Majety M, Ling X, Lam SZ, Chow RD, Ren P, Tao B, Li K, Codina A, Dai X, Shang X, Bai S, Nottoli T, Levchenko A, Booth CJ, Liu C, Fan R, Dong MB, Zhou X, Chen S. Cas12a-knock-in mice for multiplexed genome editing, disease modelling and immune-cell engineering. Nat Biomed Eng 2025:10.1038/s41551-025-01371-2. [PMID: 40114032 DOI: 10.1038/s41551-025-01371-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 02/13/2025] [Indexed: 03/22/2025]
Abstract
The pleiotropic effects of human disease and the complex nature of gene-interaction networks require knock-in mice allowing for multiplexed gene perturbations. Here we describe a series of knock-in mice with a C57BL/6 background and with the conditional or constitutive expression of LbCas12a or of high-fidelity enhanced AsCas12a, which were inserted at the Rosa26 locus. The constitutive expression of Cas12a in the mice did not lead to discernible pathology and enabled efficient multiplexed genome engineering. We used the mice for the retrovirus-based immune-cell engineering of CD4+ and CD8+ T cells, B cells and bone-marrow-derived dendritic cells, for autochthonous cancer modelling through the delivery of multiple CRISPR RNAs as a single array using adeno-associated viruses, and for the targeted genome editing of liver tissue using lipid nanoparticles. We also describe a system for simultaneous dual-gene activation and knockout (DAKO). The Cas12a-knock-in mice and the viral and non-viral delivery vehicles provide a versatile toolkit for ex vivo and in vivo applications in genome editing, disease modelling and immune-cell engineering, and for the deconvolution of complex gene interactions.
Collapse
Affiliation(s)
- Kaiyuan Tang
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Combined Program in the Biological and Biomedical Sciences, Yale University, New Haven, CT, USA
- Molecular Cell Biology, Genetics, and Development Program, Yale University, New Haven, CT, USA
| | - Liqun Zhou
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Combined Program in the Biological and Biomedical Sciences, Yale University, New Haven, CT, USA
- Immunobiology Program, Yale University, New Haven, CT, USA
| | - Xiaolong Tian
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Shao-Yu Fang
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
| | - Erica Vandenbulcke
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Yale College, Yale University, New Haven, CT, USA
| | - Andrew Du
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Yale College, Yale University, New Haven, CT, USA
| | - Johanna Shen
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Yale College, Yale University, New Haven, CT, USA
| | - Hanbing Cao
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
| | - Jerry Zhou
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Yale College, Yale University, New Haven, CT, USA
| | - Krista Chen
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Yale College, Yale University, New Haven, CT, USA
| | - Hyunu R Kim
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
| | - Zhicheng Luo
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Combined Program in the Biological and Biomedical Sciences, Yale University, New Haven, CT, USA
| | - Shan Xin
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
| | - Shawn H Lin
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Combined Program in the Biological and Biomedical Sciences, Yale University, New Haven, CT, USA
- Immunobiology Program, Yale University, New Haven, CT, USA
| | - Daniel Park
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Yale College, Yale University, New Haven, CT, USA
| | - Luojia Yang
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Combined Program in the Biological and Biomedical Sciences, Yale University, New Haven, CT, USA
- Molecular Cell Biology, Genetics, and Development Program, Yale University, New Haven, CT, USA
| | - Yueqi Zhang
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
| | - Kazushi Suzuki
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
| | - Medha Majety
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Yale College, Yale University, New Haven, CT, USA
| | - Xinyu Ling
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
| | - Stanley Z Lam
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Yale College, Yale University, New Haven, CT, USA
| | - Ryan D Chow
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- M.D.-Ph.D. Program, Yale University, West Haven, CT, USA
| | - Ping Ren
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
| | - Bo Tao
- System Biology Institute, Yale University, West Haven, CT, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Keyi Li
- Combined Program in the Biological and Biomedical Sciences, Yale University, New Haven, CT, USA
| | - Adan Codina
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Combined Program in the Biological and Biomedical Sciences, Yale University, New Haven, CT, USA
- Molecular Cell Biology, Genetics, and Development Program, Yale University, New Haven, CT, USA
| | - Xiaoyun Dai
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Center for Genome Editing, Westlake Laboratory of Life Sciences and Biomedicine, School of Medicine, Westlake University, Hangzhou, China
| | - Xingbo Shang
- System Biology Institute, Yale University, West Haven, CT, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Suxia Bai
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Timothy Nottoli
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Andre Levchenko
- System Biology Institute, Yale University, West Haven, CT, USA
- Immunobiology Program, Yale University, New Haven, CT, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, USA
| | - Carmen J Booth
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Chen Liu
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Rong Fan
- System Biology Institute, Yale University, West Haven, CT, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
- Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, CT, USA
- Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, USA
- Yale Center for RNA Science and Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Matthew B Dong
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA.
- System Biology Institute, Yale University, West Haven, CT, USA.
- M.D.-Ph.D. Program, Yale University, West Haven, CT, USA.
- Department of Medicine, Johns Hopkins Hospital, Baltimore, MD, USA.
| | - Xiaoyu Zhou
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA.
- System Biology Institute, Yale University, West Haven, CT, USA.
| | - Sidi Chen
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA.
- System Biology Institute, Yale University, West Haven, CT, USA.
- Combined Program in the Biological and Biomedical Sciences, Yale University, New Haven, CT, USA.
- Molecular Cell Biology, Genetics, and Development Program, Yale University, New Haven, CT, USA.
- Immunobiology Program, Yale University, New Haven, CT, USA.
- Yale College, Yale University, New Haven, CT, USA.
- M.D.-Ph.D. Program, Yale University, West Haven, CT, USA.
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, USA.
- Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, CT, USA.
- Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, USA.
- Yale Center for RNA Science and Medicine, Yale University School of Medicine, New Haven, CT, USA.
- Yale Liver Center, Yale University School of Medicine, New Haven, CT, USA.
- Yale Center for Biomedical Data Science, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
12
|
Kweon J, Lim W, Lee H, Kim J, Song G, Jeong W, Ham J. Cypermethrin triggers oxidative stress, apoptosis, and inflammation in bovine mammary glands by disruption of mitogen-activated protein kinase (MAPK) pathways and calcium homeostasis. Reprod Toxicol 2025; 132:108842. [PMID: 39884399 DOI: 10.1016/j.reprotox.2025.108842] [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: 09/18/2024] [Revised: 12/15/2024] [Accepted: 01/22/2025] [Indexed: 02/01/2025]
Abstract
Cyano-(3-phenoxyphenyl)methyl]3-(2,2-dichloroethenyl)-2,2-dimethylcyclopropane-1-carboxylate (cypermethrin) is a pyrethroid insecticide that is widely used to repel insects, such as cockroaches and ants. In addition to the target insects, its hazards have been outlined for carp; mice; and the nervous, reproductive, and gastrointestinal systems of humans. However, the effects of cypermethrin on the mammary tissue and milk production in dairy cattle remain unknown. Therefore, in the present study, we aimed to elucidate the impact of cypermethrin on dairy cattle using bovine mammary epithelial cells (MAC-T), which play key roles in milk yield and quality maintenance. First, we assessed the effects of cypermethrin on cell viability, proliferation, and cell cycle progression, followed by correlated gene expression analysis. Cypermethrin-treated cells exhibited G1 phase arrest and an increase in the sub G1 population. The population of MAC-T cells in both early and late apoptotic phases was increased following cypermethrin exposure. Moreover, cypermethrin caused mitochondrial calcium overload and diminished the mitochondrial membrane potential in MAC-T cells. We also observed the disruption of mitogen-activated protein kinase (MAPK) cascades and eventually, apoptotic cell death and excessive oxidative stress in cypermethrin-exposed MAC-T cells. In addition, cypermethrin affects the transcription levels related to apoptosis and inflammation, which may lead to the development of clinical morbidities, such as mastitis.
Collapse
Affiliation(s)
- Junhun Kweon
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Whasun Lim
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Hojun Lee
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Jinyoung Kim
- Department of Animal Biotechnology, Dankook University, Cheonan 31116, Republic of Korea
| | - Gwonhwa Song
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea.
| | - Wooyoung Jeong
- Department of Biomedical Sciences, Catholic Kwandong University, Gangneung 25601, Republic of Korea.
| | - Jiyeon Ham
- Division of Animal and Dairy Science, Chungnam National University, Daejeon 34134, Republic of Korea.
| |
Collapse
|
13
|
Jiang RD, Luo YZ, Lin HF, Zheng XS, Zeng WT, Liu MQ, Deng HH, Wang Q, Lai YN, Chen Y, Guo ZS, Zeng Y, Gong QC, Qiu C, Dong M, Wang X, Wang ZY, Ji LN, Hou PP, Li Q, Shen XR, Li B, Gao Y, Zhang AH, Jiang TT, Shi AM, Zhou P, Lin XH, Deng ZQ, Li JM, Shi ZL. Impaired inflammatory resolution with severe SARS-CoV-2 infection in leptin knock out obese hamster. iScience 2025; 28:111837. [PMID: 39981511 PMCID: PMC11841202 DOI: 10.1016/j.isci.2025.111837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 09/28/2024] [Accepted: 12/13/2024] [Indexed: 02/22/2025] Open
Abstract
Comorbidities, such as obesity, increase the risk of severe COVID-19. However, the mechanisms underlying severe illnesses in individuals with obesity are poorly understood. Here, we used gene-edited leptin knock out (Leptin -/-) obese hamsters to establish a severe infection model. This model exhibits robust viral replication, severe lung lesions, pronounced clinical symptoms, and fatal infection, mirroring severe COVID-19 in patients with obesity. Using single-cell transcriptomics on lung tissues pre- and post-infection, we found that monocyte-derived alveolar macrophages (MD-AM) play a key role in lung hyper-inflammation, including two unique MD-AM cell fate branches specific to Leptin -/- hamsters. Notably, reduced Trem2-dependent efferocytosis pathways in Leptin -/- hamsters indicated weakened inflammation resolution, consistent with the scRNA-seq data from patients with obesity. In summary, our study highlights the obesity-associated mechanisms underlying severe SARS-CoV-2 infections and establishes a reliable preclinical animal model for developing obesity-specific therapeutics for critical COVID-19.
Collapse
Affiliation(s)
- Ren-Di Jiang
- State Key Laboratory of Genetic Engineering, Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yun-Zhe Luo
- BGI Research, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Hao-Feng Lin
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Xiao-Shuang Zheng
- The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Wen-Tao Zeng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Mei-Qin Liu
- The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Hao-Hao Deng
- BGI Research, Beijing, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, China
| | - Qi Wang
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Ya-Na Lai
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Ying Chen
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Zi-Shuo Guo
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Ya Zeng
- BGI Research, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Qian-Chun Gong
- State Key Laboratory of Genetic Engineering, Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chen Qiu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Mei Dong
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Xi Wang
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Zi-Yi Wang
- National Engineering Research Center of Neuromodulation, School of Aerospace Engineering, Tsinghua University, Beijing, China
| | - Li-Na Ji
- School of Life Sciences, Inner Mongolia University, Hohhot, China
- Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, China
| | - Pan-Pan Hou
- Guangzhou National Laboratory, Guangzhou, China
| | - Qian Li
- The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Xu-Rui Shen
- Guangzhou National Laboratory, Guangzhou, China
| | - Bei Li
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Yun Gao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Ai-Hua Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Ting-Ting Jiang
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Ai-Min Shi
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Peng Zhou
- The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Xin-Hua Lin
- State Key Laboratory of Genetic Engineering, Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
- School of Life Sciences, Inner Mongolia University, Hohhot, China
- Shanghai Key Laboratory of Lung Inflammation and Injury, Shanghai, China
- Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, China
| | - Zi-Qing Deng
- BGI Research, Beijing, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, China
| | - Jian-Min Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Zheng-Li Shi
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- Guangzhou National Laboratory, Guangzhou, China
| |
Collapse
|
14
|
Mao H, Dumas EK, Starnbach MN. Chlamydia trachomatis impairs T cell priming by inducing dendritic cell death. Infect Immun 2025; 93:e0040224. [PMID: 39772728 PMCID: PMC11834465 DOI: 10.1128/iai.00402-24] [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: 09/11/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
The lack of effective adaptive immunity against Chlamydia trachomatis leads to chronic or repeated infection and serious disease sequelae. Dendritic cells (DCs) are professional antigen-presenting cells that are crucial for the activation of T cells during C. trachomatis infection. cDC1s and cDC2s are the two main DC subsets responsible for T cell priming, but little is known about how C. trachomatis affects their ability to prime T cells. Using a mouse model of infection, we found that C. trachomatis uptake reduced the viability of cDC1s and cDC2s both in vitro and in vivo, with cDC1s experiencing more death. DC death was mainly due to apoptosis and is alleviated in Casp3/7 or Bak1/Bax knockout DCs. In addition, we observed that C. trachomatis-specific CD8+ T cells were preferentially activated by cDC1s. Reduction in DC viability by C. trachomatis impaired the ability of infected DCs to activate T cells upon co-culture, although in the case of CD8+ T cell priming, controlling for viability was insufficient to fully rescue the defect. RNA sequencing of DCs from infected mice showed upregulation of cell death pathways, supporting our observations of DC death caused by C. trachomatis. Finally, we validated our findings with human DCs in vitro, observing C. trachomatis-induced cell death. These results indicate that C. trachomatis may evade the adaptive immune system by directly inducing cell death in DCs.
