1
|
Yousefi Rad A, Rastegari AA, Shahanipour K, Monajemi R. Moringa oleifera and Its Biochemical Compounds: Potential Multi-targeted Therapeutic Agents Against COVID-19 and Associated Cancer Progression. Biochem Genet 2025; 63:936-959. [PMID: 38583096 DOI: 10.1007/s10528-024-10758-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 02/23/2024] [Indexed: 04/08/2024]
Abstract
The Coronavirus disease-2019 (COVID-19) pandemic is a global concern, with updated pharmacological therapeutic strategies needed. Cancer patients have been found to be more susceptible to severe COVID-19 and death, and COVID-19 can also lead to cancer progression. Traditional medicinal plants have long been used as anti-infection and anti-inflammatory agents, and Moringa oleifera (M. oleifera) is one such plant containing natural products such as kaempferol, quercetin, and hesperetin, which can reduce inflammatory responses and complications associated with viral infections and multiple cancers. This review article explores the cellular and molecular mechanisms of action of M. oleifera as an anti-COVID-19 and anti-inflammatory agent, and its potential role in reducing the risk of cancer progression in cancer patients with COVID-19. The article discusses the ability of M. oleifera to modulate NF-κB, MAPK, mTOR, NLRP3 inflammasome, and other inflammatory pathways, as well as the polyphenols and flavonoids like quercetin and kaempferol, that contribute to its anti-inflammatory properties. Overall, this review highlights the potential therapeutic benefits of M. oleifera in addressing COVID-19 and associated cancer progression. However, further investigations are necessary to fully understand the cellular and molecular mechanisms of action of M. oleifera and its natural products as anti-inflammatory, anti-COVID-19, and anti-cancer strategies.
Collapse
Affiliation(s)
- Ali Yousefi Rad
- Department of Biochemistry, Falavarjan Branch, Islamic Azad University, Esfahan, Iran
| | - Ali Asghar Rastegari
- Department of Molecular and Cell Biochemistry, Falavarjan Branch, Islamic Azad University, Esfahan, Iran.
| | - Kahin Shahanipour
- Department of Biochemistry, Falavarjan Branch, Islamic Azad University, Esfahan, Iran
| | - Ramesh Monajemi
- Department of Biology, Falavarjan Branch, Islamic Azad University, Esfahan, Iran
| |
Collapse
|
2
|
Sun J, Liu J, Xue M, Zhao T, Song J, Zhang W, Chang Y, Zhan Y. Dynamic molecular responses of the sea urchin Strongylocentrotus intermedius to pathogen infection: Insights from a serial comparative transcriptome analysis. FISH & SHELLFISH IMMUNOLOGY 2025; 158:110176. [PMID: 39914794 DOI: 10.1016/j.fsi.2025.110176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 01/23/2025] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
To explore the dynamic molecular responses to pathogen infection in sea urchins, the sea urchin Strongylocentrotus intermedius were infected by a causative pathogen strain of sea urchin black peristomial membrane disease. Specimens were collected at 0, 6, 12, 24, 48, 72, and 96 h post-infection (hpi), and comparative transcriptome analysis were performed. The results showed that 1) a total of 771, 1437, 3477, 8417, 1566, and 2171 differentially expressed genes (DEGs) were identified at 6, 12, 24, 48, 72, and 96 hpi compared with the 0 hpi (as the control), respectively. 2) The number of upregulated DEGs was higher than that of downregulated DEGs at each time point after infection. The largest number of DEGs was obtained at 48 hpi. 3) Among identified DEGs, percent cellular process, binding, and metabolic process related DEGs account for 57.9 %, 49.9 %, and 45.5 %, respectively. Main Rho-GTPase family members (RhoA, Rac1, and Cdc42) exhibited a general upregulated expression trend during the examined infection process, the same as Caspase family members (Casp3, Casp6 and Casp7). 4) Cell cycle and apoptosis pathways are the most affected pathways, the DEG enrichment level of which remained in the top 30 (cell cycle pathways) and top 50 (apoptosis pathways) throughout the whole examined infection process. To sum up, all findings from this study will not only deepen our understanding of the dynamic molecular expression mechanisms of sea urchins in response to pathogen infection, but also provide new clues for elutriating the profound mechanisms of serial gene expression in innate immunity.
Collapse
Affiliation(s)
- Jingxian Sun
- Key Laboratory of Mariculture& Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian, Liaoning, 116023, PR China; College of Life Science, Liaoning Normal University, Dalian, Liaoning, 116029, PR China
| | - Jinming Liu
- Key Laboratory of Mariculture& Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian, Liaoning, 116023, PR China
| | - Mingyu Xue
- Key Laboratory of Mariculture& Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian, Liaoning, 116023, PR China
| | - Tanjun Zhao
- Key Laboratory of Mariculture& Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian, Liaoning, 116023, PR China; College of Life Science, Liaoning Normal University, Dalian, Liaoning, 116029, PR China
| | - Jian Song
- Key Laboratory of Mariculture& Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian, Liaoning, 116023, PR China
| | - Weijie Zhang
- Key Laboratory of Mariculture& Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian, Liaoning, 116023, PR China
| | - Yaqing Chang
- Key Laboratory of Mariculture& Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian, Liaoning, 116023, PR China; College of Life Science, Liaoning Normal University, Dalian, Liaoning, 116029, PR China.
| | - Yaoyao Zhan
- Key Laboratory of Mariculture& Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian, Liaoning, 116023, PR China.
| |
Collapse
|
3
|
Yang Y, He Y, Xing H, Zhao Z, Wang J, Li S, Xu X, Ma D, Hu Y. Hyaluronic acid-liposomes hybridized with HucMSC exosomes for enhanced exosomes transdermal delivery and acute skin photodamage repair. Int J Biol Macromol 2025; 306:141606. [PMID: 40024399 DOI: 10.1016/j.ijbiomac.2025.141606] [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: 10/09/2024] [Revised: 02/13/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Long-term exposure to ultraviolet (UV) radiation can damage human skin, resulting in photodamage. Repairing photodamaged skin has been a major focus of research in recent years. Extensive research has shown that human umbilical cord mesenchymal stem cell-derived exosomes (Exo) possess anti-inflammatory, pro-angiogenic, and wound healing properties, holding great potential for treating skin damage. However, due to the limitations of exosomes alone, such as poor transdermal penetration, instability, and low utilization, there is an urgent need for new delivery strategies. We designed a hybrid nanovesicle (HL@Exo) by combining ultrasonic incubation with membrane extrusion to fuse Exo with HL. HL@Exo capitalizes on the advantages of liposomal carriers and the permeation-enhancing properties of hyaluronic acid to effectively facilitate transdermal delivery of Exo. The successful fusion of HL@Exo and its skin penetration were verified by methods such as fluorescent labeling, western blotting, Transwell assays, and in vivo imaging. In vitro studies on photodamaged keratinocytes and endothelial cells demonstrated HL@Exo ability to promote cell proliferation, repair, angiogenesis, and reduce inflammation. In a laser-induced skin photodamage model, HL@Exo enhanced collagen regeneration, accelerated wound healing, and demonstrated significant anti-inflammatory effects, indicating its potential as a non-invasive treatment and offering a novel strategy for the clinical application of Exo.
Collapse
Affiliation(s)
- Yuhui Yang
- Department of Burn and Plastic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou 510235, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Yong He
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China
| | - Hui Xing
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Ziyi Zhao
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Jianjin Wang
- Honest Medical China Co., Ltd, Zhuhai 519000, China
| | - Shanying Li
- Honest Medical China Co., Ltd, Zhuhai 519000, China
| | - Xiaosong Xu
- Hengqin Perfect-Medical Laboratory Co. Ltd, Zhuhai 519000, China
| | - Dong Ma
- Department of Burn and Plastic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou 510235, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Biomedical Engineering, Jinan University, Guangzhou 510632, China; MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou 510632, China.
| | - Yunfeng Hu
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
4
|
Bai R, Wang H, Yang T, Yan Y, Zhu S, Lv C, Pei Y, Guo J, Li J, Cui X, Lv X, Zheng M. Mechanisms of Mitochondria-Mediated Apoptosis During Eimeria tenella Infection. Animals (Basel) 2025; 15:577. [PMID: 40003058 PMCID: PMC11852116 DOI: 10.3390/ani15040577] [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: 01/16/2025] [Revised: 02/10/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
Coccidiosis in chickens is a parasitic disease caused by Eimeria species, resulting in significant economic losses to the poultry industry. Among these species, Eimeria tenella is considered the most virulent pathogen, with its infection strongly associated with the apoptotic response of host cells. Eimeria tenella modulates host cell apoptosis in a stage-specific manner, suppressing apoptosis in the early phase to promote its intracellular development and triggering apoptosis in later stages to facilitate parasite egress and disease progression. This study established an in vitro infection model using 60 fifteen-day-old chick embryo cecal epithelial cells and infecting the cells with Eimeria tenella sporozoites at a 1:1 ratio of host cells to sporozoites. The aim was to examine the relationship between parasitic infection and the apoptotic response of host cells in the chick embryo cecal epithelial cells infected with E. tenella. The roles of the mitochondrial permeability transition pore (MPTP) and cytochrome c in intrinsic apoptosis were examined through the application of cyclosporine A (CsA), N, N, N', N'-tetramethyl-1,4-phenylenediamine (TMPD), and ascorbate (Asc). TUNEL staining, ELISA, and flow cytometry were performed to evaluate apoptotic rates. CsA, TMPD, and Asc significantly (p < 0.01) decreased cytochrome c release, caspase-9 activation, and apoptotic rates from 24 to 120 h post-E. tenella infection. These findings highlight the significance of cytochrome c-mediated, mitochondria-dependent apoptotic pathways in parasitized chick embryo cecal epithelial cells.
Collapse
Affiliation(s)
- Rui Bai
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China
- Shanxi Key Laboratory for Modernization of TCVM, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China
| | - Hui Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China
- Shanxi Key Laboratory for Modernization of TCVM, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China
| | - Tiantian Yang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China
| | - Yuqi Yan
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China
| | - Shuying Zhu
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China
| | - Chenyang Lv
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China
| | - Yang Pei
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China
| | - Jiale Guo
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China
| | - Jianhui Li
- College of Animal Science, Shanxi Agricultural University, Taigu, Jinzhong 030801, China
| | - Xiaozhen Cui
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China
| | - Xiaoling Lv
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China
| | - Mingxue Zheng
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China
| |
Collapse
|
5
|
Chen H, Ding B, Tan J, Meng Q, Li J, Zhang W, Zheng P, Liu B, Ma P, Lin J. Silver Molybdate Nanoparticles for Enhanced Tumor Immunotherapy through Pyroptosis Conversion and Ferroptosis Induction. Angew Chem Int Ed Engl 2025:e202501530. [PMID: 39961790 DOI: 10.1002/anie.202501530] [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: 01/19/2025] [Indexed: 02/27/2025]
Abstract
Pyroptosis holds great potential in tumor therapy due to its strong immunogenicity. Several strategies, including ion interference therapy (IIT), are developed to induce pyroptosis. However, the mechanism by which metal oxoanions induced pyroptosis remained unclear. It was reported that MoO4 2- ions could stimulate immune responses, but their pyroptosis-inducing mechanisms were not fully understood. Herein, we synthesized uniform and dispersed silver molybdate (Ag2MoO4) nanoparticles (AMO) via a solvothermal method. AMO responded to H2O2 and glutathione (GSH) stimuli, releasing Ag+ and MoO4 2- ions, generating reactive oxygen species (ROS), and depleting GSH, thereby inducing ferroptosis and pyroptosis. The MoO4 2- also inhibited cell migration and upregulated GSDME expression, converting apoptosis into caspase-3/GSDME-mediated pyroptosis. Additionally, DNA damage and ROS activated the cGAS-STING pathway, enhancing innate immunity. In vivo experiments demonstrated that the combination of AMO and the immune checkpoint inhibitor αPD-1 significantly inhibited tumor growth. This combination promoted dendritic cells (DCs) maturation, increased effector T cell numbers, induced M1 macrophage polarization, and alleviated immunosuppression. This study contributed to a deeper understanding of metal oxoanion-mediated pyroptosis, supporting its potential application in cancer immunotherapy.
Collapse
Affiliation(s)
- Hao Chen
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Binbin Ding
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Jia Tan
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Qi Meng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Jing Li
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Wenying Zhang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Pan Zheng
- Key Laboratory of Superlight Materials & Surface Technology of Ministry of Education, College of Material Sciences and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Bin Liu
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Ping'an Ma
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Jun Lin
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| |
Collapse
|
6
|
Yadav VK, Pramanik S, Alghamdi S, Atwah B, Qusty NF, Babalghith AO, Solanki VS, Agarwal N, Gupta N, Niazi P, Patel A, Choudhary N, Zairov R. Therapeutic Innovations in Nanomedicine: Exploring the Potential of Magnetotactic Bacteria and Bacterial Magnetosomes. Int J Nanomedicine 2025; 20:403-444. [PMID: 39816378 PMCID: PMC11734620 DOI: 10.2147/ijn.s462031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 12/07/2024] [Indexed: 01/18/2025] Open
Abstract
Nanotechnology has emerged as a revolutionary domain with diverse applications in medicine, and one of the noteworthy developments is the exploration of bacterial magnetosomes acquired from magnetotactic bacteria (MTB) for therapeutic purposes. The demand for natural nanomaterials in the biomedical field is continuously increasing due to their biocompatibility and eco-friendly nature. MTB produces uniform, well-ordered magnetic nanoparticles inside the magnetosomes, drawing attention due to their unique and remarkable features. MTB and magnetosomes have gained popularity in cancer treatment and diagnosis, especially in magnetic resonance imaging. Distinctive features highlighted include advancements in extraction, characterization, and functionalization techniques, alongside breakthroughs in utilizing MTB-based magnetosomes as contrast agents in imaging, biocompatible drug carriers, and tools for minimally invasive therapies. The biocompatible nature, functionalizing of the surface of bacterial magnetosomes, and response to the external magnetic field make them a potential candidate for the theragnostic purpose of MTB and magnetosomes. In the present review, emphasis has been given to the foundation of magnetosomes at a genetic level, mass production of magnetosomes, etc. Further authors have reviewed the various functionalization methods of the magnetosomes for cancer treatment. Finally, the authors have reviewed the recent advancements in MTB and magnetosome-based cancer detection, diagnosis, and treatment. Challenges such as scalability, long-term safety, and clinical translation are also discussed, presenting a roadmap for future research exploiting MTBs and magnetosomes' unique properties.
Collapse
Affiliation(s)
- Virendra Kumar Yadav
- Marwadi University Research Center, Department of Microbiology, Faculty of Sciences, Marwadi University, Rajkot, Gujarat, 360003, India
| | - Sheersha Pramanik
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu, 600036, India
| | - Saad Alghamdi
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Banan Atwah
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Naeem F Qusty
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Ahmad O Babalghith
- Medical Genetics Department, College of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Vijendra Singh Solanki
- Department of Chemistry, Institute of Science and Research (ISR), IPS Academy, Indore, India
| | - Neha Agarwal
- Department of Chemistry, Navyug Kanya Mahavidyalaya, University of Lucknow, Lucknow, Uttar Pradesh, India
| | - Nishant Gupta
- Department of Engineering and Medical Devices, River Engineering Pvt Ltd, Ecotech-III, Greater Noida, U.p., India
| | - Parwiz Niazi
- Department of Biology, Faculty of Education, Kandahar University, Kandahar, Afghanistan
| | - Ashish Patel
- Department of Lifesciences, Hemchandracharya North Gujarat University, Patan, Gujarat, 384265, India
| | - Nisha Choudhary
- Department of Lifesciences, Hemchandracharya North Gujarat University, Patan, Gujarat, 384265, India
| | - Rustem Zairov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center RAS, Kazan, Russian Federation
- Aleksander Butlerov Institute of Chemistry, Kazan Federal University, Kazan, Russian Federation
| |
Collapse
|
7
|
Zeng S, Wang J, Kang H, Li H, Peng X, Yoon J. Photon-Driven Dye Induction Pyroptosis: An Emerging Anti-Tumor Immunotherapy Paradigm. Angew Chem Int Ed Engl 2025; 64:e202417899. [PMID: 39513509 DOI: 10.1002/anie.202417899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 11/15/2024]
Abstract
Photoimmunotherapy represents a novel and promising modality in anti-tumor immunotherapy, offering new hope in the realm of cancer treatment due to its distinctive mechanism and substantial therapeutic efficacy. This innovative approach synergistically integrates photon technology with immunological principles, utilizing photon energy to activate the body's immune response. Photon-driven pyroptosis, a pivotal element of photoimmunotherapy, has significantly revitalized the advancement of this discipline. To support this critical progress, this minireview offers an exhaustive examination of the organic dyes presently employed for photon-driven pyroptosis, alongside an analysis of the prevailing challenges and opportunities in dye molecule design. It is our aspiration that this minireview will contribute to the acceleration of developments in photon-driven pyroptosis dye and the broader field of photoimmunotherapy.
