1
|
Yan W, Xiang S, Feng J, Zu X. Role of ubiquitin-specific proteases in programmed cell death of breast cancer cells. Genes Dis 2025; 12:101341. [PMID: 40083330 PMCID: PMC11904532 DOI: 10.1016/j.gendis.2024.101341] [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: 01/20/2024] [Revised: 03/26/2024] [Accepted: 04/11/2024] [Indexed: 03/16/2025] Open
Abstract
Breast cancer (BC) is the most common malignant tumor and the leading cause of cancer-related deaths among women worldwide. Great progress has been recently achieved in controlling breast cancer; however, mortality from breast cancer remains a substantial challenge, and new treatment mechanisms are being actively sought. Programmed cell death (PCD) is associated with the progression and treatment of many types of human cancers. PCD can be divided into multiple pathways including autophagy, apoptosis, mitotic catastrophe, necroptosis, ferroptosis, pyroptosis, and anoikis. Ubiquitination is a post-translational modification process in which ubiquitin, a 76-amino acid protein, is coupled to the lysine residues of other proteins. Ubiquitination is involved in many physiological events and promotes cancer development and progression. This review elaborates the role of ubiquitin-specific protease (USP) in programmed cell death, which is common in breast cancer cells, and lays the foundation for tumor diagnosis and targeted therapy.
Collapse
Affiliation(s)
| | | | - Jianbo Feng
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001 Hunan, China
| | - Xuyu Zu
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001 Hunan, China
| |
Collapse
|
2
|
Nie X, Miao S, Hou Y, Ma Y, Li M, Liu Y, Yang Y, Xu J, Wang Y. TLR4-mediated endoplasmic reticulum stress regulates pyroptosis in macrophages infected with the Bacillus Calmette-Guérin mycobacterial. Int Immunopharmacol 2025; 152:114346. [PMID: 40064059 DOI: 10.1016/j.intimp.2025.114346] [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: 11/13/2024] [Revised: 02/11/2025] [Accepted: 02/18/2025] [Indexed: 03/24/2025]
Abstract
Tuberculosis results from Mycobacterium tuberculosis (Mtb) infection. Immune responses controlled by Toll-like receptor 4 (TLR4) are closely associated with the host response to pathogens, including Mtb. NLRP3 inflammasome-mediated pyroptosis forms a significant part of the inflammatory response during Mtb infection, and endoplasmic reticulum stress (ERS) is implicated in the activation of the NLRP3 inflammasome. Here, the function of TLR4 in macrophage pyroptosis induced by infection with the Bacillus Calmette-Guérin (BCG) mycobacterial strain was investigated. It was found that infection with BCG activated TLR4 signaling, induced ERS and subsequent NLRP3 inflammasome activation, leading to pyroptosis in mouse lung tissues. The TLR4 inhibitor TAK 242 inhibited the ERS onset, NLRP3 inflammasome stimulation, and pyroptosis, while the ERS inhibitor TUDCA blocked both inflammasome activation and pyroptosis, and the NLRP3 inhibitor MCC950 specifically inhibited pyroptosis. Furthermore, TAK 242, TUDCA, and MCC950 all exacerbated lung injury caused by BCG infection and promoted BCG survival. Similarly, after in BCG-infected THP-1 macrophages, TLR4 signaling was found to mediate NLRP3 inflammasome activation through ERS, thereby inducing pyroptosis. In summary, BCG infection leads to macrophage pyroptosis via the TLR4/ERS/NLRP3 inflammasome signaling axis, providing new insights for further research into the pathogenesis and treatment of tuberculosis.