Collapse
Affiliation(s)
- Haitong Mao
- />Department of Microbiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Eric K. Dumas
- />Department of Microbiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael N. Starnbach
- />Department of Microbiology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
15
|
García-Hernández N, Calzada F, Bautista E, Sánchez-López JM, Valdes M, Hernández-Caballero ME, Ordoñez-Razo RM. Quantitative Proteomics and Molecular Mechanisms of Non-Hodgkin Lymphoma Mice Treated with Incomptine A, Part II. Pharmaceuticals (Basel) 2025; 18:242. [PMID: 40006055 PMCID: PMC11858899 DOI: 10.3390/ph18020242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/14/2024] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Incomptine A (IA) has cytotoxic activity in non-Hodgkin lymphoma (NHL) cancer cell lines. Its effects on U-937 cells include induction of apoptosis, production of reactive oxygen species, and inhibition of glycolytic enzymes. We examined the altered protein levels present in the lymph nodes of an in vivo mouse model. Methods: We induced an in vivo model with Balb/c mice with U-937 cells and treated it with IA or methotrexate, as well as healthy mice. We determined expressed proteins by TMT based on the LC-MS/MS method (Data are available via ProteomeXchange with identifier PXD060392) and a molecular docking study targeting 15 deregulated proteins. We developed analyses through the KEGG, Reactome, and Gene Ontology databases. Results: A total of 2717 proteins from the axillary and inguinal lymph nodes were analyzed and compared with healthy mice. Of 412 differentially expressed proteins, 132 were overexpressed (FC ≥ 1.5) and 117 were underexpressed (FC ≤ 0.67). This altered expression was associated with 20 significantly enriched processes, including chromatin remodeling, transcription, translation, metabolic and energetic processes, oxidative phosphorylation, glycolysis/gluconeogenesis, cell proliferation, cytoskeletal organization, and with cell death with necroptosis. Conclusions: We confirmed the previously observed dose-dependent effect of IA as a secondary metabolite with important potential as an anticancer agent for the treatment of NHL, showing that the type of drug or the anatomical location influences the response to treatment. The IA promises to be a likely safer and more effective treatment to improve outcomes, reduce toxicities, and improve survival in patients with NHL, initially targeting histones and transcription factors that will affect cell death proteins.
Collapse
Affiliation(s)
- Normand García-Hernández
- Unidad de Investigación Médica en Genética Humana, UMAE Hospital Pediatría 2° Piso, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, Mexico City 06725, Mexico;
| | - Fernando Calzada
- Unidad de Investigación Médica en Farmacología, UMAE Hospital de Especialidades, 2° Piso CORSE, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, Mexico City 06725, Mexico;
| | - Elihú Bautista
- SECIHTI-División de Biología Molecular, Instituto Potosino de Investigación Científica y Tecnológica A.C., San Luis Potosí 78216, San Luis Potosí, Mexico;
| | - José Manuel Sánchez-López
- Hospital Infantil de Tlaxcala, Investigación y Enseñanza, 20 de Noviembre S/M, San Matias Tepetomatitlan, Apetatitlan de de Antonio Carvajal 90606, Tlaxcala, Mexico;
- Phagocytes Architecture and Dynamics, IPBS, UMR5089 CNRS-Université Toulouse 3, 205 route de Narbonne, 31077 Toulouse, France
| | - Miguel Valdes
- Unidad de Investigación Médica en Farmacología, UMAE Hospital de Especialidades, 2° Piso CORSE, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, Mexico City 06725, Mexico;
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Salvador Díaz Mirón S/N, Col. Casco de Santo Tomás, Miguel Hidalgo, Mexico City 11340, Mexico
| | | | - Rosa María Ordoñez-Razo
- Unidad de Investigación Médica en Genética Humana, UMAE Hospital Pediatría 2° Piso, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, Mexico City 06725, Mexico;
| |
Collapse
|
16
|
Ji Y, Zhang Y, Si W, Guo J, Liu G, Wang C, Khan MZ, Zhao X, Liu W. Aflatoxin B1-Induced Apoptosis in Donkey Kidney via EndoG-Mediated Endoplasmic Reticulum Stress. Vet Sci 2025; 12:130. [PMID: 40005890 PMCID: PMC11860441 DOI: 10.3390/vetsci12020130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/16/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
Aflatoxin B1 (AFB1) is a prevalent environmental and forage contaminant that poses significant health risks to both humans and livestock due to its toxic effects on various organs and systems. Among its toxicological effects, nephrotoxicity is a hallmark of AFB1 exposure. However, the precise mechanisms underlying AFB1-induced kidney damage in donkeys remain poorly understood. To investigate this, we established a donkey model exposed to AFB1 by administering a diet supplemented with 1 mg AFB1/kg for 30 days. Kidney apoptosis was assessed using TUNEL staining, while gene expression and protein levels of Endonuclease G (EndoG), as well as genes related to endoplasmic reticulum (ER) stress and apoptosis, were quantified by RT-qPCR and Western blotting. Our findings indicate that AFB1 exposure resulted in significant kidney injury, apoptosis, and oxidative stress. Notably, AFB1 exposure upregulated the expression of EndoG and promoted its translocation to the ER, which subsequently induced ER stress and activated the mitochondrial apoptotic pathway. These results suggest that AFB1-induced kidney damage in donkeys is mediated through the oxidative stress and mitochondrial apoptosis pathways, primarily involving the EndoG-IRE1/ATF6-CHOP signaling axis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xia Zhao
- College of Agriculture and Biology, Liaocheng University, Liaocheng 252000, China
| | - Wenqiang Liu
- College of Agriculture and Biology, Liaocheng University, Liaocheng 252000, China
| |
Collapse
|
17
|
Wu Y, Qu H, Li X, Liu X, Wang L, Xia X, Wu X. Excessive autophagy-inducing and highly penetrable biomineralized bacteria for multimodal imaging-guided and mild hyperthermia-enhanced immunogenic cell death. J Colloid Interface Sci 2025; 679:181-196. [PMID: 39362143 DOI: 10.1016/j.jcis.2024.09.246] [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: 09/10/2024] [Revised: 09/23/2024] [Accepted: 09/29/2024] [Indexed: 10/05/2024]
Abstract
The tumor microenvironment, characterized by hypoxia, supports the efficacy of anaerobic bacteria like attenuated S. typhimurium in cancer therapies. These bacteria target and penetrate deep tumor regions, significantly reducing tumor size but often lead to tumor regrowth due to limited long-term efficacy. To enhance the therapeutic impact, a novel biohybrid system, S@UIL, has been developed by coating S. typhimurium with a zirconium-based nanoscale metal-organic framework (UiO-66-NH2) loaded with indocyanine green (ICG) and luteolin (LUT). This system maintains the bacteria's tumor-targeting ability while increasing the penetration and therapeutic effectiveness through excessive autophagy and mild hyperthermia. In a subcutaneous colon cancer model, the integration of LUT and ICG promotes autophagic cell death and photothermal sensitization, leading to the release of damage-associated molecular patterns (DAMPs). These DAMPs activate immune responses through dendritic cells and T-cells, enhancing immunogenic cell death (ICD) and potentially reducing immune evasion by tumors. This single-administration approach also integrates multimodal imaging capabilities, providing a promising strategy for improved tumor ICD induction and cancer progression inhibition.
Collapse
Affiliation(s)
- Yundi Wu
- School of Biomedical Engineering, State Key Laboratory of Marine Resource Utilization in the South China Sea, Hainan University, Haikou 570228, China; School of Life and Health Sciences, Key Laboratory of Biomedical Engineering of Hainan Province, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China; NHC Key Laboratory of Tropical Disease Control, Hainan Medical University, Haikou 571199, China
| | - Huanran Qu
- School of Biomedical Engineering, State Key Laboratory of Marine Resource Utilization in the South China Sea, Hainan University, Haikou 570228, China; School of Life and Health Sciences, Key Laboratory of Biomedical Engineering of Hainan Province, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Xiangying Li
- Department of Radiology, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou 570208, China
| | - Xiande Liu
- School of Life and Health Sciences, Key Laboratory of Biomedical Engineering of Hainan Province, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Lei Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang 453003, China
| | - Xiaojing Xia
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang 453003, China
| | - Xilong Wu
- School of Biomedical Engineering, State Key Laboratory of Marine Resource Utilization in the South China Sea, Hainan University, Haikou 570228, China; School of Life and Health Sciences, Key Laboratory of Biomedical Engineering of Hainan Province, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China.
| |
Collapse
|
18
|
Kuzu B, Arzuk E. Discovery of New Pyrazole-Tosylamide Derivatives as Apoptosis Inducers Through BCL-2 Inhibition and Caspase-3 Activation. Chem Biodivers 2025; 22:e202401673. [PMID: 39353043 DOI: 10.1002/cbdv.202401673] [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: 07/10/2024] [Revised: 09/17/2024] [Accepted: 10/01/2024] [Indexed: 10/04/2024]
Abstract
In this presented study, a series of new carbonitrile-substituted pyrazole-tosyl amide derivatives were designed and synthesized according to previous studies. The antiproliferative effects of the synthesized compounds on MDA-MB-231, MCF-7, HepG2, PC-3, and A549 cancer cell lines were assessed by MTT assay compared with non-cancerous cells. The results demonstrate that compounds 9d, 9e, and 9f had a higher antiproliferative effect (IC50 <10 μM) against both breast cancer cells. To investigate the ability of these compounds (9d-f) to induce apoptosis against breast cancer cells, BCL-2 levels and Caspase-3 activities of compound-treated breast cancer cell lines were measured by ELISA. The results revealed that these compounds significantly inhibited the levels of anti-apoptotic protein BCL-2 and increased the activity of apoptotic protein Caspase-3 in MDA-MB-231 and MCF-7 cells. Molecular docking studies confirmed that the selected compounds have high binding affinity towards the active site in the crystal structures of BCL-2 and Caspase-3. Moreover, drug-likeness and pre-ADMET evaluation showed that the compounds had suitable drug properties. This study may be a new milestone in terms of the promising importance of carbonitrile-substituted pyrazole-tosyl amide scaffolds as apoptosis-inducing agents for cancer therapy in the future.
Collapse
Affiliation(s)
- Burak Kuzu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Van Yuzuncu Yil University, Van, 65080, Türkiye
| | - Ege Arzuk
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Ege University, İzmir, 35040, Türkiye
| |
Collapse
|
19
|
Zhu L, Liu Y, Wang K, Wang N. Regulated cell death in acute myocardial infarction: Molecular mechanisms and therapeutic implications. Ageing Res Rev 2025; 104:102629. [PMID: 39644925 DOI: 10.1016/j.arr.2024.102629] [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: 09/09/2024] [Revised: 11/22/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
Acute myocardial infarction (AMI), primarily caused by coronary atherosclerosis, initiates a series of events that culminate in the obstruction of coronary arteries, resulting in severe myocardial ischemia and hypoxia. The subsequent myocardial ischemia/reperfusion (I/R) injury further aggravates cardiac damage, leading to a decline in heart function and the risk of life-threatening complications. The complex interplay of multiple regulated cell death (RCD) pathways plays a pivotal role in the pathogenesis of AMI. Each RCD pathway is orchestrated by a symphony of molecular regulatory mechanisms, highlighting the dynamic changes and critical roles of key effector molecules. Strategic disruption or inhibition of these molecular targets offers a tantalizing prospect for mitigating or even averting the onset of RCD, thereby limiting the extensive loss of cardiomyocytes and the progression of detrimental myocardial fibrosis. This review systematically summarizes the mechanisms underlying various forms of RCD, provides an in-depth exploration of the pathogenesis of AMI through the lens of RCD, and highlights a range of promising therapeutic targets that hold the potential to revolutionize the management of AMI.
Collapse
Affiliation(s)
- Lili Zhu
- Department of Pathology, Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yiyang Liu
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China; Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Kangkai Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China; Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Nian Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China; Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China.
| |
Collapse
|
20
|
Kumar Yadav A, Singh V, Acharjee S, Saha S, Kushwaha R, Dutta A, Koch B, Banerjee S. Sonodynamic Cancer Therapy by Mn(I)-tricarbonyl Complexes via Ultrasound-triggered CO Release and ROS Generation. Chemistry 2025; 31:e202403454. [PMID: 39503625 DOI: 10.1002/chem.202403454] [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: 09/20/2024] [Revised: 10/29/2024] [Accepted: 11/06/2024] [Indexed: 11/08/2024]
Abstract
A novel ferrocene conjugated Mn(I)-tricarbonyl complex viz [Mn(Fc-tpy)(CO)3Br] (Mn2) where, Fc-tpy=4'-ferrocenyl-2,2':6',2''-terpyridine was synthesized and fully characterized along with its non-ferrocene analog [Mn(Ph-tpy)(CO)3Br] Ph-tpy=4'-phenyl-2,2':6',2''-terpyridine (Mn1) for ultrasound (US) activated anticancer applications. The X-ray structure of Mn2 confirmed its distorted octahedral geometry. Mn1 and Mn2, for the first time, showed US-triggered release of CO and ROS generation (1O2 and ⋅OH) in an aqueous solution from any Mn(I)-tricarbonyl complexes, indicating its potential for synergetic CO gas therapy and sonodynamic therapy. The above-mentioned in-solution chemistry was successfully translated into in vitro cellular models. These complexes showed unprecedented US-triggered toxicity against T-cell lymphoma and human breast cancer cells (IC50 for Mn2<1 μM) while were minimally toxic without US or against normal spleen and human embryonic kidney cells. Mn2 was ca. 12 fold more anticancer active than Mn1, indicating that the ferrocene conjugation augmented the US sensitivity. The apoptotic sonotoxicity of Mn2 was due to US-promoted mitochondrial depolarization via ROS generation and CO release. The apoptosis was triggered by caspase 3 activation. This is the first report of Mn(I)-tricarbonyl-based sonosensitizers for cancer SDT. Overall, this study, for the first time, establishes the effectiveness of 3d metal carbonyls in SDT.