Collapse
Affiliation(s)
- Shuang Zeng
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, China
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, China
| | - Jingyun Wang
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, China
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, China
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Haidong Li
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, China
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, China
| | - Xiaojun Peng
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, China
| | - Juyoung Yoon
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Korea
| |
Collapse
|
8
|
Li Y, Wang X, Meng X, Xia C, Yang C, Wang J, Yang J, Wang F. Aerobic exercise inhibits GSDME-dependent myocardial cell pyroptosis to protect ischemia-reperfusion injury. Mol Med 2024; 30:273. [PMID: 39719560 DOI: 10.1186/s10020-024-01048-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 12/15/2024] [Indexed: 12/26/2024] Open
Abstract
BACKGROUND Acute myocardial infarction (AMI) remains a significant cause of global mortality, exacerbated by ischemia-reperfusion (IR) injury. Myocardial cell pyroptosis has emerged as a critical pathway influencing IR injury severity. METHODS We aimed to investigate the cardioprotective effects of aerobic exercise on IR injury by examining the modulation of IGFBP2 and its impact on GSDME-dependent myocardial cell pyroptosis. Mechanistic pathways were explored using western blot analysis, ELISA, immunofluorescence, and echocardiography. RESULTS Our findings demonstrate that aerobic exercise leads to increased circulating levels of IGFBP2, which effectively suppresses GSDME-dependent myocardial cell pyroptosis. This regulation occurs via the AKT-GSK3β signaling pathway, involving VDAC1 phosphorylation, thereby enhancing mitochondrial function and reducing oxidative stress. CONCLUSION In conclusion, our study highlights the role of IGFBP2 in mitigating GSDME-dependent pyroptosis as a mechanism through which aerobic exercise exerts cardioprotective effects against IR injury. These insights suggest potential therapeutic targets for managing acute myocardial infarction.
Collapse
Affiliation(s)
- Yi Li
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, People's Republic of China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, People's Republic of China
| | - Xiang Wang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, People's Republic of China.
| | - Xuyang Meng
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, People's Republic of China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, People's Republic of China
| | - Chenxi Xia
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, People's Republic of China
| | - Chenguang Yang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, People's Republic of China
| | - Jun Wang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, People's Republic of China
| | - Jiefu Yang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, People's Republic of China.
| | - Fang Wang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, People's Republic of China.
| |
Collapse
|
9
|
Fan H, Shen R, Yan J, Bai Y, Fu Q, Shi X, Du G, Wang D. Pyroptosis the Emerging Link Between Gut Microbiota and Multiple Sclerosis. Drug Des Devel Ther 2024; 18:6145-6164. [PMID: 39717200 PMCID: PMC11665440 DOI: 10.2147/dddt.s489454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 12/11/2024] [Indexed: 12/25/2024] Open
Abstract
This review elucidates the pivotal role of pyroptosis, triggered by gut microbiota, in the development of multiple sclerosis (MS), emphasizing its significance within the gut-brain axis. Our comprehensive analysis of recent literature reveals how dysbiosis in the gut microbiota of MS patients-characterized by reduced microbial diversity and shifts in bacterial populations-profoundly impacts immune regulation and the integrity of the central nervous system (CNS). Pyroptosis, an inflammatory form of programmed cell death, significantly exacerbates MS by promoting the release of inflammatory cytokines and causing substantial damage to CNS tissues. The gut microbiota facilitates this detrimental process through metabolites such as short-chain fatty acids and neuroactive compounds, or self-structural products like lipopolysaccharides (LPS), which modulate immune responses and influence neuronal survival. This review highlights the potential of modulating gut microbiota to regulate pyroptosis, thereby suggesting that targeting this pathway could be a promising therapeutic strategy to mitigate inflammatory responses and preserve neuronal integrity in patients with MS.
Collapse
Affiliation(s)
- Hua Fan
- The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, People’s Republic of China
| | - Ruile Shen
- The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, People’s Republic of China
| | - Junqiang Yan
- The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, People’s Republic of China
| | - Yongjie Bai
- The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, People’s Republic of China
| | - Qizhi Fu
- The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, People’s Republic of China
| | - Xiaofei Shi
- The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, People’s Republic of China
| | - Ganqin Du
- The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, People’s Republic of China
| | - Dongmei Wang
- The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, People’s Republic of China
| |
Collapse
|
10
|
David KM, Alinezhadbalalami N, Salameh ZS, Aycock KN, Coy Allen I, Davalos RV. Modulating the Cell Death Immune Response for Electroporation Treatments. Bioelectricity 2024; 6:263-271. [PMID: 39712216 PMCID: PMC11656017 DOI: 10.1089/bioe.2024.0019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2024] Open
Abstract
Irreversible electroporation (IRE) is a minimally invasive ablation technique that compromises integrity of the cell membrane through the application of short duration, high voltage electric pulses to induce cell death. Adverse effects of IRE such as muscle contractions are reduced with higher frequency biphasic pulsing, commonly known as high-frequency irreversible electroporation (H-FIRE). IRE and H-FIRE treatments have shown to increase immune activation through the induction of both immediate and delayed cell death, indicated by the release of damage-associated molecular pathways, antigens, and proteins. In this study, we demonstrated that specific modes of cell death can be elicited by modifying the applied pulse width and electric field strength of various waveforms. Several assays were performed on a human glioblastoma cell line, seeded onto a 2D monolayer for electroporation treatments. Cleavage of Caspase 3/7 and Caspase 1, well-known indicators of apoptosis and pyroptosis, respectively, was quantified. Our results indicate that apoptotic activity was increased for shorter pulse widths and stronger electric fields, whereas pyroptotic activity displayed opposite trends being significantly dominant with longer pulse widths at lower applied electric fields. When clinically applied, the activation of specific cell death mechanisms can allow for controlling the extent of an electroporation-mediated immune response and subsequently improved overall patient survival. With this information, we could use an electrode array to spatially manipulate the elicited immune response for patient-specific treatments.
Collapse
Affiliation(s)
- Kailee M. David
- Bioelectromechanical Systems Laboratory, Virginia Tech-Wake Forest School of Biomedical Engineering, Blacksburg, Virginia, USA
- Bioelectromechanical Systems Laboratory, Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech-Emory University, Atlanta, Georgia, USA
| | - Nastaran Alinezhadbalalami
- Bioelectromechanical Systems Laboratory, Virginia Tech-Wake Forest School of Biomedical Engineering, Blacksburg, Virginia, USA
| | - Zaid S. Salameh
- Bioelectromechanical Systems Laboratory, Virginia Tech-Wake Forest School of Biomedical Engineering, Blacksburg, Virginia, USA
- Bioelectromechanical Systems Laboratory, Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech-Emory University, Atlanta, Georgia, USA
| | - Kenneth N. Aycock
- Bioelectromechanical Systems Laboratory, Virginia Tech-Wake Forest School of Biomedical Engineering, Blacksburg, Virginia, USA
| | - Irving Coy Allen
- Allen Laboratory, Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, Virginia, USA
| | - Rafael V. Davalos
- Bioelectromechanical Systems Laboratory, Virginia Tech-Wake Forest School of Biomedical Engineering, Blacksburg, Virginia, USA
- Bioelectromechanical Systems Laboratory, Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech-Emory University, Atlanta, Georgia, USA
| |
Collapse
|
11
|
Shaldam MA, Mousa MHA, Tawfik HO, El-Dessouki AM, Sharaky M, Saleh MM, Alzahrani AYA, Moussa SB, Al-Karmalawy AA. Muti-target rationale design of novel substituted N-phenyl-2-((6-phenylpyridazin-3-yl)thio)acetamide candidates as telomerase/JAK1/STAT3/TLR4 inhibitors: In vitro and in vivo investigations. Bioorg Chem 2024; 153:107843. [PMID: 39332072 DOI: 10.1016/j.bioorg.2024.107843] [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: 06/11/2024] [Revised: 09/11/2024] [Accepted: 09/22/2024] [Indexed: 09/29/2024]
Abstract
In this work, additional effort was applied to design new BIBR1532-based analogues with potential inhibitory activity against telomerase and acting as multitarget antitumor candidates to overcome the resistance problem. Therefore, novel substituted N-phenyl-2-((6-phenylpyridazin-3-yl)thio)acetamide candidates (4a-n) were synthesized. Applying the lead optimization strategy of the previously designed compound 8e; compound 4l showed an improved telomerase inhibition of 64.95 % and a superior growth inhibition of 79 % suggesting its potential use as a successful "multitarget-directed drug" for cancer therapy. Accordingly, compound 4l was further selected to evaluate its additional JAK1/STAT3/TLR4 inhibitory potentials. Compound 4l represented a very promising JAK1 inhibitory potential with a 0.46-fold change, compared to that of pacritinib reference standard (0.33-fold change). Besides, it showed a superior STAT3-inhibitory potential with a 0.22-fold change compared to sorafenib (0.33-fold change). Additionally, compound 4l downregulated TLR4 protein expression by 0.81-fold change compared to that of resatorvid (0.29-fold change). Also, molecular docking was performed to investigate the binding mode and affinity of the superior candidate 4l towards the four target receptors (telomerase, JAK1, STAT3, and TLR4). Furthermore, the therapeutic potential of compound 4l as an antitumor agent was additionally explored through in vivo studies involving female mice implanted with Solid Ehrlich Carcinoma (SEC). Remarkably, compound 4l led to prominent reductions in tumor size and mass. Concurrent enhancements in biochemical, hematologic, histopathologic, and immunohistochemical parameters further confirmed the suppression of angiogenesis and inflammation, elucidating additional mechanisms by which compound 4l exerts its anticancer effects.
Collapse
Affiliation(s)
- Moataz A Shaldam
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt.
| | - Mai H A Mousa
- Pharmaceutical Chemistry Department, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo 11786, Egypt.
| | - Haytham O Tawfik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| | - Ahmed M El-Dessouki
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Ahram Canadian University, 6th of October City, Giza 12566, Egypt.
| | - Marwa Sharaky
- Cancer Biology Department, Pharmacology Unit, National Cancer Institute (NCI), Cairo University, Cairo, Egypt; Biochemistry Department, Faculty of Pharmacy, Ahram Canadian University, 6th of October City, Giza 12566, Egypt.
| | - Mohamed M Saleh
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| | | | - Sana Ben Moussa
- Department of Chemistry, Faculty of Science and Arts, King Khalid University, Mohail Assir 61421, Saudi Arabia
| | - Ahmed A Al-Karmalawy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Mashreq, Baghdad 10023, Iraq; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt.
| |
Collapse
|
12
|
Liu Y, Xiang L, Li Y, Zhang S, Zhang Y, Shi H, Liu H, Du D, Zhou B, Ye B, Li S, Yin H, Xu H, Zhang Y. Bacteria-Mediated Tumor-Targeting Delivery of Multienzyme-Mimicking Covalent Organic Frameworks Promoting Pyroptosis for Combinatorial Sono-Catalytic Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407133. [PMID: 39494618 PMCID: PMC11653599 DOI: 10.1002/advs.202407133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 10/09/2024] [Indexed: 11/05/2024]
Abstract
Pyroptosis, an inflammatory cell death, has attracted great attention for potentiating a strong immune response against tumor cells. However, developing powerful pyroptosis inducers and then activating specific pyroptosis still remains challenging. Herein, a PEG-CuP-COF@∆St nanosystem is rationally designed, consisting of PEG-CuP-COF nanozyme pyroptosis inducers and tumor-targeting bacteria of the Salmonella Typhimurium strain VNP20009 (ΔSt), with an affinity for the tumor hypoxic microenvironment. The PEG-CuP-COF nanozymes possessed excellent sonodynamic performance and multienzyme-mimicking activities to generate reactive oxygen species (ROS) and then induce potent pyroptosis. The superoxide dismutase- and peroxidase-mimicking activities of PEG-CuP-COF catalytically produced hydrogen peroxide (H2O2) and hydroxyl radicals (•OH) which have important value in triggering acute inflammatory responses and pyroptosis. Moreover, PEG-CuP-COF showed outstanding glutathione peroxidase-mimicking activities, impairing the antioxidant defense in tumor cells and enhancing sonodynamic efficiency by making them more vulnerable to ROS-induced damage. During in vivo studies, PEG-CuP-COF@∆St nanosystem with its self-driven property exhibited impressive tumor-targeting capability and activated Caspase-3/gasdermin E-dependent pyroptosis to inhibit tumor growth. More importantly, it induced a powerful immune memory effect to prevent bone metastasis. In summary, this study introduces an innovative approach for combinatorial sono-catalytic immunotherapy using bacteria-mediated tumor-targeting delivery of nanozymes as specific pyroptosis inducers.
Collapse
Affiliation(s)
- Yunyun Liu
- Department of Medical UltrasoundCenter of Minimally Invasive Treatment for TumorShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
- Ultrasound Research and Education InstituteClinical Research Center for Interventional MedicineShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and TreatmentShanghai200072P. R. China
| | - Lihua Xiang
- Department of Medical UltrasoundCenter of Minimally Invasive Treatment for TumorShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
- Ultrasound Research and Education InstituteClinical Research Center for Interventional MedicineShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and TreatmentShanghai200072P. R. China
| | - Yitong Li
- Department of Medical UltrasoundCenter of Minimally Invasive Treatment for TumorShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
- Ultrasound Research and Education InstituteClinical Research Center for Interventional MedicineShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and TreatmentShanghai200072P. R. China
| | - Shen Zhang
- Department of Medical UltrasoundCenter of Minimally Invasive Treatment for TumorShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
- Ultrasound Research and Education InstituteClinical Research Center for Interventional MedicineShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and TreatmentShanghai200072P. R. China
| | - Ying Zhang
- Department of Medical UltrasoundCenter of Minimally Invasive Treatment for TumorShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
- Ultrasound Research and Education InstituteClinical Research Center for Interventional MedicineShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and TreatmentShanghai200072P. R. China
| | - Hui Shi
- Department of Medical UltrasoundCenter of Minimally Invasive Treatment for TumorShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
- Ultrasound Research and Education InstituteClinical Research Center for Interventional MedicineShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and TreatmentShanghai200072P. R. China
| | - Hui Liu
- Department of Medical UltrasoundCenter of Minimally Invasive Treatment for TumorShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
- Ultrasound Research and Education InstituteClinical Research Center for Interventional MedicineShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and TreatmentShanghai200072P. R. China
| | - Dou Du
- Department of Medical UltrasoundCenter of Minimally Invasive Treatment for TumorShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
- Ultrasound Research and Education InstituteClinical Research Center for Interventional MedicineShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and TreatmentShanghai200072P. R. China
| | - Bangguo Zhou
- Department of Medical UltrasoundCenter of Minimally Invasive Treatment for TumorShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
- Ultrasound Research and Education InstituteClinical Research Center for Interventional MedicineShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and TreatmentShanghai200072P. R. China
| | - Beibei Ye
- Department of Medical UltrasoundCenter of Minimally Invasive Treatment for TumorShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
- Ultrasound Research and Education InstituteClinical Research Center for Interventional MedicineShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and TreatmentShanghai200072P. R. China
| | - Shaoyue Li
- Department of Medical UltrasoundCenter of Minimally Invasive Treatment for TumorShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
- Ultrasound Research and Education InstituteClinical Research Center for Interventional MedicineShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and TreatmentShanghai200072P. R. China
| | - Haohao Yin
- Department of UltrasoundZhongshan HospitalInstitute of Ultrasound in Medicine and EngineeringFudan UniversityShanghai200032P. R. China
| | - Huixiong Xu
- Department of UltrasoundZhongshan HospitalInstitute of Ultrasound in Medicine and EngineeringFudan UniversityShanghai200032P. R. China
| | - Yifeng Zhang
- Department of Medical UltrasoundCenter of Minimally Invasive Treatment for TumorShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
- Ultrasound Research and Education InstituteClinical Research Center for Interventional MedicineShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and TreatmentShanghai200072P. R. China
| |
Collapse
|
13
|
Zhao Q, Zhang S, Feng W, Zhou A, Shi L, Zhang J. Deoxynivalenol-mediated kidney injury via endoplasmic reticulum stress in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 286:117243. [PMID: 39447294 DOI: 10.1016/j.ecoenv.2024.117243] [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: 03/22/2024] [Revised: 10/12/2024] [Accepted: 10/20/2024] [Indexed: 10/26/2024]
Abstract
OBJECTIVE Deoxynivalenol (DON) is a common fungal toxin that poses significant health risks to humans and animals. The present study aimed to investigate the adverse effects and molecular mechanisms of DON-induced kidney injury. METHODS Male C57BL/6 mice aged 5-6 weeks were used to establish a DON-induced acute kidney injury model. Histological analysis, biochemical assays, molecular techniques, Western blot, RNA sequencing, and transmission electron microscopy were employed to analyze kidney damage, inflammation, oxidative stress, apoptosis, and endoplasmic reticulum (ER) stress. RESULTS DON disrupted kidney morphology, induced inflammatory cell infiltration, and triggered inflammatory responses. DON increased MDA content while decreasing antioxidant enzyme activity (SOD and CAT). It also triggered apoptosis, evidenced by elevated levels of caspase-12, cleaved caspase-3, and BAX, and reduced levels of Bcl-2. Transcriptomic analysis identified distinct expression patterns in 1756 genes in DON-exposed mouse kidneys, notably upregulating ER stress-related genes. Further investigation revealed ultrastructural changes in the ER and mitochondrial damage induced by DON, along with increased levels of p-IRE1, p-PERK, and their downstream targets, indicating unfolded protein response (UPR) activation in the kidney. The ER stress inhibitor 4-Phenylbutyric acid (4-PBA) significantly mitigated DON-induced ER stress, oxidative damage, apoptosis, tissue injury, ER expansion, and mitochondrial damage. CONCLUSION Our findings highlight the role of ER stress in DON-induced kidney injury and the protective effect of 4-PBA against these adverse effects.
