1
|
Zhang Z, Wang N, Lu J, Qu Y, Song Y, Yang X, Wei Z, Zhang Q, Herdewijn P, Chang J, Wang XN, Wang Z. Synthesis and pharmacodynamic evaluation of 2-aminoindole derivatives against influenza A virus in vitro/vivo. Eur J Med Chem 2025; 281:117044. [PMID: 39547081 DOI: 10.1016/j.ejmech.2024.117044] [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: 10/30/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
Influenza virus is a kind of respiratory pathogen with high morbidity and mortality, which still threatens human health. Existing anti-influenza drugs have various limitations, such as the inability to alleviate body injury and side effects. There remains an urgent need to develop a novel antiviral drug to efficiently inhibit viral infection while avoiding body injury. A series of 2-aminoindole derivatives were synthesized via the TMSOTf-catalyzed reactions of N-arylynamides with sulfilimines and evaluated for their anti-influenza virus activity. The experimental results showed that 2-aminoindole 3h had significant antiviral activity (EC50 = 8.37 ± 0.65 μM) and the lowest cytotoxicity (CC50 = 669.26 ± 11.42 μM) in vitro. 2-Aminoindole 3h could inhibit viral replication by effectively binding to RNA-dependent RNA polymerase (RdRp), and could also directly target host cells to inhibit cytokine storms and apoptosis induced by viral infection, thereby improving host cell survival rate. In addition, viral load and organ injury in the lung tissue of infected mice were effectively reduced by 2-aminoindole 3h with satisfactory biosafety. These findings highlight the potential of a valuable therapeutic option against influenza infection while also laying the foundation for further research and development in this area.
Collapse
Affiliation(s)
- Zhongmou Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of "Runliang" Antiviral Medicines Research and Development, Institute of Drug Discovery & Development, Zhengzhou University, Zhengzhou, 450001, China
| | - Nanfang Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Pingyuan Laboratory, State Key Laboratory of Antiviral Drugs, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China
| | - Jiejie Lu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of "Runliang" Antiviral Medicines Research and Development, Institute of Drug Discovery & Development, Zhengzhou University, Zhengzhou, 450001, China
| | - Ying Qu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of "Runliang" Antiviral Medicines Research and Development, Institute of Drug Discovery & Development, Zhengzhou University, Zhengzhou, 450001, China
| | - Yihui Song
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xinyu Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Zhanyong Wei
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake 15#, Zhengzhou 450046, China
| | - Qi Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Piet Herdewijn
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; XNA Platform, Institute of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Junbiao Chang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of "Runliang" Antiviral Medicines Research and Development, Institute of Drug Discovery & Development, Zhengzhou University, Zhengzhou, 450001, China.
| | - Xiao-Na Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Pingyuan Laboratory, State Key Laboratory of Antiviral Drugs, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China.
| | - Zhenya Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of "Runliang" Antiviral Medicines Research and Development, Institute of Drug Discovery & Development, Zhengzhou University, Zhengzhou, 450001, China; International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake 15#, Zhengzhou 450046, China; XNA Platform, Institute of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
2
|
Buzdar JA, Shah QA, Khan MZ, Zaheer A, Shah T, Ataya FS, Fouad D. Hepatoprotective effects of olive leaf extract against carbon tetrachloride-induced oxidative stress: in vivo and in-silico insights into the Nrf2-NFκB pathway. J Mol Histol 2024; 56:42. [PMID: 39673562 DOI: 10.1007/s10735-024-10325-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 11/30/2024] [Indexed: 12/16/2024]
Abstract
Olive Leaves Extract (OLE) holds therapeutic potential, traditionally used to treat hepatic ailments, though its molecular mechanisms remain unclear. This study evaluated the efficacy of ethanolic OLE against Carbon Tetrachloride (CCl4)-induced oxidative stress in a rat model. Phytochemical analysis was performed using High Performance Liquid Chromatography (HPLC). For this porous, thirty rats were divided into six groups (n = 5): Group 1 (negative control) received a standard diet, while Groups 2-6 were subjected to CCl4-induced toxicity. Group 2 served as the disease control, and Group 3 was treated with silymarin (100 mg/kg). Groups 4, 5, and 6 received OLE at 100 mg/kg, 200 mg/kg, and 300 mg/kg, respectively, for 21 days. OLE significantly modulated hepatic biomarkers (ALT, AST, ALP), increased Total Antioxidant Capacity (TAC), decreased Total Oxidation Capacity (TOC), and restored levels of SOD, GSH, and CAT compared to the CCl4 group. Malondialdehyde (MDA) levels, elevated in the disease group, however downregulated by OLE, particularly at 300 mg/kg. Histological examination revealed normal liver integrity in the OLE-treated groups. Additionally, OLE modulated the mRNA expression of IL-1β, IL-6, TNF-α, NF-kB, Bcl2, and p-53. Apoptotic markers such as Nrf2, HO-1, Cytochrome c, caspase 3, caspase 7, and Bax were normalized with OLE treatment. The inhibition of KEAP1-NRF2 protein-protein interaction showed OLE's superior efficacy compared to silymarin, with a better docking score. These findings suggest that OLE exerts significant hepatoprotective effects against CCl4-induced oxidative stress and inflammation via the Nrf2-NFκB pathway.
Collapse
Affiliation(s)
- Jameel Ahmed Buzdar
- Institute of Physiology and Pharmacology, University of Agriculture Faisalabad, Faisalabad, 38040, Pakistan.
- Disease Investigation Laboratory, Livestock and Dairy Development Department, Quetta, Government of Baluchistan, Baluchistan, 08763, Pakistan.
| | - Qurban Ali Shah
- Institute of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Muzammil Zaman Khan
- Department of Pharmacy, University of Agriculture, Faisalabad, 38040, Pakistan
| | - Azka Zaheer
- Department of Epidemiology and Public Health, University of Agriculture, Faisalabad, 38040, Pakistan
| | - Tahmina Shah
- Department of Veterinarian Physiology and Biochemistry, Sindh Agriculture University Tandojam, Tandojam, Pakistan
| | - Farid Shokry Ataya
- Department of Biochemistry, College of Science, King Saud University, PO Box 2455, Riyadh, 11451, Saudi Arabia
| | - Dalia Fouad
- Department of Zoology, College of Science, King Saud University, PO Box 22452, Riyadh, 11495, Saudi Arabia
| |
Collapse
|
3
|
Kacemi R, Campos MG. Bee Pollen as a Source of Biopharmaceuticals for Neurodegeneration and Cancer Research: A Scoping Review and Translational Prospects. Molecules 2024; 29:5893. [PMCID: PMC11677910 DOI: 10.3390/molecules29245893] [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: 09/24/2024] [Revised: 11/18/2024] [Accepted: 11/20/2024] [Indexed: 01/03/2025] Open
Abstract
Bee Pollen (BP) has many advantageous properties relying on its multitargeting potential, a new tendency in managing many challenging illnesses. In cancer and neurodegeneration, the multiple effects of BP could be of unequaled importance and need further investigation. Although still limited, available data interestingly spotlights some floral sources with promising activities in line with this investigation. Adopting scoping review methodology, we have identified many crucial bioactivities that are widely recognized to individual BP compounds but remain completely untapped in this valuable bee cocktail. A wide range of these compounds have been recently found to be endowed with great potential in modulating pivotal processes in neurodegeneration and cancer pathophysiology. In addition, some ubiquitous BP compounds have only been recently isolated, while the number of studied BPs remains extremely limited compared to the endless pool of plant species worldwide. We have also elucidated that clinical profits from these promising perspectives are still impeded by challenging hurdles such as limited bioavailability of the studied phytocompounds, diversity and lack of phytochemical standardization of BP, and the difficulty of selective targeting in some pathophysiological mechanisms. We finally present interesting insights to guide future research and pave the way for urgently needed and simplified clinical investigations.
Collapse
Affiliation(s)
- Rachid Kacemi
- Observatory of Drug-Herb Interactions, Faculty of Pharmacy, Heath Sciences Campus, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal;
| | - Maria G. Campos
- Observatory of Drug-Herb Interactions, Faculty of Pharmacy, Heath Sciences Campus, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal;
- Coimbra Chemistry Centre (CQC, FCT Unit 313) (FCTUC), University of Coimbra, Rua Larga, 3004-531 Coimbra, Portugal
| |
Collapse
|
4
|
Wang Y, Zhang R, Huang X, He X, Geng S, Pan S, Guo W, Liu X, Dang Y, Qu J, Ma H, Zhao X. CD39 inhibitor (POM-1) enhances radiosensitivity of esophageal squamous cell carcinoma (ESCC) cells by promoting apoptosis through the Bax/Bcl-2/Caspase 9/Caspase 3 pathway. Int Immunopharmacol 2024; 142:113242. [PMID: 39321701 DOI: 10.1016/j.intimp.2024.113242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 09/27/2024]
Abstract
CD39 inhibitor (sodium polyoxotungstate, POM-1) has been reported to have antitumor effects. However, the synergistic effect of POM-1 with radiotherapy requires further elucidation. This study aimed to investigate the role and the molecular mechanism of POM-1 in esophageal squamous cell carcinoma (ESCC) radiosensitization. Firstly, the expression of CD39 in ESCC cells and normal esophageal epithelial cells were detected. Then radioresistant ESCC cells (Eca109R and KYSE150R) were constructed and CD39 expression was analyzed. Furthermore, the effect of POM-1 on radiosensitivity for parent cells and radioresistant cells were observed. Then, we analyzed the effect of POM-1 and CD39 siRNA on radiotherapy-induced apoptosis and determined whether POM-1 modulated the radioresistance of ESCC cells depending on the apoptotic signaling pathway. Finally, we validated the synergistic effect of POM-1 combined with radiotherapy in vivo. Our results showed that CD39 was highly expressed in ESCC cells and radioresistant ESCC cells (p < 0.05). POM-1 reduced radioresistance and proliferation of parent cells and radioresistant cells (p < 0.05). Further mechanistic exploration showed that inhibition of CD39 promoted radiation-induced apoptosis (p < 0.05). Bax knockdown reversed the effect of POM-1 on ESCC cells (p < 0.01). Animal experiments also validated that radiotherapy combined with POM-1 enhanced tumor inhibition in vivo (p < 0.05). These results suggested that POM-1 had synergistic effect with radiotherapy by enhancing cell apoptosis through Bax/Bcl-2 signal pathway in ESCC. The combination of POM-1 and radiotherapy is expected to enhance the anti-tumor effect in ESCC.
Collapse
Affiliation(s)
- Yuchen Wang
- Department of Radiation Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ruijuan Zhang
- Department of Radiation Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xinran Huang
- Department of Radiation Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xinyu He
- Department of Radiation Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shangyi Geng
- Department of Radiation Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shupei Pan
- Department of Radiation Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wei Guo
- Department of Radiation Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaoxiao Liu
- Department of Radiation Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yongze Dang
- Department of Radiation Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jingkun Qu
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Hongbing Ma
- Department of Radiation Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Xixi Zhao
- Department of Radiation Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
5
|
Hassan MG, Hamed AA, Elmetwalli A, Abdel-Monem MO, El-Shora HM, Alsallami WM. Assessment of myco-fabricated Al 2O 3 NPs toxicity on cancer cells and pathogenic microbes by suppression of bacterial metabolic key enzymes. Int J Biol Macromol 2024; 283:137073. [PMID: 39549793 DOI: 10.1016/j.ijbiomac.2024.137073] [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: 06/07/2024] [Revised: 10/26/2024] [Accepted: 10/28/2024] [Indexed: 11/18/2024]
Abstract
There has been a recent change in global attention towards addressing antimicrobial resistance (AMR) as a result of the concerning increase in mortality rates. Nanomaterials have become highly favorable options for a wide range of industrial and biological uses. The objective of this study was to produce aluminum oxide nanoparticles (Al2O3 NPs) using a crude extract from the fungus Aspergillus sp. WAH23, and then analyze the nanoparticles using UV-analysis, electron microscopy (TEM and SEM), and (FT-IR) and X-ray diffraction (XRD). Results revealed that formed nanoparticles are spherical with an average size of 8.5 nm. XRD analysis confirmed the crystalline nature of the synthesized Al2O3 NPs. The Al2O3 nanoparticles exhibited antibacterial properties against a wide range of pathogenic microbes. The antibacterial efficacy of these nanoparticles on the examined bacterial strains was exhibited through their ability to hinder several metabolic processes, including phosphofructokinase (PFK), enolase, and glutamine synthetase. Additionally, the nanoparticles increased the activity of NADH-oxidase, the content of MDA, and the formation of H2O2. The study also examined the anticancer properties of Al2O3 nanoparticles on various types of cancer cells.
Collapse
Affiliation(s)
- Mervat G Hassan
- Botany and Microbiology Department, Faculty of Science, Benha University, P.O. Box 13511, Banha, Qalyubia, Egypt.
| | - Ahmed A Hamed
- Microbial Chemistry Department, National Research Centre, 33 El-Buhouth Street, Dokki, P.O. Box 12622, Giza, Egypt.
| | - Alaa Elmetwalli
- Department of Clinical Trial Research Unit and Drug Discovery, Egyptian Liver Research Institute and Hospital (ELRIAH), Mansoura, Egypt; Microbiology Division, Higher Technological Institute of Applied Health Sciences, Egyptian Liver Research Institute and Hospital (ELRIAH), Mansoura, Egypt
| | - Mohamed O Abdel-Monem
- Botany and Microbiology Department, Faculty of Science, Benha University, P.O. Box 13511, Banha, Qalyubia, Egypt
| | - Hamed M El-Shora
- Botany Department, Faculty of Science, Mansoura University, P.O. Box 35516, Mansoura, Dakahlia, Egypt
| | - Wamedh M Alsallami
- Botany and Microbiology Department, Faculty of Science, Benha University, P.O. Box 13511, Banha, Qalyubia, Egypt
| |
Collapse
|
6
|
Wang D, Ke H, Wang H, Shen J, Jin Y, Lu B, Wang B, Li S, Li Y, Im WT, Siddiqi MZ, Zhu H. Green Synthesis of Silver Nanoparticles (CM-AgNPs) from the Root of Chuanminshen for Improving the Cytotoxicity Effect in Cancer Cells with Antibacterial and Antioxidant Activities. Molecules 2024; 29:5682. [PMID: 39683843 DOI: 10.3390/molecules29235682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/12/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
The unique properties of silver nanoparticles (AgNPs), such as size, surface charge, and the ability to release silver ions, contribute to DNA damage, inducing of oxidative stress, and apoptosis in cancer cells. Thus, the potential application of AgNPs in the field of biomedicine, and cancer therapy are rapidly increasing day by day. Therefore, in this study, AgNPs were synthesized by extract of Chuanminshen violaceum, and then the synthesized CM-AgNPs were fully characterized. The biological activity of CM-AgNPs was investigated for antibacterial, antioxidant, and anticancer activities. The cytotoxic activity of CM-AgNPs was tested for various kinds of cancer cells including MKN45 gastric cancer cells, HCT116 human colon cancer cells, A549 human lung cancer cells, and HepG2 liver cancer cells. Among these cancer cells, the induced apoptosis activity of CM-AgNPs on HCT116 cancer cells was better and was used for further investigation. Besides, the CM-AgNPs exhibited great antioxidant activity against 1,1-diphenyl-2-picrylhydrazyl (DPPH) with 50% free radical scavenging activity, and CM-AgNPs also showed a significant antibacterial activity against Escherichia coli and Staphylococcus aureus. Thus, our pilot data demonstrated that the green synthesis of CM-AgNPs would be considered a good candidate for the treatment of HCT116 cancer cells, with its strong antioxidant activity and antibacterial effects.