Collapse
Affiliation(s)
- Ashish Kumar Yadav
- Department of Chemistry, Indian Institute of Technology (BHU) Varanasi, Uttar Pradesh, 221005, India
| | - Virendra Singh
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India
| | - Sagar Acharjee
- Department of Chemistry, Indian Institute of Technology (BHU) Varanasi, Uttar Pradesh, 221005, India
| | - Sukanta Saha
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai, Maharashtra, 400076, India
| | - Rajesh Kushwaha
- Department of Chemistry, Indian Institute of Technology (BHU) Varanasi, Uttar Pradesh, 221005, India
| | - Arnab Dutta
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai, Maharashtra, 400076, India
| | - Biplob Koch
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India
| | - Samya Banerjee
- Department of Chemistry, Indian Institute of Technology (BHU) Varanasi, Uttar Pradesh, 221005, India
| |
Collapse
|
21
|
Schwarze‐Taufiq TA, Pranoto IKA, Hui K, Kinoshita C, Yu O, Crane PK, Gray SL, Young JE. Anticholinergic drugs and dementia risk: Using stem cell-based studies to complement pharmacoepidemiology. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2025; 11:e70040. [PMID: 39911736 PMCID: PMC11795422 DOI: 10.1002/trc2.70040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 02/07/2025]
Abstract
BACKGROUND Anticholinergic (AC) use remains common in older adults despite evidence of safety risks, including increased risk in dementia. Pharmacoepidemiology studies from various populations report associations between specific anticholinergic classes - antidepressants and bladder antimuscarinics - and increased dementia incidence. However, it is difficult to determine whether these associations are directly caused by the neurotoxic effects of anticholinergic drugs or by the underlying health conditions which the medications are taken for, known as confounding by indication. Here, we leverage human induced pluripotent stem cells-derived-neurons (hiPSC-Ns) to complement the pharmacoepidemiology studies by directly examining the effects of various anticholinergic classes on dementia-related cellular phenotypes. METHODS We treated human induced pluripotent stem cell (hiPSC)-derived neurons with eight drugs representing different AC medication classes, including antidepressants, bladder antimuscarinics, antihistamines, and antispasmodics. We analyzed these neurons for cytotoxicity, amyloid beta (Aβ) peptide levels in the conditioned medium, and the level of intracellular phosphorylated tau from these cultures. RESULTS We observed that antidepressants and bladder antimuscarinics were consistently cytotoxic, whereas antihistamines and antispasmodics did not show overt cytotoxicity at the times and concentrations that we tested. Some of the cytotoxic medications altered the amounts of Aβ1-42 peptides, but there were no significant differences in the intracellular ratio of phosphorylated tau/total tau between AC drug treatments. CONCLUSIONS These results corroborate population-based studies and suggest a molecular basis for the differences in dementia risk observed according to AC class. This warrants future work examining the effect of AC medications on hiPSC-derived cells from multiple subjects and examining other molecular outcomes including synaptic function and neuroinflammation in hiPSC-based models. Highlights Certain classes of anticholinergic (AC) medications are linked to dementia.Human-induced pluripotent stem cell (hiPSC) models are used to directly test the cytotoxicity of AC medications.AC classes that are associated with dementia are more neurotoxic.
Collapse
Affiliation(s)
- Tiara A. Schwarze‐Taufiq
- Department of Laboratory Medicine and PathologyUniversity of WashingtonSeattleWashingtonUSA
- Institute for Stem Cell and Regenerative MedicineUniversity of WashingtonSeattleWashingtonUSA
| | - Inez K. A. Pranoto
- Department of Laboratory Medicine and PathologyUniversity of WashingtonSeattleWashingtonUSA
- Institute for Stem Cell and Regenerative MedicineUniversity of WashingtonSeattleWashingtonUSA
| | - Katherine Hui
- Department of Laboratory Medicine and PathologyUniversity of WashingtonSeattleWashingtonUSA
- Institute for Stem Cell and Regenerative MedicineUniversity of WashingtonSeattleWashingtonUSA
| | - Chizuru Kinoshita
- Department of Laboratory Medicine and PathologyUniversity of WashingtonSeattleWashingtonUSA
- Institute for Stem Cell and Regenerative MedicineUniversity of WashingtonSeattleWashingtonUSA
| | - Onchee Yu
- Kaiser Permanente Washington Health Research InstituteSeattleWashingtonUSA
| | - Paul K. Crane
- Department of MedicineUniversity of WashingtonSeattleWashingtonUSA
| | - Shelly L. Gray
- School of PharmacyUniversity of WashingtonSeattleWashingtonUSA
| | - Jessica E. Young
- Department of Laboratory Medicine and PathologyUniversity of WashingtonSeattleWashingtonUSA
- Institute for Stem Cell and Regenerative MedicineUniversity of WashingtonSeattleWashingtonUSA
| |
Collapse
|
22
|
Algieri C, Cugliari A, Glogowski PA, Granata S, Fabbri M, Trombetti F, Bacci ML, Nesci S. Inside-out submitochondrial particles affect the mitochondrial permeability transition pore opening under conditions of mitochondrial dysfunction. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2025; 1866:149528. [PMID: 39615732 DOI: 10.1016/j.bbabio.2024.149528] [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: 08/29/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 12/10/2024]
Abstract
The inside-out submitochondrial particles (IO-SMPs) showed a strong protective effect against mitochondrial permeability transition pore (mPTP) opening in mitochondria isolated from swine hearts 3 h after explantation. The latter condition was used to emulate situation of mitochondrial damage. We identified that the protective effect of IO-SMPs cannot be attributed to a functional modulation of the enzymatic complexes involved in mPTP formation. Indeed, oxidative phosphorylation and F1FO-ATPase activity were not affected. Conversely, mPTP desensitization might be caused by structural modification. IO-SMP incorporation into the mitochondria can modulate the membrane-bound enzyme complexes' functionality, inducing F1FO-ATPase to be unable to carry out the conformational changes useful for mPTP opening. Thus, the data are a valid starting point for IO-SMP application in the treatment of impaired cardiovascular conditions supported by mPTP opening.
Collapse
Affiliation(s)
- Cristina Algieri
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano Emilia, Italy
| | - Antonia Cugliari
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano Emilia, Italy
| | | | | | - Micaela Fabbri
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano Emilia, Italy
| | - Fabiana Trombetti
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano Emilia, Italy
| | - Maria Laura Bacci
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano Emilia, Italy
| | - Salvatore Nesci
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano Emilia, Italy.
| |
Collapse
|
23
|
Bitgen N, Cakir M, Akkoc S, Donmez-Altuntas H. A Newly Synthesized Benzimidazolium Salt: 1-(2-Cyanobenzyl)-3-(4-Vinylbenzyl)-1H-Benzo[D]imidazol-3-ium Chloride as a Potential Anticancer Agent for Colon Cancer Treatment, In Vitro Study. J Biochem Mol Toxicol 2025; 39:e70112. [PMID: 39749430 DOI: 10.1002/jbt.70112] [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: 08/20/2024] [Revised: 10/23/2024] [Accepted: 12/21/2024] [Indexed: 01/04/2025]
Abstract
Colon cancer is one of the most common cancer-related deaths. Drug resistance is one of the biggest challenges in cancer treatment. Numerous pharmacological and biochemical investigations have documented the benzimidazole ring's anticancer, anti-inflammatory, and antioxidant properties. Within the scope of our project, the effect of newly synthesized benzimidazolium salt (BS) on cell proliferation was tested with MTT assay, and its effect on apoptosis and cell cycle was tested with annexin V and PI in the two different colon cancer cell lines (HT-29 and DLD-1). Our study examined the expressions of some genes related to apoptosis and, additionally, caspase activities with the multicaspase kit. BS showed an antiproliferative effect at lower doses in HT-29 colon cancer cells. When HT-29 cells were exposed to a 20 µM dosage, they showed increased caspase activity and apoptosis compared to DLD-1 cells. HT-29 accumulated in the G2/M phase of the cell cycle, whereas DLD-1 cells accumulated more in the S phase. In HT-29 cells, colony formation was inhibited; however, in DLD-1 cells, this effect was insufficient. Based on the apoptosis-death pathway, BS is expected to have anti-cancer effects. As a result of this work, this chemical was thoroughly examined in two different colon cancer cell lines, and additional, more comprehensive initiatives are being planned in light of the information obtained from this study.
Collapse
Affiliation(s)
- Nazmiye Bitgen
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
- Genome and Stem Cell Center, Erciyes University, Kayseri, Türkiye
| | - Mustafa Cakir
- Department of Medical Biology, Faculty of Medicine, Van Yuzuncu Yil University, Van, Türkiye
| | - Senem Akkoc
- Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Suleyman Demirel University, Isparta, Türkiye
- Faculty of Engineering and Natural Sciences, Bahçeşehir University, Istanbul, Türkiye
| | | |
Collapse
|
24
|
Qian S, Long Y, Tan G, Li X, Xiang B, Tao Y, Xie Z, Zhang X. Programmed cell death: molecular mechanisms, biological functions, diseases, and therapeutic targets. MedComm (Beijing) 2024; 5:e70024. [PMID: 39619229 PMCID: PMC11604731 DOI: 10.1002/mco2.70024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 11/02/2024] [Accepted: 11/11/2024] [Indexed: 01/12/2025] Open
Abstract
Programmed cell death represents a precisely regulated and active cellular demise, governed by a complex network of specific genes and proteins. The identification of multiple forms of programmed cell death has significantly advanced the understanding of its intricate mechanisms, as demonstrated in recent studies. A thorough grasp of these processes is essential across various biological disciplines and in the study of diseases. Nonetheless, despite notable progress, the exploration of the relationship between programmed cell death and disease, as well as its clinical application, are still in a nascent stage. Therefore, further exploration of programmed cell death and the development of corresponding therapeutic methods and strategies holds substantial potential. Our review provides a detailed examination of the primary mechanisms behind apoptosis, autophagy, necroptosis, pyroptosis, and ferroptosis. Following this, the discussion delves into biological functions and diseases associated dysregulated programmed cell death. Finally, we highlight existing and potential therapeutic targets and strategies focused on cancers and neurodegenerative diseases. This review aims to summarize the latest insights on programmed cell death from mechanisms to diseases and provides a more reliable approach for clinical transformation.
Collapse
Affiliation(s)
- Shen'er Qian
- Department of Otolaryngology Head and Neck SurgeryThe Third Xiangya Hospital, Central South UniversityChangshaHunanChina
| | - Yao Long
- Cancer Research InstituteSchool of Basic MedicineCentral South UniversityChangshaHunanChina
- Department of PathologyXiangya Hospital, Central South UniversityChangshaHunanChina
| | - Guolin Tan
- Department of Otolaryngology Head and Neck SurgeryThe Third Xiangya Hospital, Central South UniversityChangshaHunanChina
| | - Xiaoguang Li
- Department of Otolaryngology Head and Neck SurgeryShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Ear InstituteShanghai Jiao Tong University School of Medicine, Shanghai Key LabShanghaiChina
| | - Bo Xiang
- Cancer Research InstituteSchool of Basic MedicineCentral South UniversityChangshaHunanChina
- Furong LaboratoryCentral South UniversityChangshaHunanChina
| | - Yongguang Tao
- Cancer Research InstituteSchool of Basic MedicineCentral South UniversityChangshaHunanChina
| | - Zuozhong Xie
- Department of Otolaryngology Head and Neck SurgeryThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Xiaowei Zhang
- Department of Otolaryngology Head and Neck SurgeryThe Third Xiangya Hospital, Central South UniversityChangshaHunanChina
| |
Collapse
|
25
|
Gungordu S, Aptullahoglu E. Targeting MDM2-mediated suppression of p53 with idasanutlin: a promising therapeutic approach for acute lymphoblastic leukemia. Invest New Drugs 2024; 42:603-611. [PMID: 39305365 DOI: 10.1007/s10637-024-01473-9] [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: 07/17/2024] [Accepted: 09/17/2024] [Indexed: 12/08/2024]
Abstract
Despite available treatments for acute lymphoblastic leukemia (ALL), the disease's high clinical variability necessitates new therapeutic strategies, particularly for patients with high-risk features. The tumor suppressor protein p53, encoded by the TP53 gene and known as the guardian of the genome, plays a crucial role in preventing tumor development. Over 90% of ALL cases initially harbor wild-type TP53. Reactivation of p53, which is encoded from the wild type TP53 but lost its function for several reasons, is an attractive therapeutic approach in cancer treatment. p53 can be activated in a non-genotoxic manner by targeting its primary repressor, the MDM2 protein. Clinical trials involving MDM2 inhibitors are currently being conducted in a growing body of investigation, reflecting of the interest in incorporating these treatments into cancer treatment strategies. Early-phase clinical trials have demonstrated the promise of idasanutlin (RG7388), one of the developed compounds. It is a second-generation MDM2-p53 binding antagonist with enhanced potency, selectivity, and bioavailability. The aim of this study is to evaluate the efficacy of RG7388 as a therapeutic strategy for ALL and to investigate its potential impact on improving treatment outcomes for high-risk patients. RG7388 potently decreased the viability in five out of six ALL cell lines with diverse TP53 mutation profiles, whereas only one cell line exhibited high resistance. RG7388 induced a pro-apoptotic gene expression signature with upregulation of p53-target genes involved in the intrinsic and extrinsic pathways of apoptosis. Consequently, RG7388 led to a concentration-dependent increase in caspase-3/7 activity and cleaved poly (ADP-ribose) polymerase. In this research, RG7388 was investigated with pre-clinical methods in ALL cells as a novel treatment strategy. This study suggests further functional research and in-vivo evaluation, and it highlights the prospect of treating p53-functional ALL with MDM2 inhibitors.