Collapse
Affiliation(s)
- Qingbo Zhao
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming & Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Siyi Zhang
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming & Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Weili Feng
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming & Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Ao Zhou
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming & Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Liangyu Shi
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming & Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Jing Zhang
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming & Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China.
| |
Collapse
|
14
|
Joghataie P, Ardakani MB, Sabernia N, Salary A, Khorram S, Sohbatzadeh T, Goodarzi V, Amiri BS. The Role of Circular RNA in the Pathogenesis of Chemotherapy-Induced Cardiotoxicity in Cancer Patients: Focus on the Pathogenesis and Future Perspective. Cardiovasc Toxicol 2024; 24:1151-1167. [PMID: 39158829 DOI: 10.1007/s12012-024-09914-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 08/11/2024] [Indexed: 08/20/2024]
Abstract
Cardiotoxicity is a serious challenge cancer patients face today. Various factors are involved in cardiotoxicity. Circular RNAs (circRNAs) are one of the effective factors in the occurrence and prevention of cardiotoxicity. circRNAs can lead to increased proliferation, apoptosis, and regeneration of cardiomyocytes by regulating the molecular pathways, as well as increasing or decreasing gene expression; some circRNAs have a dual role in cardiomyocyte regeneration or death. Identifying each of the pathways related to these processes can be effective on managing patients and preventing cardiotoxicity. In this study, an overview of the molecular pathways involved in cardiotoxicity by circRNAs and their effects on the downstream factors have been discussed.
Collapse
Affiliation(s)
- Pegah Joghataie
- Department of Cardiology, School of Medicine, Hazrat-E Rasool General Hospital, Iran University of Medical Sciences, Tehran, Iran
| | | | - Neda Sabernia
- Department of Internal Medicine, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | | | - Tooba Sohbatzadeh
- Student Research Committee, School of Medicine, Alborz University of Medical Sciences, Alborz, Iran
| | - Vahid Goodarzi
- Department of Anesthesiology, Rasoul-Akram Medical Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Bahareh Shateri Amiri
- Assistant Professor of Internal Medicine, Department of Internal Medicine, School of Medicine, Hazrat-E Rasool General Hospital, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
15
|
Abreu H, Lallukka M, Raineri D, Leigheb M, Ronga M, Cappellano G, Spriano S, Chiocchetti A. Evaluation of the immune response of peripheral blood mononuclear cells cultured on Ti6Al4V-ELI polished or etched surfaces. Front Bioeng Biotechnol 2024; 12:1458091. [PMID: 39439551 PMCID: PMC11493608 DOI: 10.3389/fbioe.2024.1458091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024] Open
Abstract
INTRODUCTION While titanium and its alloys exhibit excellent biocompatibility and corrosion resistance, their polished surfaces can hinder fast and effective osseointegration and other biological processes, such as angiogenesis, due to their inert and hydrophobic properties. Despite being commonly used for orthopedic implants, research focuses on developing surface treatments to improve osseointegration, promoting cell adhesion and proliferation, as well as increasing protein adsorption capacity. This study explores a chemical treatment intended for titanium-based implants that enhances tissue integration without compromising the mechanical properties of the Ti6Al4V substrate. However, recognizing that inflammation contributes to nearly half of early implant failures, we assessed the impact of this treatment on T-cell viability, cytokine production, and phenotype. METHODS Ti6Al4V with extra low interstitial (ELI) content discs were treated with hydrofluoric acid followed by a controlled oxidation step in hydrogen peroxide that creates a complex surface topography with micro- and nano-texture and modifies the chemistry of the surface oxide layer. The acid etched surface contains an abundance of hydroxyl groups, crucial for promoting bone growth and apatite precipitation, while also enabling further functionalization with biomolecules. RESULTS While cell viability remained high in both groups, untreated discs triggered an increase in Th2 cells and a decrease of the Th17 subset. Furthermore, peripheral blood mononuclear cells exposed to untreated discs displayed a rise in various pro-inflammatory and anti-inflammatory cytokines compared to the control and treated groups. Conversely, the treated discs showed a similar profile to the control, both in terms of immune cell subset frequencies and cytokine secretion. DISCUSSION The dysregulation of the cytokine profile upon contact with untreated Ti6Al4V-ELI discs, namely upregulation of IL-2 could be responsible for the decrease in Th17 frequency, and thus might contribute to implant-associated bacterial infection. Interestingly, the chemical treatment restores the immune response to levels comparable to the control condition, suggesting the treatment's potential to mitigate inflammation by enhancing biocompatibility.
Collapse
Affiliation(s)
- Hugo Abreu
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Diseases-CAAD, Università del Piemonte Orientale, Novara, Italy
| | - Mari Lallukka
- Applied Science and Technology Department, Politecnico di Torino, Torino, Italy
| | - Davide Raineri
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Diseases-CAAD, Università del Piemonte Orientale, Novara, Italy
| | - Massimiliano Leigheb
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, Novara, Italy
- Orthopaedics and Traumatology Unit, “Maggiore della Carità” Hospital, Novara, Italy
| | - Mario Ronga
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, Novara, Italy
- Orthopaedics and Traumatology Unit, “Maggiore della Carità” Hospital, Novara, Italy
| | - Giuseppe Cappellano
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Diseases-CAAD, Università del Piemonte Orientale, Novara, Italy
| | - Silvia Spriano
- Applied Science and Technology Department, Politecnico di Torino, Torino, Italy
| | - Annalisa Chiocchetti
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Diseases-CAAD, Università del Piemonte Orientale, Novara, Italy
| |
Collapse
|
16
|
Kasana S, Kumar S, Patel P, Kurmi BD, Jain S, Sahu S, Vaidya A. Caspase inhibitors: a review on recently patented compounds (2016-2023). Expert Opin Ther Pat 2024; 34:1047-1072. [PMID: 39206873 DOI: 10.1080/13543776.2024.2397732] [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: 03/06/2024] [Accepted: 08/24/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION Caspases are a family of protease enzymes that play a crucial role in apoptosis. Dysregulation of caspase activity has been implicated in various pathological conditions, making caspases an important focus of research in understanding cell death mechanisms and developing therapeutic strategies for diseases associated with abnormal apoptosis. AREAS COVERED It is a comprehensive review of caspase inhibitors that have been comprising recently granted patents from 2016 to 2023. It includes peptide and non-peptide caspase inhibitors with their application for different diseases. EXPERT OPINION This review categorizes and analyses recently patented caspase inhibitors on various diseases. Diseases linked to caspase dysregulation, including neurodegenerative disorders, and autoimmune conditions, are highlighted to accentuate the therapeutic relevance of the patented caspase inhibitors. This paper serves as a valuable resource for researchers, clinicians, and pharmaceutical developers seeking an up-to-date understanding of recently patented caspase inhibitors. The integration of recent patented compounds, structural insights, and mechanistic details provides a holistic view of the progress in caspase inhibitor research and its potential impact on addressing various diseases.
Collapse
Affiliation(s)
- Shivani Kasana
- Department of Pharmaceutical Chemistry and Analysis, ISF College of Pharmacy, Moga, India
| | - Shivam Kumar
- Department of Pharmaceutical Chemistry and Analysis, ISF College of Pharmacy, Moga, India
| | - Preeti Patel
- Department of Pharmaceutical Chemistry and Analysis, ISF College of Pharmacy, Moga, India
| | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, India
| | - Shweta Jain
- Sir Madanlal Institute of Pharmacy, Etawah, India
| | - Sanjeev Sahu
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Ankur Vaidya
- Faculty of Pharmacy, Uttar Pradesh University of Medical Sciences, Etawah, India
| |
Collapse
|
17
|
Fang M, Lin Y, Xue C, Sheng K, Guo Z, Han Y, Lin H, Wu Y, Sang Y, Chen X, Howell SB, Lin X, Lin X. The AKT inhibitor AZD5363 elicits synthetic lethality in ARID1A-deficient gastric cancer cells via induction of pyroptosis. Br J Cancer 2024; 131:1080-1091. [PMID: 39003371 PMCID: PMC11405682 DOI: 10.1038/s41416-024-02778-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 06/11/2024] [Accepted: 06/20/2024] [Indexed: 07/15/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is a deadly disease with poor overall survival and limited therapeutic options. Genetic alterations such as mutations and/or deletions in chromatin remodeling gene AT-rich interactive domain 1 A (ARID1A) occur frequently in GC. Although ARID1A mutations/deletions are not a druggable target for conventional treatments, novel therapeutic strategies based on a synthetic lethal approach may be effective for the treatment of ARID1A-deficient cancers. METHODS A kinase inhibitor library containing 551 compounds was screened in ARID1A isogenic GC cells for the ability to induce synthetic lethality effect. Selected hits' activity was validated, and the mechanism of the most potent candidate drug, AKT inhibitor AD5363 (capivasertib), on induction of the synthetic lethality with ARID1A deficiency was investigated. RESULTS After robust vulnerability screening of 551 diverse protein kinase inhibitors, we identified the AKT inhibitor AZD5363 as being the most potent lead compound in inhibiting viability of ARID1A-/- cells. A synthetic lethality between loss of ARID1A expression and AKT inhibition by AZD5363 was validated in both GC cell model system and xenograft model. Mechanistically, AZD5363 treatment induced pyroptotic cell death in ARID1A-deficient GC cells through activation of the Caspase-3/GSDME pathway. Furthermore, ARID1A occupied the AKT gene promoter and regulated its transcription negatively, thus the GC cells deficient in ARID1A showed increased expression and phosphorylation of AKT. CONCLUSIONS Our study demonstrates a novel synthetic lethality interaction and unique mechanism between ARID1A loss and AKT inhibition, which may provide a therapeutic and mechanistic rationale for targeted therapy on patients with ARID1A-defective GC who are most likely to be beneficial to AZD5363 treatment.
Collapse
Affiliation(s)
- Menghan Fang
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Youfen Lin
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Department of Endocrinology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Chaorong Xue
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Kaiqin Sheng
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Zegeng Guo
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Yuting Han
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Hanbin Lin
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Yuecheng Wu
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Yuchao Sang
- Scientific Research Center, Anxi County Hospital, Quanzhou, China
| | - Xintan Chen
- Scientific Research Center, Anxi County Hospital, Quanzhou, China
| | - Stephen B Howell
- Department of Medicine and the Moores Cancer Center, University of California, San Diego, CA, USA
| | - Xu Lin
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China.
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China.
| | - Xinjian Lin
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China.
- Scientific Research Center, Anxi County Hospital, Quanzhou, China.
| |
Collapse
|
18
|
Zhou L, Li J, Chen J, Yao X, Zeng X, Liu Y, Wang Y, Wang X. Anticancer activity and mechanism studies of photoactivated iridium(III) complexes toward lung cancer A549 cells. Dalton Trans 2024; 53:15176-15189. [PMID: 39221457 DOI: 10.1039/d4dt01677g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Cyclometalated iridium(III) compounds have been widely explored due to their outstanding photo-physical properties and multiple anticancer activities. In this paper, three cyclometalated iridium(III) compounds [Ir(ppy)2(DBDIP)]PF6 (5a), [Ir(bzq)2(DBDIP)]PF6 (5b), and [Ir(piq)2(DBDIP)]PF6 (5c) (ppy: 2-phenylpyridine; bzq: benzo[h]quinoline; piq: 1-phenylisoquinoline, and DBDIP: 2-(2,3-dihydrobenzo[b][1,4]dioxin-6-yl)-1H-imidazo[4,5-f][1,10]phenanthroline) were synthesized and the mechanism of antitumor activity was investigated. Compounds photoactivated by visible light show strong cytotoxicity against tumor cells, especially toward A549 cells. Biological experiments such as migration, cellular localization, mitochondrial membrane potential and permeability, reactive oxygen species (ROS) and calcium ion level detection were performed, and they demonstrated that the compounds induced the apoptosis of A549 cells through a mitochondrial pathway. At the same time, oxidative stress caused by ROS production increases the release of damage-related molecules and the expression of porogen gasdermin D (GSDMD), and the content of LDH released from damaged cell membranes also increased. Besides, the content of the lipid peroxidation product, malondialdehyde (MDA), increased and the expression of GPX4 decreased. These indicate that the compounds promote cell death by combining ferroptosis and pyroptosis. The results reveal that cyclometalated iridium(III) compounds 5a-5c may be a potential chemotherapeutic agent for photodynamic therapy of cancers.
Collapse
Affiliation(s)
- Lin Zhou
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Jiongbang Li
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Ju Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Xin Yao
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Xiandong Zeng
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Yunjun Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Yi Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Xiuzhen Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| |
Collapse
|
19
|
Zeng Q, Jiang T, Wang J. Role of LMO7 in cancer (Review). Oncol Rep 2024; 52:117. [PMID: 38994754 PMCID: PMC11267500 DOI: 10.3892/or.2024.8776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 06/17/2024] [Indexed: 07/13/2024] Open
Abstract
Cancer constitutes a multifaceted ailment characterized by the dysregulation of numerous genes and pathways. Among these, LIM domain only 7 (LMO7) has emerged as a significant player in various cancer types, garnering substantial attention for its involvement in tumorigenesis and cancer progression. This review endeavors to furnish a comprehensive discourse on the functional intricacies and mechanisms of LMO7 in cancer, with a particular emphasis on its potential as both a therapeutic target and prognostic indicator. It delves into the molecular attributes of LMO7, its implications in cancer etiology and the underlying mechanisms propelling its oncogenic properties. Furthermore, it underscores the extant challenges and forthcoming prospects in targeting LMO7 for combating cancer.
Collapse
Affiliation(s)
- Qun Zeng
- Hunan Key Laboratory of The Research and Development of Novel Pharmaceutical Preparations, The Hunan Provincial University Key Laboratory of The Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical University, Changsha, Hunan 410000, P.R. China
- Department of Biochemistry and Molecular Biology, Hengyang Medical School, University of South China, Hengyang, Hunan 421000, P.R. China
| | - Tingting Jiang
- Hunan Key Laboratory of The Research and Development of Novel Pharmaceutical Preparations, The Hunan Provincial University Key Laboratory of The Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical University, Changsha, Hunan 410000, P.R. China
- Department of Clinical Laboratory, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421000, P.R. China
| | - Jing Wang
- Hunan Key Laboratory of The Research and Development of Novel Pharmaceutical Preparations, The Hunan Provincial University Key Laboratory of The Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical University, Changsha, Hunan 410000, P.R. China
| |
Collapse
|
20
|
Yang X, Cui X, Wang G, Zhou M, Wu Y, Du Y, Li X, Xu T. HDAC inhibitor regulates the tumor immune microenvironment via pyroptosis in triple negative breast cancer. Mol Carcinog 2024; 63:1800-1813. [PMID: 38860600 DOI: 10.1002/mc.23773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/29/2024] [Accepted: 06/01/2024] [Indexed: 06/12/2024]
Abstract
Pyroptosis, an inflammatory form of cell death, promotes the release of immunogenic substances and stimulates immune cell recruitment, a process, which could turn cold tumors into hot ones. Thus, instigating pyroptosis in triple-negative breast cancer (TNBC) serves as a viable method for restoring antitumor immunity. We analyzed the effects of Histone Deacetylase Inhibitors (HDACi) on TNBC cells using the Cell Counting Kit-8 and colony formation assay. Apoptosis and lactate dehydrogenase (LDH) release assays were utilized to determine the form of cell death. The pyroptotic executor was validated by quantitative real-time polymerase chain reaction and western blot. Transcriptome was analyzed to investigate pyroptosis-inducing mechanisms. A subcutaneously transplanted tumor model was generated in BALB/c mice to evaluate infiltration of immune cells. HDACi significantly diminished cell proliferation, and pyroptotic "balloon"-like cells became apparent. HDACi led to an intra and extracellular material exchange, signified by the release of LDH and the uptake of propidium iodide. Among the gasdermin family, TNBC cells expressed maximum quantities of GSDME, and expression of GSDMA, GSDMB, and GSDME were augmented post HDACi treatment. Pyroptosis was instigated via the activation of the caspase 3-GSDME pathway with the potential mechanisms being cell cycle arrest and altered intracellular REDOX balance due to aberrant glutathione metabolism. In vivo experiments demonstrated that HDACi can activate pyroptosis, limit tumor growth, and escalate CD8+ lymphocyte and CD11b+ cell infiltration along with an increased presence of granzyme B in tumors. HDACi can instigate pyroptosis in TNBC, promoting infiltration of immune cells and consequently intensifying the efficacy of anticancer immunity.