Collapse
Affiliation(s)
- Dandan Wang
- College of Life Sciences, Yantai University, Yantai 264005, China
| | - Haijing Ke
- College of Life Sciences, Yantai University, Yantai 264005, China
| | - Hongtao Wang
- College of Life Sciences, Yantai University, Yantai 264005, China
| | - Jingyu Shen
- College of Life Sciences, Yantai University, Yantai 264005, China
| | - Yan Jin
- School of Life Science, Nantong University, Nantong 226019, China
| | - Bo Lu
- College of Life Sciences, Yantai University, Yantai 264005, China
| | - Bingju Wang
- College of Life Sciences, Yantai University, Yantai 264005, China
| | - Shuang Li
- College of Life Sciences, Yantai University, Yantai 264005, China
| | - Yao Li
- College of Life Sciences, Yantai University, Yantai 264005, China
| | - Wan Taek Im
- Department of Biotechnology, Hankyong National University, 327 Jungang-ro, Anseong-si 17579, Gyeonggi-do, Republic of Korea
| | - Muhammad Zubair Siddiqi
- Department of Biotechnology, Hankyong National University, 327 Jungang-ro, Anseong-si 17579, Gyeonggi-do, Republic of Korea
| | - Haibo Zhu
- School of Public Health and Management, Binzhou Medical University, Yantai 264005, China
| |
Collapse
|
7
|
Elizalde-Velázquez GA, Gómora-Martínez O, Raldua D, Herrera-Vázquez SE, Gómez-Oliván LM. Understanding the impact of environmentally relevant alkyl C12-16 dimethylbenzyl ammonium chloride concentrations on zebrafish health. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 953:175984. [PMID: 39244042 DOI: 10.1016/j.scitotenv.2024.175984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/30/2024] [Accepted: 08/31/2024] [Indexed: 09/09/2024]
Abstract
Alkyldimethylbenzylammonium chlorides (ADBACs), classified as second-generation quaternary ammonium compounds, are extensively employed across various sectors, encompassing veterinary medicine, food production, pharmaceuticals, cosmetics, ophthalmology, and agriculture. Consequently, significant volumes of ADBAC C12-C16 are discharged into the environment, posing a threat to aquatic organisms. Regrettably, comprehensive data regarding the toxicological characteristics of these compounds remain scarce. This research aimed to determine whether or not ADBAC C12-C16, at environmentally relevant concentrations (0.4, 0.8, and 1.6 μg/L), may instigate oxidative stress and alter the expression of apoptosis-related genes in the liver, brain, gut, and gills of Danio rerio adults (5-6 months). The findings revealed that ADBAC C12-C16 elicited an oxidative stress response across all examined organs following 96 h of exposure. Nonetheless, the magnitude of this response varied among organs, with the gills exhibiting the highest degree of susceptibility, followed by the gut, liver, and brain, in descending order. Only the gut and gills of the examined organs displayed a concentration-dependent reduction in the activity of superoxide dismutase (SOD) and catalase (CAT). Akin to the oxidative stress response, all organs exhibited a marked increase in bax, blc2, casp3, and p53 expression levels. However, the gills and gut manifested a distinctive suppression in the expression of nrf1 and nrf2. Our Principal Component Analysis (PCA) confirmed that SOD, CAT, nrf1, and nrf2 were negatively correlated to oxidative damage biomarkers and apoptosis-related genes in the gills and gut; meanwhile, in the remaining organs, all biomarkers were extensively correlated. From the above, it can be concluded that ADBAC C12-C16 in low and environmental concentrations may threaten the health of freshwater fish.
Collapse
Affiliation(s)
- Gustavo Axel Elizalde-Velázquez
- Laboratorio de Toxicología Ambiental, Facultad de Química, Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan, Colonia Residencial Colón, CP 50120 Toluca, Estado de México, Mexico
| | - Omar Gómora-Martínez
- Laboratorio de Toxicología Ambiental, Facultad de Química, Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan, Colonia Residencial Colón, CP 50120 Toluca, Estado de México, Mexico
| | - Demetrio Raldua
- Institute of Environmental Assessment and Water Research (IDAEA-CSIC), Jordi Girona 18, 08034 Barcelona, Spain
| | - Selene Elizabeth Herrera-Vázquez
- Laboratorio de Toxicología Ambiental, Facultad de Química, Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan, Colonia Residencial Colón, CP 50120 Toluca, Estado de México, Mexico
| | - Leobardo Manuel Gómez-Oliván
- Laboratorio de Toxicología Ambiental, Facultad de Química, Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan, Colonia Residencial Colón, CP 50120 Toluca, Estado de México, Mexico.
| |
Collapse
|
8
|
Yuan R, Gao D, Yang G, Zhuoma D, Pu Z, Ciren Y, Li B, Yu J. Oxysophocarpine Prevents the Glutamate-Induced Apoptosis of HT-22 Cells via the Nrf2/HO-1 Signaling Pathway. Curr Issues Mol Biol 2024; 46:13035-13049. [PMID: 39590371 PMCID: PMC11593028 DOI: 10.3390/cimb46110777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/09/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Oxysophocarpine (OSC), a quinolizidine alkaloid, shows neuroprotective potential, though its mechanisms are unclear. The aim of the present study was to investigate the neuroprotective effects of OSC through the nuclear factor erythroid 2-related factor 2 (Nrf2)/ heme oxygenase-1 (HO-1) signaling pathway using the HT-22 cell line. Assessments of cell viability were conducted utilizing the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Assessments of oxidative stress (OS) were conducted through the quantification of reactive oxygen species (ROS). The integrity of the mitochondrial membrane potential (MMP) was scrutinized using fluorescent probe technology. Apoptosis levels were quantified using terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining. The trafficking of Nrf2 within the cell nucleus was examined through immunofluorescence analysis. Furthermore, Western blotting (WB) was applied to evaluate the expression levels of proteins implicated in apoptosis and the Nrf2/HO-1 pathway. To further probe the influence of OSC on the overexpression of antioxidant enzymes, cells were subjected to transfection with HO-1 siRNA. The results showed that OSC inhibited glutamate-induced OS, as evidenced by reduced cell viability and ROS levels. Furthermore, the apoptotic condition induced by glutamate in HT-22 cells was significantly reduced following OSC treatment. More interestingly, the Nrf2/HO-1 signaling pathway was upregulated following OSC treatment. These results suggest that OSC can exert neuroprotective effects by regulating the Nrf2/HO-1 pathway to inhibit neuronal cell apoptosis, potentially aiding in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Ruiying Yuan
- Department of Pharmaceutical Sciences, School of Medicine, Tibet University, Lhasa 850000, China; (R.Y.); (D.G.); (G.Y.); (D.Z.); (Z.P.); (Y.C.)
- College of Pharmacy, Wuhan University, Wuhan 430000, China;
| | - Dan Gao
- Department of Pharmaceutical Sciences, School of Medicine, Tibet University, Lhasa 850000, China; (R.Y.); (D.G.); (G.Y.); (D.Z.); (Z.P.); (Y.C.)
| | - Guibing Yang
- Department of Pharmaceutical Sciences, School of Medicine, Tibet University, Lhasa 850000, China; (R.Y.); (D.G.); (G.Y.); (D.Z.); (Z.P.); (Y.C.)
| | - Dongzhi Zhuoma
- Department of Pharmaceutical Sciences, School of Medicine, Tibet University, Lhasa 850000, China; (R.Y.); (D.G.); (G.Y.); (D.Z.); (Z.P.); (Y.C.)
| | - Zhen Pu
- Department of Pharmaceutical Sciences, School of Medicine, Tibet University, Lhasa 850000, China; (R.Y.); (D.G.); (G.Y.); (D.Z.); (Z.P.); (Y.C.)
| | - Yangzhen Ciren
- Department of Pharmaceutical Sciences, School of Medicine, Tibet University, Lhasa 850000, China; (R.Y.); (D.G.); (G.Y.); (D.Z.); (Z.P.); (Y.C.)
| | - Bin Li
- Department of Pharmaceutical Sciences, School of Medicine, Tibet University, Lhasa 850000, China; (R.Y.); (D.G.); (G.Y.); (D.Z.); (Z.P.); (Y.C.)
| | - Jianqing Yu
- College of Pharmacy, Wuhan University, Wuhan 430000, China;
| |
Collapse
|
9
|
Zhang T, Huang Q, Lu L, Zhou K, Hu K, Gan K. ROS-responsive Hydrogel Loaded with Allicin Suppresses Cell Apoptosis for the Treatment of Intervertebral Disc Degeneration in a Rat Model. World Neurosurg 2024; 193:675-686. [PMID: 39490768 DOI: 10.1016/j.wneu.2024.10.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Intervertebral disc degeneration (IVDD) is a common cause of lower back pain, and cell apoptosis plays a key role in its progression. This study explores the therapeutic potential of a reactive oxygen species (ROS)-responsive hydrogel loaded with allicin for treating IVDD. METHODS Allicin was encapsulated in an ROS-responsive hydrogel, and its controlled release was studied in vitro. Nucleus pulposus cells were treated with hydrogen peroxide to induce apoptosis, and the effects of the hydrogel were examined using quantitative polymerase chain reaction and Western blotting. An in vivo rat model of IVDD was also established to assess the efficacy of the treatment. RESULTS The ROS-responsive hydrogel effectively inhibited apoptosis in nucleus pulposus cells by reducing ROS levels and modulating the expression of apoptotic and antiapoptotic genes. In the rat model, the hydrogel loaded with allicin significantly reduced IVDD, preserving disc morphology and matrix integrity. CONCLUSIONS ROS-responsive hydrogel loaded with allicin shows potential as a therapeutic approach for IVDD by inhibiting cell apoptosis and reducing disc degeneration in vivo.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Orthopaedics, Li Huili Hospital Affiliated to Ningbo University, Ningbo, Zhejiang, China
| | - Qing Huang
- Department of Gynecology, Li Huili Hospital Affiliated to Ningbo University, Ningbo, Zhejiang, China
| | - Liangjie Lu
- Department of Orthopaedics, Li Huili Hospital Affiliated to Ningbo University, Ningbo, Zhejiang, China
| | - Ke Zhou
- Department of Orthopaedics, Li Huili Hospital Affiliated to Ningbo University, Ningbo, Zhejiang, China
| | - Keqi Hu
- Department of Orthopaedics, Li Huili Hospital Affiliated to Ningbo University, Ningbo, Zhejiang, China
| | - Kaifeng Gan
- Department of Orthopaedics, Li Huili Hospital Affiliated to Ningbo University, Ningbo, Zhejiang, China.
| |
Collapse
|
10
|
Wang K, Zhou Y, Wen C, Du L, Li L, Cui Y, Luo H, Liu Y, Zeng L, Li S, Xiong L, Yue R. Protective effects of tetramethylpyrazine on myocardial ischemia/reperfusion injury involve NLRP3 inflammasome suppression by autophagy activation. Biochem Pharmacol 2024; 229:116541. [PMID: 39284501 DOI: 10.1016/j.bcp.2024.116541] [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: 03/05/2024] [Revised: 08/27/2024] [Accepted: 09/13/2024] [Indexed: 09/21/2024]
Abstract
Tetramethylpyrazine (TMP) belongs to the active ingredients of the traditional Chinese medicine Chuanxiong, which has a certain protective effect in myocardial ischemia-reperfusion (I/R) injury. It can improve postoperative cardiac function and alleviate ventricular remodeling in acute myocardial infarction patients. However, its specific protective mechanism is still unclear. In this study, a certain concentration of TMP was introduced into I/R mice or H9C2 cells after oxygen-glucose deprivation/reoxygenation (OGD/R) treatment to observe the effects of TMP on cardiomyocyte activity, cytotoxicity, apoptosis, autophagy, pyroptosis, and NLRP3 inflammasome activation. The results displayed that TMP intervention could reduce OGD/R and I/R-induced cardiomyocyte apoptosis, accelerate cellular activity and autophagy levels, and ameliorate myocardial tissue necrosis in I/R mice in a dose-dependent manner. Further, TMP prevented the formation of NLRP3 inflammasomes to suppress pyroptosis by increasing the level of cardiomyocyte autophagy after I/R and OGD/R modelling, the introduction of chloroquine to suppress autophagic activity in vivo and in vitro was further analyzed to confirm whether TMP inhibits NLRP3 inflammasome activation and pyroptosis by increasing autophagy, and we found the inhibitory effect of TMP on NLRP3 inflammasomes and its protective effect against myocardial injury were blocked when autophagy was inhibited with chloroquine. In conclusion, this experiment demonstrated that TMP unusually attenuated I/R injury in mice, and this protective effect was achieved by inhibiting the activation of NLRP3 inflammasomes through enhancing autophagic activity.
Collapse
Affiliation(s)
- Kun Wang
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China.
| | - Yang Zhou
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China.
| | - Cong Wen
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China.
| | - Linqin Du
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China.
| | - Lan Li
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China.
| | - Yangyang Cui
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China.
| | - Hao Luo
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China.
| | - Yanxu Liu
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China.
| | - Lang Zeng
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China.
| | - Shikang Li
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China.
| | - Lijuan Xiong
- Department of Cardiology, People's Hospital of Guang'an District, Guang'an 638550, China.
| | - Rongchuan Yue
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Cardiology, People's Hospital of Guang'an District, Guang'an 638550, China.
| |
Collapse
|
11
|
Green DR. Cell death: Revisiting the roads to ruin. Dev Cell 2024; 59:2523-2531. [PMID: 39378838 PMCID: PMC11469552 DOI: 10.1016/j.devcel.2024.08.008] [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: 06/08/2024] [Revised: 07/17/2024] [Accepted: 08/08/2024] [Indexed: 10/10/2024]
Abstract
A paradigm shift in the study of cell death is currently occurring: whereas previously we had always considered that there were "points of no return" in any cell death pathway, we now realize that in many types of active, regulated cell death, this is not the case. We are also learning that cells that "almost die," but nevertheless survive, can transiently take on an altered state, with potential implications for understanding cancer therapies and relapse. In this perspective, we parse the many forms of cell death by analogy to suicide, sabotage, and murder, and consider how cells that might be "instructed" to engage a cell death pathway might nevertheless survive.
Collapse
Affiliation(s)
- Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
12
|
Hartley B, Bassiouni W, Roczkowsky A, Fahlman R, Schulz R, Julien O. N-Terminomic Identification of Intracellular MMP-2 Substrates in Cardiac Tissue. J Proteome Res 2024; 23:4188-4202. [PMID: 38647137 PMCID: PMC11460328 DOI: 10.1021/acs.jproteome.3c00755] [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: 11/11/2023] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/25/2024]
Abstract
Proteases are enzymes that induce irreversible post-translational modifications by hydrolyzing amide bonds in proteins. One of these proteases is matrix metalloproteinase-2 (MMP-2), which has been shown to modulate extracellular matrix remodeling and intracellular proteolysis during myocardial injury. However, the substrates of MMP-2 in heart tissue are limited, and lesser known are the cleavage sites. Here, we used degradomics to investigate the substrates of intracellular MMP-2 in rat ventricular extracts. First, we designed a novel, constitutively active MMP-2 fusion protein (MMP-2-Fc) that we expressed and purified from mammalian cells. Using this protease, we proteolyzed ventricular extracts and used subtiligase-mediated N-terminomic labeling which identified 95 putative MMP-2-Fc proteolytic cleavage sites using mass spectrometry. The intracellular MMP-2 cleavage sites identified in heart tissue extracts were enriched for proteins primarily involved in metabolism, as well as the breakdown of fatty acids and amino acids. We further characterized the cleavage of three of these MMP-2-Fc substrates based on the gene ontology analysis. We first characterized the cleavage of sarco/endoplasmic reticulum calcium ATPase (SERCA2a), a known MMP-2 substrate in myocardial injury. We then characterized the cleavage of malate dehydrogenase (MDHM) and phosphoglycerate kinase 1 (PGK1), representing new cardiac tissue substrates. Our findings provide insights into the intracellular substrates of MMP-2 in cardiac cells, suggesting that MMP-2 activation plays a role in cardiac metabolism.