Collapse
Affiliation(s)
- Seyda Gungordu
- Biotechnology Application and Research Centre, Bilecik Şeyh Edebali University, 11100, Bilecik, Turkey
| | - Erhan Aptullahoglu
- Biotechnology Application and Research Centre, Bilecik Şeyh Edebali University, 11100, Bilecik, Turkey.
- Department of Molecular Biology and Genetics, Faculty of Science, Bilecik Şeyh Edebali University, 11100, Bilecik, Turkey.
| |
Collapse
|
26
|
Campasino K, Yourick MR, Zhao Y, Sepehr E, Vaught C, Yourick JJ, Sprando RL, Gao X. Effect of cannabidiol and hemp extract on viability and function of hepatocytes derived from human induced pluripotent stem cells. Toxicol In Vitro 2024; 101:105933. [PMID: 39233107 DOI: 10.1016/j.tiv.2024.105933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/20/2024] [Accepted: 09/01/2024] [Indexed: 09/06/2024]
Abstract
Since the passage of the 2018 Agriculture Improvement Act (2018 Farm Bill), the number of products containing cannabis-derived compounds available to consumers have rapidly increased. Potential effects on liver function as a result from consumption of products containing cannabidiol (CBD), including hemp extracts, have been observed but the mechanisms for the effects are not fully understood. In this study, hepatocytes derived from human induced pluripotent stem cells (iPSCs) were used to evaluate potential hepatic effects of CBD and hemp extract at exposure concentrations ranging from 0.1 to 30 μM. Despite that a significant reduction in cell viability occurred only in the 30 μM group for both CBD and hemp extract, significant changes to cytochrome P450 activity, mitochondrial membrane potential, and lipid accumulation occurred within the concentration range of 0.1-3 μM for both CBD and hemp extract. Albumin and urea production, caspase 3/7 activity, and intracellular glutathione were significantly affected within the concentration range of 3-30 μM by CBD or hemp extract. These findings indicate that CBD and hemp extract can alter hepatic function and metabolism. The current study contributes data to help inform the evaluation of potential hepatotoxic effects of products containing cannabis-derived compounds.
Collapse
Affiliation(s)
- Kayla Campasino
- Division of Toxicology, Office of Applied Research and Safety Assessment, US FDA/CFSAN, Laurel, MD 20708, USA
| | - Miranda R Yourick
- Division of Toxicology, Office of Applied Research and Safety Assessment, US FDA/CFSAN, Laurel, MD 20708, USA
| | - Yang Zhao
- Division of Toxicology, Office of Applied Research and Safety Assessment, US FDA/CFSAN, Laurel, MD 20708, USA
| | - Estatira Sepehr
- Division of Toxicology, Office of Applied Research and Safety Assessment, US FDA/CFSAN, Laurel, MD 20708, USA
| | - Cory Vaught
- Division of Toxicology, Office of Applied Research and Safety Assessment, US FDA/CFSAN, Laurel, MD 20708, USA
| | - Jeffrey J Yourick
- Division of Toxicology, Office of Applied Research and Safety Assessment, US FDA/CFSAN, Laurel, MD 20708, USA
| | - Robert L Sprando
- Division of Toxicology, Office of Applied Research and Safety Assessment, US FDA/CFSAN, Laurel, MD 20708, USA
| | - Xiugong Gao
- Division of Toxicology, Office of Applied Research and Safety Assessment, US FDA/CFSAN, Laurel, MD 20708, USA.
| |
Collapse
|
27
|
Ahmadi Somaghian S, Pajouhi N, Dezfoulian O, Pirnia A, Kaeidi A, Rasoulian B. The protective effects of hyperoxic pre-treatment in human-derived adipose tissue mesenchymal stem cells against in vitro oxidative stress and a rat model of renal ischaemia-reperfusion. Arch Physiol Biochem 2024; 130:606-615. [PMID: 37506037 DOI: 10.1080/13813455.2023.2238918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/23/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023]
Abstract
OBJECTIVE Improvement of cell survival is essential for achieving better clinical outcomes in stem cell therapy. We investigated the effects of hyperoxic pre-treatment (HP) on the viability of human adipose stromal stem cells (ASCs). MATERIALS AND METHODS MTT and Western blot tests were used to assess cell viability and the expression of apoptosis-related proteins, respectively. For the in-vivo trial, the rats were subjected to renal ischaemia-reperfusion (IR). RESULTS The results showed that HP could significantly increase the viability of ASCs and decrease apoptotic markers (Bax/BCL-2 ratio and Caspase-3) compared with control cells. There were some additional effects with regard to the improvement of renal structure and function in the animal model. However, the difference between the treated and non-treated transplanted ASCs failed to reach significance. CONCLUSION These results suggested that HP could increase the survival of ASCs against oxidative stress-induced damages in the in-vitro condition, but this strategy was not highly effective in renal IR.
Collapse
Affiliation(s)
- Shahram Ahmadi Somaghian
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Naser Pajouhi
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Omid Dezfoulian
- Department of Pathobiology, School of Veterinary Medicine, Lorestan University, Khorramabad, Iran
| | - Afshin Pirnia
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Ayat Kaeidi
- Physiology-Pharmacology Research Center, Research Institute of Basic Medical Science, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Bahram Rasoulian
- Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| |
Collapse
|
28
|
Montesdeoca N, Johannknecht L, Efanova E, Heinen-Weiler J, Karges J. Ferroptosis Inducing Co(III) Polypyridine Sulfasalazine Complex for Therapeutically Enhanced Anticancer Therapy. Angew Chem Int Ed Engl 2024; 63:e202412585. [PMID: 39136323 DOI: 10.1002/anie.202412585] [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: 07/04/2024] [Indexed: 11/01/2024]
Abstract
Despite significant improvements in the treatment of cancerous tumors in the last decades, cancer remains one of the deadliest diseases worldwide. To overcome the shortcomings of currently applied chemotherapeutic treatments, much research efforts have been devoted towards the development of ferroptosis inducing anticancer agents. Ferroptosis is a newly described form of regulated, non-apoptotic cell death that is associated with high potential inside the clinics. Herein, the chemical synthesis and biological evaluation of a Co(III) polypyridine sulfasalazine complex as a ferroptosis inducer is reported. Upon entering the cancerous cells, the metal complex primarily accumulated in the mitochondria, triggering the production of hydroxy radicals and lipid peroxides, ultimately causing cell death by ferroptosis. The compound demonstrated to eradicate various monolayer cancer cells as well as colon carcinoma multicellular tumor spheroids. To the best of our knowledge this study reports on the first example of a Co(III) complex that is capable of inducing ferroptosis.
Collapse
Affiliation(s)
- Nicolás Montesdeoca
- Faculty of Chemistry and Biochemistry, Ruhr-University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany
| | - Lukas Johannknecht
- Faculty of Chemistry and Biochemistry, Ruhr-University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany
| | - Elizaveta Efanova
- Faculty of Chemistry and Biochemistry, Ruhr-University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany
| | - Jacqueline Heinen-Weiler
- Medical Imaging Center (MIC), Electron Microscopy Medical Analysis - Core Facility (EMMACF) Faculty of Medicine, Ruhr-University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany
| | - Johannes Karges
- Faculty of Chemistry and Biochemistry, Ruhr-University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany
| |
Collapse
|
29
|
Wei F, Cheng F, Li H, You J. Imidacloprid affects human cells through mitochondrial dysfunction and oxidative stress. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 951:175422. [PMID: 39128528 DOI: 10.1016/j.scitotenv.2024.175422] [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: 06/06/2024] [Revised: 08/07/2024] [Accepted: 08/07/2024] [Indexed: 08/13/2024]
Abstract
Given their relatively low persistence and mammalian toxicity, neonicotinoid pesticides have been extensively used worldwide and are omnipresent in the environment. Recent studies have shown that neonicotinoids may pose adverse effects on non-target organisms other than the known neurotoxicity, raising emerging concerns that these insecticides might pose human health risk through additional toxicity pathways. In the present study, the mitochondria function, oxidative stress, DNA damages, and genes transcription levels were examined in the human neuroblastoma SH-SY5Y cells after 48-h exposure to imidacloprid at concentrations from 0.05 to 200 μmol/L. Results showed that imidacloprid induced mitochondrial dysfunction with the degradation of adenosine triphosphate (ATP) and mitochondrial membrane potential (MMP) levels. In addition, imidacloprid caused oxidative stress by stimulating the generation of reactive oxygen species (ROS) and hydrogen peroxide (H2O2) via the disruption of calcium ion level and mitochondrial function. Ultimately, the oxidative stress continued to produce DNA damage and apoptosis in SH-SY5Y cells at imidacloprid concentrations above 47.6 μmol/L. Among the evaluated endpoints, ATP was the most sensitive, with a median activity concentration of 0.74 μmol/L. The 5 % hazard concentration of imidacloprid was estimated to be 0.69 μmol/L, which can be used as a threshold for human health risk assessment for imidacloprid. Collectively, our results provide an important support for further research on potential toxicity of neonicotinoids related to mitochondrial toxicity in humans.
Collapse
Affiliation(s)
- Fenghua Wei
- School of Chemistry and Environment, Jiaying University, Meizhou 514015, China; Guangdong Provincial Key Laboratory of Environmental Pollution and Health, College of Environment and Climate, Jinan University, Guangzhou 510632, China
| | - Fei Cheng
- Guangdong Provincial Key Laboratory of Environmental Pollution and Health, College of Environment and Climate, Jinan University, Guangzhou 510632, China; State Key Laboratory of Organic Geochemistry, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China
| | - Huizhen Li
- Guangdong Provincial Key Laboratory of Environmental Pollution and Health, College of Environment and Climate, Jinan University, Guangzhou 510632, China
| | - Jing You
- Guangdong Provincial Key Laboratory of Environmental Pollution and Health, College of Environment and Climate, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
30
|
Al-Othman R, Al-Jarallah A, Babiker F. High-density lipoprotein protects normotensive and hypertensive rats against ischemia-reperfusion injury through differential regulation of mTORC1 and mTORC2 signaling. Front Pharmacol 2024; 15:1398630. [PMID: 39611167 PMCID: PMC11603114 DOI: 10.3389/fphar.2024.1398630] [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: 03/10/2024] [Accepted: 10/29/2024] [Indexed: 11/30/2024] Open
Abstract
Background High-density lipoprotein (HDL) protects against myocardial ischemia-reperfusion (I/R) injury. Mammalian target of rapamycin complexes 1 and 2 (mTORC1 and mTORC2) play opposing roles in protecting against I/R injury, whereby mTORC1 appears to be detrimental while mTORC2 is protective. However, the role of HDL and mTORC signaling in protecting against I/R in hypertensive rodents is not clearly understood. In this study, we investigated the involvement of mTORC1 and mTORC2 in HDL-mediated protection against myocardial I/R injury in normotensive Wistar Kyoto (WKY) rats and spontaneously hypertensive rats (SHR). Methods Hearts from WKY and SHR were subjected to I/R injury using a modified Langendorff system. Hemodynamics data were collected, and infarct size was measured. Rapamycin and JR-AB2-011 were used to test the role of mTORC1 and mTORC2, respectively. MK-2206 was used to test the role of Akt in HDL-mediated cardiac protection. The expression levels and the activation states of mediators of mTORC1 and mTORC2 signaling and myocardial apoptosis were measured by immunoblotting and/or enzyme-linked immunosorbent assay (ELISA). Results HDL protected hearts from WKY and SHR against I/R injury as indicated by significant improvements in cardiac hemodynamics and reduction in infarct size. HDL induced greater protection in WKY compared to SHR. HDL treatment attenuated mTORC1 signaling in WKY by reducing the phosphorylation of P70S6K (mTORC1 substrate). In SHR however, HDL attenuated mTORC1 signaling by reducing the levels of phospho-mTORC1, Rag C (mTORC1 activator), and phospho-PRAS40 (mTORC1 inhibitor). HDL increased the phosphorylation of mTORC2 substrate Akt, specifically the Akt2 isoform in SHR and to a greater extent in WKY. HDL-induced protection was abolished in the presence of Akt antagonist and involved attenuation of GSK, caspases 7 and 8 activation, and cytochrome C release. Conclusion HDL mediates cardiac protection via attenuation of mTORC1, activation of mTORC2-Akt2, and inhibition of myocardial apoptosis. HDL regulates mTORC1 and mTORC2 signaling via distinct mechanisms in normotensive and hypertensive rats. HDL attenuation of mTORC1 and activation of mTORC2-Akt2 signaling could be a mechanism by which HDL protects against myocardial I/R injury in hypertension.