Collapse
Affiliation(s)
- Xue Yang
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
- Laboratory of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
- Laboratory of General Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
| | - Xiaoqing Cui
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
- Laboratory of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
- Laboratory of General Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
| | - Ge Wang
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
- Laboratory of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
- Laboratory of General Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
| | - Mengying Zhou
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Yonglin Wu
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
- Laboratory of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
- Laboratory of General Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
| | - Yaying Du
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
- Laboratory of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
- Laboratory of General Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
| | - Xingrui Li
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
- Laboratory of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
- Laboratory of General Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
| | - Tao Xu
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
- Laboratory of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
- Laboratory of General Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
- Department of Obstetrics and Gynecology, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
| |
Collapse
|
21
|
Dou H, Yu PY, Liu YQ, Zhu Y, Li FC, Wang YY, Chen XY, Xiao M. Recent advances in caspase-3, breast cancer, and traditional Chinese medicine: a review. J Chemother 2024; 36:370-388. [PMID: 37936479 DOI: 10.1080/1120009x.2023.2278014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 10/25/2023] [Accepted: 10/27/2023] [Indexed: 11/09/2023]
Abstract
Caspases (cysteinyl aspartate-specific proteinases) are a group of structurally similar proteases in the cytoplasm that can be involved in cell differentiation, programmed death, proliferation, and inflammatory generation. Experts have found that caspase-3 can serve as a terminal splicing enzyme in apoptosis and participate in the mechanism by which cytotoxic drugs kill cancer cells. Breast cancer (BC) has become the most common cancer among women worldwide, posing a severe threat to their lives. Finding new therapeutic targets for BC is the primary task of contemporary physicians. Numerous studies have revealed the close association between caspase-3 expression and BC. Caspase-3 is essential in BC's occurrence, invasion, and metastasis. In addition, Caspase-3 exerts anticancer effects by regulating cell death mechanisms. Traditional Chinese medicine acting through caspase-3 expression is increasingly used in clinical treatment. This review summarizes the biological mechanism of caspase-3 and research progress on BC. It introduces a variety of traditional Chinese medicine related to caspase-3 to provide new ideas for the clinical treatment of BC.
Collapse
Affiliation(s)
- He Dou
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, P. R. China
| | - Ping Yang Yu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, P. R. China
| | - Yu Qi Liu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, P. R. China
| | - Yue Zhu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, P. R. China
| | - Fu Cheng Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, P. R. China
| | - You Yu Wang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, P. R. China
| | - Xing Yan Chen
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, P. R. China
| | - Min Xiao
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, P. R. China
| |
Collapse
|
22
|
Guo Z, Zhang J, Li M, Xing Z, Li X, Qing J, Zhang Y, Zhu L, Qi M, Zou X. Mechanism of action of quercetin in regulating cellular autophagy in multiple organs of Goto-Kakizaki rats through the PI3K/Akt/mTOR pathway. Front Med (Lausanne) 2024; 11:1442071. [PMID: 39211336 PMCID: PMC11357923 DOI: 10.3389/fmed.2024.1442071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
Objective This experimental study investigated the protective function of quercetin on the liver, spleen, and kidneys of Goto-Kakizaki (GK) rats and explores its mechanism of action on autophagy-related factors and pathways. Materials and methods GK rats were randomly divided into three groups: DM, DM + L-Que, and DM + H-Que, with age-matched Wistar rats serving as the control group. The control and DM groups were gavaged with saline, and the quercetin-treated group was gavaged with quercetin for 8 weeks each. Weekly blood glucose levels were monitored. Upon conclusion of the experiment, blood samples were gathered for lipid and hepatic and renal function analyses. The histopathologic morphology and lipid deposition in rats were examined. Disease-related targets were identified using molecular docking methods and network pharmacology analysis. Subsequently, immunohistochemical analysis was performed, followed by Western blotting to evaluate the levels of autophagy-related proteins and proteins in the AKT/PI3K/mTOR pathway, as well as their phosphorylation levels. Results The results showed that, compared with the control group, the DM group exhibited significant increases in blood glucose, serum liver and kidney markers, liver fat vacuoles, and inflammatory cell infiltration. Immunohistochemistry (IHC) results indicated that quercetin reduced the extensive expression of AKT, P62, and mTOR in the liver and spleen of diabetic rats. The expression of autophagy and pathway-related proteins, such as P62, PI3K, P-PI3K, Akt, P-AKT, mTOR, and P-mTOR, was upregulated, while the expression of LC3A/LC3B, Beclin-1, Pink-1, and Parkin was downregulated. Conversely, the quercetin group showed a reduction in liver and kidney injury serum markers by decreasing lipid deposition and cell necrosis, indicating that quercetin has protective effects on the liver, spleen, and kidneys of GK rats. Additionally, in the quercetin group, the expression of autophagy and pathway-related proteins such as LC3A/LC3B, Beclin-1, Pink-1, and Parkin was upregulated, while the expression of P62, PI3K, P-PI3K, Akt, P-AKT, mTOR, and P-mTOR was downregulated, with statistically significant correlations. Conclusion Quercetin markedly ameliorates liver, spleen, and kidney damage in GK rats, potentially through the inhibition of the PI3K/Akt/mTOR pathway, promoting autophagy. This research offers a rationale to the therapeutic potential of quercetin in mitigating organ damage associated with diabetes.
Collapse
Affiliation(s)
- Zhiqun Guo
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha, China
- School of Public Health, Changsha Medical University, Changsha, China
| | - Jingyu Zhang
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha, China
- School of Public Health, Changsha Medical University, Changsha, China
| | - Mianxin Li
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha, China
- School of Public Health, Changsha Medical University, Changsha, China
| | - Zengwei Xing
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha, China
- School of Public Health, Changsha Medical University, Changsha, China
| | - Xi Li
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha, China
- School of Public Health, Changsha Medical University, Changsha, China
| | - Jiaqi Qing
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha, China
- School of Public Health, Changsha Medical University, Changsha, China
| | - Yuan Zhang
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha, China
- School of Public Health, Changsha Medical University, Changsha, China
| | - Lemei Zhu
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha, China
- School of Public Health, Changsha Medical University, Changsha, China
| | - Mingxu Qi
- Department of Cardiovascular Medicine, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan, China
| | - Xuemin Zou
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha, China
- School of Public Health, Changsha Medical University, Changsha, China
| |
Collapse
|
23
|
El-Gamil DS, Zaky MY, Maximous PM, Sharaky M, El-Dessouki AM, Riad NM, Shaaban S, Abdel-Halim M, Al-Karmalawy AA. Exploring chromone-2-carboxamide derivatives for triple-negative breast cancer targeting EGFR, FGFR3, and VEGF pathways: Design, synthesis, and preclinical insights. Drug Dev Res 2024; 85:e22228. [PMID: 38952003 DOI: 10.1002/ddr.22228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 03/27/2024] [Accepted: 06/17/2024] [Indexed: 07/03/2024]
Abstract
Chromone-based compounds have established cytotoxic, antiproliferative, antimetastatic, and antiangiogenic effects on various cancer cell types via modulating different molecular targets. Herein, 17 novel chromone-2-carboxamide derivatives were synthesized and evaluated for their in vitro anticancer activity against 15 human cancer cell lines. Among the tested cell lines, MDA-MB-231, the triple-negative breast cancer cell line, was found to be the most sensitive, where the N-(2-furylmethylene) (15) and the α-methylated N-benzyl (17) derivatives demonstrated the highest growth inhibition with GI50 values of 14.8 and 17.1 μM, respectively. In vitro mechanistic studies confirmed the significant roles of compounds 15 and 17 in the induction of apoptosis and suppression of EGFR, FGFR3, and VEGF protein levels in MDA-MB-231 cancer cells. Moreover, compound 15 exerted cell cycle arrest at both the G0-G1 and G2-M phases. The in vivo efficacy of compound 15 as an antitumor agent was further investigated in female mice bearing Solid Ehrlich Carcinoma. Notably, administration of compound 15 resulted in a marked decrease in both tumor weight and volume, accompanied by improvements in biochemical, hematological, histological, and immunohistochemical parameters that verified the repression of both angiogenesis and inflammation as additional Anticancer mechanisms. Moreover, the binding interactions of compounds 15 and 17 within the binding sites of all three target receptors (EGFR, FGFR3, and VEGF) were clearly illustrated using molecular docking.
Collapse
Affiliation(s)
- Dalia S El-Gamil
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt
| | - Mohamed Y Zaky
- Zoology Department, Molecular Physiology Division, Faculty of Science, Beni-Suef University, Beni Suef, Egypt
| | - Patrick M Maximous
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Marwa Sharaky
- Cancer Biology Department, Pharmacology Unit, National Cancer Institute (NCI), Cairo University, Cairo, Egypt
- Biochemistry Department, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt
| | - Ahmed M El-Dessouki
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt
| | - Noura M Riad
- Department of Chemistry, School of Life and Medical Sciences, New Administrative Capital, University of Hertfordshire hosted by Global Academic Foundation, Cairo, Egypt
| | - Saad Shaaban
- Department of Chemistry, College of Science, King Faisal University, Al-Ahsa, Saudi Arabia
- Organic Chemistry Division, Department of Chemistry, College of Science, Mansoura University, Mansoura, Egypt
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Ahmed A Al-Karmalawy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta, Egypt
| |
Collapse
|
24
|
Meng X, Chen H, Tan Z, Yan W, Liu Y, Lv J, Han M. USP53 Affects the Proliferation and Apoptosis of Breast Cancer Cells by Regulating the Ubiquitination Level of ZMYND11. Biol Proced Online 2024; 26:24. [PMID: 39044157 PMCID: PMC11264418 DOI: 10.1186/s12575-024-00251-4] [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: 05/20/2024] [Accepted: 07/05/2024] [Indexed: 07/25/2024] Open
Abstract
Breast cancer is the most common female malignancy worldwide. Ubiquitin-specific peptidase 53 (USP53) has been shown to exert cancer-suppressing functions in several solid tumors, but its role and the underlying mechanism in breast cancer has not been clearly elucidated. Therefore, we have carried out a series of detailed studies on this matter at the levels of bioinformatics, clinical tissue, cell function and animal model. We found that USP53 expression was downregulated in breast cancer specimens and was negatively correlated with the clinical stages. Gain- and loss-of-function experiments demonstrated USP53 inhibited proliferation, clonogenesis, cell cycle and xenograft growth, as well as induced apoptosis and mitochondrial damage of breast cancer cells. Co-immunoprecipitation data suggested that USP53 interacted with zinc finger MYND-type containing 11 (ZMYND11), and catalyzed its deubiquitination and stabilization. The 33-50 amino acid Cys-box domain was key for USP53 enzyme activity, but not essential for its binding with ZMYND11. The rescue experiments revealed that the anti-tumor role of USP53 in breast cancer cells was at least partially mediated by ZMYND11. Both USP53 and ZMYND11 were prognostic protective factors for breast cancer. USP53-ZMYND11 axis may be a good potential biomarker or therapeutic target for breast cancer, which can provide novel insights into the diagnosis, treatment and prognosis.
Collapse
Affiliation(s)
- Xiangchao Meng
- Breast Disease Diagnosis and Treatment Center, First Hospital of Qinhuangdao, Qinhuangdao, 066000, P. R. China
| | - Hongye Chen
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, 100000, P. R. China
| | - Zhihui Tan
- Breast Disease Diagnosis and Treatment Center, First Hospital of Qinhuangdao, Qinhuangdao, 066000, P. R. China
| | - Weitao Yan
- Breast Disease Diagnosis and Treatment Center, First Hospital of Qinhuangdao, Qinhuangdao, 066000, P. R. China
| | - Yinfeng Liu
- Breast Disease Diagnosis and Treatment Center, First Hospital of Qinhuangdao, Qinhuangdao, 066000, P. R. China
| | - Ji Lv
- Breast Disease Diagnosis and Treatment Center, First Hospital of Qinhuangdao, Qinhuangdao, 066000, P. R. China.
| | - Meng Han
- Breast Disease Diagnosis and Treatment Center, First Hospital of Qinhuangdao, Qinhuangdao, 066000, P. R. China.
| |
Collapse
|
25
|
Ansari N, Salesi M. The association between primary Sjogren's syndrome and non-Hodgkin's lymphoma: a systematic review and meta-analysis of cohort studies. Clin Rheumatol 2024; 43:2177-2186. [PMID: 38722505 DOI: 10.1007/s10067-024-06993-6] [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: 02/28/2024] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 06/19/2024]
Abstract
Primary Sjögren's syndrome (pSS), a chronic autoimmune condition, has been associated with an increased risk of several cancers. This study aims to delve into the relationship between pSS and the potential development of non-Hodgkin's lymphoma (NHL) utilizing an in-depth systematic review and meta-analysis approach. To thoroughly explore the topic, we conducted a thorough examination of the literature, drawing from reputable databases such as ProQuest, PubMed, Web of Science, Cochrane, and Google Scholar. Our data collection spanned until February 8, 2024, with no time limitation. Data were analyzed with Stata 14 software at a significance threshold of p < 0.05. We examined 15 cohort studies encompassing a total of 50,308 individuals from 1997 to 2023. The findings revealed a substantial link between pSS and the risk of NHL, evident across all demographics. Specifically, the standardized incidence ratio (SIR) was generally 8.78 (95% CI 5.51, 13.99), with similar trends observed in both men (SIR, 6.29; 95% CI 1.93, 20.51) and women (SIR, 9.60; 95% CI 5.89, 15.63). Additionally, the SIR (10.50 (95% CI 7, 15.75)), HR (2.82 (95% CI 1.28, 6.18)), and OR (10.50 (95% CI 3.04, 36.28)) indices further supported this association. Furthermore, the risk of non-NHL associated with pSS was noticeable across different age groups of 40-49 years (SIR, 30.13; 95% CI 14.62, 62.08), 50-59 years (SIR, 9.12; 95% CI 5.13, 16.19), and 60-69 years (SIR, 9; 95% CI 4.68, 17.32). pSS substantively augments the likelihood of NHL manifestation. It notably impacts females and those in earlier stages of adulthood with more acuity than males and older cohorts.
Collapse
Affiliation(s)
- Narges Ansari
- Isfahan University of Medical Science, Isfahan, Iran
| | - Mansour Salesi
- Department of Rheumatology, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
26
|
Kombe Kombe AJ, Fotoohabadi L, Nanduri R, Gerasimova Y, Daskou M, Gain C, Sharma E, Wong M, Kelesidis T. The Role of the Nrf2 Pathway in Airway Tissue Damage Due to Viral Respiratory Infections. Int J Mol Sci 2024; 25:7042. [PMID: 39000157 PMCID: PMC11241721 DOI: 10.3390/ijms25137042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 06/14/2024] [Accepted: 06/20/2024] [Indexed: 07/16/2024] Open
Abstract
Respiratory viruses constitute a significant cause of illness and death worldwide. Respiratory virus-associated injuries include oxidative stress, ferroptosis, inflammation, pyroptosis, apoptosis, fibrosis, autoimmunity, and vascular injury. Several studies have demonstrated the involvement of the nuclear factor erythroid 2-related factor 2 (Nrf2) in the pathophysiology of viral infection and associated complications. It has thus emerged as a pivotal player in cellular defense mechanisms against such damage. Here, we discuss the impact of Nrf2 activation on airway injuries induced by respiratory viruses, including viruses, coronaviruses, rhinoviruses, and respiratory syncytial viruses. The inhibition or deregulation of Nrf2 pathway activation induces airway tissue damage in the presence of viral respiratory infections. In contrast, Nrf2 pathway activation demonstrates protection against tissue and organ injuries. Clinical trials involving Nrf2 agonists are needed to define the effect of Nrf2 therapeutics on airway tissues and organs damaged by viral respiratory infections.
Collapse
Affiliation(s)
- Arnaud John Kombe Kombe
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (A.J.K.K.)
| | - Leila Fotoohabadi
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (A.J.K.K.)
| | - Ravikanth Nanduri
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (A.J.K.K.)
| | - Yulia Gerasimova
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (A.J.K.K.)
| | - Maria Daskou
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Chandrima Gain
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Eashan Sharma
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Michael Wong
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Theodoros Kelesidis
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (A.J.K.K.)