Collapse
Affiliation(s)
- Bridgette Hartley
- Department
of Biochemistry, University of Alberta, Edmonton T6G 2H7, Canada
| | - Wesam Bassiouni
- Department
of Pharmacology, University of Alberta, Edmonton T6G 2S2, Canada
| | - Andrej Roczkowsky
- Department
of Pharmacology, University of Alberta, Edmonton T6G 2S2, Canada
| | - Richard Fahlman
- Department
of Biochemistry, University of Alberta, Edmonton T6G 2H7, Canada
| | - Richard Schulz
- Department
of Pharmacology, University of Alberta, Edmonton T6G 2S2, Canada
- Department
of Pediatrics, University of Alberta, Edmonton T6G 2S2, Canada
| | - Olivier Julien
- Department
of Biochemistry, University of Alberta, Edmonton T6G 2H7, Canada
| |
Collapse
|
13
|
Das T, Mondal S, Das S, Das S, Das Saha K. Enhanced anticancer activity of (-)-epigallocatechin-3-gallate (EGCG) encapsulated NPs toward colon cancer cell lines. Free Radic Res 2024; 58:565-582. [PMID: 38810269 DOI: 10.1080/10715762.2024.2360013] [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: 11/03/2023] [Revised: 04/08/2024] [Accepted: 05/15/2024] [Indexed: 05/31/2024]
Abstract
(-)-Epigallocatechin-3-gallate (EGCG), a bioactive polyphenol of green tea, has chemo-preventive effects against various cancer cells. Nanoparticles (NPs) carrying different ligands are able to specifically interact with their receptors on different cancer cells that can provide effective release of cytotoxic drugs. In the present study, we have prepared EGCG entrapped NPs using PLGA (poly(d,l-lactide-co-glycolide)). Polyethylene glycol (PEG) and folic acid (FA) via double emulsion solvent evaporation (DESE) method obtained PLGA-EGCG (P-E), PLGA-PEG-EGCG (PP-E), and PLGA-PEG-FA-EGCG (PPF-E). Nanoformulations had been characterized with 1H NMR and FT-IR techniques, AFM, and DLS. PPF-E NPs showed an average size of 220 nm. Analysis of zeta potential confirmed the stability of NPs. HCT-116, HT-29, HCT-15, and HEK 293 cells were treated with both the prepared NPs and free EGCG (0-140 μM). Result showed PPF-E NPs had improved delivery, uptake and cell cytotoxicity toward human folic acid receptor-positive (FR+) colorectal cancer (CRC) cells as mainly on HCT-116 compared to HT-29, but not on the folic acid-negative cells (FR-) as HCT-15. PPF-E NPs enhanced intracellular reactive oxygen species (ROS) level in absence of N-acetyl-l-cysteine (NAC), elevated DNA fragmentation level, and increased apoptotic cell death at higher doses compared to other two NPs and free EGCG. In conclusion, PPF-E NPs exerted greater efficacy than PP-E, P-E, and free EGCG in HCT-116 cells.
Collapse
Affiliation(s)
- Tanushree Das
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Sanchaita Mondal
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Sujata Das
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Sanjib Das
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Krishna Das Saha
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
14
|
Wang WG, Li SL, Liu B, Tang N, Zhang C, Jiang XF, Tao LM, Xu WP, Zhang Y. Natural pyrethrins induce cytotoxicity in SH-SY5Y cells and neurotoxicity in zebrafish embryos (Danio rerio). ENVIRONMENTAL RESEARCH 2024; 257:119267. [PMID: 38815718 DOI: 10.1016/j.envres.2024.119267] [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: 03/14/2024] [Revised: 05/21/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
Natural pyrethrins are widely used in agriculture because of their good insecticidal activity. Meanwhile, natural pyrethrins play an important role in the safety evaluation of pyrethroids as precursors for structural development of pyrethroid insecticides. However, there are fewer studies evaluating the neurological safety of natural pyrethrins on non-target organisms. In this study, we used SH-SY5Y cells and zebrafish embryos to explore the neurotoxicity of natural pyrethrins. Natural pyrethrins were able to induce SH-SY5Y cells damage, as evidenced by decreased viability, cycle block, apoptosis and DNA damage. The apoptotic pathway may be related to the involvement of mitochondria and the results showed that natural pyrethrins induced a rise in Capase-3 viability, Ca2+ overload, a decrease in adenosine triphosphate (ATP) and a collapse of mitochondrial membrane potential in SH-SY5Y cells. Natural pyrethrins may mediate DNA damage in SH-SY5Y cells through oxidative stress. The results showed that natural pyrethrins induced an increase in reactive oxygen species (ROS) levels, superoxide dismutase (SOD) activity, malondialdehyde (MDA) content and catalase (CAT) activity, and induced a decrease in glutathione peroxidase (GPx) activity in SH-SY5Y cells. In vivo, natural pyrethrins induced developmental malformations in zebrafish embryos, which were mainly characterized by pericardial edema and yolk sac edema. Meanwhile, the results showed that natural pyrethrins induced damage to the Huc-GFP axis and disturbed lipid metabolism in the head of zebrafish embryos. Further results showed elevated ROS levels and apoptosis in the head of zebrafish embryos, which corroborated with the results of the cell model. Finally, the results of mRNA expression assay of neurodevelopment-related genes indicated that natural pyrethrins exposure interfered with their expression and led to neurodevelopmental damage in zebrafish embryos. Our study may raise concerns about the neurological safety of natural pyrethrins on non-target organisms.
Collapse
Affiliation(s)
- Wei-Guo Wang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Shou-Lin Li
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Bin Liu
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China; Qingpu District Agro-Technology Extension Service Center, Shanghai, 201799, China
| | - Ning Tang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Cheng Zhang
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, 75390, United States
| | - Xu-Feng Jiang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Li-Ming Tao
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Wen-Ping Xu
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yang Zhang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
| |
Collapse
|
15
|
Gomez‐Cardona E, Dehkordi MH, Van Baar K, Vitkauskaite A, Julien O, Fearnhead HO. An atlas of caspase cleavage events in differentiating muscle cells. Protein Sci 2024; 33:e5156. [PMID: 39180494 PMCID: PMC11344277 DOI: 10.1002/pro.5156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 08/02/2024] [Accepted: 08/11/2024] [Indexed: 08/26/2024]
Abstract
Executioner caspases, such as caspase-3, are known to induce apoptosis, but in other contexts, they can control very different fates, including cell differentiation and neuronal plasticity. While hundreds of caspase substrates are known to be specifically targeted during cell death, we know very little about how caspase activity brings about non-apoptotic fates. Here, we report the first proteome identification of cleavage events in C2C12 cells undergoing myogenic differentiation and its comparison to undifferentiated or dying C2C12 cells. These data have identified new caspase substrates, including caspase substrates specifically associated with differentiation, and show that caspases are regulating proteins involved in myogenesis in myotubes, several days after caspase-3 initiated differentiation. Cytoskeletal proteins emerged as a major group of non-apoptotic caspase substrates. We also identified proteins with well-established roles in muscle differentiation as substrates cleaved in differentiating cells.
Collapse
Affiliation(s)
- Erik Gomez‐Cardona
- Department of Biochemistry, Faculty of Medicine and DentistryUniversity of AlbertaAlbertaCanada
| | - Mahshid H. Dehkordi
- Pharmacology and Therapeutics, School of MedicineUniversity of GalwayGalwayIreland
| | - Kolden Van Baar
- Department of Biochemistry, Faculty of Medicine and DentistryUniversity of AlbertaAlbertaCanada
| | - Aiste Vitkauskaite
- Pharmacology and Therapeutics, School of MedicineUniversity of GalwayGalwayIreland
| | - Olivier Julien
- Department of Biochemistry, Faculty of Medicine and DentistryUniversity of AlbertaAlbertaCanada
| | - Howard O. Fearnhead
- Pharmacology and Therapeutics, School of MedicineUniversity of GalwayGalwayIreland
| |
Collapse
|
16
|
Smith AJ, Hergenrother PJ. Raptinal: a powerful tool for rapid induction of apoptotic cell death. Cell Death Discov 2024; 10:371. [PMID: 39164225 PMCID: PMC11335860 DOI: 10.1038/s41420-024-02120-1] [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: 05/15/2024] [Revised: 07/25/2024] [Accepted: 07/30/2024] [Indexed: 08/22/2024] Open
Abstract
Chemical inducers of apoptosis have been utilized for decades as tools to uncover steps of the apoptotic cascade and to treat various diseases, most notably cancer. While there are several useful compounds available, limitations in potency, universality, or speed of cell death of these pro-apoptotic agents have meant that no single compound is suitable for all (or most) purposes. Raptinal is a recently described small molecule that induces intrinsic pathway apoptosis rapidly and reliably, and consequently, has been utilized in cell culture and whole organisms for a wide range of biological studies. Its distinct mechanism of action complements the current arsenal of cytotoxic compounds, making it useful as a probe for the apoptosis pathway and other cellular processes. The rapid induction of cell death by Raptinal and its widespread commercial availability make it the pro-apoptotic agent of choice for many applications.
Collapse
Affiliation(s)
- Amanda J Smith
- Department of Chemistry, Carl R. Woese Institute for Genomic Biology, and Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Paul J Hergenrother
- Department of Chemistry, Carl R. Woese Institute for Genomic Biology, and Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
17
|
Říhová K, Lapčík P, Veselá B, Knopfová L, Potěšil D, Pokludová J, Šmarda J, Matalová E, Bouchal P, Beneš P. Caspase-9 Is a Positive Regulator of Osteoblastic Cell Migration Identified by diaPASEF Proteomics. J Proteome Res 2024; 23:2999-3011. [PMID: 38498986 PMCID: PMC11301665 DOI: 10.1021/acs.jproteome.3c00641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 02/21/2024] [Accepted: 03/07/2024] [Indexed: 03/20/2024]
Abstract
Caspase-9 is traditionally considered the initiator caspase of the intrinsic apoptotic pathway. In the past decade, however, other functions beyond initiation/execution of cell death have been described including cell type-dependent regulation of proliferation, differentiation/maturation, mitochondrial, and endosomal/lysosomal homeostasis. As previous studies revealed nonapoptotic functions of caspases in osteogenesis and bone homeostasis, this study was performed to identify proteins and pathways deregulated by knockout of caspase-9 in mouse MC3T3-E1 osteoblasts. Data-independent acquisition-parallel accumulation serial fragmentation (diaPASEF) proteomics was used to compare protein profiles of control and caspase-9 knockout cells. A total of 7669 protein groups were quantified, and 283 upregulated/141 downregulated protein groups were associated with the caspase-9 knockout phenotype. The deregulated proteins were mainly enriched for those associated with cell migration and motility and DNA replication/repair. Altered migration was confirmed in MC3T3-E1 cells with the genetic and pharmacological inhibition of caspase-9. ABHD2, an established regulator of cell migration, was identified as a possible substrate of caspase-9. We conclude that caspase-9 acts as a modulator of osteoblastic MC3T3-E1 cell migration and, therefore, may be involved in bone remodeling and fracture repair.
Collapse
Affiliation(s)
- Kamila Říhová
- Department
of Experimental Biology, Faculty of Science, Masaryk University, Brno 625 00, Czech Republic
- International
Clinical Research Center, St. Anne’s
University Hospital, Brno 602 00, Czech Republic
| | - Petr Lapčík
- Department
of Biochemistry, Faculty of Science, Masaryk
University, Brno 625 00, Czech Republic
| | - Barbora Veselá
- Laboratory
of Odontogenesis and Osteogenesis, Institute of Animal Physiology
and Genetics, Czech Academy of Sciences, Brno 602 00, Czech Republic
| | - Lucia Knopfová
- Department
of Experimental Biology, Faculty of Science, Masaryk University, Brno 625 00, Czech Republic
- International
Clinical Research Center, St. Anne’s
University Hospital, Brno 602 00, Czech Republic
| | - David Potěšil
- Proteomics
Core Facility, Central European Institute for Technology, Masaryk University, Brno 625 00, Czech Republic
| | - Jana Pokludová
- Department
of Experimental Biology, Faculty of Science, Masaryk University, Brno 625 00, Czech Republic
- International
Clinical Research Center, St. Anne’s
University Hospital, Brno 602 00, Czech Republic
| | - Jan Šmarda
- Department
of Experimental Biology, Faculty of Science, Masaryk University, Brno 625 00, Czech Republic
| | - Eva Matalová
- Laboratory
of Odontogenesis and Osteogenesis, Institute of Animal Physiology
and Genetics, Czech Academy of Sciences, Brno 602 00, Czech Republic
- Department
of Physiology, Faculty of Veterinary Medicine, University of Veterinary Sciences, Brno 612 42, Czech Republic
| | - Pavel Bouchal
- Department
of Biochemistry, Faculty of Science, Masaryk
University, Brno 625 00, Czech Republic
| | - Petr Beneš
- Department
of Experimental Biology, Faculty of Science, Masaryk University, Brno 625 00, Czech Republic
- International
Clinical Research Center, St. Anne’s
University Hospital, Brno 602 00, Czech Republic
| |
Collapse
|
18
|
de Oliveira G, de Andrade Rodrigues L, Souza da Silva AA, Gouvea LC, Silva RCL, Sasso-Cerri E, Cerri PS. Reduction of osteoclast formation and survival following suppression of cytokines by diacerein in periodontitis. Biomed Pharmacother 2024; 177:117086. [PMID: 39013222 DOI: 10.1016/j.biopha.2024.117086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/29/2024] [Accepted: 07/01/2024] [Indexed: 07/18/2024] Open
Abstract
Periodontitis causes an increase in several bioactive agents such as interleukins (IL), tumor necrosis factor (TNF)-α and receptor activator of NF-kB ligand (RANKL), which induce the osteoclast formation and activity. Since diacerein exerts anti-TNF-α and anti-IL-1 effects, alleviating bone destruction in osteoarthritis, we investigated whether this drug inhibits the formation and survival of osteoclast in the periodontitis. Rats were distributed into 3 groups: 1) group with periodontitis treated with 100 mg/kg diacerein (PDG), 2) group with periodontitis treated with saline (PSG) and group control (CG) without any treatment. After 7, 15 and 30 days, the maxillae were collected for light and transmission electron microscopy analyses. Gingiva samples were collected to evaluate the mRNA levels for Tnf, Il1b, Tnfsf11 and Tnfrsf11b by RT-qPCR. In PDG, the expression of Tnf and Il1b genes reduced significantly compared to PSG, except for Tnf expression at 7 days. The number of osteoclasts reduced significantly in the PDG in comparison with PSG at 7 and 15 days. In all periods, the IL-6 immunoexpression, RANKL/OPG immunoexpression and mRNA levels of Tnfsf11/Tnfrsf11b ratio were significantly lower in PDG than in PSG. PDG exhibited significantly higher frequency of TUNEL-positive osteoclasts than in PSG and CG at all time points. Osteoclasts with caspase-3-immunolabelled cytoplasm and nuclei with masses of condensed chromatin were observed in PDG, confirming osteoclast apoptosis. Diacerein inhibits osteoclastogenesis by decreasing Tnf and Il1b mRNA levels, resulting in decreased RANKL/OPG ratio, and induces apoptosis in osteoclasts of alveolar process of rat molars with periodontitis.
Collapse
Affiliation(s)
- Gabriella de Oliveira
- São Paulo State University (UNESP), School of Dentistry, Laboratory of Histology and Embryology, Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry, Araraquara, SP, Brazil
| | - Lucas de Andrade Rodrigues
- São Paulo State University (UNESP), School of Dentistry, Laboratory of Histology and Embryology, Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry, Araraquara, SP, Brazil
| | | | - Lays Cristina Gouvea
- São Paulo State University (UNESP), School of Dentistry, Laboratory of Histology and Embryology, Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry, Araraquara, SP, Brazil
| | - Renata Cristina Lima Silva
- São Paulo State University (UNESP), School of Dentistry, Laboratory of Histology and Embryology, Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry, Araraquara, SP, Brazil
| | - Estela Sasso-Cerri
- São Paulo State University (UNESP), School of Dentistry, Laboratory of Histology and Embryology, Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry, Araraquara, SP, Brazil
| | - Paulo Sérgio Cerri
- São Paulo State University (UNESP), School of Dentistry, Laboratory of Histology and Embryology, Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry, Araraquara, SP, Brazil.
| |
Collapse
|
19
|
Yao CG, Zhao ZJ, Tan T, Yan JN, Chen ZW, Xiong JT, Li HL, Wei YH, Hu KH. Lindqvist-type Polyoxometalates Act as Anti-breast Cancer Drugs via Mitophagy-induced Apoptosis. Curr Med Sci 2024; 44:809-819. [PMID: 39096476 DOI: 10.1007/s11596-024-2910-2] [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: 12/08/2023] [Accepted: 06/11/2024] [Indexed: 08/05/2024]
Abstract
OBJECTIVE Lindqvist-type polyoxometalates (POMs) exhibit potential antitumor activities. This study aimed to examine the effects of Lindqvist-type POMs against breast cancer and the underlying mechanism. METHODS Using different cancer cell lines, the present study evaluated the antitumor activities of POM analogues that were modified at the body skeleton based on molybdenum-vanadium-centered negative oxygen ion polycondensations with different side strains. Cell colony formation assay, autophagy detection, mitochondrial observation, qRT-PCR, Western blotting, and animal model were used to evaluate the antitumor activities of POMs against breast cancer cells and the related mechanism. RESULTS MO-4, a Lindqvist-type POM linking a proline at its side strain, was selected for subsequent experiments due to its low half maximal inhibitory concentration in the inhibition of proliferation of breast cancer cells. It was found that MO-4 induced the apoptosis of multiple types of breast cancer cells. Mechanistically, MO-4 activated intracellular mitophagy by elevating mitochondrial reactive oxygen species (ROS) levels and resulting in apoptosis. In vivo, breast tumor growth and distant metastasis were significantly reduced following MO-4 treatment. CONCLUSION Collectively, the results of the present study demonstrated that the novel Lindqvist-type POM MO-4 may exhibit potential in the treatment of breast cancer.