Collapse
Affiliation(s)
- Reham Al-Othman
- Department of Biochemistry, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Aishah Al-Jarallah
- Department of Biochemistry, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Fawzi Babiker
- Department of Physiology, College of Medicine, Kuwait University, Kuwait City, Kuwait
| |
Collapse
|
31
|
Chen JH, Raman V, Kuehne SA, Camilleri J, Hirschfeld J. Chemical, Antibacterial, and Cytotoxic Properties of Four Different Endodontic Sealer Leachates Over Time. J Endod 2024; 50:1612-1621. [PMID: 39197739 DOI: 10.1016/j.joen.2024.08.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 06/25/2024] [Accepted: 08/18/2024] [Indexed: 09/01/2024]
Abstract
INTRODUCTION The management of apical periodontitis involves the elimination of bacteria to achieve healing of the periapical tissues. To this end, understanding of the antimicrobial properties and cytotoxicity of root canal sealers over time is important. This study aimed to assess the cytocompatibility and antibacterial activity of leachates obtained from selected endodontic sealers. METHODS Four sealers were used in this study; AH Plus, an epoxy resin-based sealer, and three hydraulic calcium silicate-based sealers: AH Plus Bioceramic, BioRoot RCS, and BioRoot Flow. Sealer chemistry was assessed by scanning electron microscopy and energy dispersive spectrum analysis. Leachates of the tested sealers were prepared by immersing sealer discs in Hank's balanced salt solution for 28 and 90 days. Element release was assessed using inductively coupled plasma mass spectroscopy. Enterococcus faecalis and Fusobacterium nucleatum were exposed to the leachates followed by colony-forming unit determination, and the human osteoblast-like cell line Saos-2 was assessed with regard to cell death, caspase expression and activity of alkaline phosphatase (ALP) after stimulation with the leachates. RESULTS Calcium leaching was detected in the tricalcium silicate-based sealers, which increased the leachate pH significantly (P < .0001). AH Plus Bioceramic leachate displayed antimicrobial activity at 28 days against E. faecalis (P < .01). ALP levels were reduced in response to AH Plus Bioceramic and BioRoot RCS 1-day leachates (P < .05). High cell viability was observed after exposure to all sealers. CONCLUSIONS The leachates of the tested sealers were cytocompatible; however, their antimicrobial activity as well as their potential to induce the bone formation marker ALP was minimal.
Collapse
Affiliation(s)
- Jo-Hsun Chen
- School of Dentistry, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Veksina Raman
- School of Dentistry, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Sarah A Kuehne
- Nottingham Trent University, School of Science and Technology, Nottingham, United Kingdom
| | - Josette Camilleri
- School of Dentistry, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom.
| | - Josefine Hirschfeld
- School of Dentistry, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
32
|
Liu G, Zhou L, Mu K, Peng L, Ran F, Feng G, Liu Y. Based on Spectrum-Effect Relationship, Network Pharmacology, and Molecular Docking, the Material Basis and Mechanism of Antioxidant Effect of Miao Medicine Indigofera stachyoides Lindl Were Studied. ACS OMEGA 2024; 9:42199-42211. [PMID: 39431090 PMCID: PMC11483384 DOI: 10.1021/acsomega.4c04212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 09/10/2024] [Accepted: 09/25/2024] [Indexed: 10/22/2024]
Abstract
Indigofera stachyoides Lindl (IS.Lindl, Xuerensen in Chinese) is a traditional medicine frequently utilized by ethnic minorities; nevertheless, the chemicals responsible for these effects have not been identified. Ultraperformance liquid chromatography (UPLC) was utilized to establish the fingerprints of various origins. Free radical scavenging in 1,1-diphenyl-2-trinitrophenylhydrazine (DPPH) and 2,2-biazobis(3-ethyl-phenylpropylthiazole-6-sulfonate) diammonium salt (ABTS) assays was used to evaluate the antioxidant activity. Partial least-squares regression analysis (PLSR) and gray correlation analysis (GRA) were utilized to determine the spectral-effect relationship in order to screen the antioxidant pharmacodynamic components. The corresponding targets were obtained from Traditional Chinese Medicine Systems Pharmacology (TCMSP) and Integrated Traditional Chinese Medicine (ITCM). Disease-related targets for antioxidants were collected from GeneCards and Online Mendelian Inheritance in Man (OMIM) databases. The PPI interaction network analysis, GO enrichment analysis, and KEGG pathway analysis were established using the online analysis platform, and Autodock software assisted in aligning the components with important targets. The fingerprint profile revealed 32 common peaks, and eight standards were identified using standard comparison, with similarity ranging from 0.920 to 0.995. The primary antioxidant components include proanthocyanidin B1, epicatechin, and proanthocyanidin B3. Important targets include EGFR, CASP3, IL6, PTGS2, and TNF. Important signaling pathways include the AGE-RAGE signaling pathways in diabetic complications, the MAPK signaling pathway, the IL-17 signaling pathway, and the pathways in cancer. The results of molecular docking technology showed that the main active ingredients of the drug could bind well to the core target. In this study, we successfully established the spectral-effect relationship of IS.Lindl and clarified the effective substances of IS.Lindl. Through network pharmacology and molecular docking methods, it was clear that IS.Lindl plays an antioxidant role through multicomponent, multitarget, and multipathway synergy.
Collapse
Affiliation(s)
- Gang Liu
- Guizhou University of Traditional
Chinese Medicine, Guiyang 550025, Guizhou, China
| | | | - Kailang Mu
- Guizhou University of Traditional
Chinese Medicine, Guiyang 550025, Guizhou, China
| | - Leqiang Peng
- Guizhou University of Traditional
Chinese Medicine, Guiyang 550025, Guizhou, China
| | - Fei Ran
- Guizhou University of Traditional
Chinese Medicine, Guiyang 550025, Guizhou, China
| | - Guo Feng
- Guizhou University of Traditional
Chinese Medicine, Guiyang 550025, Guizhou, China
| | - Yuchen Liu
- Guizhou University of Traditional
Chinese Medicine, Guiyang 550025, Guizhou, China
| |
Collapse
|
33
|
Ma M, Wang Q, Liu Y, Li G, Liu L, Wang G, Guo Y, Huang S, Ma Q, Ji C, Zhao L. Bacillus CotA laccase improved the intestinal health, amino acid metabolism and hepatic metabolic capacity of Pekin ducks fed naturally contaminated AFB 1 diet. J Anim Sci Biotechnol 2024; 15:138. [PMID: 39385285 PMCID: PMC11465776 DOI: 10.1186/s40104-024-01091-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 08/20/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Aflatoxin B1 (AFB1) is a prevalent contaminant in agricultural products, presenting significant risks to animal health. CotA laccase from Bacillus licheniformis has shown significant efficacy in degrading mycotoxins in vitro test. The efficacy of Bacillus CotA laccase in animals, however, remains to be confirmed. A 2 × 2 factorial design was used to investigate the effects of Bacillus CotA laccase level (0 or 1 U/kg), AFB1 challenge (challenged or unchallenged) and their interactions on ducks. The purpose of this study was to evaluate the efficacy of Bacillus CotA laccase in alleviating AFB1 toxicosis of ducks. RESULTS Bacillus CotA laccase alleviated AFB1-induced declines in growth performance of ducks accompanied by improved average daily gain (ADG) and lower feed/gain ratio (F/G). Bacillus CotA laccase ameliorated AFB1-induced gut barrier dysfunctions and inflammation testified by increasing the jejunal villi height/crypt depth ratio (VH/CD) and the mRNA expression of tight junction protein 1 (TJP1) and zonula occluden-1 (ZO-1) as well as decreasing the expression of inflammation-related genes in the jejunum of ducks. Amino acid metabolome showed that Bacillus CotA laccase ameliorated AFB1-induced amino acid metabolism disorders evidenced by increasing the level of glutamic acid in serum and upregulating the expression of amino acid transport related genes in jejunum of ducks. Bacillus CotA laccase ameliorated AFB1-induced liver injury testified by suppressing oxidative stress, inhibiting apoptosis, and downregulating the expression of hepatic metabolic enzyme related genes of ducks. Moreover, Bacillus CotA laccase degraded AFB1 in digestive tract of ducks, resulting in the reduced absorption level of AFB1 across intestinal epithelium testified by the decreased level of AFB1-DNA adduct in the liver, and the reduced content of AFB1 residues in liver and feces of ducks. CONCLUSIONS Bacillus CotA laccase effectively improved the growth performance, intestinal health, amino acid metabolism and hepatic aflatoxin metabolism of ducks fed AFB1 diets, highlighting its potential as an efficient and safe feed enzyme for AFB1 degradation in animal production.
Collapse
Affiliation(s)
- Mingxin Ma
- State Key Laboratory of Animal Nutrition and Feeding, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, No. 2. Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Qianqian Wang
- State Key Laboratory of Animal Nutrition and Feeding, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, No. 2. Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Yanrong Liu
- State Key Laboratory of Animal Nutrition and Feeding, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, No. 2. Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Guiming Li
- Poultry Institute, Shandong Academy of Agricultural Sciences, Jinan, 250100, China
| | - Limeng Liu
- State Key Laboratory of Animal Nutrition and Feeding, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, No. 2. Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Gaigai Wang
- State Key Laboratory of Animal Nutrition and Feeding, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, No. 2. Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Yongpeng Guo
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
| | - Shimeng Huang
- State Key Laboratory of Animal Nutrition and Feeding, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, No. 2. Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Qiugang Ma
- State Key Laboratory of Animal Nutrition and Feeding, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, No. 2. Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Cheng Ji
- State Key Laboratory of Animal Nutrition and Feeding, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, No. 2. Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Lihong Zhao
- State Key Laboratory of Animal Nutrition and Feeding, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, No. 2. Yuanmingyuan West Road, Beijing, 100193, People's Republic of China.
| |
Collapse
|
34
|
Shi J, Jin Y, Lin S, Li X, Zhang D, Wu J, Qi Y, Li Y. Mitochondrial non-energetic function and embryonic cardiac development. Front Cell Dev Biol 2024; 12:1475603. [PMID: 39435335 PMCID: PMC11491369 DOI: 10.3389/fcell.2024.1475603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 09/20/2024] [Indexed: 10/23/2024] Open
Abstract
The initial contraction of the heart during the embryonic stage necessitates a substantial energy supply, predominantly derived from mitochondrial function. However, during embryonic heart development, mitochondria influence beyond energy supplementation. Increasing evidence suggests that mitochondrial permeability transition pore opening and closing, mitochondrial fusion and fission, mitophagy, reactive oxygen species production, apoptosis regulation, Ca2+ homeostasis, and cellular redox state also play critical roles in early cardiac development. Therefore, this review aims to describe the essential roles of mitochondrial non-energetic function embryonic cardiac development.
Collapse
Affiliation(s)
- Jingxian Shi
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuxi Jin
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Sha Lin
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xing Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, China
| | - Jinlin Wu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yan Qi
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, China
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
35
|
Ansari WA, Khan MA, Hasan SMM, Siddiqui Z, Ahmad S, Khan MS, Khan MF. Computational screening of chemical constituents derived from berry fruits as allosteric caspace-3/-7 inhibitors. 3 Biotech 2024; 14:234. [PMID: 39297056 PMCID: PMC11405617 DOI: 10.1007/s13205-024-04067-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 08/23/2024] [Indexed: 09/21/2024] Open
Abstract
With the aim of finding the plant-derived allosteric inhibitors of caspase-3/-7, we conducted computational investigations of bioactive compounds present in various berry fruits. In a molecular docking study, perulactone demonstrated excellent binding affinity scores of -12.1 kcal/mol and -9.1 kcal/mol for caspase 7 and 3, respectively, whereas FDA-approved allosteric inhibitors (DICA and FICA) were found to show lower docking scores (-5.6 and -6.1 kcal/mol) against caspase 7 while (-5.0 and -5.1 kcal/mol) for caspase 3, respectively. MD simulations were used to validate the binding stability of perulactone in the active sites of caspase-7/-3, and the results showed outstanding stability with lower ligand RMSDs of 1.270-3.088 Å and 2.426-9.850 Å against the targeted receptor. Furthermore, we performed MMGBSA free binding energy, where the perulactone values of ΔG Bind were determined to be -63.98 kcal/mol and -66.32 kcal/mol for both receptors (3IBF and 1NME), which are significantly better than the -45.16 kcal/mol and -39.51 kcal/mol for DICA as well as -26.37 kcal/mol and -15.50 kcal/mol for FICA, respectively. The drug resemblance of perulactone was effectively evaluated by ADMET. Thus, our findings indicated that perulactone could be an orally administered therapeutic candidate for regulating apoptosis in a variety of disorders. However, there may be an urgent need to study using in vitro and in vivo experiments. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-024-04067-7.