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
27
|
Li X, Jiang X, Gao F, Zhou L, Wang G, Li B, Gu S, Huang W, Duan H. Study and evaluation of a gelatin- silver oxide nanoparticles releasing nitric oxide production of wound healing dressing for diabetic ulcer. PLoS One 2024; 19:e0298124. [PMID: 38885218 PMCID: PMC11182517 DOI: 10.1371/journal.pone.0298124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 01/19/2024] [Indexed: 06/20/2024] Open
Abstract
This study aimed to develop a novel Gelatin silver oxide material for releasing nitric oxide bionanocomposite wound dressing with enhanced mechanical, chemical, and antibacterial properties for the treatment of diabetic wounds. The gelatin- silver oxide nanoparticles (Ag2O-NP) bio nanocomposite was prepared using chitosan and gelatin polymers incorporated with silver oxide nanoparticles through the freeze-drying method. The samples were characterized using scanning electron microscopy (SEM) and X-ray diffraction (XRD) analysis. Results showed that the Ag2O-NP nanoparticles increased porosity, decreased pore size, and improved elastic modulus. The Ag2O-NP wound dressing exhibited the most effective antibacterial properties against Staphylococcus aureus and Escherichia coli. Among the samples, the wound dressing containing silver oxide nanoparticles demonstrated superior physical and mechanical properties, with 48% porosity, a tensile strength of 3.2 MPa, and an elastic modulus of 51.7 MPa. The fabricated wound dressings had a volume ratio of empty space to total volume ranging from 40% to 60%. In parallel, considering the complications of diabetes and its impact on the vascular system, another aspect of the research focused on developing a per2mediated wound dressing capable of releasing nitric oxide gas to regenerate damaged vessels and accelerate diabetic wound healing. Chitosan, a biocompatible and biodegradable polymer, was selected as the substrate for the wound dressing, and beta-glycerophosphate (GPβ), tripolyphosphate (TPP), and per2mediated alginate (AL) were used as crosslinkers. The chitosan-alginate (CS-AL) wound dressing exhibited optimal characteristics in terms of hole count and uniformity in the scanning electron microscope test. It also demonstrated superior water absorption (3854%) and minimal air permeability. Furthermore, the CS-AL sample exhibited an 80% degradation rate after 14 days, indicating its suitability as a wound dressing. The wound dressing was loaded with S-nitrosoglutathione (GSNO) powder, and the successful release of nitric oxide gas was confirmed through the grease test, showing a peak at a wavelength of 540 nm. Subsequent investigations revealed that the treatment of human umbilical vein endothelial cells (HUVECs) with high glucose led to a decrease in the expression of PER2 and SIRT1, while the expression of PER2 increased, which may subsequently enhance the expression of SIRT1 and promote cell proliferation activity. However, upon treatment of the cells with the modified materials, an increase in the expression of PER2 and SIRT1 was observed, resulting in a partial restoration of cell proliferative activity. This comprehensive study successfully developed per2-mediated bio-nanocomposite wound dressings with improved physical, mechanical, chemical, and antibacterial properties. The incorporation of silver oxide nanoparticles enhanced the antimicrobial activity, while the released nitric oxide gas from the dressing demonstrated the ability to mitigate vascular endothelial cell damage induced by high glucose levels. These advancements show promising potential for facilitating the healing process of diabetic wounds by addressing complications associated with diabetes and enhancing overall wound healing.
Collapse
Affiliation(s)
- Xian Li
- Department of Orthopedics, Dongguan Songshan Lake Tungwah Hospital, Dongguan City, Guangdong Province, China
| | - Xin Jiang
- Department of Orthopedics, Dongguan Songshan Lake Tungwah Hospital, Dongguan City, Guangdong Province, China
| | - Fei Gao
- Department of Orthopedics, Dongguan Songshan Lake Tungwah Hospital, Dongguan City, Guangdong Province, China
| | - Lifeng Zhou
- Department of Orthopedics, Dongguan Songshan Lake Tungwah Hospital, Dongguan City, Guangdong Province, China
| | - Guosheng Wang
- Department of Orthopedics, Dongguan Songshan Lake Tungwah Hospital, Dongguan City, Guangdong Province, China
| | - Bingfa Li
- Department of Orthopedics, Dongguan Songshan Lake Tungwah Hospital, Dongguan City, Guangdong Province, China
| | - Shihao Gu
- Department of Orthopedics, Dongguan Songshan Lake Tungwah Hospital, Dongguan City, Guangdong Province, China
| | - Wei Huang
- Department of Orthopedics, Dongguan Songshan Lake Tungwah Hospital, Dongguan City, Guangdong Province, China
| | - Hongkai Duan
- Department of Orthopedics, Dongguan Songshan Lake Tungwah Hospital, Dongguan City, Guangdong Province, China
| |
Collapse
|
28
|
Ji F, Shi C, Shu Z, Li Z. Nanomaterials Enhance Pyroptosis-Based Tumor Immunotherapy. Int J Nanomedicine 2024; 19:5545-5579. [PMID: 38882539 PMCID: PMC11178094 DOI: 10.2147/ijn.s457309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 05/22/2024] [Indexed: 06/18/2024] Open
Abstract
Pyroptosis, a pro-inflammatory and lytic programmed cell death pathway, possesses great potential for antitumor immunotherapy. By releasing cellular contents and a large number of pro-inflammatory factors, tumor cell pyroptosis can promote dendritic cell maturation, increase the intratumoral infiltration of cytotoxic T cells and natural killer cells, and reduce the number of immunosuppressive cells within the tumor. However, the efficient induction of pyroptosis and prevention of damage to normal tissues or cells is an urgent concern to be addressed. Recently, a wide variety of nanoplatforms have been designed to precisely trigger pyroptosis and activate the antitumor immune responses. This review provides an update on the progress in nanotechnology for enhancing pyroptosis-based tumor immunotherapy. Nanomaterials have shown great advantages in triggering pyroptosis by delivering pyroptosis initiators to tumors, increasing oxidative stress in tumor cells, and inducing intracellular osmotic pressure changes or ion imbalances. In addition, the challenges and future perspectives in this field are proposed to advance the clinical translation of pyroptosis-inducing nanomedicines.
Collapse
Affiliation(s)
- Fujian Ji
- Department of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People’s Republic of China
| | - Chunyu Shi
- Department of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People’s Republic of China
| | - Zhenbo Shu
- Department of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People’s Republic of China
| | - Zhongmin Li
- Department of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People’s Republic of China
| |
Collapse
|
29
|
Han L, Xu S, Zhou D, Chen R, Ding Y, Zhang M, Bao M, He B, Li S. Unveiling the causal link between metabolic factors and ovarian cancer risk using Mendelian randomization analysis. Front Endocrinol (Lausanne) 2024; 15:1401648. [PMID: 38899007 PMCID: PMC11185996 DOI: 10.3389/fendo.2024.1401648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
Background Metabolic abnormalities are closely tied to the development of ovarian cancer (OC), yet the relationship between anthropometric indicators as risk indicators for metabolic abnormalities and OC lacks consistency. Method The Mendelian randomization (MR) approach is a widely used methodology for determining causal relationships. Our study employed summary statistics from the genome-wide association studies (GWAS), and we used inverse variance weighting (IVW) together with MR-Egger and weighted median (WM) supplementary analyses to assess causal relationships between exposure and outcome. Furthermore, additional sensitivity studies, such as leave-one-out analyses and MR-PRESSO were used to assess the stability of the associations. Result The IVW findings demonstrated a causal associations between 10 metabolic factors and an increased risk of OC. Including "Basal metabolic rate" (OR= 1.24, P= 6.86×10-4); "Body fat percentage" (OR= 1.22, P= 8.20×10-3); "Hip circumference" (OR= 1.20, P= 5.92×10-4); "Trunk fat mass" (OR= 1.15, P= 1.03×10-2); "Trunk fat percentage" (OR= 1.25, P= 8.55×10-4); "Waist circumference" (OR= 1.23, P= 3.28×10-3); "Weight" (OR= 1.21, P= 9.82×10-4); "Whole body fat mass" (OR= 1.21, P= 4.90×10-4); "Whole body fat-free mass" (OR= 1.19, P= 4.11×10-3) and "Whole body water mass" (OR= 1.21, P= 1.85×10-3). Conclusion Several metabolic markers linked to altered fat accumulation and distribution are significantly associated with an increased risk of OC.
Collapse
Affiliation(s)
- Li Han
- Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical University, Changsha, China
- Hunan Key Laboratory of The Research and Development of Novel Pharmaceutical Preparations, School of Pharmaceutical Science, Changsha Medical University, Changsha, China
| | - Shuling Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Dongqi Zhou
- Department of Traditional Chinese Medicine, Sichuan Taikang Hospital, Chengdu, Sichuan, China
| | - Rumeng Chen
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yining Ding
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Mengling Zhang
- School of Stomatology, Changsha Medical University, Changsha, China
| | - Meihua Bao
- Hunan Key Laboratory of The Research and Development of Novel Pharmaceutical Preparations, School of Pharmaceutical Science, Changsha Medical University, Changsha, China
- The Hunan Provincial Key Laboratory of the TCM Agricultural Biogenomics, Changsha Medical University, Changsha, China
| | - Binsheng He
- The Hunan Provincial Key Laboratory of the TCM Agricultural Biogenomics, Changsha Medical University, Changsha, China
| | - Sen Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
30
|
Bhat AA, Kukreti N, Afzal M, Goyal A, Thapa R, Ali H, Shahwan M, Almalki WH, Kazmi I, Alzarea SI, Singh SK, Dua K, Gupta G. Ferroptosis and circular RNAs: new horizons in cancer therapy. EXCLI JOURNAL 2024; 23:570-599. [PMID: 38887390 PMCID: PMC11180955 DOI: 10.17179/excli2024-7005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 04/09/2024] [Indexed: 06/20/2024]
Abstract
Cancer poses intricate challenges to treatment due to its complexity and diversity. Ferroptosis and circular RNAs (circRNAs) are emerging as innovative therapeutic avenues amid the evolving landscape of cancer therapy. Extensive investigations into circRNAs reveal their diverse roles, ranging from molecular regulators to pivotal influencers of ferroptosis in cancer cell lines. The results underscore the significance of circRNAs in modulating molecular pathways that impact crucial aspects of cancer development, including cell survival, proliferation, and metastasis. A detailed analysis delineates these pathways, shedding light on the molecular mechanisms through which circRNAs influence ferroptosis. Building upon recent experimental findings, the study evaluates the therapeutic potential of targeting circRNAs to induce ferroptosis. By identifying specific circRNAs associated with the etiology of cancer, this analysis paves the way for the development of targeted therapeutics that exploit vulnerabilities in cancer cells. This review consolidates the existing understanding of ferroptosis and circRNAs, emphasizing their role in cancer therapy and providing impetus for ongoing research in this dynamic field. See also the graphical abstract(Fig. 1).
Collapse
Affiliation(s)
- Asif Ahmad Bhat
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Neelima Kukreti
- School of Pharmacy, Graphic Era Hill University, Dehradun 248007, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, U. P., India
| | - Riya Thapa
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Haider Ali
- Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India
- Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - Moyad Shahwan
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman, 346, United Arab Emirates
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, Ajman, 346, United Arab Emirates
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Sami I. Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, 72341, Sakaka, Al-Jouf, Saudi Arabia
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India
- Faculty of Health, Australian Research Center in Complementary and Integrative Medicine, University of Technology, Sydney, Ultimo-NSW 2007, Australia
- School of Medical and Life Sciences, Sunway University, Sunway, Malaysia
| | - Kamal Dua
- Faculty of Health, Australian Research Center in Complementary and Integrative Medicine, University of Technology, Sydney, Ultimo-NSW 2007, Australia
- Discipline of Pharmacy, Graduate School of Health, University of Technology, Sydney, Ultimo-NSW 2007, Australia
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, Ajman, 346, United Arab Emirates
| |
Collapse
|
31
|
Lagzian A, Askari M, Haeri MS, Sheikhi N, Banihashemi S, Nabi-Afjadi M, Malekzadegan Y. Increased V-ATPase activity can lead to chemo-resistance in oral squamous cell carcinoma via autophagy induction: new insights. Med Oncol 2024; 41:108. [PMID: 38592406 DOI: 10.1007/s12032-024-02313-9] [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: 11/27/2023] [Accepted: 01/23/2024] [Indexed: 04/10/2024]
Abstract
Oral squamous cell carcinoma (OSCC) is a cancer type with a high rate of recurrence and a poor prognosis. Tumor chemo-resistance remains an issue for OSCC patients despite the availability of multimodal therapy options, which causes an increase in tumor invasiveness. Vacuolar ATPase (V-ATPase), appears to be one of the most significant molecules implicated in MDR in tumors like OSCC. It is primarily responsible for controlling the acidity in the solid tumors' microenvironment, which interferes with the absorption of chemotherapeutic medications. However, the exact cellular and molecular mechanisms V-ATPase plays in OSCC chemo-resistance have not been understood. Uncovering these mechanisms can contribute to combating OSCC chemo-resistance and poor prognosis. Hence, in this review, we suggest that one of these underlying mechanisms is autophagy induced by V-ATPase which can potentially contribute to OSCC chemo-resistance. Finally, specialized autophagy and V-ATPase inhibitors may be beneficial as an approach to reduce drug resistance to anticancer therapies in addition to serving as coadjuvants in antitumor treatments. Also, V-ATPase could be a prognostic factor for OSCC patients. However, in the future, more investigations are required to demonstrate these suggestions and hypotheses.
Collapse
Affiliation(s)
- Ahmadreza Lagzian
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Marziye Askari
- Department of Immunology, School of Medicine, Hamedan University of Medical Sciences, Hamedan, Iran
| | - Melika Sadat Haeri
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Nastaran Sheikhi
- Biotechnology Department, Biological Sciences Faculty, Alzahra University, Tehran, Iran
| | - Sara Banihashemi
- Department of Bioscience, School of Science and Technology, Nottingham Trend University, Nottingham, UK
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Yalda Malekzadegan
- Department of Microbiology, Saveh University of Medical Sciences, Saveh, Iran.
| |
Collapse
|
32
|
Xu M, Zhao M, Zhu M, Yuan H, Li Z, Yan P, Ma C, Zhao H, Wang S, Wan R, Wang L, Yu G. Hibiscus manihot L. flower extract induces anticancer activity through modulation of apoptosis and autophagy in A549 cells. Sci Rep 2024; 14:8102. [PMID: 38582921 PMCID: PMC10998869 DOI: 10.1038/s41598-024-58439-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 03/29/2024] [Indexed: 04/08/2024] Open
Abstract
Lung cancer is a major public health issue and heavy burden in China and worldwide due to its high incidence and mortality without effective treatment. It's imperative to develop new treatments to overcome drug resistance. Natural products from food source, given their wide-ranging and long-term benefits, have been increasingly used in tumor prevention and treatment. This study revealed that Hibiscus manihot L. flower extract (HML) suppressed the proliferation and migration of A549 cells in a dose and time dependent manner and disrupting cell cycle progression. HML markedly enhanced the accumulation of ROS, stimulated the dissipation of mitochondrial membrane potential (MMP) and that facilitated mitophagy through the loss of mitochondrial function. In addition, HML induced apoptosis by activation of the PTEN-P53 pathway and inhibition of ATG5/7-dependent autophagy induced by PINK1-mediated mitophagy in A549 cells. Moreover, HML exert anticancer effects together with 5-FU through synergistic effect. Taken together, HML may serve as a potential tumor prevention and adjuvant treatment for its functional attributes.