Collapse
Affiliation(s)
- Chen-Guang Yao
- Sino-German Biomedical Center, Hubei Provincial Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, 430068, China
| | - Zi-Jia Zhao
- Sino-German Biomedical Center, Hubei Provincial Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, 430068, China
| | - Ting Tan
- Sino-German Biomedical Center, Hubei Provincial Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, 430068, China
| | - Jiang-Ning Yan
- Sino-German Biomedical Center, Hubei Provincial Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, 430068, China
| | - Zhong-Wei Chen
- Sino-German Biomedical Center, Hubei Provincial Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, 430068, China
| | - Jun-Tao Xiong
- Center for Evaluation of Hubei Medical Products Administration, Wuhan, 430068, China
| | - Han-Luo Li
- Sino-German Biomedical Center, Hubei Provincial Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, 430068, China
| | - Yan-Hong Wei
- Sino-German Biomedical Center, Hubei Provincial Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, 430068, China
| | - Kang-Hong Hu
- Sino-German Biomedical Center, Hubei Provincial Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, 430068, China.
| |
Collapse
|
20
|
Exconde PM, Bourne CM, Kulkarni M, Discher BM, Taabazuing CY. Inflammatory caspase substrate specificities. mBio 2024; 15:e0297523. [PMID: 38837391 PMCID: PMC11253702 DOI: 10.1128/mbio.02975-23] [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] [Indexed: 06/07/2024] Open
Abstract
Caspases are a family of cysteine proteases that act as molecular scissors to cleave substrates and regulate biological processes such as programmed cell death and inflammation. Extensive efforts have been made to identify caspase substrates and to determine factors that dictate substrate specificity. Thousands of putative substrates have been identified for caspases that regulate an immunologically silent type of cell death known as apoptosis, but less is known about substrates of the inflammatory caspases that regulate an immunostimulatory type of cell death called pyroptosis. Furthermore, much of our understanding of caspase substrate specificities is derived from work done with peptide substrates, which do not often translate to native protein substrates. Our knowledge of inflammatory caspase biology and substrates has recently expanded and here, we discuss the recent advances in our understanding of caspase substrate specificities, with a focus on inflammatory caspases. We highlight new substrates that have been discovered and discuss the factors that engender specificity. Recent evidence suggests that inflammatory caspases likely utilize two binding interfaces to recognize and process substrates, the active site and a conserved exosite.
Collapse
Affiliation(s)
- Patrick M. Exconde
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Christopher M. Bourne
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Madhura Kulkarni
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Bohdana M. Discher
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Cornelius Y. Taabazuing
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
21
|
Cai XQ, Yang H, Liang BQ, Deng CC, Xue HY, Zhang JJ, Wang XZ. Glutamate rescues heat stress-induced apoptosis of Sertoli cells by enhancing the activity of antioxidant enzymes and activating the Trx1-Akt pathway in vitro. Theriogenology 2024; 223:1-10. [PMID: 38642435 DOI: 10.1016/j.theriogenology.2024.04.005] [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: 06/17/2023] [Revised: 04/09/2024] [Accepted: 04/15/2024] [Indexed: 04/22/2024]
Abstract
Heat stress reduces the number of Sertoli cells, which is closely related to an imbalanced redox status. Glutamate functions to maintain the equilibrium of redox homeostasis. However, the role of glutamate in heat treated Sertoli cells remains unclear. Herein, Sertoli cells from 3-week-old piglets were treated at 44 °C for 30 min (heat stress). Glutamate levels increased significantly following heat stress treatment, followed by a gradual decrease during recovery, while glutathione (GSH) showed a gradual increase. The addition of exogenous glutamate (700 μM) to Sertoli cells before heat stress significantly reduced the heat stress-induced apoptosis rate, mediated by enhanced levels of antioxidant substances (superoxide dismutase (SOD), total antioxidant capacity (TAC), and GSH) and reduced levels of oxidative substances (reactive oxygen species (ROS) and malondialdehyde (MDA)). Glutamate addition to Sertoli cells before heat stress upregulated the levels of glutamate-cysteine ligase, modifier subunit (Gclm), glutathione synthetase (Gss), thioredoxin (Trx1) and B-cell leukemia/lymphoma 2 (Bcl-2), and the ratio of phosphorylated Akt (protein kinase B)/total Akt. However, it decreased the levels of Bcl2-associated X protein (Bax) and cleaved-caspase 3. Addition of the inhibitor of glutaminase (Gls1), Bptes (Bis-2-(5-phenylacetamido-1,3,4-thiadiazol-2-yl)ethyl sulfide, 30 μM)to Sertoli cells before heat stress reversed these effects. These results inferred that glutamate rescued heat stress-induced apoptosis in Sertoli cells by enhancing activity of antioxidant enzymes and activating the Trx1-Akt pathway. Thus, glutamate supplementation might represent a novel strategy to alleviate the negative effect of heat stress.
Collapse
Affiliation(s)
- Xia-Qing Cai
- Chongqing Key Laboratory of Forage & Herbivore, College of Veterinary Medicine, Southwest University, Beibei, Chongqing, 400715, PR China.
| | - Huan Yang
- Chongqing Key Laboratory of Forage & Herbivore, College of Veterinary Medicine, Southwest University, Beibei, Chongqing, 400715, PR China.
| | - Bing-Qian Liang
- Chongqing Key Laboratory of Forage & Herbivore, College of Veterinary Medicine, Southwest University, Beibei, Chongqing, 400715, PR China.
| | - Cheng-Chen Deng
- Chongqing Key Laboratory of Forage & Herbivore, College of Veterinary Medicine, Southwest University, Beibei, Chongqing, 400715, PR China.
| | - Hong-Yan Xue
- Chongqing Key Laboratory of Forage & Herbivore, College of Veterinary Medicine, Southwest University, Beibei, Chongqing, 400715, PR China.
| | - Jiao-Jiao Zhang
- Chongqing Key Laboratory of Forage & Herbivore, College of Veterinary Medicine, Southwest University, Beibei, Chongqing, 400715, PR China.
| | - Xian-Zhong Wang
- Chongqing Key Laboratory of Forage & Herbivore, College of Veterinary Medicine, Southwest University, Beibei, Chongqing, 400715, PR China.
| |
Collapse
|
22
|
Peng J, Song X, Yu W, Pan Y, Zhang Y, Jian H, He B. The role and mechanism of cinnamaldehyde in cancer. J Food Drug Anal 2024; 32:140-154. [PMID: 38934689 PMCID: PMC11210466 DOI: 10.38212/2224-6614.3502] [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: 11/20/2023] [Accepted: 03/15/2024] [Indexed: 06/28/2024] Open
Abstract
As cancer continues to rise globally, there is growing interest in discovering novel methods for prevention and treatment. Due to the limitations of traditional cancer therapies, there has been a growing emphasis on investigating herbal remedies and exploring their potential synergistic effects when combined with chemotherapy drugs. Cinnamaldehyde, derived from cinnamon, has gained significant attention for its potential role in cancer prevention and treatment. Extensive research has demonstrated that cinnamaldehyde exhibits promising anticancer properties by modulating various cellular processes involved in tumor growth and progression. However, challenges and unanswered questions remain regarding the precise mechanisms for its effective use as an anticancer agent. This article aims to explore the multifaceted effects of cinnamaldehyde on cancer cells and shed light on these existing issues. Cinnamaldehyde has diverse anti-cancer mechanisms, including inducing apoptosis by activating caspases and damaging mitochondrial function, inhibiting tumor angiogenesis, anti-proliferation, anti-inflammatory and antioxidant. In addition, cinnamaldehyde also acts as a reactive oxygen species scavenger, reducing oxidative stress and preventing DNA damage and genomic instability. This article emphasizes the promising therapeutic potential of cinnamaldehyde in cancer treatment and underscores the need for future research to unlock novel mechanisms and strategies for combating cancer. By providing valuable insights into the role and mechanism of cinnamaldehyde in cancer, this comprehensive understanding paves the way for its potential as a novel therapeutic agent. Overall, cinnamaldehyde holds great promise as an anticancer agent, and its comprehensive exploration in this article highlights its potential as a valuable addition to cancer treatment options.
Collapse
Affiliation(s)
- Jiahua Peng
- Department of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Institute of Obstetrics and Gynecology, Nanchang, Jiangxi,
China
| | - Xin Song
- Department of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Institute of Obstetrics and Gynecology, Nanchang, Jiangxi,
China
| | - Wenbin Yu
- Jiangxi Key Laboratory of Bioprocess Engineering, College of Life Sciences, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi,
China
| | - Yuhan Pan
- School of Finance, Shanghai University of Finance and Economics, Shanghai,
China
| | - Yufei Zhang
- Jiangxi Key Laboratory of Bioprocess Engineering, College of Life Sciences, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi,
China
| | - Hui Jian
- Department of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Institute of Obstetrics and Gynecology, Nanchang, Jiangxi,
China
| | - Bin He
- Jiangxi Key Laboratory of Bioprocess Engineering, College of Life Sciences, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi,
China
| |
Collapse
|
23
|
Zhang X, Ge R, Wu J, Cai X, Deng G, Lv J, Ma M, Yu N, Yao L, Peng D. Structural characterization and improves cognitive disorder in ageing mice of a glucomannan from Dendrobium huoshanense. Int J Biol Macromol 2024; 269:131995. [PMID: 38692529 DOI: 10.1016/j.ijbiomac.2024.131995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 04/01/2024] [Accepted: 04/28/2024] [Indexed: 05/03/2024]
Abstract
In the present work, a neutral polysaccharide (DHP-2W) with attenuating cognitive disorder was identified from Dendrobium huoshanense and its structure was clarified. The polysaccharide was successfully purified from D. huoshanense by column chromatography and its activity was evaluated. With a molecular weight of 508.934kDa, this polysaccharide is composed of mannose and glucose at a molar ratio of 75.81: 24.19. Structural characterization revealed that DHP-2W has a backbone consisting of 4)-β-D-Manp-(1 and 4)-β-D-Glcp-(1. In vivo experiments revealed that DHP-2W improved cognitive disorder in D-galactose treated mice and relieved oxidative stress and inflammation. DHP-2W attenuates D-galactose-induced cognitive disorder by inhibiting the BCL2/BAX/CASP3 pathway and activating the AMPK/SIRT pathway, thereby inhibiting apoptosis. Furthermore, DHP-2W had a significant effect on regulating the serum levels of Flavin adenine dinucleotide, Shikimic acid, and Kynurenic acid in aged mice. These, in turn, had a positive impact on AMPK/SIRT1 and BCL2/BAX/CASP3, resulting in protective effects against cognitive disorder.
Collapse
Affiliation(s)
- Xiaoqian Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China; Anhui Academy of Chinese Medicine, Hefei, China.
| | - Ruipeng Ge
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China; Anhui Academy of Chinese Medicine, Hefei, China
| | - Jing Wu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China; Anhui Academy of Chinese Medicine, Hefei, China
| | - Xiao Cai
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China; Anhui Academy of Chinese Medicine, Hefei, China
| | - Guanghui Deng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China; Anhui Academy of Chinese Medicine, Hefei, China
| | - Jiahui Lv
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China; Anhui Academy of Chinese Medicine, Hefei, China
| | - Mengzhen Ma
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China; Anhui Academy of Chinese Medicine, Hefei, China
| | - Nianjun Yu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China; Anhui Academy of Chinese Medicine, Hefei, China; MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China; Anhui Province Key Laboratory for Research and Development of Research & Development of Chinese Medicine, Hefei, China.
| | - Liang Yao
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China; Anhui Academy of Chinese Medicine, Hefei, China.
| | - Daiyin Peng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China; Anhui Academy of Chinese Medicine, Hefei, China; MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China; Anhui Province Key Laboratory for Research and Development of Research & Development of Chinese Medicine, Hefei, China.
| |
Collapse
|
24
|
Golmohammadi M, Meibodi SAA, Al-Hawary SIS, Gupta J, Sapaev IB, Najm MAA, Alwave M, Nazifi M, Rahmani M, Zamanian MY, Moriasi G. Neuroprotective effects of resveratrol on retinal ganglion cells in glaucoma in rodents: A narrative review. Animal Model Exp Med 2024; 7:195-207. [PMID: 38808561 PMCID: PMC11228121 DOI: 10.1002/ame2.12438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 04/22/2024] [Indexed: 05/30/2024] Open
Abstract
Glaucoma, an irreversible optic neuropathy, primarily affects retinal ganglion cells (RGC) and causes vision loss and blindness. The damage to RGCs in glaucoma occurs by various mechanisms, including elevated intraocular pressure, oxidative stress, inflammation, and other neurodegenerative processes. As the disease progresses, the loss of RGCs leads to vision loss. Therefore, protecting RGCs from damage and promoting their survival are important goals in managing glaucoma. In this regard, resveratrol (RES), a polyphenolic phytoalexin, exerts antioxidant effects and slows down the evolution and progression of glaucoma. The present review shows that RES plays a protective role in RGCs in cases of ischemic injury and hypoxia as well as in ErbB2 protein expression in the retina. Additionally, RES plays protective roles in RGCs by promoting cell growth, reducing apoptosis, and decreasing oxidative stress in H2O2-exposed RGCs. RES was also found to inhibit oxidative stress damage in RGCs and suppress the activation of mitogen-activated protein kinase signaling pathways. RES could alleviate retinal function impairment by suppressing the hypoxia-inducible factor-1 alpha/vascular endothelial growth factor and p38/p53 axes while stimulating the PI3K/Akt pathway. Therefore, RES might exert potential therapeutic effects for managing glaucoma by protecting RGCs from damage and promoting their survival.
Collapse
Affiliation(s)
- Maryam Golmohammadi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | - Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Ibrohim B Sapaev
- Tashkent Institute of Irrigation and Agricultural Mechanization Engineers, Tashkent, Uzbekistan
- New Uzbekistan University, Tashkent, Uzbekistan
| | - Mazin A A Najm
- Pharmaceutical Chemistry Department, College of Pharmacy, Al-Ayen University, Thi-Qar, Iraq
| | - Marim Alwave
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | - Mozhgan Nazifi
- Department of Neurology, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammadreza Rahmani
- Physiology-Pharmacology Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Physiology and Pharmacology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Yasin Zamanian
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Gervason Moriasi
- Department of Medical Biochemistry, School of Medicine, College of Health Sciences, Mount Kenya University, Thika, Kenya
| |
Collapse
|
25
|
Li G, Chen L, Bai H, Zhang L, Wang J, Li W. Depletion of squalene epoxidase in synergy with glutathione peroxidase 4 inhibitor RSL3 overcomes oxidative stress resistance in lung squamous cell carcinoma. PRECISION CLINICAL MEDICINE 2024; 7:pbae011. [PMID: 38779359 PMCID: PMC11109822 DOI: 10.1093/pcmedi/pbae011] [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: 03/12/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024] Open
Abstract
Background Lung squamous cell carcinoma (LUSC) lacks effective targeted therapies and has a poor prognosis. Disruption of squalene epoxidase (SQLE) has been implicated in metabolic disorders and cancer. However, the role of SQLE as a monooxygenase involved in oxidative stress remains unclear. Methods We analyzed the expression and prognosis of lung adenocarcinoma (LUAD) and LUSC samples from GEO and TCGA databases. The proliferative activity of the tumors after intervention of SQLE was verified by cell and animal experiments. JC-1 assay, flow cytometry, and Western blot were used to show changes in apoptosis after intervention of SQLE. Flow cytometry and fluorescence assay of ROS levels were used to indicate oxidative stress status. Results We investigated the unique role of SQLE expression in the diagnosis and prognosis prediction of LUSC. Knockdown of SQLE or treatment with the SQLE inhibitor terbinafine can suppress the proliferation of LUSC cells by inducing apoptosis and reactive oxygen species accumulation. However, depletion of SQLE also results in the impairment of lipid peroxidation and ferroptosis resistance such as upregulation of glutathione peroxidase 4. Therefore, prevention of SQLE in synergy with glutathione peroxidase 4 inhibitor RSL3 effectively mitigates the proliferation and growth of LUSC. Conclusion Our study indicates that the low expression of SQLE employs adaptive survival through regulating the balance of apoptosis and ferroptosis resistance. In future, the combinational therapy of targeting SQLE and ferroptosis could be a promising approach in treating LUSC.