Collapse
Affiliation(s)
- Waseem Ahmad Ansari
- Department of Biotechnology, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Hardoi Road, Lucknow, 226003 India
| | - Mohsin Ali Khan
- Center for Disease Mapping and Therapeutic Research, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Hardoi Road, Lucknow, 226003 India
| | - S. M. Mahfooz Hasan
- Center for Disease Mapping and Therapeutic Research, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Hardoi Road, Lucknow, 226003 India
| | - Zainab Siddiqui
- Center for Disease Mapping and Therapeutic Research, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Hardoi Road, Lucknow, 226003 India
| | - Saheem Ahmad
- Department of Medical Laboratory Science, College of Applied Medical Sciences, University of Hail, 2440 Hail, Saudi Arabia
| | - Mohd Shahnawaz Khan
- Department of Biochemistry, College of Sciences, King Saud University, 12371 Riyadh, Saudi Arabia
| | - Mohammad Faheem Khan
- Department of Biotechnology, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Hardoi Road, Lucknow, 226003 India
- Department of Chemistry, Era University, Sarfarazganj, Hardoi Road, Lucknow, 226003 India
| |
Collapse
|
36
|
El Gaafary M, Abdel-Baki PM, El-Halawany AM, Mohamed HM, Duweb A, Abdallah HM, Mohamed GA, Ibrahim SRM, Simmet T, Syrovets T. Prenylated xanthones from mangosteen (Garcinia mangostana) target oxidative mitochondrial respiration in cancer cells. Biomed Pharmacother 2024; 179:117365. [PMID: 39217837 DOI: 10.1016/j.biopha.2024.117365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
Mangosteen (Garcinia mangostana) is well-known for its nutritional value and health benefits. Breast cancer is the most common cancer and the leading cause of cancer-related mortality among females worldwide. Here we show that the prenylated xanthones, α-mangostin, γ-mangostin, 9-hydroxycalabaxanthone (9-HCX), and garcinone E from the mangosteen pericarp exhibit cytotoxicity against a panel of human cancer cell lines including lung adenocarcinoma (A549), cervical carcinoma (HeLa), prostatic carcinoma (DU 145), pancreatic carcinoma (MIA PaCa-2), hepatocellular carcinoma (Hep G2), bladder urothelial cancer (5637), as well as the triple-negative breast cancer cells MDA-MB-231. In line with its higher predicted bioactivity score compared to other prenylated xanthones, 9-HCX induced the strongest antiproliferative and proapoptotic effects in MDA-MB-231 breast cancer xenografts in vivo. In different in vitro models, we demonstrate that prenylated xanthones from G. mangostana target mitochondria in cancer cells by inhibition of the mitochondrial respiratory chain complex II (α-mangostin, γ-mangostin, and garcinone E) and complex III (9-HCX) as shown in isolated mitochondria. Accordingly, oxidative mitochondrial respiration (OXPHOS) was inhibited, mitochondrial proton leak increased, and adenosine triphosphate (ATP) synthesis decreased as analyzed by Seahorse assay in MDA-MB-231 cells. Hence, the prenylated xanthones increased mitochondrial superoxide levels, induced mitochondrial membrane permeabilization, and initiated caspase 3/7-mediated apoptosis in MDA-MB-231 triple-negative breast cancer cells. Thus, prenylated xanthones from Garcinia mangostana exhibit anticancer activity based on interference with the mitochondrial respiration.
Collapse
Affiliation(s)
- Menna El Gaafary
- Institute of Experimental and Clinical Pharmacology, Toxicology, and Pharmacology of Natural Products, Ulm University, D-89081 Ulm, Germany; Department of Analytical Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt.
| | - Passent M Abdel-Baki
- Department of Analytical Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt.
| | - Ali M El-Halawany
- Department of Analytical Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt.
| | - Heba M Mohamed
- Department of Analytical Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt; Faculty of Health Sciences, Higher Colleges of Technology, Dubai, United Arab Emirates.
| | - Amira Duweb
- Institute of Experimental and Clinical Pharmacology, Toxicology, and Pharmacology of Natural Products, Ulm University, D-89081 Ulm, Germany; Department of Pharmacology, Faculty of Medicine, University of Tripoli, Tripoli, Libya.
| | - Hossam M Abdallah
- Institute of Experimental and Clinical Pharmacology, Toxicology, and Pharmacology of Natural Products, Ulm University, D-89081 Ulm, Germany; Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Gamal A Mohamed
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Sabrin R M Ibrahim
- Preparatory Year Program, Department of Chemistry, Batterjee Medical College, Jeddah 21442, Saudi Arabia; Department of Pharmacognosy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt.
| | - Thomas Simmet
- Institute of Experimental and Clinical Pharmacology, Toxicology, and Pharmacology of Natural Products, Ulm University, D-89081 Ulm, Germany.
| | - Tatiana Syrovets
- Institute of Experimental and Clinical Pharmacology, Toxicology, and Pharmacology of Natural Products, Ulm University, D-89081 Ulm, Germany.
| |
Collapse
|
37
|
Wu Z, Xiang H, Wang X, Zhang R, Guo Y, Qu L, Zhou J, Xiao Y. Integrating network pharmacology, molecular docking and experimental verification to explore the therapeutic effect and potential mechanism of nomilin against triple-negative breast cancer. Mol Med 2024; 30:166. [PMID: 39342122 PMCID: PMC11439318 DOI: 10.1186/s10020-024-00928-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/06/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Nomilin is a limonoid compound known for its multiple biological activities, but its role in triple negative breast cancer (TNBC) remains unclear. This study aims to uncover the potential therapeutic effect of nomilin on TNBC and elucidate the specific mechanism of its action. METHODS We employed weighted gene co-expression network analysis (WGCNA), differential expression analysis, and the GeneCards database to identify potential targets for TNBC. Simultaneously, we utilized the Swiss Target Prediction, ChEMBL, and STITCH databases to identify potential targets of nomilin. The core targets and mechanisms of nomilin against TNBC were predicted through protein-protein interaction (PPI) network analysis, molecular docking, and enrichment analysis. The results of the network pharmacology were corroborated by conducting experiments. RESULTS A total of 17,204 TNBC targets were screened, and 301 potential targets of nomilin were identified. Through the PPI network, eight core targets of nomilin against TNBC were pinpointed, namely BCL2, Caspase3, CyclinD1, EGFR, HSP90AA1, KRAS, PARP1, and TNF. Molecular docking, molecular dynamics simulation and proteome microarray revealed that nomilin exhibits strong binding activity to these core proteins. Enrichment analysis results indicated that the anti-TNBC effect of nomilin is associated with PI3K/Akt pathway. In vitro and in vivo experiments have demonstrated that nomilin inhibits TNBC cell proliferation and migration while promoting cell apoptosis through the PI3K/Akt pathway. CONCLUSION For the first time, the research effectively discovered the objectives and mechanisms of nomilin in combating TNBC using network pharmacology, molecular docking, molecular dynamics simulation, proteome microarray and experimental confirmation, presenting a hopeful approach for treating TNBC.
Collapse
Affiliation(s)
- Zhixuan Wu
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, 325035, China
| | - Haoyi Xiang
- Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310016, China
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital of Zhejiang University, Hangzhou, Zhejiang Province, 310016, China
| | - Xiaowu Wang
- Department of Burns and Skin Repair Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Ruian, 325200, China
| | - Rongrong Zhang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, 325035, China
| | - Yangyang Guo
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, 325035, China
| | - Liangchen Qu
- Emergency Department, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, 318000, China.
| | - Jingyao Zhou
- Pharmacy Department, Taizhou Central Hospital, Taizhou, Zhejiang Province, 318000, China.
| | - Yanyi Xiao
- The Dingli Clinical College of Wenzhou Medical University, Wenzhou, Zhejiang Province, 325000, China.
- Department of Thyroid and Breast Surgery, Wenzhou Central Hospital, The Second Affiliated Hospital of Shanghai University, Wenzhou, Zhejiang Province, 325000, China.
| |
Collapse
|
38
|
Ye YX, Pan JC, Wang HC, Zhang XT, Zhu HL, Liu XH. Advances in small-molecule fluorescent probes for the study of apoptosis. Chem Soc Rev 2024; 53:9133-9189. [PMID: 39129564 DOI: 10.1039/d4cs00502c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Apoptosis, as type I cell death, is an active death process strictly controlled by multiple genes, and plays a significant role in regulating various activities. Mounting research indicates that the unique modality of cell apoptosis is directly or indirectly related to different diseases including cancer, autoimmune diseases, viral diseases, neurodegenerative diseases, etc. However, the underlying mechanisms of cell apoptosis are complicated and not fully clarified yet, possibly due to the lack of effective chemical tools for the nondestructive and real-time visualization of apoptosis in complex biological systems. In the past 15 years, various small-molecule fluorescent probes (SMFPs) for imaging apoptosis in vitro and in vivo have attracted broad interest in related disease diagnostics and therapeutics. In this review, we aim to highlight the recent developments of SMFPs based on enzyme activity, plasma membranes, reactive oxygen species, reactive sulfur species, microenvironments and others during cell apoptosis. In particular, we generalize the mechanisms commonly used to design SMFPs for studying apoptosis. In addition, we discuss the limitations of reported probes, and emphasize the potential challenges and prospects in the future. We believe that this review will provide a comprehensive summary and challenging direction for the development of SMFPs in apoptosis related fields.
Collapse
Affiliation(s)
- Ya-Xi Ye
- Institute of Pharmaceutical Biotechnology, School of Biology and Food Engineering, Suzhou University, Suzhou 234000, P. R. China.
| | - Jian-Cheng Pan
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210023, P. R. China.
| | - Hai-Chao Wang
- Institute of Pharmaceutical Biotechnology, School of Biology and Food Engineering, Suzhou University, Suzhou 234000, P. R. China.
| | - Xing-Tao Zhang
- Institute of Pharmaceutical Biotechnology, School of Biology and Food Engineering, Suzhou University, Suzhou 234000, P. R. China.
| | - Hai-Liang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210023, P. R. China.
| | - Xin-Hua Liu
- Institute of Pharmaceutical Biotechnology, School of Biology and Food Engineering, Suzhou University, Suzhou 234000, P. R. China.
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei 230032, P. R. China
| |
Collapse
|
39
|
Zhang Y, Bejaoui M, Linh TN, Arimura T, Isoda H. A novel amphiphilic squalene-based compound with open-chain polyethers reduces malignant melanoma metastasis in-vitro and in-vivo. Cell Commun Signal 2024; 22:437. [PMID: 39261954 PMCID: PMC11389383 DOI: 10.1186/s12964-024-01813-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 08/31/2024] [Indexed: 09/13/2024] Open
Abstract
Squalene (SQ) is a well-known antioxidant and anti-inflammatory agent that provides promising anti-aging and UV-protective roles on human skin. However, its strong hydrophobic nature, accompanied by issues such as poor solubility and limited tissue permeation, has created challenges for scientists to investigate its untapped potential in more complex conditions, including cancer progression. The present study assessed the potent anti-metastatic properties of a newly synthesized amphiphilic ethylene glycol SQ derivative (SQ-diEG) in melanoma, the most fatal skin cancer. In vitro and in vivo experiments have discovered that SQ-diEG may exert its potential on melanoma malignancy through the mitochondria-mediated caspase activation apoptotic signaling pathway. The potent anti-metastatic effect of SQ-diEG was observed in vitro using highly proliferative and aggressive melanoma cells. Administration of SQ-diEG (25 mg/kg) significantly decreased the tumor burden on the lung and inhibited the metastasis-associated proteins and gene markers in B16F10 lung colonization mice model. Furthermore, global gene profiling also revealed a promising role of SQ-diEG in tumor microenvironment. We anticipated that the amphiphilic nature of the SQ compound bearing ethylene glycol oligomers could potentially augment its ability to reach the pathology site, thus enhancing its therapeutic potential in melanoma.
Collapse
Affiliation(s)
- Yaman Zhang
- Tsukuba Life Science Innovation Program (T-LSI), University of Tsukuba, Tsukuba, Japan
| | - Meriem Bejaoui
- Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
- Alliance for Research On the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Japan
| | - Tran Ngoc Linh
- Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Takashi Arimura
- Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Hiroko Isoda
- Tsukuba Life Science Innovation Program (T-LSI), University of Tsukuba, Tsukuba, Japan.
- Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan.
- Alliance for Research On the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Japan.
- Institution of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan.
| |
Collapse
|
40
|
Sarkar R, Choudhury SM, Kanneganti TD. Classical apoptotic stimulus, staurosporine, induces lytic inflammatory cell death, PANoptosis. J Biol Chem 2024; 300:107676. [PMID: 39151726 PMCID: PMC11418131 DOI: 10.1016/j.jbc.2024.107676] [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: 03/09/2024] [Revised: 07/27/2024] [Accepted: 08/03/2024] [Indexed: 08/19/2024] Open
Abstract
Innate immunity is the body's first line of defense against disease, and regulated cell death is a central component of this response that balances pathogen clearance and inflammation. Cell death pathways are generally categorized as non-lytic and lytic. While non-lytic apoptosis has been extensively studied in health and disease, lytic cell death pathways are also increasingly implicated in infectious and inflammatory diseases and cancers. Staurosporine (STS) is a well-known inducer of non-lytic apoptosis. However, in this study, we observed that STS also induces lytic cell death at later timepoints. Using biochemical assessments with genetic knockouts, pharmacological inhibitors, and gene silencing, we identified that STS triggered PANoptosis via the caspase-8/RIPK3 axis, which was mediated by RIPK1. PANoptosis is a lytic, innate immune cell death pathway initiated by innate immune sensors and driven by caspases and RIPKs through PANoptosome complexes. Deletion of caspase-8 and RIPK3, core components of the PANoptosome complex, protected against STS-induced lytic cell death. Overall, our study identifies STS as a time-dependent inducer of lytic cell death, PANoptosis. These findings emphasize the importance of understanding trigger- and time-specific activation of distinct cell death pathways to advance our understanding of the molecular mechanisms of innate immunity and cell death for clinical translation.