Collapse
Affiliation(s)
- Minglu Xu
- School of Chemistry and Chemical Engineering, Henan Institute of Science and Technology, Xinxiang, 453-003, Henan, China
| | - Mengxia Zhao
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Institute of Biomedical Science, College of Life Science, Henan Normal university, 46 Jianshe Road, Xinxiang, 453007, Henan, China
| | - Miaomiao Zhu
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Institute of Biomedical Science, College of Life Science, Henan Normal university, 46 Jianshe Road, Xinxiang, 453007, Henan, China
| | - Hongmei Yuan
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Institute of Biomedical Science, College of Life Science, Henan Normal university, 46 Jianshe Road, Xinxiang, 453007, Henan, China
| | - Zhongzheng Li
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Institute of Biomedical Science, College of Life Science, Henan Normal university, 46 Jianshe Road, Xinxiang, 453007, Henan, China
| | - Peishuo Yan
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Institute of Biomedical Science, College of Life Science, Henan Normal university, 46 Jianshe Road, Xinxiang, 453007, Henan, China
| | - Chi Ma
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Institute of Biomedical Science, College of Life Science, Henan Normal university, 46 Jianshe Road, Xinxiang, 453007, Henan, China
| | - Huabin Zhao
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Institute of Biomedical Science, College of Life Science, Henan Normal university, 46 Jianshe Road, Xinxiang, 453007, Henan, China
| | - Shenghui Wang
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Institute of Biomedical Science, College of Life Science, Henan Normal university, 46 Jianshe Road, Xinxiang, 453007, Henan, China
| | - Ruyan Wan
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Institute of Biomedical Science, College of Life Science, Henan Normal university, 46 Jianshe Road, Xinxiang, 453007, Henan, China
| | - Lan Wang
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Institute of Biomedical Science, College of Life Science, Henan Normal university, 46 Jianshe Road, Xinxiang, 453007, Henan, China.
| | - Guoying Yu
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Institute of Biomedical Science, College of Life Science, Henan Normal university, 46 Jianshe Road, Xinxiang, 453007, Henan, China.
| |
Collapse
|
33
|
Syed RU, Afsar S, Aboshouk NAM, Salem Alanzi S, Abdalla RAH, Khalifa AAS, Enrera JA, Elafandy NM, Abdalla RAH, Ali OHH, Satheesh Kumar G, Alshammari MD. LncRNAs in necroptosis: Deciphering their role in cancer pathogenesis and therapy. Pathol Res Pract 2024; 256:155252. [PMID: 38479121 DOI: 10.1016/j.prp.2024.155252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 03/02/2024] [Accepted: 03/04/2024] [Indexed: 04/14/2024]
Abstract
Necroptosis, a controlled type of cell death that is different from apoptosis, has become a key figure in the aetiology of cancer and offers a possible target for treatment. A growing number of biological activities, including necroptosis, have been linked to long noncoding RNAs (lncRNAs), a varied family of RNA molecules with limited capacity to code for proteins. The complex interactions between LncRNAs and important molecular effectors of necroptosis, including mixed lineage kinase domain-like pseudokinase (MLKL) and receptor-interacting protein kinase 3 (RIPK3), will be investigated. We will explore the many methods that LncRNAs use to affect necroptosis, including protein-protein interactions, transcriptional control, and post-transcriptional modification. Additionally, the deregulation of certain LncRNAs in different forms of cancer will be discussed, highlighting their dual function in influencing necroptotic processes as tumour suppressors and oncogenes. The goal of this study is to thoroughly examine the complex role that LncRNAs play in controlling necroptotic pathways and how that regulation affects the onset and spread of cancer. In the necroptosis for cancer treatment, this review will also provide insight into the possible therapeutic uses of targeting LncRNAs. Techniques utilising LncRNA-based medicines show promise in controlling necroptotic pathways to prevent cancer from spreading and improve the effectiveness of treatment.
Collapse
Affiliation(s)
- Rahamat Unissa Syed
- Department of Pharmaceutics, College of Pharmacy, University of Ha'il, Hail 81442, Saudi Arabia.
| | - S Afsar
- Department of Virology, Sri Venkateswara University, Tirupathi, Andhra Pradesh 517502, India.
| | - Nayla Ahmed Mohammed Aboshouk
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | | | | | - Amna Abakar Suleiman Khalifa
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | - Jerlyn Apatan Enrera
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | - Nancy Mohammad Elafandy
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | - Randa Abdeen Husien Abdalla
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | - Omar Hafiz Haj Ali
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | - G Satheesh Kumar
- Department of Pharmaceutical Chemistry, College of Pharmacy, Seven Hills College of Pharmacy, Venkataramapuram, Tirupati, India
| | - Maali D Alshammari
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hail, Hail 81442, Saudi Arabia
| |
Collapse
|
34
|
Hussain MS, Moglad E, Afzal M, Bansal P, Kaur H, Deorari M, Ali H, Shahwan M, Hassan Almalki W, Kazmi I, Alzarea SI, Singh SK, Dua K, Gupta G. Circular RNAs in the KRAS pathway: Emerging players in cancer progression. Pathol Res Pract 2024; 256:155259. [PMID: 38503004 DOI: 10.1016/j.prp.2024.155259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 03/07/2024] [Accepted: 03/08/2024] [Indexed: 03/21/2024]
Abstract
Circular RNAs (circRNAs) have been recognized as key components in the intricate regulatory network of the KRAS pathway across various cancers. The KRAS pathway, a central signalling cascade crucial in tumorigenesis, has gained substantial emphasis as a possible therapeutic target. CircRNAs, a subgroup of non-coding RNAs known for their closed circular arrangement, play diverse roles in gene regulation, contributing to the intricate landscape of cancer biology. This review consolidates existing knowledge on circRNAs within the framework of the KRAS pathway, emphasizing their multifaceted functions in cancer progression. Notable circRNAs, such as Circ_GLG1 and circITGA7, have been identified as pivotal regulators in colorectal cancer (CRC), influencing KRAS expression and the Ras signaling pathway. Aside from their significance in gene regulation, circRNAs contribute to immune evasion, apoptosis, and drug tolerance within KRAS-driven cancers, adding complexity to the intricate interplay. While our comprehension of circRNAs in the KRAS pathway is evolving, challenges such as the diverse landscape of KRAS mutant tumors and the necessity for synergistic combination therapies persist. Integrating cutting-edge technologies, including deep learning-based prediction methods, holds the potential for unveiling disease-associated circRNAs and identifying novel therapeutic targets. Sustained research efforts are crucial to comprehensively unravel the molecular mechanisms governing the intricate interplay between circRNAs and the KRAS pathway, offering insights that could potentially revolutionize cancer diagnostics and treatment strategies.
Collapse
Affiliation(s)
- Md Sadique Hussain
- School of Pharmaceutical Sciences, Jaipur National University, Jagatpura, Jaipur, Rajasthan 302017, India
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Pooja Bansal
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Bengaluru, Karnataka 560069, India; Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Harpreet Kaur
- School of Basic & Applied Sciences, Shobhit University, Gangoh, Uttar Pradesh 247341, India; Department of Health & Allied Sciences, Arka Jain University, Jamshedpur, Jharkhand 831001, India
| | - Mahamedha Deorari
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - Moyad Shahwan
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman 346, United Arab Emirates; Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman 346, United Arab Emirates
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology, Ultimo, Sydney, NSW 2007, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology, Ultimo, Sydney, NSW 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology, Ultimo, Sydney, NSW 2007, Australia
| | - Gaurav Gupta
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman 346, United Arab Emirates; School of Pharmacy, Graphic Era Hill University, Dehradun 248007, India.
| |
Collapse
|
35
|
Hussain MS, Moglad E, Bansal P, Kaur H, Deorari M, Almalki WH, Kazmi I, Alzarea SI, Singh M, Kukreti N. Exploring the oncogenic and tumor-suppressive roles of Circ-ADAM9 in cancer. Pathol Res Pract 2024; 256:155257. [PMID: 38537524 DOI: 10.1016/j.prp.2024.155257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 04/14/2024]
Abstract
Circular RNAs (circRNAs) constitute a recently identified category of closed continuous loop RNA transcripts, serving as a subset of competing endogenous RNAs (ceRNAs) with the capacity to modulate genes by acting as microRNA sponges. In the context of cancer growth, numerous investigations have explored the potential functions of circRNAs, revealing their diverse functions either as oncogenes, promoting cancer progression, or as tumor suppressors, mitigating disease development. Among these, circRNA ADAM9 (Circ-ADAM9) is now recognized as an important player in a variety of mechanisms, both physiological and pathological, especially in cancer. The aberrant expression of Circ-ADAM9 has been observed across multiple human malignancies, implying a significant involvement in tumorigenesis. This comprehensive review aims to synthesize recent findings elucidating the function of Circ-ADAM9 in many malignancies. Additionally, the review explores the possibility of Circ-ADAM9 as a valuable biomarker, offering insights into its prognostic, diagnostic, and therapeutic implications. By summarizing the latest discoveries in this field, the review contributes to our understanding of the multifaceted contribution of Circ-ADAM9 in tumor biology and its potential applications in clinical settings.
Collapse
Affiliation(s)
- Md Sadique Hussain
- School of Pharmaceutical Sciences, Jaipur National University, Jagatpura, Jaipur, Rajasthan 302017, India
| | - Ehssan Moglad
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Pooja Bansal
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka 560069, India; Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Harpreet Kaur
- School of Basic & Applied Sciences, Shobhit University, Gangoh, Uttar Pradesh 247341, India; Department of Health & Allied Sciences, Arka Jain University, Jamshedpur, Jharkhand 831001, India
| | - Mahamedha Deorari
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia.
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, 72341, Sakaka, Aljouf, Saudi Arabia
| | - Mahaveer Singh
- School of Pharmacy and Technology Management, SVKMs, NMIMS University, Shirpur campus, Maharastra 425405, India
| | - Neelima Kukreti
- School of Pharmacy, Graphic Era Hill University, Dehradun 248007, India
| |
Collapse
|
36
|
Su H, Peng C, Liu Y. Regulation of ferroptosis by PI3K/Akt signaling pathway: a promising therapeutic axis in cancer. Front Cell Dev Biol 2024; 12:1372330. [PMID: 38562143 PMCID: PMC10982379 DOI: 10.3389/fcell.2024.1372330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
The global challenge posed by cancer, marked by rising incidence and mortality rates, underscores the urgency for innovative therapeutic approaches. The PI3K/Akt signaling pathway, frequently amplified in various cancers, is central in regulating essential cellular processes. Its dysregulation, often stemming from genetic mutations, significantly contributes to cancer initiation, progression, and resistance to therapy. Concurrently, ferroptosis, a recently discovered form of regulated cell death characterized by iron-dependent processes and lipid reactive oxygen species buildup, holds implications for diseases, including cancer. Exploring the interplay between the dysregulated PI3K/Akt pathway and ferroptosis unveils potential insights into the molecular mechanisms driving or inhibiting ferroptotic processes in cancer cells. Evidence suggests that inhibiting the PI3K/Akt pathway may sensitize cancer cells to ferroptosis induction, offering a promising strategy to overcome drug resistance. This review aims to provide a comprehensive exploration of this interplay, shedding light on the potential for disrupting the PI3K/Akt pathway to enhance ferroptosis as an alternative route for inducing cell death and improving cancer treatment outcomes.
Collapse
Affiliation(s)
- Hua Su
- Xingyi People’s Hospital, Xinyi, China
| | - Chao Peng
- Xingyi People’s Hospital, Xinyi, China
| | - Yang Liu
- The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
37
|
Tan W, Li Y, Ma L, Fu X, Long Q, Yan F, Li W, Liu X, Ding H, Wang Y, Zhang W. Exosomes of endothelial progenitor cells repair injured vascular endothelial cells through the Bcl2/Bax/Caspase-3 pathway. Sci Rep 2024; 14:4465. [PMID: 38396011 PMCID: PMC10891177 DOI: 10.1038/s41598-024-55100-x] [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: 10/31/2023] [Accepted: 02/20/2024] [Indexed: 02/25/2024] Open
Abstract
The main objective of this study is to evaluate the influence of exosomes derived from endothelial progenitor cells (EPC-Exo) on neointimal formation induced by balloon injury in rats. Furthermore, the study aims to investigate the potential of EPC-Exo to promote proliferation, migration, and anti-apoptotic effects of vascular endothelial cells (VECs) in vitro. The underlying mechanisms responsible for these observed effects will also be thoroughly explored and analyzed. Endothelial progenitor cells (EPCs) was isolated aseptically from Sprague-Dawley (SD) rats and cultured in complete medium. The cells were then identified using immunofluorescence and flow cytometry. The EPC-Exo were isolated and confirmed the identities by western-blot, transmission electron microscope, and nanoparticle analysis. The effects of EPC-Exo on the rat carotid artery balloon injury (BI) were detected by hematoxylin and eosin (H&E) staining, ELISA, immunohistochemistry, immunofluorescence, western-blot and qPCR. LPS was used to establish an oxidative damage model of VECs. The mechanism of EPC-Exo repairing injured vascular endothelial cells was detected by measuring the proliferation, migration, and tube function of VECs, actin cytoskeleton staining, TUNEL staining, immunofluorescence, western-blot and qPCR. In vivo, EPC-Exo exhibit inhibitory effects on neointima formation following carotid artery injury and reduce the levels of inflammatory factors, including TNF-α and IL-6. Additionally, EPC-Exo downregulate the expression of adhesion molecules on the injured vascular wall. Notably, EPC-Exo can adhere to the injured vascular area, promoting enhanced endothelial function and inhibiting vascular endothelial hyperplasia Moreover, they regulate the expression of proteins and genes associated with apoptosis, including B-cell lymphoma-2 (Bcl2), Bcl2-associated x (Bax), and Caspase-3. In vitro, experiments further confirmed that EPC-Exo treatment significantly enhances the proliferation, migration, and tube formation of VECs. Furthermore, EPC-Exo effectively attenuate lipopolysaccharides (LPS)-induced apoptosis of VECs and regulate the Bcl2/Bax/Caspase-3 signaling pathway. This study demonstrates that exosomes derived from EPCs have the ability to inhibit excessive carotid intimal hyperplasia after BI, promote the repair of endothelial cells in the area of intimal injury, and enhance endothelial function. The underlying mechanism involves the suppression of inflammation and anti-apoptotic effects. The fundamental mechanism for this anti-apoptotic effect involves the regulation of the Bcl2/Bax/Caspase-3 signaling pathway.
Collapse
Affiliation(s)
- Wei Tan
- College of Integrated Chinese and Western Medicine, Key Laboratory of Hunan Provincial for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Hunan, 410208, China
| | - Yanling Li
- College of Integrated Chinese and Western Medicine, Key Laboratory of Hunan Provincial for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Hunan, 410208, China
| | - Lu Ma
- College of Integrated Chinese and Western Medicine, Key Laboratory of Hunan Provincial for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Hunan, 410208, China
| | - Xinying Fu
- College of Integrated Chinese and Western Medicine, Key Laboratory of Hunan Provincial for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Hunan, 410208, China
| | - Qingyin Long
- College of Integrated Chinese and Western Medicine, Key Laboratory of Hunan Provincial for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Hunan, 410208, China
| | - Fanchen Yan
- College of Integrated Chinese and Western Medicine, Key Laboratory of Hunan Provincial for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Hunan, 410208, China
| | - Wanyu Li
- College of Integrated Chinese and Western Medicine, Key Laboratory of Hunan Provincial for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Hunan, 410208, China
| | - Xiaodan Liu
- College of Integrated Chinese and Western Medicine, Key Laboratory of Hunan Provincial for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Hunan, 410208, China
| | - Huang Ding
- College of Integrated Chinese and Western Medicine, Key Laboratory of Hunan Provincial for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Hunan, 410208, China
| | - Yang Wang
- Institute of Integrative Medicine, Key Laboratory of Hunan Province for Liver Manifestation of Traditional Chinese Medicine, Xiangya Hospital, Central South University, Hunan, 410008, China
| | - Wei Zhang
- College of Integrated Chinese and Western Medicine, Key Laboratory of Hunan Provincial for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Hunan, 410208, China.
| |
Collapse
|
38
|
Khan F, Pandey P, Verma M, Upadhyay TK. Terpenoid-Mediated Targeting of STAT3 Signaling in Cancer: An Overview of Preclinical Studies. Biomolecules 2024; 14:200. [PMID: 38397437 PMCID: PMC10886526 DOI: 10.3390/biom14020200] [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: 01/08/2024] [Revised: 02/01/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
Cancer has become one of the most multifaceted and widespread illnesses affecting human health, causing substantial mortality at an alarming rate. After cardiovascular problems, the condition has a high occurrence rate and ranks second in terms of mortality. The development of new drugs has been facilitated by increased research and a deeper understanding of the mechanisms behind the emergence and advancement of the disease. Numerous preclinical and clinical studies have repeatedly demonstrated the protective effects of natural terpenoids against a range of malignancies. Numerous potential bioactive terpenoids have been investigated in natural sources for their chemopreventive and chemoprotective properties. In practically all body cells, the signaling molecule referred to as signal transducer and activator of transcription 3 (STAT3) is widely expressed. Numerous studies have demonstrated that STAT3 regulates its downstream target genes, including Bcl-2, Bcl-xL, cyclin D1, c-Myc, and survivin, to promote the growth of cells, differentiation, cell cycle progression, angiogenesis, and immune suppression in addition to chemotherapy resistance. Researchers viewed STAT3 as a primary target for cancer therapy because of its crucial involvement in cancer formation. This therapy primarily focuses on directly and indirectly preventing the expression of STAT3 in tumor cells. By explicitly targeting STAT3 in both in vitro and in vivo settings, it has been possible to explain the protective effect of terpenoids against malignant cells. In this study, we provide a complete overview of STAT3 signal transduction processes, the involvement of STAT3 in carcinogenesis, and mechanisms related to STAT3 persistent activation. The article also thoroughly summarizes the inhibition of STAT3 signaling by certain terpenoid phytochemicals, which have demonstrated strong efficacy in several preclinical cancer models.