Collapse
Affiliation(s)
- Guo Li
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing 100021, China
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lu Chen
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hua Bai
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing 100021, China
| | - Li Zhang
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Chengdu 610041, China
| | - Jie Wang
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing 100021, China
| | - Weimin Li
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Chengdu 610041, China
- The Research Units of West China, Chinese Academy of Medical Sciences, West China Hospital, Chengdu 610041, China
| |
Collapse
|
26
|
Lu Z, Wang Y, Liu C, Fan H. Efficacy and Safety of Asparagusic Acid against Echinococcus multilocularis In Vitro and in a Murine Infection Model. Trop Med Infect Dis 2024; 9:110. [PMID: 38787043 PMCID: PMC11126102 DOI: 10.3390/tropicalmed9050110] [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: 04/02/2024] [Revised: 05/05/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
Alveolar echinococcosis (AE) stands as a perilous zoonotic affliction caused by the larvae of Echinococcus multilocularis. There is an imperative need to explore novel therapeutic agents or lead compounds for the treatment of AE. Asparagusic acid, characterized by its low toxicity and possessing antimicrobial, antioxidant, and anti-parasitic attributes, emerges as a promising candidate. The aim of this study was to investigate the in vivo and in vitro efficacy of asparagusic acid against E. multilocularis. Morphological observations, scanning electron microscopy, ROS assays, mitochondrial membrane potential assays, and Western blot were used to evaluate the in vitro effects of asparagusic acid on protoscoleces. The effects of asparagusic acid on vesicles were assessed via PGI release, γ-GGT release, and transmission electron microscopy observations. CellTiter-Glo assays, Caspase3 activity assays, flow cytometry, and Western blot were used for an evaluation of the effect of asparaginic acid on the proliferation and apoptosis of germinal cells. The in vivo efficacy of asparagusic acid was evaluated in a murine AE model. Asparagusic acid exhibited a pronounced killing effect on the protoscoleces post-treatment. Following an intervention with asparagusic acid, there was an increase in ROS levels and a decline in mitochondrial membrane potential in the protoscolex. Moreover, asparagusic acid treatment resulted in the upregulation of PGI and γ-GGT release in metacestode vesicles, concomitant with the inhibition of germinal cell viability. Furthermore, asparagusic acid led to an enhanced relative expression of Caspase3 in the culture supernatant of both the protoscoleces and germinal cells, accompanied by an increase in the proportion of apoptotic germinal cells. Notably, asparagusic acid induced an augmentation in Bax and Caspase3 protein expression while reducing Bcl2 protein expression in both the protoscoleces and germinal cells. In vitro cytotoxicity assessments demonstrated the low toxicity of asparagusic acid towards normal human hepatocytes and HFF cells. Additionally, in vivo experiments revealed that asparagusic acid administration at doses of 10 mg/kg and 40 mg/kg significantly reduced metacestode wet weight. A histopathological analysis displayed the disruption of the germinal layer structure within lesions post-asparagusic acid treatment, alongside the preservation of laminated layer structures. Transmission electron microscopy further revealed mitochondrial swelling and heightened cell necrosis subsequent to the asparagusic acid treatment. Furthermore, asparagusic acid promoted Caspase3 and Bax protein expression while decreasing Bcl2 protein expression in perilesional tissues. Subsequently, it inhibited the expression of Ki67, MMP2, and MMP9 proteins in the perilesional tissues and curbed the activation of the PI3K/Akt signaling pathway within the lesion-host microenvironmental tissues. Asparagusic acid demonstrated a pronounced killing effect on E. multilocularis, suggesting its potential as a promising therapeutic agent for the management of AE.
Collapse
Affiliation(s)
- Zhuanhong Lu
- School of Clinical Medicine, Qinghai University Affiliated Hospital, Xining 810001, China; (Z.L.); (Y.W.)
| | - Yating Wang
- School of Clinical Medicine, Qinghai University Affiliated Hospital, Xining 810001, China; (Z.L.); (Y.W.)
| | - Chuanchuan Liu
- School of Clinical Medicine, Qinghai University Affiliated Hospital, Xining 810001, China; (Z.L.); (Y.W.)
- Qinghai Key Laboratory of Echinococcosis Research, Xining 810001, China
| | - Haining Fan
- School of Clinical Medicine, Qinghai University Affiliated Hospital, Xining 810001, China; (Z.L.); (Y.W.)
- Qinghai Key Laboratory of Echinococcosis Research, Xining 810001, China
| |
Collapse
|
27
|
Li X, Liu H, Wang Y, Crabbe MJC, Wang L, Ma W, Ren Z. Preparation of a novel metallothionein-AuNP composite material by genetic modification and AuS covalent combination. Int J Biol Macromol 2024; 262:129960. [PMID: 38325687 DOI: 10.1016/j.ijbiomac.2024.129960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 01/22/2024] [Accepted: 02/01/2024] [Indexed: 02/09/2024]
Abstract
Metallothionein (MTs) can be used in the prevention and treatment of tumors and diabetes due to its antioxidant properties. However, it is necessary to solve its non-transmembrane properties and further improve its antioxidant activity, increase its fluorescence visualization and enhance its stability to meet practical applications in the biomedical field. Here, we report the preparation of a novel metallothionein-AuNP composite material with high transmembrane ability, fluorescence visualization, antioxidant activity, and stability by genetic modification (introducing transduction peptide TAT, fluorescence tag GFP and increasing sulfydryl groups) and immobilization technology (covalently bonding with AuNPs). The transmembrane activity of modified proteins was verified by immunofluorescence. Increasing the sulfhydryl content within a certain range can enhance the antioxidant activity of the protein. In addition, GFP were used to further simplify the imaging of the metallothionein-AuNP composite in cells. XPS results indicated that AuNPs can immobilize metallothionein through AuS covalent bonds. TGA characterization and degradation experiments showed that thermal and degradation stability of the immobilized material was significantly improved. This work provides new ideas to construct metallothionein composites with high transmembrane ability, antioxidant activity, fluorescence visualization and stability to meet novel applications in the biomedical field.
Collapse
Affiliation(s)
- Xuefen Li
- School of Life Science, Shanxi University, Taiyuan 030006, PR China
| | - Hui Liu
- School of Life Science, Shanxi University, Taiyuan 030006, PR China
| | - Yuxia Wang
- School of Life Science, Shanxi University, Taiyuan 030006, PR China
| | - M James C Crabbe
- School of Life Science, Shanxi University, Taiyuan 030006, PR China; Wolfson College, University of Oxford, Oxford OX2 6UD, UK; Institute of Biomedical and Environmental Science & Technology, School of Life Sciences, Faculty of Creative Arts, Technologies and Science, University of Bedfordshire, University Square, Luton LU1 3JU, UK
| | - Lan Wang
- School of Life Science, Shanxi University, Taiyuan 030006, PR China
| | - Wenli Ma
- School of Life Science, Shanxi University, Taiyuan 030006, PR China.
| | - Zhumei Ren
- School of Life Science, Shanxi University, Taiyuan 030006, PR China.
| |
Collapse
|
28
|
Hartley B, Bassiouni W, Roczkowsky A, Fahlman R, Schulz R, Julien O. Data-Independent Acquisition Proteomics and N-Terminomics Methods Reveal Alterations in Mitochondrial Function and Metabolism in Ischemic-Reperfused Hearts. J Proteome Res 2024; 23:844-856. [PMID: 38264990 PMCID: PMC10846531 DOI: 10.1021/acs.jproteome.3c00754] [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: 11/11/2023] [Revised: 01/06/2024] [Accepted: 01/10/2024] [Indexed: 01/25/2024]
Abstract
Myocardial ischemia-reperfusion (IR) (stunning) injury triggers changes in the proteome and degradome of the heart. Here, we utilize quantitative proteomics and comprehensive degradomics to investigate the molecular mechanisms of IR injury in isolated rat hearts. The control group underwent aerobic perfusion, while the IR injury group underwent 20 min of ischemia and 30 min of reperfusion to induce a stunning injury. As MMP-2 activation has been shown to contribute to myocardial injury, hearts also underwent IR injury with ARP-100, an MMP-2-preferring inhibitor, to dissect the contribution of MMP-2 to IR injury. Using data-independent acquisition (DIA) and mass spectroscopy, we quantified 4468 proteins in ventricular extracts, whereby 447 proteins showed significant alterations among the three groups. We then used subtiligase-mediated N-terminomic labeling to identify more than a hundred specific cleavage sites. Among these protease substrates, 15 were identified following IR injury. We identified alterations in numerous proteins involved in mitochondrial function and metabolism following IR injury. Our findings provide valuable insights into the biochemical mechanisms of myocardial IR injury, suggesting alterations in reactive oxygen/nitrogen species handling and generation, fatty acid metabolism, mitochondrial function and metabolism, and cardiomyocyte contraction.
Collapse
Affiliation(s)
- Bridgette Hartley
- Department
of Biochemistry, University of Alberta, Edmonton T6G 2H7, Canada
| | - Wesam Bassiouni
- Department
of Pharmacology, University of Alberta, Edmonton T6G 2S2, Canada
| | - Andrej Roczkowsky
- Department
of Pharmacology, University of Alberta, Edmonton T6G 2S2, Canada
| | - Richard Fahlman
- Department
of Biochemistry, University of Alberta, Edmonton T6G 2H7, Canada
| | - Richard Schulz
- Department
of Pharmacology, University of Alberta, Edmonton T6G 2S2, Canada
- Department
of Pediatrics, University of Alberta, Edmonton T6G 2S2, Canada
| | - Olivier Julien
- Department
of Biochemistry, University of Alberta, Edmonton T6G 2H7, Canada
| |
Collapse
|
29
|
Ziegler AR, Dufour A, Scott NE, Edgington-Mitchell LE. Ion Mobility-Based Enrichment-Free N-Terminomics Analysis Reveals Novel Legumain Substrates in Murine Spleen. Mol Cell Proteomics 2024; 23:100714. [PMID: 38199506 PMCID: PMC10862022 DOI: 10.1016/j.mcpro.2024.100714] [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: 07/28/2023] [Revised: 12/19/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024] Open
Abstract
Aberrant levels of the asparaginyl endopeptidase legumain have been linked to inflammation, neurodegeneration, and cancer, yet our understanding of this protease is incomplete. Systematic attempts to identify legumain substrates have been previously confined to in vitro studies, which fail to mirror physiological conditions and obscure biologically relevant cleavage events. Using high-field asymmetric waveform ion mobility spectrometry (FAIMS), we developed a streamlined approach for proteome and N-terminome analyses without the need for N-termini enrichment. Compared to unfractionated proteomic analysis, we demonstrate FAIMS fractionation improves N-termini identification by >2.5 fold, resulting in the identification of >2882 unique N-termini from limited sample amounts. In murine spleens, this approach identifies 6366 proteins and 2528 unique N-termini, with 235 cleavage events enriched in WT compared to legumain-deficient spleens. Among these, 119 neo-N-termini arose from asparaginyl endopeptidase activities, representing novel putative physiological legumain substrates. The direct cleavage of selected substrates by legumain was confirmed using in vitro assays, providing support for the existence of physiologically relevant extra-lysosomal legumain activity. Combined, these data shed critical light on the functions of legumain and demonstrate the utility of FAIMS as an accessible method to improve depth and quality of N-terminomics studies.
Collapse
Affiliation(s)
- Alexander R Ziegler
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Antoine Dufour
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada; McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, Alberta, Canada
| | - Nichollas E Scott
- Department of Microbiology and Immunology, Peter Doherty Institute, The University of Melbourne, Parkville, Victoria, Australia.
| | - Laura E Edgington-Mitchell
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
30
|
Wang H, Lian X, Wang K, Wang S. WWP2 binds to NKRF, enhances the NF-κB signaling, and promotes malignant phenotypes of acute myeloid leukemia cells. Biochem Cell Biol 2024; 102:85-95. [PMID: 37921219 DOI: 10.1139/bcb-2022-0360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023] Open
Abstract
Acute myeloid leukemia (AML) is one of the hematological malignancies with a high recurrence rate. WW domain-containing E3 ubiquitin protein ligase 2 (WWP2) is identified as a pivotal regulator of tumor progression. This study aimed to assess the possible role of WWP2 in AML. Analysis of the GEPIA database indicated an elevated WWP2 expression in AML. We established stable WWP2-overexpressed or WWP2-silenced cells using lentivirus loaded with cDNA encoding WWP2 mRNA or shRNA targeting WWP2. Notably, WWP2 overexpression facilitated cell proliferation and cell cycle progression, which was manifested as the increase of colony formation number, S-phase percentage and cell cycle related protein levels. As observed, WWP2 knockdown presented opposite effects, leading to inhibition of tumorigenicity. Strikingly, WWP2 knockdown induced apoptosis, accompanied by upregulation of pro-apoptosis proteins cleaved caspase-9, Bax and cleaved caspase-3 and downregulation of anti-apoptosis protein Bcl-2. Functionally, we further confirmed that WWP2 overexpression enhanced the NF-κB signaling and upregulated the levels of downstream genes, which may contribute to aggravating the development of AML. More importantly, by co-immunoprecipitation assay, we verified that WWP2 bound to NF-κB-repressing factor (NKRF) and promoted NKRF ubiquitylation. Dramatically, NKRF overexpression abolished the role of WWP2 in facilitating the process of AML. Overall, our observations confirm that WWP2 exerts a critical role in the tumorigenicity of AML, and NKRF is regarded as an essential factor in the WWP2-mediated AML progression. WWP2 may be proposed as a promising target of AML.
Collapse
Affiliation(s)
- Hongjia Wang
- Department of Hematology, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xin Lian
- Department of Hematology, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Kexin Wang
- Department of Hematology, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Shuye Wang
- Department of Hematology, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
31
|
Baena-Lopez LA, Wang L, Wendler F. Cellular stress management by caspases. Curr Opin Cell Biol 2024; 86:102314. [PMID: 38215516 DOI: 10.1016/j.ceb.2023.102314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/14/2023] [Accepted: 12/18/2023] [Indexed: 01/14/2024]
Abstract
Cellular stress plays a pivotal role in the onset of numerous human diseases. Consequently, the removal of dysfunctional cells, which undergo excessive stress-induced damage via various cell death pathways, including apoptosis, is essential for maintaining organ integrity and function. The evolutionarily conserved family of cysteine-aspartic-proteases, known as caspases, has been a key player in orchestrating apoptosis. However, recent research has unveiled the capability of these enzymes to govern fundamental cellular processes without triggering cell death. Remarkably, some of these non-lethal functions of caspases may contribute to restoring cellular equilibrium in stressed cells. This manuscript discusses how caspases can function as cellular stress managers and their potential impact on human health and disease. Additionally, it sheds light on the limitations of caspase-based therapies, given our still incomplete understanding of the biology of these enzymes, particularly in non-apoptotic contexts.