Collapse
Affiliation(s)
- Roman Sarkar
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Sk Mohiuddin Choudhury
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | |
Collapse
|
41
|
Theofilas P, Wang C, Butler D, Morales DO, Petersen C, Ambrose A, Chin B, Yang T, Khan S, Ng R, Kayed R, Karch CM, Miller BL, Gestwicki JE, Gan L, Temple S, Arkin MR, Grinberg LT. iPSC-induced neurons with the V337M MAPT mutation are selectively vulnerable to caspase-mediated cleavage of tau and apoptotic cell death. Mol Cell Neurosci 2024; 130:103954. [PMID: 39032719 PMCID: PMC11866097 DOI: 10.1016/j.mcn.2024.103954] [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: 02/19/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND Tau post-translational modifications (PTMs) result in the gradual build-up of abnormal tau and neuronal degeneration in tauopathies, encompassing variants of frontotemporal lobar degeneration (FTLD) and Alzheimer's disease (AD). Tau proteolytically cleaved by active caspases, including caspase-6, may be neurotoxic and prone to self-aggregation. Also, our recent findings show that caspase-6 truncated tau represents a frequent and understudied aspect of tau pathology in AD in addition to phospho-tau pathology. In AD and Pick's disease, a large percentage of caspase-6 associated cleaved-tau positive neurons lack phospho-tau, suggesting that many vulnerable neurons to tau pathology go undetected when using conventional phospho-tau antibodies and possibly will not respond to phospho-tau based therapies. Therefore, therapeutic strategies against caspase cleaved-tau pathology could be necessary to modulate the extent of tau abnormalities in AD and other tauopathies. METHODS To understand the timing and progression of caspase activation, tau cleavage, and neuronal death, we created two mAbs targeting caspase-6 tau cleavage sites and probed postmortem brain tissue from an individual with FTLD due to the V337M MAPT mutation. We then assessed tau cleavage and apoptotic stress response in cortical neurons derived from induced pluripotent stem cells (iPSCs) carrying the FTD-related V337M MAPT mutation. Finally, we evaluated the neuroprotective effects of caspase inhibitors in these iPSC-derived neurons. RESULTS FTLD V337M MAPT postmortem brain showed positivity for both cleaved tau mAbs and active caspase-6. Relative to isogenic wild-type MAPT controls, V337M MAPT neurons cultured for 3 months post-differentiation showed a time-dependent increase in pathogenic tau in the form of caspase-cleaved tau, phospho-tau, and higher levels of tau oligomers. Accumulation of toxic tau species in V337M MAPT neurons was correlated with increased vulnerability to pro-apoptotic stress. Notably, this mutation-associated cell death was pharmacologically rescued by the inhibition of effector caspases. CONCLUSIONS Our results suggest an upstream, time-dependent accumulation of caspase-6 cleaved tau in V337M MAPT neurons promoting neurotoxicity. These processes can be reversed by caspase inhibition. These results underscore the potential of developing caspase-6 inhibitors as therapeutic agents for FTLD and other tauopathies. Additionally, they highlight the promise of using caspase-cleaved tau as biomarkers for these conditions.
Collapse
Affiliation(s)
- Panos Theofilas
- Memory and Aging Center, Department of Neurology, UCSF, San Francisco, CA, USA
| | - Chao Wang
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | | | - Dulce O Morales
- Memory and Aging Center, Department of Neurology, UCSF, San Francisco, CA, USA
| | - Cathrine Petersen
- Memory and Aging Center, Department of Neurology, UCSF, San Francisco, CA, USA
| | - Andrew Ambrose
- Department of Pharmaceutical Chemistry and Small Molecule Discovery Center, UCSF, San Francisco, CA, USA
| | | | | | - Shireen Khan
- ChemPartner San Francisco, South San Francisco, CA, USA
| | - Raymond Ng
- ChemPartner San Francisco, South San Francisco, CA, USA
| | - Rakez Kayed
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | | | - Bruce L Miller
- Memory and Aging Center, Department of Neurology, UCSF, San Francisco, CA, USA
| | - Jason E Gestwicki
- Department of Pharmaceutical Chemistry, UCSF, San Francisco, CA, USA
| | - Li Gan
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA; Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | | | - Michelle R Arkin
- Department of Pharmaceutical Chemistry and Small Molecule Discovery Center, UCSF, San Francisco, CA, USA.
| | - Lea T Grinberg
- Memory and Aging Center, Department of Neurology, UCSF, San Francisco, CA, USA; Department of Pathology, University of Sao Paulo Medical School, Brazil.
| |
Collapse
|
42
|
Chen Y, Jiang B, Qu C, Jiang C, Zhang C, Wang Y, Chen F, Sun X, Su L, Luo Y. Bioactive components in prunella vulgaris for treating Hashimoto's disease via regulation of innate immune response in human thyrocytes. Heliyon 2024; 10:e36103. [PMID: 39253271 PMCID: PMC11382315 DOI: 10.1016/j.heliyon.2024.e36103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/05/2024] [Accepted: 08/09/2024] [Indexed: 09/11/2024] Open
Abstract
Background Hashimoto's thyroiditis (HT) is a thyroid autoimmune disease characterized by lymphocytic infiltration and thyroid destruction. Prunella vulgaris (PV) is a traditional Chinese herbal medicine with documented clinical efficacy in treating HT. We previously reported an immunoregulatory effect of PV in thyrocytes; however, the bioactive components of PV remained unclear. This study aimed to elucidate key components of PV for treating HT and their acting mechanisms. Methods Network pharmacology was used to predict key PV components for HT. The predicted components were tested to determine whether they could exert an immunoregulatory effect of PV in human thyrocytes. Limited proteolysis-mass spectrometry (Lip-MS) was used to explore interacting proteins with PV components in human thyrocytes. Microscale thermophoresis binding assay was used to evaluate the affinity of PV components with the target protein. Results Eleven PV components with 192 component targets and 3415 HT-related genes were gathered from public databases. With network pharmacology, a 'component-target-disease' network was established wherein four flavonoids including quercetin, luteolin, kaempferol, morin, and a phytosterol, β-sitosterol were predicted as key components in PV for HT. In stimulated primary human thyrocytes or Nthy-ori-31 cells, key components inhibited gene expressions of inflammatory cytokines including tumor necrosis factor α (TNF-α), interleukin-6 (IL-6), and interferon-β (IFN-β), cellular apoptosis, and activation of nuclear factor κB (NF-κB) and interferon regulatory factor 3 (IRF-3). Heat shock protein 90 alpha, class A, member 1 (HSP90AA1), was identified to interact with flavonoids in PV by Lip-MS. Morin had the highest affinity with HSP90AA1 (KD = 122.74 μM), followed by kaempferol (KD = 168.53 μM), luteolin (KD = 293.94 μM), and quercetin (KD = 356.86 μM). Conclusion Quercetin, luteolin, kaempferol, morin, and β-sitosterol reproduced an anti-inflammatory and anti-apoptosis effect of PV in stimulated human thyrocytes, which potentially contributed to the treatment efficacy of PV in HT.
Collapse
Affiliation(s)
- Yongzhao Chen
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University of Chinese Medicine, Zhongshan Road 321, Nanjing, 210008, China
| | - Bo Jiang
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Zhongshan Road 321, Nanjing, 210008, China
| | - Cheng Qu
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Zhongshan Road 321, Nanjing, 210008, China
| | - Chaoyu Jiang
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Zhongshan Road 321, Nanjing, 210008, China
| | - Chen Zhang
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Zhongshan Road 321, Nanjing, 210008, China
| | - Yanxue Wang
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Zhongshan Road 321, Nanjing, 210008, China
| | - Fei Chen
- General Surgery Center, Department of Thyroid Surgery, Zhujiang Hospital, Southern Medical University 253 Gongye Middle Avenue, Haizhu District, Guangzhou, 510280, China
| | - Xitai Sun
- Division of Pancreas and Metabolism Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China, Zhongshan Road 321, Nanjing, 210008, China
| | - Lei Su
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Zhongshan Road 321, Nanjing, 210008, China
| | - Yuqian Luo
- Clinical Medicine Research Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China, Zhongshan Road 321, Nanjing, 210008, China
| |
Collapse
|
43
|
Pospíšilová J, Heger T, Kurka O, Kvasnicová M, Chládková A, Nemec I, Rárová L, Cankař P. Atropisomeric 1-phenylbenzimidazoles affecting microtubule organization: influence of axial chirality. Org Biomol Chem 2024; 22:6966-6980. [PMID: 38988246 DOI: 10.1039/d4ob00863d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
Benzimidazoles are frequently used in medicinal chemistry. Their anticancer effect is among the most prominent biological activities exhibited by this scaffold. Although numerous benzimidazole derivatives have been synthesized, possible atropisomerism of ortho-substituted 1-phenylbenzimidazoles has been largely overlooked. The aim of this research was to synthesize a small library of novel atropisomeric benzimidazole derivatives and explore their biological activity in various cancer and normal human cell lines. The new unique structural motif provides an interesting 3D architecture with axial chirality, which further contributes to molecular complexity and specificity. Racemates and their separated atropisomers arrested the cell cycle, caused apoptosis, and affected microtubule organization in cancer cells in vitro at different intensities. Moreover, this phenomenon was also verified by the inhibition of endothelial cell migration. These results showed that (+)-atropisomers, especially 5n, exhibit a stronger effect and show promise as agents for cancer therapy.
Collapse
Affiliation(s)
- Jana Pospíšilová
- Department of Organic Chemistry, Faculty of Science, Palacký University Olomouc, 17. listopadu 1192/12, 77900 Olomouc, Czech Republic.
| | - Tomáš Heger
- Department of Experimental Biology, Faculty of Science, Palacký University Olomouc, Slechtitelu 27, 77900 Olomouc, Czech Republic.
| | - Ondřej Kurka
- Laboratory of Growth Regulators, Institute of Experimental Botany of the Czech Academy of Sciences and Faculty of Science, Palacký University, Slechtitelu 27, Olomouc CZ-77900, Czech Republic
| | - Marie Kvasnicová
- Department of Experimental Biology, Faculty of Science, Palacký University Olomouc, Slechtitelu 27, 77900 Olomouc, Czech Republic.
- Laboratory of Growth Regulators, Institute of Experimental Botany of the Czech Academy of Science, Palacký University, Slechtitelu 27, 77900 Olomouc, Czech Republic
| | - Anna Chládková
- Department of Organic Chemistry, Faculty of Science, Palacký University Olomouc, 17. listopadu 1192/12, 77900 Olomouc, Czech Republic.
| | - Ivan Nemec
- Department of Inorganic Chemistry, Faculty of Science, Palacký University Olomouc, 17. listopadu 1192/12, 77900 Olomouc, Czech Republic
| | - Lucie Rárová
- Department of Experimental Biology, Faculty of Science, Palacký University Olomouc, Slechtitelu 27, 77900 Olomouc, Czech Republic.
- Laboratory of Growth Regulators, Institute of Experimental Botany of the Czech Academy of Science, Palacký University, Slechtitelu 27, 77900 Olomouc, Czech Republic
| | - Petr Cankař
- Department of Organic Chemistry, Faculty of Science, Palacký University Olomouc, 17. listopadu 1192/12, 77900 Olomouc, Czech Republic.
| |
Collapse
|
44
|
Liu X, Xu Y, Wang Y, Peng X, Jian J, Wang X, Wang T. RETRACTED ARTICLE: Administration of methylprednisolone do not affect the spinal scar component of spinal cord injury. J Spinal Cord Med 2024:1-10. [PMID: 39167368 DOI: 10.1080/10790268.2024.2352929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/23/2024] Open
Abstract
We, the Editors and Publisher of the journal The Journal of Spinal Cord Medicine, have retracted the following article:Liu, X., Xu, Y., Wang, Y., Peng, X., Jian, J., Wang, X., & Wang, T. (2024). Administration of methylprednisolone do not affect the spinal scar component of spinal cord injury. The Journal of Spinal Cord Medicine, 1-10. https://doi.org/10.1080/10790268.2024.2352929Following publication, in November 2024, the authors notified the journal that they had identified areas in the article which required improvement, and requested retraction.Upon investigation by the journal, significant concerns were identified regarding the animal model and the integrity of Figure(s) 2 and 4.When approached for an explanation following their request to retract the article, the authors did not respond.As verifying the validity of published work is core to the integrity of the scholarly record, we are therefore retracting the article. The corresponding author listed in this publication has been informed. The author(s) agree with the retraction.We have been informed in our decision-making by our editorial policies and the COPE guidelines.The retracted article will remain online to maintain the scholarly record, but it will be digitally watermarked on each page as 'Retracted'.