Collapse
Affiliation(s)
- Fahad Khan
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 602105, India;
| | - Pratibha Pandey
- University Centre for Research and Development, Chandigarh University, Gharuan, Mohali 140413, India
| | - Meenakshi Verma
- University Centre for Research and Development, Chandigarh University, Gharuan, Mohali 140413, India
- Department of Chemistry, University Institute of Sciences, Chandigarh University, Gharuan, Mohali 140413, India
| | - Tarun Kumar Upadhyay
- Department of Biotechnology, Parul Institute of Applied Sciences and Research and Development Cell, Parul University, Vadodara 391760, India;
| |
Collapse
|
39
|
Mao Y, Kong X, Liang Z, Yang C, Wang S, Fan H, Ning C, Xiao W, Wu Y, Wu J, Yuan L, Yuan Z. Viola yedoensis Makino alleviates heat stress-induced inflammation, oxidative stress, and cell apoptosis in the spleen and thymus of broilers. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117350. [PMID: 37907144 DOI: 10.1016/j.jep.2023.117350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/12/2023] [Accepted: 10/23/2023] [Indexed: 11/02/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Viola yedoensis Makino (VYM) is a traditional Chinese herbal medicine widely distributed in China. It has many pharmacological effects such as anti-inflammatory, immune regulation and anti-oxidation. However, the protective effect of VYM on the spleen and thymus of broilers induced by heat stress has rarely been reported. AIM OF THE STUDY We established a heat stress model of broilers to explore the protective effect of VYM on spleen and thymus of broilers. MATERIALS AND METHODS In this experiment, a heat stress model was made by adjusting the feeding temperature of broilers. The protective effect of VYM on the spleen and thymus of heat-stressed broilers were evaluated by detecting immune organ coefficient, histological observation, Enzyme-Linked Immunosorbent Assay, production of antioxidant enzymes and peroxides, TUNEL Staining, Quantitative Real-time PCR. RESULTS In this study, 60 healthy male AA broilers were divided into 6 groups: Control, 4.5% VYM, HS, HS + 0.5% VYM, HS + 1.5% VYM, HS + 4.5% VYM. After 42 days of feeding, serum, spleen and thymus were collected for detection and analysis. The study revealed that heat stress can lead to pathological damage in the spleen and thymus of broilers, reduce the content of immunoglobulin and newcastle disease (ND), infectious bursal disease (IBD) antibody levels, increase the expression of inflammatory factors IL-1β, INF-γ, heat shock 70 kDa protein (HSP70), heat shock 90 kDa protein (HSP90). Heat stress inhibits the activity of antioxidant enzymes CAT and SOD, promotes the production of MDA, and then lead to oxidative damage of the spleen and thymus. In addition, apoptotic cells and the ratio of Bax/Bcl-2 was increased. However, the addition of VYM to the feed can alleviate the adverse effects of heat stress on the spleen and thymus of broilers. CONCLUSIONS This study showed that the addition of VYM to the diet could inhibit oxidative stress and apoptosis, and reduce the inflammatory damage of heat stress on the spleen and thymus of broilers. This study provides a basis for further exploring the regulatory role of VYM in heat stress-induced immune imbalance in broilers. In addition, this study also provides a theoretical basis for the development of VYM as a feed additive with immunomodulatory effects.
Collapse
Affiliation(s)
- Yan Mao
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, PR China
| | - Xiangyi Kong
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, PR China
| | - Zengenni Liang
- Department of Hunan Agricultural Product Processing Institute, Hunan Academy of Agricultural Sciences, Changsha 410128, PR China
| | - Chenglin Yang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, PR China
| | - Siqi Wang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, PR China
| | - Hui Fan
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, PR China
| | - Can Ning
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, PR China
| | - Wenguang Xiao
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, PR China
| | - You Wu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, PR China
| | - Jing Wu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, PR China
| | - Liyun Yuan
- Xiangyang Vocational and Technical College, Xiangyang 441050, PR China
| | - Zhihang Yuan
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, PR China.
| |
Collapse
|
40
|
Albalawi E, Thakur A, Ramakrishna MT, Bhatia Khan S, SankaraNarayanan S, Almarri B, Hadi TH. Oral squamous cell carcinoma detection using EfficientNet on histopathological images. Front Med (Lausanne) 2024; 10:1349336. [PMID: 38348235 PMCID: PMC10859441 DOI: 10.3389/fmed.2023.1349336] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 12/28/2023] [Indexed: 02/15/2024] Open
Abstract
Introduction Oral Squamous Cell Carcinoma (OSCC) poses a significant challenge in oncology due to the absence of precise diagnostic tools, leading to delays in identifying the condition. Current diagnostic methods for OSCC have limitations in accuracy and efficiency, highlighting the need for more reliable approaches. This study aims to explore the discriminative potential of histopathological images of oral epithelium and OSCC. By utilizing a database containing 1224 images from 230 patients, captured at varying magnifications and publicly available, a customized deep learning model based on EfficientNetB3 was developed. The model's objective was to differentiate between normal epithelium and OSCC tissues by employing advanced techniques such as data augmentation, regularization, and optimization. Methods The research utilized a histopathological imaging database for Oral Cancer analysis, incorporating 1224 images from 230 patients. These images, taken at various magnifications, formed the basis for training a specialized deep learning model built upon the EfficientNetB3 architecture. The model underwent training to distinguish between normal epithelium and OSCC tissues, employing sophisticated methodologies including data augmentation, regularization techniques, and optimization strategies. Results The customized deep learning model achieved significant success, showcasing a remarkable 99% accuracy when tested on the dataset. This high accuracy underscores the model's efficacy in effectively discerning between normal epithelium and OSCC tissues. Furthermore, the model exhibited impressive precision, recall, and F1-score metrics, reinforcing its potential as a robust diagnostic tool for OSCC. Discussion This research demonstrates the promising potential of employing deep learning models to address the diagnostic challenges associated with OSCC. The model's ability to achieve a 99% accuracy rate on the test dataset signifies a considerable leap forward in earlier and more accurate detection of OSCC. Leveraging advanced techniques in machine learning, such as data augmentation and optimization, has shown promising results in improving patient outcomes through timely and precise identification of OSCC.
Collapse
Affiliation(s)
- Eid Albalawi
- Department of Computer Science, College of Computer Science and Information Technology, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Arastu Thakur
- Department of Computer Science and Engineering, Faculty of Engineering and Technology, JAIN (Deemed-to-be University), Bangalore, India
| | - Mahesh Thyluru Ramakrishna
- Department of Computer Science and Engineering, Faculty of Engineering and Technology, JAIN (Deemed-to-be University), Bangalore, India
| | - Surbhi Bhatia Khan
- Department of Data Science, School of Science, Engineering and Environment, University of Salford, Salford, United Kingdom
- Department of Electrical and Computer Engineering, Lebanese American University, Byblos, Lebanon
| | - Suresh SankaraNarayanan
- Department of Computer Science, College of Computer Science and Information Technology, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Badar Almarri
- Department of Computer Science, College of Computer Science and Information Technology, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Theyazn Hassn Hadi
- Applied College in Abqaiq, King Faisal University, Al-Ahsa, Saudi Arabia
| |
Collapse
|
41
|
Liu B, Duan H, Liu Z, Liu Y, Chu H. DNA-functionalized metal or metal-containing nanoparticles for biological applications. Dalton Trans 2024; 53:839-850. [PMID: 38108230 DOI: 10.1039/d3dt03614f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
The conjugation of DNA molecules with metal or metal-containing nanoparticles (M/MC NPs) has resulted in a number of new hybrid materials, enabling a diverse range of novel biological applications in nanomaterial assembly, biosensor development, and drug/gene delivery. In such materials, the molecular recognition, gene therapeutic, and structure-directing functions of DNA molecules are coupled with M/MC NPs. In turn, the M/MC NPs have optical, catalytic, pore structure, or photodynamic/photothermal properties, which are beneficial for sensing, theranostic, and drug loading applications. This review focuses on the different DNA functionalization protocols available for M/MC NPs, including gold NPs, upconversion NPs, metal-organic frameworks, metal oxide NPs and quantum dots. The biological applications of DNA-functionalized M/MC NPs in the treatment or diagnosis of cancers are discussed in detail.
Collapse
Affiliation(s)
- Bei Liu
- College of Science, Minzu University of China, 27 Zhongguancun South Avenue, Beijing 100081, China
| | - Huijuan Duan
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China.
| | - Zechao Liu
- College of Science, Minzu University of China, 27 Zhongguancun South Avenue, Beijing 100081, China
| | - Yuechen Liu
- College of Science, Minzu University of China, 27 Zhongguancun South Avenue, Beijing 100081, China
| | - Hongqian Chu
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China.
| |
Collapse
|
42
|
Sarani M, Roostaee M, Adeli-Sardou M, Kalantar-Neyestanaki D, Mousavi SAA, Amanizadeh A, Barani M, Amirbeigi A. Green synthesis of Ag and Cu-doped Bismuth oxide nanoparticles: Revealing synergistic antimicrobial and selective cytotoxic potentials for biomedical advancements. J Trace Elem Med Biol 2024; 81:127325. [PMID: 37922658 DOI: 10.1016/j.jtemb.2023.127325] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/15/2023] [Accepted: 10/19/2023] [Indexed: 11/07/2023]
Abstract
BACKGROUND Nanotechnology has emerged as a transformative realm of exploration across diverse scientific domains. A particular focus lies on metal oxide nanoparticles, which boast distinctive physicochemical attributes on the nanoscale. Of note, green synthesis has emerged as a promising avenue, leveraging plant extracts as both reduction and capping agents. This approach offers environmentally friendly and cost-effective avenues for generating monodispersed nanoparticles with precise morphologies. METHODS In this investigation, we embarked on the synthesis of Bismuth oxide nanoparticles, both in their pure form and doped with silver (Ag) and copper (Cu). This synthesis harnessed the potential of Biebersteinia multifida extract as a versatile reducing agent. To comprehensively characterize the synthesized nanoparticles, a suite of analytical techniques was employed, including energy-dispersive X-ray spectroscopy, field-emission scanning electron microscopy (FESEM), X-ray diffraction (XRD), Fourier-transform infrared spectroscopy (FT-IR), UV-Vis spectroscopy, and Raman spectroscopy. RESULTS The synthesized nanoparticles underwent a rigorous assessment. Their antibacterial attributes were probed, revealing a pronounced enhancement in antibiofilm activity against Pseudomonas aeruginosa and Staphylococcus aureus bacteria upon metal nanoparticle doping. Furthermore, their potential for combating cancer was scrutinized, with the nanoparticles exhibiting selective cytotoxicity towards cancer cells, U87, compared to normal 3T3 cells. Notably, among the doped nanoparticles, Cu-doped variants demonstrated the highest potency, further underscoring their promising potential. CONCLUSION In conclusion, the present study underscores the efficacy of green synthesized Bismuth oxide nanoparticles, particularly those doped with Ag and Cu, in augmenting antibacterial efficacy, bolstering biofilm inhibition, and manifesting selective cytotoxicity against cancer cells. These findings portend a promising trajectory for these nanoparticles in the spheres of biomedicine and therapeutics. As we look ahead, a deeper elucidation of their mechanistic underpinnings and in vivo investigations are essential to fully unlock their potential for forthcoming biomedical applications.
Collapse
Affiliation(s)
- Mina Sarani
- Zabol Medicinal Plants Research Center, Zabol University of Medical Sciences, Zabol, Iran
| | - Maryam Roostaee
- Department of Chemistry, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Mahboubeh Adeli-Sardou
- Herbal and Traditional Medicines Research Center, Kerman University of Medical Sciences, Kerman, Iran; Department of Biotechnology, Institute of Science and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| | - Davood Kalantar-Neyestanaki
- Medical Mycology and Bacteriology Research Center, Kerman University of Medical Sciences, Kerman, Iran; Department of Medical Microbiology (Bacteriology and virology), Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman 7616913555, Iran
| | - Seyed Amin Ayatollahi Mousavi
- Medical Mycology and Bacteriology Research Center, Kerman University of Medical Sciences, Kerman, Iran; Department of Medical Mycology and Parasitology, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Azam Amanizadeh
- Department of Medical Mycology and Parasitology, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahmood Barani
- Medical Mycology and Bacteriology Research Center, Kerman University of Medical Sciences, Kerman, Iran.
| | - Alireza Amirbeigi
- Department of General Surgery, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
43
|
Fan H, Zhou Y, Zhang Z, Zhou G, Yuan C. ROR1-AS1: A Meaningful Long Noncoding RNA in Oncogenesis. Mini Rev Med Chem 2024; 24:1884-1893. [PMID: 38859780 DOI: 10.2174/0113895575294482240530154620] [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: 12/31/2023] [Revised: 04/27/2024] [Accepted: 05/06/2024] [Indexed: 06/12/2024]
Abstract
Long noncoding RNA (lncRNA) is a non-coding RNA with a length of more than 200 nucleotides, involved in multiple regulatory processes in vivo, and is related to the physiology and pathology of human diseases. An increasing number of experimental results suggest that when lncRNA is abnormally expressed, it results in the development of tumors. LncRNAs can be divided into five broad categories: sense, antisense, bidirectional, intronic, and intergenic. Studies have found that some antisense lncRNAs are involved in a variety of human tumorigenesis. The newly identified ROR1-AS1, which functions as an antisense RNA of ROR1, is located in the 1p31.3 region of the human genome. Recent studies have reported that abnormal expression of lncRNA ROR1-AS1 can affect cell growth, proliferation, invasion, and metastasis and increase oncogenesis and tumor spread, indicating lncRNA ROR1-AS1 as a promising target for many tumor biological therapies. In this study, the pathophysiology and molecular mechanism of ROR1-AS1 in various malignancies are discussed by retrieving the related literature. ROR1-AS1 is a cancer-associated lncRNA, and studies have found that it is either over- or underexpressed in multiple malignancies, including liver cancer, colon cancer, osteosarcoma, glioma, cervical cancer, bladder cancer, lung adenocarcinoma, and mantle cell lymphoma. Furthermore, it has been demonstrated that lncRNA ROR1-AS1 participates in proliferation, migration, invasion, and suppression of apoptosis of cancer cells. Furthermore, lncRNA ROR1-AS1 promotes the development of tumors by up-regulating or downregulating ROR1-AS1 conjugates and various pathways and miR-504, miR-4686, miR-670-3p, and miR-375 sponges, etc., suggesting that lncRNA ROR1-AS1 may be used as a marker in tumors or a potential therapeutic target for a variety of tumors.
Collapse
Affiliation(s)
- Hong Fan
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, China
- College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, China
| | - Yunxi Zhou
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, China
- College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, China
| | - Ziyan Zhang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, China
- College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, China
| | - Gang Zhou
- College of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- Yichang Hospital of Traditional Chinese Medicine, Yichang, 443002, China
| | - Chengfu Yuan
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, China
- College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, China
| |
Collapse
|
44
|
Shakoor A, Alam A, Jan F, Khan M, Ali M, Ullah S, Khan A, AlAsmari AF, Alasmari F, Al-Ghafri A, Al-Harrasi A. Novel benzimidazole derivatives as effective inhibitors of prolyl oligopeptidase: synthesis, in vitro and in silico analysis. Future Med Chem 2024; 16:43-58. [PMID: 38054466 DOI: 10.4155/fmc-2023-0267] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 11/10/2023] [Indexed: 12/07/2023] Open
Abstract
Background: This research aims to discover novel derivatives having potential therapeutic applications in treating conditions related to prolyl oligopeptidase (POP) dysfunction. Method: Novel benzimidazole derivatives have been synthesized, characterized and screened for their in vitro POP inhibition. Results: All these derivatives showed excellent-to-good inhibitory activities in the range of IC50 values of 3.61 ± 0.15 to 43.72 ± 1.18 μM, when compared with standard Z-prolyl-prolinal. The docking analysis revealed the strong interactions between our compounds and the target enzyme, providing critical insights into their binding affinities and potential implications for drug development. Conclusion: The significance of these compounds in targeting POP enzyme offers promising prospects for future research in the field of neuropharmacology.