Collapse
Affiliation(s)
| | - Li Wang
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX13RE, UK
| | - Franz Wendler
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX13RE, UK. https://twitter.com/wendlerfranz
| |
Collapse
|
32
|
Gomez-Cardona E, Eskandari-Sedighi G, Fahlman R, Westaway D, Julien O. Application of N-Terminal Labeling Methods Provide Novel Insights into Endoproteolysis of the Prion Protein in Vivo. ACS Chem Neurosci 2024; 15:134-146. [PMID: 38095594 PMCID: PMC10768724 DOI: 10.1021/acschemneuro.3c00533] [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: 08/12/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 01/04/2024] Open
Abstract
Alternative α- and β-cleavage events in the cellular prion protein (PrPC) central region generate fragments with distinct biochemical features that affect prion disease pathogenesis, but the assignment of precise cleavage positions has proven challenging. Exploiting mouse transgenic models expressing wild-type (WT) PrPC and an octarepeat region mutant allele (S3) with increased β-fragmentation, cleavage sites were defined using LC-MS/MS in conjunction with N-terminal enzymatic labeling and chemical in-gel acetylation. Our studies profile the net proteolytic repertoire of the adult brain, as deduced from defining hundreds of proteolytic events in other proteins, and position individual cleavage events in PrPC α- and β-target areas imputed from earlier, lower resolution methods; these latter analyses established site heterogeneity, with six cleavage sites positioned in the β-cleavage region of WT PrPC and nine positions for S3 PrPC. Regarding α-cleavage, aside from reported N-termini at His110 and Val111, we identified a total of five shorter fragments in the brain of both mice lines. We infer that aminopeptidase activity in the brain could contribute to the ragged N-termini observed around PrPC's α- and β-cleavage sites, with this work providing a point of departure for further in vivo studies of brain proteases.
Collapse
Affiliation(s)
- Erik Gomez-Cardona
- Department
of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Ghazaleh Eskandari-Sedighi
- Department
of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
- Center
for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta T6G 2M8, Canada
| | - Richard Fahlman
- Department
of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - David Westaway
- Department
of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
- Center
for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta T6G 2M8, Canada
- Department
of Medicine, University of Alberta, Edmonton, Alberta T6G 2G3, Canada
| | - Olivier Julien
- Department
of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| |
Collapse
|
33
|
Zhai Z, Mu T, Zhao L, Zhu D, Zhong X, Li Y, Liang C, Li W, Zhou Q. Stachydrine represses the proliferation and enhances cell cycle arrest and apoptosis of breast cancer cells via PLA2G2A/DCN axis. Chem Biol Drug Des 2024; 103:e14429. [PMID: 38230769 DOI: 10.1111/cbdd.14429] [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: 08/14/2023] [Revised: 12/03/2023] [Accepted: 12/11/2023] [Indexed: 01/18/2024]
Abstract
Considering the therapeutic efficacy of Stachydrine on breast cancer (BC), this study aims to decipher the relevant mechanism. The effects of Stachydrine on BC cell viability, proliferation and apoptosis were firstly investigated. Then, Bioinformatics was applied to sort out the candidate interacting with Stachydrine as well as its expression and downstream target in BC. Relative expressions of genes of interest as well as proliferation- and apoptosis-related factors in BC cells were quantified through quantitative reverse-transcription PCR and western blot as appropriate. As a result, Stachydrine inhibited the proliferation, down-regulated the expressions of proliferating cell nuclear antigen and CyclinD1, enhanced cell cycle arrest and apoptosis, and up-regulated the levels of Cleaved caspase-3 and Cleaved caspase-9 in BC cells. Phospholipase A2 Group IIA (PLA2G2A) was predicted as the candidate interacting with Stachydrine and to be lowly expressed in BC. PLA2G2A silencing reversed while PLA2G2A overexpression reinforced the effects of Stachydrine. Decorin (DCN) was the downstream target of PLA2G2A and also lowly expressed in BC. PLA2G2A silencing counteracted yet overexpressed PLA2G2A strengthened the promoting effects of Stachydrine on DCN level. Collectively, Stachydrine inhibits the growth of BC cells to promote cell cycle arrest and apoptosis via PLA2G2A/DCN axis.
Collapse
Affiliation(s)
- Zhen Zhai
- Mammary Department, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Tianlong Mu
- Pathology Department, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Lina Zhao
- Mammary Department, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Dongsheng Zhu
- Mammary Department, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Xin Zhong
- Mammary Department, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Yiliang Li
- Mammary Department, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Chen Liang
- Mammary Department, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Wei Li
- Mammary Department, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Qingyuan Zhou
- Mammary Department, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
34
|
Sever AIM, Alderson TR, Rennella E, Aramini JM, Liu ZH, Harkness RW, Kay LE. Activation of caspase-9 on the apoptosome as studied by methyl-TROSY NMR. Proc Natl Acad Sci U S A 2023; 120:e2310944120. [PMID: 38085782 PMCID: PMC10743466 DOI: 10.1073/pnas.2310944120] [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: 06/28/2023] [Accepted: 10/23/2023] [Indexed: 12/18/2023] Open
Abstract
Mitochondrial apoptotic signaling cascades lead to the formation of the apoptosome, a 1.1-MDa heptameric protein scaffold that recruits and activates the caspase-9 protease. Once activated, caspase-9 cleaves and activates downstream effector caspases, triggering the onset of cell death through caspase-mediated proteolysis of cellular proteins. Failure to activate caspase-9 enables the evasion of programmed cell death, which occurs in various forms of cancer. Despite the critical apoptotic function of caspase-9, the structural mechanism by which it is activated on the apoptosome has remained elusive. Here, we used a combination of methyl-transverse relaxation-optimized NMR spectroscopy, protein engineering, and biochemical assays to study the activation of caspase-9 bound to the apoptosome. In the absence of peptide substrate, we observed that both caspase-9 and its isolated protease domain (PD) only very weakly dimerize with dissociation constants in the millimolar range. Methyl-NMR spectra of isotope-labeled caspase-9, within the 1.3-MDa native apoptosome complex or an engineered 480-kDa apoptosome mimic, reveal that the caspase-9 PD remains monomeric after recruitment to the scaffold. Binding to the apoptosome, therefore, organizes caspase-9 PDs so that they can rapidly and extensively dimerize only when substrate is present, providing an important layer in the regulation of caspase-9 activation. Our work highlights the unique role of NMR spectroscopy to structurally characterize protein domains that are flexibly tethered to large scaffolds, even in cases where the molecular targets are in excess of 1 MDa, as in the present example.
Collapse
Affiliation(s)
- Alexander I. M. Sever
- Department of Chemistry, University of Toronto, Toronto, ONM5S 3H6, Canada
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ONM5G 0A4, Canada
| | - T. Reid Alderson
- Department of Chemistry, University of Toronto, Toronto, ONM5S 3H6, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ONM5S 1A8, Canada
- Department of Biochemistry, University of Toronto, Toronto, ONM5S 1A8, Canada
| | - Enrico Rennella
- Department of Chemistry, University of Toronto, Toronto, ONM5S 3H6, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ONM5S 1A8, Canada
- Department of Biochemistry, University of Toronto, Toronto, ONM5S 1A8, Canada
| | - James M. Aramini
- Department of Chemistry, University of Toronto, Toronto, ONM5S 3H6, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ONM5S 1A8, Canada
- Department of Biochemistry, University of Toronto, Toronto, ONM5S 1A8, Canada
| | - Zi Hao Liu
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ONM5G 0A4, Canada
- Department of Biochemistry, University of Toronto, Toronto, ONM5S 1A8, Canada
| | - Robert W. Harkness
- Department of Chemistry, University of Toronto, Toronto, ONM5S 3H6, Canada
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ONM5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ONM5S 1A8, Canada
- Department of Biochemistry, University of Toronto, Toronto, ONM5S 1A8, Canada
| | - Lewis E. Kay
- Department of Chemistry, University of Toronto, Toronto, ONM5S 3H6, Canada
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ONM5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ONM5S 1A8, Canada
- Department of Biochemistry, University of Toronto, Toronto, ONM5S 1A8, Canada
| |
Collapse
|
35
|
Wang L, Xu Y, Zhao X, Zhu X, He X, Sun A, Zhuang G. Antagonistic effects of N-acetylcysteine on lead-induced apoptosis and oxidative stress in chicken embryo fibroblast cells. Heliyon 2023; 9:e21847. [PMID: 38034812 PMCID: PMC10682149 DOI: 10.1016/j.heliyon.2023.e21847] [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: 05/21/2023] [Revised: 10/30/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
Lead (Pb) is a heavy metal that can have harmful effects on the environment, which has severe cytotoxicity in many animal tissues. N-acetylcysteine (NAC) has antioxidant activity, reducing lead-induced oxidative stress and apoptosis, but its role in chicken cells is unknown. The current study explored the antagonistic effect of NAC on lead-induced apoptosis and oxidative stress in chicken embryo fibroblast (CEF). In this study, CEF was used as a model to measure the cytotoxic effects of lead nitrate at different concentrations, demonstrating a dose-dependent effect on CEF activity. Employing inverted microscopy, the investigation of morphological alterations in CEF cells was conducted. Fluorescence staining methodology enabled the assessment of reactive oxygen species (ROS) levels within CEF cells. Moreover, an enzyme-linked immunosorbent assay was utilized to detect the presence of oxidative damage indicators encompassing superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), catalase (CAT) activity, malondialdehyde (MDA) content, and total antioxidant capacity (T-AOC) within CEF cells. Furthermore, the determination of the apoptosis rate of CEF cells was accomplished through the utilization of the Hoechst 33258 staining method in combination with the Annexin V-FITC dual staining method. By using RT-qPCR for detection, lead treatment increased expression of pro-apoptotic genes, caspase-3, and caspase-9, and reduced expression of anti-apoptotic genes, Bcl-2, and BI-1. Reduced antioxidant capacity was shown by increased ROS and MDA levels in CEF cells after lead treatment. The results showed that NAC inhibited the expression of caspase-3 and caspase-9 in lead-treated CEF cells, while NAC had a certain inhibitory effect on the relative expression of Bcl-2 and BI-1 mRNA in lead-induced CEF cells. NAC significantly reduced lead-induced oxidative damage and apoptosis. Overall, our results demonstrate a novel protective effect of NAC against lead-induced injury in chicken cells, providing a theoretical basis for future investigations of drugs that are effective in preventing lead poisoning in animals.
Collapse
Affiliation(s)
- Lele Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, Henan, People's Republic of China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, Henan, People's Republic of China
| | - Yijie Xu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, Henan, People's Republic of China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, Henan, People's Republic of China
| | - Xuyang Zhao
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, Henan, People's Republic of China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, Henan, People's Republic of China
| | - Xiaojing Zhu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, Henan, People's Republic of China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, Henan, People's Republic of China
| | - Xiuyuan He
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, Henan, People's Republic of China
| | - Aijun Sun
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, Henan, People's Republic of China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, Henan, People's Republic of China
| | - Guoqing Zhuang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, Henan, People's Republic of China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, Henan, People's Republic of China
| |
Collapse
|
36
|
Yin Q, Yang H, Fang L, Wu Q, Gao S, Wu Y, Zhou L. Fibroblast growth factor 23 regulates hypoxia‑induced osteoblast apoptosis through the autophagy‑signaling pathway. Mol Med Rep 2023; 28:199. [PMID: 37711045 PMCID: PMC10540001 DOI: 10.3892/mmr.2023.13086] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 08/18/2023] [Indexed: 09/16/2023] Open
Abstract
Hypoxia can lead to programmed osteoblast death. Prevention of osteoblast apoptosis caused by hypoxia is of great significance in the study of the occurrence and development of bone necrosis. The present study aimed to investigate the effects and mechanism of fibroblast growth factor 23 (FGF23) on hypoxia‑induced apoptosis in primary osteoblasts and MC3T3‑E1 cells osteoblasts. Cells were transfected with a plasmid carrying the FGF23 gene and a cell model of hypoxia‑induced apoptosis was established. FGF23 mRNA levels were measured using reverse transcription‑quantitative (RT‑q) PCR and western blotting was used to assess protein levels. Apoptosis was analyzed by MTT assay, fluorescein diacetate and ethidium bromide staining, flow cytometry and RT‑qPCR and western blotting were used to verify the mRNA and protein levels of apoptosis‑ and autophagy‑related gene mRNA. The targeted relationship between miR‑17‑5p and FGF23 was confirmed using the StarBase database, TargetScan database and a luciferase reporter assay. FGF23 decreased cell survival and increased the rate of apoptosis. The mRNA and protein expression of the pro‑apoptotic genes Bax and caspases 3 and 9 increased, whereas that of the anti‑apoptotic Bcl‑2 decreased. The expressions of the autophagy‑associated proteins beclin‑1, light chain 3‑II (LC3‑II) and the LC3‑II/LC3‑I ratio were significantly increased. In addition, a luciferase reporter assay confirmed that FGF23 directly regulated micro RNA (miR)‑17‑5p. The effects of FGF23 silencing were reversed by miR‑17‑5p inhibition. FGF23 may regulate hypoxia‑induced osteoblast apoptosis by targeting miR‑17‑5p through the autophagy‑signaling pathway. This provides a rationale for FGF23 as a potential therapeutic target for osteonecrosis of the femoral head.
Collapse
Affiliation(s)
- Qipu Yin
- Institute of Sports Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong 271016, P.R. China
| | - Hongxia Yang
- School of Nursing, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong 271016, P.R. China
| | - Lun Fang
- Institute of Sports Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong 271016, P.R. China
| | - Qi Wu
- Department of Rehabilitation and Physiotherapy, Taian Maternal and Child Health Hospital, Taian, Shandong 271000, P.R. China
| | - Shan Gao
- School of Pharmaceutical Science, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong 271016, P.R. China
| | - Yadi Wu
- Institute of Sports Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong 271016, P.R. China
| | - Lu Zhou
- Institute of Sports Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong 271016, P.R. China
| |
Collapse
|
37
|
Jiang Q, Wang H, Qiao Z, Hou Y, Sui Z, Zhao B, Liang Z, Jiang B, Zhang Y, Zhang L. Metal organic layers enabled cell surface engineering coupling biomembrane fusion for dynamic membrane proteome profiling. Chem Sci 2023; 14:11727-11736. [PMID: 37920345 PMCID: PMC10619618 DOI: 10.1039/d3sc03725h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 09/30/2023] [Indexed: 11/04/2023] Open
Abstract
Systematically dissecting the highly dynamic and tightly communicating membrane proteome of living cells is essential for the system-level understanding of fundamental cellular processes and intricate relationship between membrane-bound organelles constructed through membrane traffic. While extensive efforts have been made to enrich membrane proteins, their comprehensive analysis with high selectivity and deep coverage remains a challenge, especially at the living cell state. To address this problem, we developed the cell surface engineering coupling biomembrane fusion method to map the whole membrane proteome from the plasma membrane to various organelle membranes taking advantage of the exquisite interaction between two-dimensional metal-organic layers and phospholipid bilayers on the membrane. This approach, which bypassed conventional biochemical fractionation and ultracentrifugation, facilitated the enrichment of membrane proteins in their native phospholipid bilayer environment, helping to map the membrane proteome with a specificity of 77% and realizing the deep coverage of the HeLa membrane proteome (5087 membrane proteins). Furthermore, membrane N-phosphoproteome was profiled by integrating the N-phosphoproteome analysis strategy, and the dynamic membrane proteome during apoptosis was deciphered in combination with quantitative proteomics. The features of membrane protein N-phosphorylation modifications and many differential proteins during apoptosis associated with mitochondrial dynamics and ER homeostasis were found. The method provided a simple and robust strategy for efficient analysis of membrane proteome, offered a reliable platform for research on membrane-related cell dynamic events and expanded the application of metal-organic layers.
Collapse
Affiliation(s)
- Qianqian Jiang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R & A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences Dalian 116023 China
- University of Chinese Academy of Sciences Beijing 100049 China
| | - He Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R & A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences Dalian 116023 China
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Zichun Qiao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R & A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences Dalian 116023 China
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Yutong Hou
- Dalian Medical University Dalian 116044 China
| | - Zhigang Sui
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R & A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences Dalian 116023 China
| | - Baofeng Zhao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R & A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences Dalian 116023 China
| | - Zhen Liang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R & A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences Dalian 116023 China
| | - Bo Jiang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R & A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences Dalian 116023 China
| | - Yukui Zhang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R & A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences Dalian 116023 China
| | - Lihua Zhang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R & A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences Dalian 116023 China
| |
Collapse
|
38
|
Sklifasovskaya AP, Blagonravov M, Ryabinina A, Goryachev V, Syatkin S, Chibisov S, Akhmetova K, Prokofiev D, Agostinelli E. The role of heat shock proteins in the pathogenesis of heart failure (Review). Int J Mol Med 2023; 52:106. [PMID: 37772383 PMCID: PMC10558216 DOI: 10.3892/ijmm.2023.5309] [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: 07/11/2023] [Accepted: 09/11/2023] [Indexed: 09/30/2023] Open
Abstract
The influence of heat shock proteins (HSPs) on protein quality control systems in cardiomyocytes is currently under investigation. The effect of HSPs on the regulated cell death of cardiomyocytes (CMCs) is of great importance, since they play a major role in the implementation of compensatory and adaptive mechanisms in the event of cardiac damage. HSPs mediate a number of mechanisms that activate the apoptotic cascade, playing both pro‑ and anti‑apoptotic roles depending on their location in the cell. Another type of cell death, autophagy, can in some cases lead to cell death, while in other situations it acts as a cell survival mechanism. The present review considered the characteristics of the expression of HSPs of different molecular weights in CMCs in myocardial damage caused by heart failure, as well as their role in the realization of certain types of regulated cell death.