Collapse
Affiliation(s)
- Xin Liu
- Institute of Base Medicine, Jinzhou Medical University, Jinzhou, People's Republic of China
| | - Yang Xu
- Institute of Neurological Disease, West China Hospital, Sichuan University & The Research Units of West China, Chinese Academy of Medical Sciences, Chengdu, People's Republic of China
| | - Yangyang Wang
- Institute of Neurological Disease, West China Hospital, Sichuan University & The Research Units of West China, Chinese Academy of Medical Sciences, Chengdu, People's Republic of China
| | - Xia Peng
- Institute of Neuroscience, Laboratory Zoology Department, Kunming Medical University, Kunming, People's Republic of China
| | - Jiao Jian
- Institute of Neuroscience, Laboratory Zoology Department, Kunming Medical University, Kunming, People's Republic of China
| | - Xuefang Wang
- Institute of Neurological Disease, West China Hospital, Sichuan University & The Research Units of West China, Chinese Academy of Medical Sciences, Chengdu, People's Republic of China
| | - Tinghua Wang
- Institute of Base Medicine, Jinzhou Medical University, Jinzhou, People's Republic of China
- Institute of Neurological Disease, West China Hospital, Sichuan University & The Research Units of West China, Chinese Academy of Medical Sciences, Chengdu, People's Republic of China
- Institute of Neuroscience, Laboratory Zoology Department, Kunming Medical University, Kunming, People's Republic of China
| |
Collapse
|
45
|
Lê HG, Hwang BS, Choi JS, Jeong YT, Kang JM, Võ TC, Oh YT, Na BK. Iris setosa Pall. ex Link Extract Reveals Amoebicidal Activity against Acanthamoeba castellanii and Acanthamoeba polyphaga with Low Toxicity to Human Corneal Cells. Microorganisms 2024; 12:1658. [PMID: 39203500 PMCID: PMC11356916 DOI: 10.3390/microorganisms12081658] [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: 07/25/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 09/03/2024] Open
Abstract
Acanthamoeba keratitis (AK) is a sight-threatening and difficult-to-treat ocular infection. The significant side effects of current AK treatments highlight the urgent need to develop a safe and effective AK medication. In this study, the amoebicidal activity of Iris setosa Pall. ex Link extract (ISE) against Acanthamoeba was examined and its specific amoebicidal mechanism was explored. ISE induced significant morphological changes in Acanthamoeba trophozoites and exhibited amoebicidal activity against A. castellanii and A. polyphaga. ISE was further fractionated into five subfractions by sequential extraction with n-hexane, chloroform, ethyl acetate, n-butanol, and water, and their amoebicidal activities and underlying amoebicidal mechanisms were investigated. The n-butanol subfraction of ISE (ISE-BuOH) displayed selective amoebicidal activity against the Acanthamoeba species with minimal cytotoxicity in human corneal cells (HCE-2). ISE-BuOH triggered apoptosis-like programmed cell death (PCD) in amoebae, characterized by DNA fragmentation, increased ROS production, and caspase-3 activity elevation. ISE-BuOH also demonstrated a partial cysticidal effect against the amoeba species. ISE-BuOH could be a promising candidate in the development of therapeutic drugs for AK.
Collapse
Affiliation(s)
- Hương Giang Lê
- Department of Parasitology and Tropical Medicine, and Institute of Health Science, Gyeongsang National University College of Medicine, Jinju 52727, Republic of Korea; (H.G.L.); (J.-M.K.); (T.C.V.)
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Buyng Su Hwang
- Nakdonggang National Institute of Biological Resources, Sangju 37242, Republic of Korea; (B.S.H.); (J.-S.C.); (Y.T.J.)
| | - Ji-Su Choi
- Nakdonggang National Institute of Biological Resources, Sangju 37242, Republic of Korea; (B.S.H.); (J.-S.C.); (Y.T.J.)
| | - Yong Tae Jeong
- Nakdonggang National Institute of Biological Resources, Sangju 37242, Republic of Korea; (B.S.H.); (J.-S.C.); (Y.T.J.)
| | - Jung-Mi Kang
- Department of Parasitology and Tropical Medicine, and Institute of Health Science, Gyeongsang National University College of Medicine, Jinju 52727, Republic of Korea; (H.G.L.); (J.-M.K.); (T.C.V.)
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Tuấn Cường Võ
- Department of Parasitology and Tropical Medicine, and Institute of Health Science, Gyeongsang National University College of Medicine, Jinju 52727, Republic of Korea; (H.G.L.); (J.-M.K.); (T.C.V.)
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Young Taek Oh
- Nakdonggang National Institute of Biological Resources, Sangju 37242, Republic of Korea; (B.S.H.); (J.-S.C.); (Y.T.J.)
| | - Byoung-Kuk Na
- Department of Parasitology and Tropical Medicine, and Institute of Health Science, Gyeongsang National University College of Medicine, Jinju 52727, Republic of Korea; (H.G.L.); (J.-M.K.); (T.C.V.)
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Republic of Korea
| |
Collapse
|
46
|
Svandova E, Vesela B, Janeckova E, Chai Y, Matalova E. Exploring caspase functions in mouse models. Apoptosis 2024; 29:938-966. [PMID: 38824481 PMCID: PMC11263464 DOI: 10.1007/s10495-024-01976-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2024] [Indexed: 06/03/2024]
Abstract
Caspases are enzymes with protease activity. Despite being known for more than three decades, caspase investigation still yields surprising and fascinating information. Initially associated with cell death and inflammation, their functions have gradually been revealed to extend beyond, targeting pathways such as cell proliferation, migration, and differentiation. These processes are also associated with disease mechanisms, positioning caspases as potential targets for numerous pathologies including inflammatory, neurological, metabolic, or oncological conditions. While in vitro studies play a crucial role in elucidating molecular pathways, they lack the context of the body's complexity. Therefore, laboratory animals are an indispensable part of successfully understanding and applying caspase networks. This paper aims to summarize and discuss recent knowledge, understanding, and challenges in caspase knock-out mice.
Collapse
Affiliation(s)
- Eva Svandova
- Laboratory of Odontogenesis and Osteogenesis, Institute of Animal Physiology and Genetic, Brno, Czech Republic.
| | - Barbora Vesela
- Laboratory of Odontogenesis and Osteogenesis, Institute of Animal Physiology and Genetic, Brno, Czech Republic
| | - Eva Janeckova
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, USA
| | - Yang Chai
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, USA
| | - Eva Matalova
- Laboratory of Odontogenesis and Osteogenesis, Institute of Animal Physiology and Genetic, Brno, Czech Republic
- Department of Physiology, University of Veterinary Sciences, Brno, Czech Republic
| |
Collapse
|
47
|
Zhang X, Lei Y, Zhou H, Liu H, Xu P. The Role of PKM2 in Multiple Signaling Pathways Related to Neurological Diseases. Mol Neurobiol 2024; 61:5002-5026. [PMID: 38157121 DOI: 10.1007/s12035-023-03901-y] [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: 09/09/2023] [Accepted: 12/18/2023] [Indexed: 01/03/2024]
Abstract
Pyruvate kinase M2 (PKM2) is a key rate-limiting enzyme in glycolysis. It is well known that PKM2 plays a vital role in the proliferation of tumor cells. However, PKM2 can also exert its biological functions by mediating multiple signaling pathways in neurological diseases, such as Alzheimer's disease (AD), cognitive dysfunction, ischemic stroke, post-stroke depression, cerebral small-vessel disease, hypoxic-ischemic encephalopathy, traumatic brain injury, spinal cord injury, Parkinson's disease (PD), epilepsy, neuropathic pain, and autoimmune diseases. In these diseases, PKM2 can exert various biological functions, including regulation of glycolysis, inflammatory responses, apoptosis, proliferation of cells, oxidative stress, mitochondrial dysfunction, or pathological autoimmune responses. Moreover, the complexity of PKM2's biological characteristics determines the diversity of its biological functions. However, the role of PKM2 is not entirely the same in different diseases or cells, which is related to its oligomerization, subcellular localization, and post-translational modifications. This article will focus on the biological characteristics of PKM2, the regulation of PKM2 expression, and the biological role of PKM2 in neurological diseases. With this review, we hope to have a better understanding of the molecular mechanisms of PKM2, which may help researchers develop therapeutic strategies in clinic.
Collapse
Affiliation(s)
- Xiaoping Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yihui Lei
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Hongyan Zhou
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Haijun Liu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Ping Xu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
48
|
Chen B, Zhang Y, Song G, Wei X. RNF135 Promotes Human Osteosarcoma Cell Growth and Inhibits Apoptosis by Upregulating the PI3K/AKT Pathway. Cancer Rep (Hoboken) 2024; 7:e2159. [PMID: 39118262 PMCID: PMC11310095 DOI: 10.1002/cnr2.2159] [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: 02/10/2024] [Revised: 06/24/2024] [Accepted: 07/16/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Ring finger protein 135 (RNF135) is an E3 ubiquitin ligase that has been implicated in the tumorigenesis of multiple human malignancies. However, whether RNF135 plays a role in the development of human osteosarcoma (OS) remains unknown. METHODS RNF135 expression in 20 human OS and 20 human osteochondroma specimens were evaluated by means of immunohistochemistry staining. The effects of shRNA-mediated RNF135 knockdown on human OS cell growth and apoptosis were evaluated through a panel of in vitro studies on cell proliferation, colony formation, exposure of phosphatidylserine on the cell surface, and caspase 3/7 activation. The protein levels of PI3K, AKT, and p-AKT were determined by western blot analysis. RESULTS We detected significantly higher RNF135 levels in human OS tissues than human osteochondroma tissues. In in vitro studies, shRNA-mediated RNF135 knockdown in human OS cells inhibited proliferation and induced apoptosis. In addition, RNF135 knockdown reduced PI3K and p-AKT protein levels and activated caspase 3 and 7. CONCLUSIONS These results supported that RNF135 contributes to human OS development through PI3K/AKT-dependent mechanisms. Targeting RNF135 may provide a new therapeutic approach for treating this human malignancy.
Collapse
Affiliation(s)
- Bingyao Chen
- Second Department of OrthopedicsBeijing Daxing District People's HospitalBeijingChina
- Department of OrthopedicsAerospace Central HospitalBeijingChina
- Senior Department of OrthopedicsThe Fourth Medical Center of PLA General HospitalBeijingChina
| | - Yinglong Zhang
- Senior Department of OrthopedicsThe Fourth Medical Center of PLA General HospitalBeijingChina
| | - Guangze Song
- Department of OrthopedicsAerospace Central HospitalBeijingChina
- Senior Department of OrthopedicsThe Fourth Medical Center of PLA General HospitalBeijingChina
| | - Xing Wei
- Department of OrthopedicsAerospace Central HospitalBeijingChina
- Senior Department of OrthopedicsThe Fourth Medical Center of PLA General HospitalBeijingChina
| |
Collapse
|
49
|
Jung YS, Cho CH, Yoo G, Kim M, Kurniawati UD, Ha SK, Lee SH. Phlorofucofuroeckol-A from Ecklonia Cava attenuates methylglyoxal-induced nephrotoxicity by regulating RAGE and inhibiting the formation of advanced glycosylated end products. FOOD BIOSCI 2024; 60:104413. [DOI: 10.1016/j.fbio.2024.104413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
50
|
Morris JL, Letson HL, Dobson GP. Safety evaluation of adenosine, lidocaine and magnesium (ALM) intranasal therapy toward human nasal epithelial cells in vitro. Basic Clin Pharmacol Toxicol 2024; 135:98-108. [PMID: 38784974 DOI: 10.1111/bcpt.14036] [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: 01/31/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024]
Abstract
Adenosine, lidocaine and Mg2+ (ALM) solution is an emerging therapy that reduces secondary injury after intravenous administration in experimental models of traumatic brain injury (TBI). Intranasal delivery of ALM may offer an alternative route for rapid, point-of-care management of TBI. As a preliminary safety screen, we evaluated whether ALM exerts cytotoxic or inflammatory effects on primary human nasal epithelial cells (pHNEC) in vitro. Submerged monolayers and air-liquid interface cultures of pHNEC were exposed to media only, normal saline only, therapeutic ALM or supratherapeutic ALM for 15 or 60 min. Safety was measured through viability, cytotoxicity, apoptosis, cellular and mitochondrial stress, and inflammatory mediator secretion assays. No differences were found in viability or cytotoxicity in cultures exposed to saline or ALM for up to 60 min, with no evidence of apoptosis after exposure to supratherapeutic ALM concentrations. Despite comparable inflammatory cytokine secretion profiles and mitochondrial activity, cellular stress responses were significantly lower in cultures exposed to ALM than saline. In summary, data show ALM therapy has neither adverse toxic nor inflammatory effects on human nasal epithelial cells, setting the stage for in vivo toxicity studies and possible clinical translation of intranasal ALM therapy for TBI treatment.
Collapse
Affiliation(s)
- Jodie L Morris
- Heart and Trauma Research Laboratory, College of Medicine and Dentistry, James Cook University, Townsville, Australia
| | - Hayley L Letson
- Heart and Trauma Research Laboratory, College of Medicine and Dentistry, James Cook University, Townsville, Australia
| | - Geoffrey P Dobson
- Heart and Trauma Research Laboratory, College of Medicine and Dentistry, James Cook University, Townsville, Australia
| |
Collapse
|