Collapse
Affiliation(s)
- Abdul Shakoor
- Department of Chemistry, Abdul Wali Khan University, Mardan, 23200, Pakistan
| | - Aftab Alam
- Department of Chemistry, University of Malakand, PO Box 18800, Khyber Pakhtunkhwa, Pakistan
| | - Faheem Jan
- Shenyang National Laboratory for Materials Science, Institute of Metal Research Chinese Academy of Sciences, Shenyang, Liaoning, 110016, People's Republic of China
| | - Momin Khan
- Department of Chemistry, Abdul Wali Khan University, Mardan, 23200, Pakistan
| | - Mumtaz Ali
- Department of Chemistry, University of Malakand, PO Box 18800, Khyber Pakhtunkhwa, Pakistan
| | - Saeed Ullah
- Natural and Medical Sciences Research Center, University of Nizwa, PO Box 33, Nizwa, Oman
| | - Ajmal Khan
- Natural and Medical Sciences Research Center, University of Nizwa, PO Box 33, Nizwa, Oman
| | - Abdullah F AlAsmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Fawaz Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Ahmed Al-Ghafri
- Natural and Medical Sciences Research Center, University of Nizwa, PO Box 33, Nizwa, Oman
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa, PO Box 33, Nizwa, Oman
| |
Collapse
|
45
|
Ayaz M, Alam A, Zainab, Assad M, Javed A, Islam MS, Rafiq H, Ali M, Ahmad W, Khan A, Latif A, Al-Harrasi A, Ahmad M. Biooriented Synthesis of Ibuprofen-Clubbed Novel Bis-Schiff Base Derivatives as Potential Hits for Malignant Glioma: In Vitro Anticancer Activity and In Silico Approach. ACS OMEGA 2023; 8:49228-49243. [PMID: 38173864 PMCID: PMC10764114 DOI: 10.1021/acsomega.3c07216] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/18/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024]
Abstract
This research work is based on the synthesis of bis-Schiff base derivatives of the commercially available ibuprofen drug in outstanding yields through multistep reactions. Structures of the synthesized compounds were confirmed by the help of modern spectroscopic techniques including high-resolution electrospray ionization mass spectrometry (HR-ESI-MS), 1H NMR, and 13C NMR. The synthesized compounds were evaluated for their anticancer activity using a normal human embryonic kidney HEK293 cell and U87-malignant glioma (ATCC-HTB-14) as a cancer cell line. All of the synthesized compounds among the series exhibited excellent to less antiproliferative activity having IC50 values ranging from 5.75 ± 0.43 to 150.45 ± 0.20 μM. Among them, compound 5e (IC50 = 5.75 ± 0.43 μM) was found as the most potent antiprolifarative agent, while 5f, 5b, 5a, 5n, 5r, 5s, 5g, 5q, 5i, and 5j exhibited good activity with IC50 values from 24.17 ± 0.46 to 43.71 ± 0.07 μM. These findings suggest that these cells (HEK293) are less cytotoxic to the activities of compounds and increase the cancer cell death in brain, while the lower cytotoxicity of the potent compounds in noncancerous cells suggests that these derivatives will provide promising treatment for patients suffering from brain cancer. The results of the docking study exposed a promising affinity of the active compounds toward casein kinase-2 enzyme, which shows green signal for cancer treatment.
Collapse
Affiliation(s)
- Muhammad Ayaz
- Department
of Chemistry, University of Malakand, Dir Lower, Khyber Pakhtunkhwa 18800, Pakistan
| | - Aftab Alam
- Department
of Chemistry, University of Malakand, Dir Lower, Khyber Pakhtunkhwa 18800, Pakistan
| | - Zainab
- College
of Chemistry and Materials Science, Hebei
Normal University, Shijiazhuang 050024, China
| | - Mohammad Assad
- Department
of Biochemistry, Abdul Wali Khan University
Mardan, Khyber
Pakhtunkhwa 23200, Pakistan
| | - Aneela Javed
- Molecular
Immunology Laboratory, Department of Healthcare Biotechnology Atta-Ur-Rahman
School of Applied Biosciences, National
University of Sciences and Technology, H-12 Campus, Islamabad 44000, Pakistan
| | - Mohammad Shahidul Islam
- Department
of Chemistry, College of Science, King Saud
University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Huma Rafiq
- Molecular
Immunology Laboratory, Department of Healthcare Biotechnology Atta-Ur-Rahman
School of Applied Biosciences, National
University of Sciences and Technology, H-12 Campus, Islamabad 44000, Pakistan
| | - Mumtaz Ali
- Department
of Chemistry, University of Malakand, Dir Lower, Khyber Pakhtunkhwa 18800, Pakistan
| | - Waqar Ahmad
- Department
of Chemistry, University of Malakand, Dir Lower, Khyber Pakhtunkhwa 18800, Pakistan
| | - Ajmal Khan
- Natural and
Medical Sciences Research Center, University
of Nizwa, P.O. Box 33, Birkat Al Mauz, PC 616 Nizwa, Sultanate of Oman
| | - Abdul Latif
- Department
of Chemistry, University of Malakand, Dir Lower, Khyber Pakhtunkhwa 18800, Pakistan
| | - Ahmed Al-Harrasi
- Natural and
Medical Sciences Research Center, University
of Nizwa, P.O. Box 33, Birkat Al Mauz, PC 616 Nizwa, Sultanate of Oman
| | - Manzoor Ahmad
- Department
of Chemistry, University of Malakand, Dir Lower, Khyber Pakhtunkhwa 18800, Pakistan
| |
Collapse
|
46
|
Hu Y, Liu Y, Zong L, Zhang W, Liu R, Xing Q, Liu Z, Yan Q, Li W, Lei H, Liu X. The multifaceted roles of GSDME-mediated pyroptosis in cancer: therapeutic strategies and persisting obstacles. Cell Death Dis 2023; 14:836. [PMID: 38104141 PMCID: PMC10725489 DOI: 10.1038/s41419-023-06382-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 12/01/2023] [Accepted: 12/06/2023] [Indexed: 12/19/2023]
Abstract
Pyroptosis is a novel regulated cell death (RCD) mode associated with inflammation and innate immunity. Gasdermin E (GSDME), a crucial component of the gasdermin (GSDM) family proteins, has the ability to convert caspase-3-mediated apoptosis to pyroptosis of cancer cells and activate anti-tumor immunity. Accumulating evidence indicates that GSDME methylation holds tremendous potential as a biomarker for early detection, diagnosis, prognosis, and treatment of tumors. In fact, GSDME-mediated pyroptosis performs a dual role in anti-tumor therapy. On the one side, pyroptotic cell death in tumors caused by GSDME contributes to inflammatory cytokines release, which transform the tumor immune microenvironment (TIME) from a 'cold' to a 'hot' state and significantly improve anti-tumor immunotherapy. However, due to GSDME is expressed in nearly all body tissues and immune cells, it can exacerbate chemotherapy toxicity and partially block immune response. How to achieve a balance between the two sides is a crucial research topic. Meanwhile, the potential functions of GSDME-mediated pyroptosis in anti-programmed cell death protein 1 (PD-1) therapy, antibody-drug conjugates (ADCs) therapy, and chimeric antigen receptor T cells (CAR-T cells) therapy have not yet been fully understood, and how to improve clinical outcomes persists obscure. In this review, we systematically summarize the latest research regarding the molecular mechanisms of pyroptosis and discuss the role of GSDME-mediated pyroptosis in anti-tumor immunity and its potential applications in cancer treatment.
Collapse
Affiliation(s)
- Yixiang Hu
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Ya Liu
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Lijuan Zong
- Department of Rehabilitation Medicine, Zhongda Hospital of Southeast University, Nanjing, 210096, China
| | - Wenyou Zhang
- Department of Pharmacy, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Renzhu Liu
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Qichang Xing
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Zheng Liu
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Qingzi Yan
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Wencan Li
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Haibo Lei
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China.
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China.
| | - Xiang Liu
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China.
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China.
| |
Collapse
|
47
|
Wu Y, Wang X, Yang L, Kang S, Yan G, Han Y, Fang H, Sun H. Potential of alisols as cancer therapeutic agents: Investigating molecular mechanisms, pharmacokinetics and metabolism. Biomed Pharmacother 2023; 168:115722. [PMID: 37865991 DOI: 10.1016/j.biopha.2023.115722] [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: 08/14/2023] [Revised: 10/11/2023] [Accepted: 10/11/2023] [Indexed: 10/24/2023] Open
Abstract
Albeit remarkable achievements in anti-cancer endeavors, the prevention and treatment of cancer remain unresolved challenges. Hence, there is an urgent need to explore new and efficacious natural compounds with potential anti-cancer therapeutic agents. One such group of compounds is alisols, tetracyclic triterpene alcohols extracted from alisma orientale. Alisols play a significant role in cancer therapy as they can suppress cancer cell proliferation and migration by regulating signaling pathways such as mTOR, Bax/Bcl-2, CHOP, caspase, NF-kB and IRE1. Pharmacokinetic studies showed that alisols can be absorbed entirely, rapidly, and evenly distributed in vivo. Moreover, alisols are low in toxicity and relatively safe to take. Remarkably, each alisol can be converted into many compounds with different pathways to their anti-cancer effects in the body. Thus, alisols are regarded as promising anti-cancer agents with minimal side effects and low drug resistance. This review will examine and discuss alisols' anti-cancer molecular mechanism, pharmacokinetics and metabolism. Based on a comprehensive analysis of nearly 20 years of research, we evaluate the therapeutic potential of alisols for various types of cancer and offer insights and strategies for developing new cancer treatments.
Collapse
Affiliation(s)
- Yinqi Wu
- State key laboratory of Integration and Innovation of Classical formula and modern Chinese medicine, National Chinmedomics Research Center, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Xijun Wang
- State key laboratory of Integration and Innovation of Classical formula and modern Chinese medicine, National Chinmedomics Research Center, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau; State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou, China.
| | - Le Yang
- State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou, China
| | - Shuyu Kang
- State key laboratory of Integration and Innovation of Classical formula and modern Chinese medicine, National Chinmedomics Research Center, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Guangli Yan
- State key laboratory of Integration and Innovation of Classical formula and modern Chinese medicine, National Chinmedomics Research Center, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Ying Han
- State key laboratory of Integration and Innovation of Classical formula and modern Chinese medicine, National Chinmedomics Research Center, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Heng Fang
- State key laboratory of Integration and Innovation of Classical formula and modern Chinese medicine, National Chinmedomics Research Center, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Hui Sun
- State key laboratory of Integration and Innovation of Classical formula and modern Chinese medicine, National Chinmedomics Research Center, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China.
| |
Collapse
|
48
|
Kumar M, Kumar S, Chakrabartty S, Poulose A, Mostafa H, Goyal B. Dispersive Modeling of Normal and Cancerous Cervical Cell Responses to Nanosecond Electric Fields in Reversible Electroporation Using a Drift-Step Rectifier Diode Generator. MICROMACHINES 2023; 14:2136. [PMID: 38138305 PMCID: PMC10745406 DOI: 10.3390/mi14122136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/16/2023] [Accepted: 11/18/2023] [Indexed: 12/24/2023]
Abstract
This paper creates an approximate three-dimensional model for normal and cancerous cervical cells using image processing and computer-aided design (CAD) tools. The model is then exposed to low-frequency electric pulses to verify the work with experimental data. The transmembrane potential, pore density, and pore radius evolution are analyzed. This work adds a study of the electrodeformation of cells under an electric field to investigate cytoskeleton integrity. The Maxwell stress tensor is calculated for the dispersive bi-lipid layer plasma membrane. The solid displacement is calculated under electric stress to observe cytoskeleton integrity. After verifying the results with previous experiments, the cells are exposed to a nanosecond pulsed electric field. The nanosecond pulse is applied using a drift-step rectifier diode (DSRD)-based generator circuit. The cells' transmembrane voltage (TMV), pore density, pore radius evolution, displacement of the membrane under electric stress, and strain energy are calculated. A thermal analysis of the cells under a nanosecond pulse is also carried out to prove that it constitutes a non-thermal process. The results showed differences in normal and cancerous cell responses to electric pulses due to changes in morphology and differences in the cells' electrical and mechanical properties. This work is a model-driven microdosimetry method that could be used for diagnostic and therapeutic purposes.
Collapse
Affiliation(s)
- Mayank Kumar
- Technical Research Analyst (TRA), Electronics/Biomedical Engineering, Aranca, Mumbai 400076, Maharastra, India;
| | - Sachin Kumar
- Department of Electronics and Communication Engineering, Galgotias College of Engineering and Technology, Greater Noida 201310, Uttar Pradesh, India;
| | - Shubhro Chakrabartty
- School of Computer Science Engineering and Applications, D Y Patil International University, Pune 411044, Maharastra, India
| | - Alwin Poulose
- School of Data Science, Indian Institute of Science Education and Research Thiruvananthapuram (IISER TVM), Vithura, Thiruvananthapuram 695551, Kerala, India
| | - Hala Mostafa
- Department of Information Technology, College of Computer and Information Sciences, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia;
| | - Bhawna Goyal
- University Centre for Research and Development, Chandigarh University, Gharuan, Mohali 140413, Punjab, India;
| |
Collapse
|
49
|
He SY, Liu RP, Wang CR, Wang XQ, Wang J, Xu YN, Kim NH, Han DW, Li YH. Improving the developmental competences of porcine parthenogenetic embryos by Notoginsenoside R1-induced enhancement of mitochondrial activity and alleviation of proapoptotic events. Reprod Domest Anim 2023; 58:1583-1594. [PMID: 37696770 DOI: 10.1111/rda.14474] [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: 05/13/2023] [Revised: 08/08/2023] [Accepted: 09/01/2023] [Indexed: 09/13/2023]
Abstract
Notoginsenoside R1 (NGR1), derived from the Panax notoginseng root and rhizome, exhibits diverse pharmacological influences on the brain, neurons, and osteoblasts, such as antioxidant effects, mitochondrial function protection, energy metabolism regulation, and inhibition of oxygen radicals, apoptosis, and cellular autophagy. However, its effect on early porcine embryonic development remains unclear. Therefore, we investigated NGR1's effects on blastocyst quality, reactive oxygen species (ROS) levels, glutathione (GSH) levels, mitochondrial function, and embryonic development-related gene expression in porcine embryos by introducing NGR1 during the in vitro culture (IVC) of early porcine embryos. Our results indicate that an addition of 1 μM NGR1 significantly increased glutathione (GSH) levels, blastocyst formation rate, and total cell number and proliferation capacity; decreased ROS levels and apoptosis rates in orphan-activated porcine embryos; and improved intracellular mitochondrial distribution, enhanced membrane potential, and reduced autophagy. In addition, pluripotency-related factor levels were elevated (NANOG and octamer-binding transcription factor 4 [OCT4]), antioxidant-related genes were upregulated (nuclear factor-erythroid 2-related factor 2 [NRF2]), and apoptosis- (caspase 3 [CAS3]) and autophagy-related genes (light chain 3 [LC3B]) were downregulated. These results indicate that NGR1 can enhance early porcine embryonic development by protecting mitochondrial function.
Collapse
Affiliation(s)
- Sheng-Yan He
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Rong-Ping Liu
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Chao-Rui Wang
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Xin-Qin Wang
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Jing Wang
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Yong-Nan Xu
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Nam-Hyung Kim
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Dong-Wook Han
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Ying-Hua Li
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| |
Collapse
|
50
|
Wang S, Guo Q, Xu R, Lin P, Deng G, Xia X. Combination of ferroptosis and pyroptosis dual induction by triptolide nano-MOFs for immunotherapy of Melanoma. J Nanobiotechnology 2023; 21:383. [PMID: 37858186 PMCID: PMC10585872 DOI: 10.1186/s12951-023-02146-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 10/09/2023] [Indexed: 10/21/2023] Open
Abstract
Immunotherapy has good potential to eradicate tumors in the long term. However, due to the low immunogenicity of tumor cells, current cancer immunotherapies are not effective. To address this limitation, we constructed a BSA-FA functionalized iron-containing metal-organic framework (TPL@TFBF) that triggers a potent systemic anti-tumor immune response by inducing ferroptosis and pyroptosis in tumor cells and releasing large quantities of damage-associated molecular patterns (DAMPs) to induce immunogenicity, and showing excellent efficacy against melanoma lung metastases in vivo. This nanoplatform forms a metal-organic framework through the coordination between tannic acid (TA) and Fe3+ and is then loaded with triptolide (TPL), which is coated with FA-modified BSA. The nanoparticles target melanoma cells by FA modification, releasing TPL, Fe3+ and TA. Fe3+ is reduced to Fe2+ by TA, triggering the Fenton reaction and resulting in ROS production. Moreover, TPL increases the production of intracellular ROS by inhibiting the expression of nuclear factor erythroid-2 related factor (Nrf2). Such simultaneous amplification of intracellular ROS induces the cells to undergo ferroptosis and pyroptosis, releasing large amounts of DAMPs, which stimulate antigen presentation of dendritic cells (DCs) and the proliferation of cytotoxic T lymphocytes (CD4+/CD8 + T cells) to inhibit tumor and lung metastasis. In addition, combining nanoparticle treatment with immune checkpoint blockade (ICB) further inhibits melanoma growth. This work provides a new strategy for tumor immunotherapy based on various combinations of cell death mechanisms.
Collapse
Affiliation(s)
- Shengmei Wang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Qiuyan Guo
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Rubing Xu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Peng Lin
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Guoyan Deng
- The First Hospital of Hunan University of Chinese Medicine, Changsha, 410007, Hunan, China
| | - Xinhua Xia
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
| |
Collapse
|