Collapse
Affiliation(s)
| | | | - Anna Ryabinina
- Institute of Medicine, RUDN University, 117198 Moscow, Russia, Italy
| | | | - Sergey Syatkin
- Institute of Medicine, RUDN University, 117198 Moscow, Russia, Italy
| | - Sergey Chibisov
- Institute of Medicine, RUDN University, 117198 Moscow, Russia, Italy
| | - Karina Akhmetova
- Institute of Medicine, RUDN University, 117198 Moscow, Russia, Italy
| | - Daniil Prokofiev
- Institute of Medicine, RUDN University, 117198 Moscow, Russia, Italy
| | - Enzo Agostinelli
- Department of Sensory Organs, Faculty of Medicine and Dentistry, Sapienza University of Rome, University Hospital Policlinico Umberto I, I-00161 Rome, Italy
- International Polyamines Foundation, ETS-ONLUS, I-00159 Rome, Italy
| |
Collapse
|
39
|
Campesi I, Capobianco G, Cano A, Lodde V, Cruciani S, Maioli M, Sotgiu G, Idda ML, Puci MV, Ruoppolo M, Costanzo M, Caterino M, Cambosu F, Montella A, Franconi F. Stratification of Amniotic Fluid Cells and Amniotic Fluid by Sex Opens Up New Perspectives on Fetal Health. Biomedicines 2023; 11:2830. [PMID: 37893203 PMCID: PMC10604128 DOI: 10.3390/biomedicines11102830] [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: 08/28/2023] [Revised: 09/29/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Amniotic fluid is essential for fetus wellbeing and is used to monitor pregnancy and predict fetal outcomes. Sex affects health and medicine from the beginning of life, but knowledge of its influence on cell-depleted amniotic fluid (AF) and amniotic fluid cells (AFCs) is still neglected. We evaluated sex-related differences in AF and in AFCs to extend personalized medicine to prenatal life. AFCs and AF were obtained from healthy Caucasian pregnant women who underwent amniocentesis at the 16th-18th week of gestation for advanced maternal age. In the AF, inflammation biomarkers (TNFα, IL6, IL8, and IL4), malondialdehyde, nitrites, amino acids, and acylcarnitines were measured. Estrogen receptors and cell fate (autophagy, apoptosis, senescence) were measured in AFCs. TNFα, IL8, and IL4 were higher in female AF, whereas IL6, nitrites, and MDA were similar. Valine was higher in male AF, whereas several acylcarnitines were sexually different, suggesting a mitochondrial involvement in establishing sex differences. Female AFCs displayed higher expression of ERα protein and a higher ERα/ERβ ratio. The ratio of LC3II/I, an index of autophagy, was higher in female AFCs, while LC3 gene was similar in both sexes. No significant sex differences were found in the expression of the lysosomal protein LAMP1, while p62 was higher in male AFCs. LAMP1 gene was upregulated in male AFCs, while p62 gene was upregulated in female ones. Finally, caspase 9 activity and senescence linked to telomeres were higher in female AFCs, while caspase 3 and β-galactosidase activities were similar. This study supports the idea that sex differences start very early in prenatal life and influence specific parameters, suggesting that it may be relevant to appreciate sex differences to cover knowledge gaps. This might lead to improving the diagnosis of risk prediction for pregnancy complications and achieving a more satisfactory monitoring of fetus health, even preventing future diseases in adulthood.
Collapse
Affiliation(s)
- Ilaria Campesi
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (A.C.); (V.L.); (S.C.); (M.M.); (A.M.)
- Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, 07100 Sassari, Italy;
| | - Giampiero Capobianco
- Gynecologic and Obstetric Clinic, Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy
| | - Antonella Cano
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (A.C.); (V.L.); (S.C.); (M.M.); (A.M.)
| | - Valeria Lodde
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (A.C.); (V.L.); (S.C.); (M.M.); (A.M.)
| | - Sara Cruciani
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (A.C.); (V.L.); (S.C.); (M.M.); (A.M.)
| | - Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (A.C.); (V.L.); (S.C.); (M.M.); (A.M.)
| | - Giovanni Sotgiu
- Clinical Epidemiology and Medical Statistics Unit, Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (G.S.); (M.V.P.)
| | - Maria Laura Idda
- Institute of Genetics and Biomedical Research, 07100 Sassari, Italy;
| | - Mariangela Valentina Puci
- Clinical Epidemiology and Medical Statistics Unit, Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (G.S.); (M.V.P.)
| | - Margherita Ruoppolo
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (M.R.); (M.C.); (M.C.)
- CEINGE—Biotecnologie Avanzate s.c.ar.l., 80145 Naples, Italy
| | - Michele Costanzo
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (M.R.); (M.C.); (M.C.)
- CEINGE—Biotecnologie Avanzate s.c.ar.l., 80145 Naples, Italy
| | - Marianna Caterino
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (M.R.); (M.C.); (M.C.)
- CEINGE—Biotecnologie Avanzate s.c.ar.l., 80145 Naples, Italy
| | - Francesca Cambosu
- Genetics and Developmental Biology Unit, Azienda Ospedaliera Universitaria Sassari, 07100 Sassari, Italy;
| | - Andrea Montella
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (A.C.); (V.L.); (S.C.); (M.M.); (A.M.)
| | - Flavia Franconi
- Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, 07100 Sassari, Italy;
| |
Collapse
|
40
|
Li Y, Zhu J, Yu Z, Zhai F, Li H, Jin X. Regulation of apoptosis by ubiquitination in liver cancer. Am J Cancer Res 2023; 13:4832-4871. [PMID: 37970337 PMCID: PMC10636691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/04/2023] [Indexed: 11/17/2023] Open
Abstract
Apoptosis is a programmed cell death process critical to cell development and tissue homeostasis in multicellular organisms. Defective apoptosis is a crucial step in the malignant transformation of cells, including hepatocellular carcinoma (HCC), where the apoptosis rate is higher than in normal liver tissues. Ubiquitination, a post-translational modification process, plays a precise role in regulating the formation and function of different death-signaling complexes, including those involved in apoptosis. Aberrant expression of E3 ubiquitin ligases (E3s) in liver cancer (LC), such as cellular inhibitors of apoptosis proteins (cIAPs), X chromosome-linked IAP (XIAP), and linear ubiquitin chain assembly complex (LUBAC), can contribute to HCC development by promoting cell survival and inhibiting apoptosis. Therefore, the review introduces the main apoptosis pathways and the regulation of proteins in these pathways by E3s and deubiquitinating enzymes (DUBs). It summarizes the abnormal expression of these regulators in HCC and their effects on cancer inhibition or promotion. Understanding the role of ubiquitination in apoptosis and LC can provide insights into potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Yuxuan Li
- Department of Hepatobiliary and Pancreatic Surgery, Ningbo Medical Center of LiHuiLi Hospital, Ningbo UniversityNingbo 315040, Zhejiang, P. R. China
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo UniversityNingbo 315211, Zhejiang, P. R. China
| | - Jie Zhu
- Department of Hepatobiliary and Pancreatic Surgery, Ningbo Medical Center of LiHuiLi Hospital, Ningbo UniversityNingbo 315040, Zhejiang, P. R. China
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo UniversityNingbo 315211, Zhejiang, P. R. China
| | - Zongdong Yu
- Department of Hepatobiliary and Pancreatic Surgery, Ningbo Medical Center of LiHuiLi Hospital, Ningbo UniversityNingbo 315040, Zhejiang, P. R. China
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo UniversityNingbo 315211, Zhejiang, P. R. China
| | - Fengguang Zhai
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo UniversityNingbo 315211, Zhejiang, P. R. China
| | - Hong Li
- Department of Hepatobiliary and Pancreatic Surgery, Ningbo Medical Center of LiHuiLi Hospital, Ningbo UniversityNingbo 315040, Zhejiang, P. R. China
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo UniversityNingbo 315211, Zhejiang, P. R. China
| | - Xiaofeng Jin
- Department of Hepatobiliary and Pancreatic Surgery, Ningbo Medical Center of LiHuiLi Hospital, Ningbo UniversityNingbo 315040, Zhejiang, P. R. China
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo UniversityNingbo 315211, Zhejiang, P. R. China
| |
Collapse
|
41
|
Hartley B, Bassiouni W, Schulz R, Julien O. The roles of intracellular proteolysis in cardiac ischemia-reperfusion injury. Basic Res Cardiol 2023; 118:38. [PMID: 37768438 DOI: 10.1007/s00395-023-01007-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023]
Abstract
Ischemic heart disease remains a leading cause of human mortality worldwide. One form of ischemic heart disease is ischemia-reperfusion injury caused by the reintroduction of blood supply to ischemic cardiac muscle. The short and long-term damage that occurs due to ischemia-reperfusion injury is partly due to the proteolysis of diverse protein substrates inside and outside of cardiomyocytes. Ischemia-reperfusion activates several diverse intracellular proteases, including, but not limited to, matrix metalloproteinases, calpains, cathepsins, and caspases. This review will focus on the biological roles, intracellular localization, proteolytic targets, and inhibitors of these proteases in cardiomyocytes following ischemia-reperfusion injury. Recognition of the intracellular function of each of these proteases includes defining their activation, proteolytic targets, and their inhibitors during myocardial ischemia-reperfusion injury. This review is a step toward a better understanding of protease activation and involvement in ischemic heart disease and developing new therapeutic strategies for its treatment.
Collapse
Affiliation(s)
- Bridgette Hartley
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Wesam Bassiouni
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| | - Richard Schulz
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada.
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada.
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB, Canada.
- Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada.
| | - Olivier Julien
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
42
|
Fan Y, Shen J, Liu X, Cui J, Liu J, Peng D, Jin Y. β-Sitosterol Suppresses Lipopolysaccharide-Induced Inflammation and Lipogenesis Disorder in Bovine Mammary Epithelial Cells. Int J Mol Sci 2023; 24:14644. [PMID: 37834091 PMCID: PMC10572156 DOI: 10.3390/ijms241914644] [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: 08/27/2023] [Revised: 09/21/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
β-sitosterol, a natural plant steroid, has been shown to promote anti-inflammatory and antioxidant activities in the body. In this study, β-sitosterol was used to protect against lipopolysaccharide (LPS)-induced cell damage in bovine mammary epithelial cells, which are commonly studied as a cell model of mammary inflammatory response and lipogenesis. Results showed that treatment with a combination of LPS and β-sitosterol significantly reduced oxidative stress and inflammation, while increasing the expression of anti-apoptotic proteins and activating the hypoxia-inducible factor-1(HIF-1α)/mammalian target of rapamycin(mTOR) signaling pathway to inhibit apoptosis and improve lipid synthesis-related gene expression. Our finding suggests that β-sitosterol has the potential to alleviate inflammation in the mammary gland.
Collapse
Affiliation(s)
| | | | | | | | | | - Dongqiao Peng
- Jilin Provincial Key Laboratory of Livestock and Poultry Feed and Feeding in Northeastern Frigid Area, Department of Animal Science, College of Animal Science, Jilin University, Changchun 130062, China; (Y.F.); (J.S.); (X.L.); (J.C.); (J.L.)
| | - Yongcheng Jin
- Jilin Provincial Key Laboratory of Livestock and Poultry Feed and Feeding in Northeastern Frigid Area, Department of Animal Science, College of Animal Science, Jilin University, Changchun 130062, China; (Y.F.); (J.S.); (X.L.); (J.C.); (J.L.)
| |
Collapse
|
43
|
Lu C, Lubin JH, Sarma VV, Stentz SZ, Wang G, Wang S, Khare SD. Prediction and design of protease enzyme specificity using a structure-aware graph convolutional network. Proc Natl Acad Sci U S A 2023; 120:e2303590120. [PMID: 37729196 PMCID: PMC10523478 DOI: 10.1073/pnas.2303590120] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 08/14/2023] [Indexed: 09/22/2023] Open
Abstract
Site-specific proteolysis by the enzymatic cleavage of small linear sequence motifs is a key posttranslational modification involved in physiology and disease. The ability to robustly and rapidly predict protease-substrate specificity would also enable targeted proteolytic cleavage by designed proteases. Current methods for predicting protease specificity are limited to sequence pattern recognition in experimentally derived cleavage data obtained for libraries of potential substrates and generated separately for each protease variant. We reasoned that a more semantically rich and robust model of protease specificity could be developed by incorporating the energetics of molecular interactions between protease and substrates into machine learning workflows. We present Protein Graph Convolutional Network (PGCN), which develops a physically grounded, structure-based molecular interaction graph representation that describes molecular topology and interaction energetics to predict enzyme specificity. We show that PGCN accurately predicts the specificity landscapes of several variants of two model proteases. Node and edge ablation tests identified key graph elements for specificity prediction, some of which are consistent with known biochemical constraints for protease:substrate recognition. We used a pretrained PGCN model to guide the design of protease libraries for cleaving two noncanonical substrates, and found good agreement with experimental cleavage results. Importantly, the model can accurately assess designs featuring diversity at positions not present in the training data. The described methodology should enable the structure-based prediction of specificity landscapes of a wide variety of proteases and the construction of tailor-made protease editors for site-selectively and irreversibly modifying chosen target proteins.
Collapse
Affiliation(s)
- Changpeng Lu
- Institute for Quantitative Biomedicine, Rutgers–The State University of New Jersey, Piscataway, NJ08854
| | - Joseph H. Lubin
- Department of Chemistry and Chemical Biology, Rutgers–The State University of New Jersey, Piscataway, NJ08854
| | - Vidur V. Sarma
- Institute for Quantitative Biomedicine, Rutgers–The State University of New Jersey, Piscataway, NJ08854
| | | | - Guanyang Wang
- Department of Statistics, Rutgers–The State University of New Jersey, Piscataway, NJ08854
| | - Sijian Wang
- Institute for Quantitative Biomedicine, Rutgers–The State University of New Jersey, Piscataway, NJ08854
- Department of Statistics, Rutgers–The State University of New Jersey, Piscataway, NJ08854
| | - Sagar D. Khare
- Institute for Quantitative Biomedicine, Rutgers–The State University of New Jersey, Piscataway, NJ08854
- Department of Chemistry and Chemical Biology, Rutgers–The State University of New Jersey, Piscataway, NJ08854
| |
Collapse
|
44
|
Cho UH, Hetzer MW. Caspase-mediated nuclear pore complex trimming in cell differentiation and endoplasmic reticulum stress. eLife 2023; 12:RP89066. [PMID: 37665327 PMCID: PMC10476967 DOI: 10.7554/elife.89066] [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] [Indexed: 09/05/2023] Open
Abstract
During apoptosis, caspases degrade 8 out of ~30 nucleoporins to irreversibly demolish the nuclear pore complex. However, for poorly understood reasons, caspases are also activated during cell differentiation. Here, we show that sublethal activation of caspases during myogenesis results in the transient proteolysis of four peripheral Nups and one transmembrane Nup. 'Trimmed' NPCs become nuclear export-defective, and we identified in an unbiased manner several classes of cytoplasmic, plasma membrane, and mitochondrial proteins that rapidly accumulate in the nucleus. NPC trimming by non-apoptotic caspases was also observed in neurogenesis and endoplasmic reticulum stress. Our results suggest that caspases can reversibly modulate nuclear transport activity, which allows them to function as agents of cell differentiation and adaptation at sublethal levels.
Collapse
Affiliation(s)
- Ukrae H Cho
- Molecular and Cell Biology Laboratory, Salk Institute for Biological StudiesLa JollaUnited States
| | - Martin W Hetzer
- Molecular and Cell Biology Laboratory, Salk Institute for Biological StudiesLa JollaUnited States
- Institute of Science and Technology Austria (IST Austria)KlosterneuburgAustria
| |
Collapse
|
45
|
Kang XF, Lu XL, Bi CF, Hu XD, Li Y, Li JK, Yang LS, Liu J, Ma L, Zhang JF. Xuebijing injection protects sepsis induced myocardial injury by mediating TLR4/NF-κB/IKKα and JAK2/STAT3 signaling pathways. Aging (Albany NY) 2023; 15:8501-8517. [PMID: 37650558 PMCID: PMC10496990 DOI: 10.18632/aging.204990] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 07/20/2023] [Indexed: 09/01/2023]
Abstract
OBJECTIVE Compelling evidence has demonstrated that Xuebijing (XBJ) exerted protective effects against SIMI. The aims of this study were to investigate whether TLR4/IKKα-mediated NF-κB and JAK2/STAT3 pathways were involved in XBJ's cardio-protection during sepsis and the mechanisms. METHODS In this study, rats were randomly assigned to three groups: Sham group; CLP group; XBJ group. Rats were treated with XBJ or sanitary saline after CLP. Echocardiography, myocardial enzymes and HE were used to detect cardiac function. IL-1β, IL-6 and TNF-α in serum were measured using ELISA kits. Cardiomyocyte apoptosis were tested by TUNEL staining. The protein levels of Bax, Bcl-2, Bcl-xl, Cleaved-Caspase 3, Cleaved-Caspase 9, Cleaved-PARP, TLR4, p-NF-κB, p-IKKα, p-JAK2 and p-STAT3 in the myocardium were assayed by western blotting. And finally, immunofluorescence was used to assess the level of p-JAK2 and p-STAT3 in heart tissue. RESULTS The results of echocardiography, myocardial enzyme and HE test showed that XBJ could significantly improve SIMI. The IL-1β, IL-6 and TNF-α levels in the serum were markedly lower in the XBJ group than in the CLP group (p<0.05). TUNEL staining's results showed that XBJ ameliorated CLP-induced cardiomyocyte apoptosis. Meanwhile, XBJ downregulated the protein levels of Bax, Cleaved-Caspase 3, Cleaved-Caspase 9, Cleaved-PARP, TLR4, p-NF-κB, p-IKKα, p-JAK2 and p-STAT3, as well as upregulated the protein levels of Bcl-2, Bcl-xl (p <0.05). CONCLUSIONS In here, we observed that XBJ's cardioprotective advantages may be attributable to its ability to suppress inflammation and apoptosis via inhibiting the TLR4/ IKKα-mediated NF-κB and JAK2/STAT3 pathways during sepsis.
Collapse
Affiliation(s)
- Xiang-Fei Kang
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan 750000, Ningxia, China
| | - Xiao-Li Lu
- Laboratory Animal Centre, Ningxia Medical University, Yinchuan 750000, Ningxia, China
| | - Cheng-Fei Bi
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan 750000, Ningxia, China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan 750000, Ningxia, China
| | - Xiao-Dong Hu
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan 750000, Ningxia, China
| | - Ying Li
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan 750000, Ningxia, China
| | - Jin-Kui Li
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan 750000, Ningxia, China
| | - Li-Shan Yang
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan 750000, Ningxia, China
| | - Jia Liu
- Medical Experimental Center, General Hospital of Ningxia Medical University, Yinchuan 750000, Ningxia, China
| | - Lei Ma
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan 750000, Ningxia, China
| | - Jun-Fei Zhang
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan 750000, Ningxia, China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan 750000, Ningxia, China
| |
Collapse
|
46
|
Yang M, Gao X, Hu C, Wang S, Sheng H, Ma Y. Bta-miR-484 Targets SFRP1 and Affects Preadipocytes Proliferation, Differentiation, and Apoptosis. Int J Mol Sci 2023; 24:12710. [PMID: 37628891 PMCID: PMC10454478 DOI: 10.3390/ijms241612710] [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: 06/07/2023] [Revised: 08/02/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
MicroRNAs (miRNAs) are essential regulators of numerous biological processes in animals, including adipogenesis. Despite the abundance of miRNAs associated with adipogenesis, their exact mechanisms of action remain largely unknown. Our study highlights the role of bta-miR-484 as a major regulator of adipocyte proliferation, apoptosis, and differentiation. Here, we demonstrated that the expression of bta-miR-484 initially increased during adipogenesis before decreasing. Overexpression of bta-miR-484 in adipocytes ultimately inhibited cell proliferation and differentiation, reduced the number of EdU fluorescence-stained cells, increased the number of G1 phase cells, reduced the number of G2 and S phase cells, and downregulated the expression of proliferation markers (CDK2 and PCNA) and differentiation markers (CEBPA, FABP4, and LPL). Additionally, overexpression of bta-miR-484 promoted the expression of apoptosis-related genes (Caspase 3, Caspase 9, and BAX), and increased the number of apoptotic cells observed via flow cytometry. In contrast, bta-miR-484 inhibition in adipocytes yielded opposite effects to those observed during bta-miR-484 overexpression. Moreover, luciferase reporter assays confirmed SFRP1 as a target gene of bta-miR-484, and revealed that bta-miR-484 downregulates SFRP1 mRNA expression. These findings offer compelling evidence that bta-miR-484 targets SFRP1, inhibits proliferation and differentiation, and promotes apoptosis. Therefore, these results offer novel insights into the bta-miR-484 regulation of adipocyte growth and development.
Collapse
Affiliation(s)
| | | | | | | | | | - Yun Ma
- Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| |
Collapse
|
47
|
Khalil MI, Agamy AF, Elshewemi SS, Sultan AS, Abdelmeguid NE. Pterostilbene induces apoptosis in hepatocellular carcinoma cells: Biochemical, pathological, and molecular markers. Saudi J Biol Sci 2023; 30:103717. [PMID: 37483838 PMCID: PMC10359945 DOI: 10.1016/j.sjbs.2023.103717] [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: 04/14/2023] [Revised: 06/13/2023] [Accepted: 06/23/2023] [Indexed: 07/25/2023] Open
Abstract
Worldwide, hepatocellular carcinoma (HCC) is considered the sixth most prevalent cancer and ranked third in causes leading to death. Pterostilbene (PTE), a dimethylated analog of resveratrol, is a phytochemical found in fruits such as blueberries and grapes, and is known for its anticancer effect. The current study intended to investigate the effect of PTE on HepG2 cells. Cell viability, colony-forming potential, lipid peroxidation, catalase enzyme (CAT), superoxide dismutase (SOD), and caspase 3 activities, histone release, and expression levels of mTOR, S6K1, p53, and STAT3 proteins were assessed in PTE-treated HepG2 cells. In addition, the cellular and ultrastructural alterations were evaluated by light and transmission electron microscopy. PTE induced a significant reduction in HepG2 viability in a dose-dependent manner (IC50 of PTE = 74 ± 6 μM), accompanied by a decrease in colony formation potential. PTE-treated cancer cells exhibited a decrease in lipid peroxidation and CAT activity, and an increase in histone release, caspase-3, and SOD activities. Ultrastructurally, PTE-treated cells exhibited notable cell shrinkage, reduced number of filopodia, increased vacuolization, apoptotic bodies, accumulation of lipid droplets, enlarged mitochondria, dilated endoplasmic reticulum, pyknotic nuclei, and cellular fragmentation. mTOR, S6K1, and STAT3 levels were downregulated, however p53 level was modulated in PTE-treated cells. The anticancer potential of PTE might be related to its ability to alter the ultrastructure morphology, reduce mitotic activity, and modulate some key protein required for cell proliferation, suggesting its potential to trigger cancer cells towards apoptosis.
Collapse
Affiliation(s)
- Mahmoud I. Khalil
- Department of Biological Sciences, Faculty of Science, Beirut Arab University, Lebanon
- Molecular Biology Unit, Zoology Department, Faculty of Science, Alexandria University, Egypt
| | - Alaa F. Agamy
- Molecular Biology Unit, Zoology Department, Faculty of Science, Alexandria University, Egypt
| | | | - Ahmed S. Sultan
- Biochemistry Department, Faculty of Science, Alexandria University, Egypt
| | | |
Collapse
|
48
|
Jin C, Yuan S, Piao L, Ren M, Liu Q. Propofol synergizes with circAPBB2 to protect against hypoxia/reoxygenation-induced oxidative stress, inflammation, and apoptosis of human cardiomyocytes. Immun Inflamm Dis 2023; 11:e952. [PMID: 37647434 PMCID: PMC10408373 DOI: 10.1002/iid3.952] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/05/2023] [Accepted: 07/08/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Myocardial injury is the main manifestation of cardiovascular diseases, and previous studies have shown that propofol (PPF) regulates myocardial injury. However, the mechanism of PPF in regulating myocardial injury remains to be further explored. This work aims to analyze the effects of PPF on human cardiomyocyte injury and the underlying mechanism. METHODS The regulatory and functional role of PPF and circAPBB2 in human cardiomyocyte injury were analyzed using an in vitro hypoxia/reoxygenation (H/R) cell model, which was established by treating human cardiomyocytes (AC16 cells) with H/R. The study evaluated AC16 cell injury by analyzing cytotoxicity, oxidative stress, inflammation and apoptosis of H/R-induced AC16 cells. Quantitative real-time polymerase chain reaction was performed to detect circAPBB2, miR-18a-5p and dual specificity phosphatase 14 (DUSP14) expression. Protein expression was analyzed by Western blot analysis assay. Dual-luciferase reporter assay, RNA pull-down assay and RNA immunoprecipitation assay were performed to identify the associations among circAPBB2, miR-18a-5p and DUSP14. Cytotoxicity was investigated by cell counting kit-8 assay and lactate dehydrogenase activity detection kit. Oxidative stress was evaluated by cellular reactive oxygen species assay kit and superoxide dismutase activity assay kit. The production of tumor necrosis factor-α and interleukin-1β was evaluated by enzyme-linked immunosorbent assays. RESULTS The expression of circAPBB2 and DUSP14 was significantly decreased, while miR-18a-5p was increased in H/R-induced AC16 cells when compared with controls. H/R treatment-induced cytotoxicity, oxidative stress, inflammation and cell apoptosis were attenuated after circAPBB2 overexpression or PPF treatment, whereas these effects were restored by increasing miR-18a-5p expression. PPF treatment improved the inhibitory effect of ectopic circAPBB2 expression on H/R-induced cell injury. MiR-18a-5p silencing ameliorated H/R-induced AC16 damage by interacting with DUSP14. Mechanically, circAPBB2 acted as a miR-18a-5p sponge, and miR-18a-5p targeted DUSP14 in AC16 cells. CONCLUSION PPF synergized with circAPBB2 to protect AC16 cells against H/R-induced oxidative stress, inflammation and apoptosis through the miR-18a-5p/DUSP14 pathway.
Collapse
Affiliation(s)
- Chenghao Jin
- Department of AnesthesiologyBeijing Tongren Hospital, Capital Medical UniversityBeijingChina
| | - Shunnv Yuan
- Laboratory MedicineThe Affiliated Hospital of Yanbian UniversityJilinChina
| | - Longyi Piao
- Department of OncologyJilin Central Hospital of Jilin UniversityJilinChina
| | - Mingcheng Ren
- Department of OncologyDandong Central Hospital DandongLiaoningChina
| | - Qiang Liu
- Department of AnesthesiologyBeijing Tongren Hospital, Capital Medical UniversityBeijingChina
| |
Collapse
|
49
|
Hanna R, Rozenberg A, Saied L, Ben-Yosef D, Lavy T, Kleifeld O. In-Depth Characterization of Apoptosis N-terminome Reveals a Link Between Caspase-3 Cleavage and Post-Translational N-terminal Acetylation. Mol Cell Proteomics 2023:100584. [PMID: 37236440 PMCID: PMC10362333 DOI: 10.1016/j.mcpro.2023.100584] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/16/2023] [Accepted: 05/22/2023] [Indexed: 05/28/2023] Open
Abstract
The N-termini of proteins contain information about their biochemical properties and functions. These N-termini can be processed by proteases, and can undergo other co- or post-translational modifications. We have developed LATE (LysN Amino Terminal Enrichment), a method that uses selective chemical derivatization of α-amines to isolate the N-terminal peptides, in order to improve N-terminome identification in conjunction with other enrichment strategies. We applied LATE alongside another N-terminomic method to study caspase-3 mediated proteolysis both in vitro and during apoptosis in cells. This has enabled us to identify many unreported caspase-3 cleavages, some of which cannot be identified by other methods. Moreover, we have found direct evidence that neo-N-termini generated by caspase-3 cleavage can be further modified by Nt-acetylation. Some of these neo-Nt-acetylation events occur in the early phase of the apoptotic process and may have a role in translation inhibition. This has provided a comprehensive overview of the caspase-3 degradome and has uncovered previously unrecognized crosstalk between post-translational Nt-acetylation and caspase proteolytic pathways.
Collapse
Affiliation(s)
- Rawad Hanna
- Faculty of Biology, Technion-Israel Institute of Technology, Technion City, Haifa 3200003, Israel
| | - Andrey Rozenberg
- Faculty of Biology, Technion-Israel Institute of Technology, Technion City, Haifa 3200003, Israel
| | - Layla Saied
- Faculty of Biology, Technion-Israel Institute of Technology, Technion City, Haifa 3200003, Israel
| | - Daniel Ben-Yosef
- Faculty of Biology, Technion-Israel Institute of Technology, Technion City, Haifa 3200003, Israel
| | - Tali Lavy
- Faculty of Biology, Technion-Israel Institute of Technology, Technion City, Haifa 3200003, Israel
| | - Oded Kleifeld
- Faculty of Biology, Technion-Israel Institute of Technology, Technion City, Haifa 3200003, Israel.
| |
Collapse
|
50
|
Luo SY, Moussa EW, Lopez-Orozco J, Felix-Lopez A, Ishida R, Fayad N, Gomez-Cardona E, Wang H, Wilson JA, Kumar A, Hobman TC, Julien O. Identification of Human Host Substrates of the SARS-CoV-2 M pro and PL pro Using Subtiligase N-Terminomics. ACS Infect Dis 2023; 9:749-761. [PMID: 37011043 PMCID: PMC10081575 DOI: 10.1021/acsinfecdis.2c00458] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Indexed: 04/04/2023]
Abstract
The recent emergence of SARS-CoV-2 in the human population has caused a global pandemic. The virus encodes two proteases, Mpro and PLpro, that are thought to play key roles in the suppression of host protein synthesis and immune response evasion during infection. To identify the specific host cell substrates of these proteases, active recombinant SARS-CoV-2 Mpro and PLpro were added to A549 and Jurkat human cell lysates, and subtiligase-mediated N-terminomics was used to capture and enrich protease substrate fragments. The precise location of each cleavage site was identified using mass spectrometry. Here, we report the identification of over 200 human host proteins that are potential substrates for SARS-CoV-2 Mpro and PLpro and provide a global mapping of proteolysis for these two viral proteases in vitro. Modulating proteolysis of these substrates will increase our understanding of SARS-CoV-2 pathobiology and COVID-19.
Collapse
Affiliation(s)
- Shu Y. Luo
- Department
of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Eman W. Moussa
- Department
of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Joaquin Lopez-Orozco
- Department
of Cell Biology, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Alberto Felix-Lopez
- Department
of Medical Microbiology & Immunology, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Ray Ishida
- Department
of Medical Microbiology & Immunology, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Nawell Fayad
- Department
of Cell Biology, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Erik Gomez-Cardona
- Department
of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Henry Wang
- Department
of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Joyce A. Wilson
- Department
of Biochemistry, Microbiology & Immunology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Anil Kumar
- Department
of Biochemistry, Microbiology & Immunology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Tom C. Hobman
- Department
of Cell Biology, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
- Department
of Medical Microbiology & Immunology, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
- Li
Ka Shing Institute of Virology, Edmonton, Alberta T6G
2E1, Canada
| | - Olivier Julien
- Department
of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
- Li
Ka Shing Institute of Virology, Edmonton, Alberta T6G
2E1, Canada
| |
Collapse
|