Collapse
Affiliation(s)
- Xueyi Nie
- School of Life Sciences, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China
| | - Shen'ao Miao
- School of Life Sciences, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China
| | - Yuxin Hou
- School of Life Sciences, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China
| | - Yabo Ma
- School of Life Sciences, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China
| | - Mengyuan Li
- School of Life Sciences, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China
| | - Yueyang Liu
- School of Life Sciences, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China
| | - Yi Yang
- School of Life Sciences, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China
| | - Jinrui Xu
- School of Life Sciences, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China.
| | - Yujiong Wang
- School of Life Sciences, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China.
| |
Collapse
|
3
|
Chen Q, Feng G, Shen Y, Li X, Pei Q, Wang H, Tian L, Cao Y, Wu J, Yang H, Mu L. An Anionic Cathelicidin Exerts Antimelanoma Effects in Mice by Promoting Pyroptosis. J Med Chem 2025. [PMID: 40207383 DOI: 10.1021/acs.jmedchem.5c00281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
While cationic antimicrobial peptides (AMPs) are extensively studied for antitumor effects, anionic AMPs remain underexplored. Notably, no amphibian-derived anionic cathelicidins with antitumor activity have been reported. This study identifies Boma-CATH, a novel anionic cathelicidin (net charge-3) from Bombina maxima skin, which suppresses melanoma growth in mice and triggers pyroptosis-like morphological changes in A375 cells via the NLRP3/Caspase-1/GSDMD pathway. Further investigation revealed that ROS played a crucial role in promoting pyroptosis, as NAC (ROS scavenger) and Ac-YVAD-cmk (Caspase-1 inhibitor) reversed cell death and reduced LDH/IL-1β release in vitro and in vivo. GSDMD knockdown further validated its role. Additionally, Boma-CATH inhibited A375 cell proliferation, migration, and invasion, demonstrating dual antitumor mechanisms: pyroptosis induction and metastasis suppression. Importantly, Boma-CATH caused no adverse effects in mice, highlighting its therapeutic safety. These findings position Boma-CATH as a promising melanoma treatment and expand the mechanistic understanding of anionic AMPs in oncology.
Collapse
Affiliation(s)
- Qian Chen
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Guizhu Feng
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Yan Shen
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Xiang Li
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Qiqi Pei
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Hanying Wang
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Li Tian
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Yuanyuan Cao
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Jing Wu
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Hailong Yang
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Lixian Mu
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| |
Collapse
|
4
|
Owida HA, Abed AY, Altalbawy FMA, H M, Abbot V, Jakhonkulovna SM, Mohammad SI, Vasudevan A, Khalaf RM, Zwamel AH. NLRP3 inflammasome-based therapies by natural products: a new development in the context of cancer therapy. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04030-0. [PMID: 40116873 DOI: 10.1007/s00210-025-04030-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/06/2025] [Indexed: 03/23/2025]
Abstract
The leucine-rich repeat containing protein (NLR) canonical inflammasome family includes Nod-like receptor protein 3 (NLRP3). Via the mediation of apoptosis proteins and immunological reactions, it controls the pathogenesis of malignancy. Experimental studies showed a relationship among lymphogenesis, cancer metastasis, and NLRP3 expression. Natural products have also been used as lead-based substances in a number of investigations to speed up the creation of novel, specific NLRP3 inhibitors. Via the mediation of apoptotic proteins and immunological responses, it controls the pathogenesis of malignancy. Moreover, it was recently noted that among human cancers, chemotherapy activates NLRP3. Induction of NLRP3 could encourage the generation of IL-1β and IL-22 to facilitate the propagation of malignancy. Additionally, prior research has demonstrated that the usage of NLRP3 in cancer therapy may result in resistance to drugs. The depletion of NLRP3 could affect the survival of cells. Natural products have been used as lead materials in a number of studies to help generate novel, specific NLRP3 antagonists more quickly. In the present review, we examine the mechanism behind the beneficial effects of the natural substances on the inhibition of cancer growth and progression, with special focus on NLRP3 regulation.
Collapse
Affiliation(s)
- Hamza Abu Owida
- Department of Medical Engineering, Faculty of Engineering, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Ahmed Yaseen Abed
- Department of Medical Laboratories Techniques, College of Health and Medical Technology, University of Al Maarif, Ramadi, Al Anbar, 31001, Iraq.
| | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia
| | - Malathi H
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Vikrant Abbot
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjeri, Mohali, 140307, Punjab, India
| | | | - Suleiman Ibrahim Mohammad
- Electronic Marketing and Social Media, Economic and Administrative Sciences, Zarqa University, Zarqa, Jordan
- Faculty of Business and Communications, INTI International University, 71800, Negeri Sembilan, Malaysia
| | - Asokan Vasudevan
- Faculty of Business and Communications, INTI International University, 71800, Negeri Sembilan, Malaysia
| | | | - Ahmed Hussein Zwamel
- Department of Medical Analysis, Medical Laboratory Technique College, the Islamic University, Najaf, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, the Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
5
|
Kuriakose BB, Zwamel AH, Mutar AA, Uthirapathy S, Bishoyi AK, Naidu KS, Hjazi A, Nakash P, Arya R, Almalki SG. The critical role of NLRP3 in drug resistance of cancers: Focus on the molecular mechanisms and possible therapeutics. Semin Oncol 2025; 52:27-40. [PMID: 40037148 DOI: 10.1016/j.seminoncol.2025.152337] [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: 12/24/2024] [Revised: 02/08/2025] [Accepted: 02/12/2025] [Indexed: 03/06/2025]
Abstract
Nod-like receptor protein 3 (NLRP3) is a member of the leucine-rich repeat-containing protein (NLR) canonical inflammasome family. It regulates the pathophysiology of cancer by facilitating immune responses and apoptotic proteins. Furthermore, it has been observed that chemotherapy activates NLRP3 in human malignancies. The secretion of IL-1β and IL-22 to promote cancer spread may be triggered by NLRP3 activation. Furthermore, earlier studies have exhibited that NLRP3 may cause medication resistance when used in cancer treatments given that cell viability may be regulated by NLRP3 depletion. Additionally, clinical studies have demonstrated correlation between NLRP3 expression, lymphogenesis, and cancer metastasis. Various NLRP3 agonists may cause the EMT process, stimulate IL-1β and Wnt/β-catenin signaling, and alter miRNA function in drug-resistant cells. This review seeks to clarify the possibility involvement of NLRP3-related pathways in the control of cancer cells' resistance to widely used treatment approaches, such as chemotherapy. In the end, an improved perception of the corresponding mechanisms behind NLRP3's tumor-supporting activities will help NLRP3-based treatments advance in the future.
Collapse
Affiliation(s)
- Beena Briget Kuriakose
- Department of Basic Medical Sciences, College of Applied Medical Sciences, King khalid University, Khamis Mushayt, Kingdom of Saudi Arabia
| | - Ahmed Hussein Zwamel
- Department of medical analysis, Medical laboratory technique college, the Islamic University, Najaf, Iraq; Department of medical analysis, Medical laboratory technique college, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq; Department of medical analysis, Medical laboratory technique college, the Islamic University of Babylon, Babylon, Iraq
| | - Ayad Abdulrazzaq Mutar
- Medical Laboratory Techniques department, College of Health and medical technology, Al-maarif University, Anbar, Iraq.
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Ashok Kumar Bishoyi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, Gujarat, India
| | - K Satyam Naidu
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, India
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Princse Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Prashant Nakash
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, India
| | - Renu Arya
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, India
| | - Sami G Almalki
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah, Saudi Arabia
| |
Collapse
|
6
|
Guo Y, Min J, Chang B, Chen Z, Chen W. Exploring the Role of TRAF6-TAK1 Pathway in Podocyte Pyroptosis and Its Implications for Primary Membranous Nephropathy Therapy. Inflammation 2025:10.1007/s10753-025-02249-w. [PMID: 39883393 DOI: 10.1007/s10753-025-02249-w] [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: 08/17/2024] [Revised: 01/16/2025] [Accepted: 01/16/2025] [Indexed: 01/31/2025]
Abstract
Primary membranous nephropathy (PMN) is a prevalent renal disorder characterized by immune-mediated damage to the glomerular basement membrane, with recent studies highlighting the significant role of pyroptosis in its progression. In this study, we investigate the molecular mechanisms underlying PMN, focusing on the role of Tumor necrosis factor receptor-associated factor 6 (TRAF6) in promoting disease advancement. Specifically, we examine how TRAF6 facilitates PMN progression by inducing the ubiquitination of Transforming growth factor-beta-activated kinase 1 (TAK1), which in turn activates the Gasdermin D (GSDMD)/Caspase-1 axis, leading to podocyte pyroptosis. Utilizing transcriptomic data from the gene expression omnibus database, we identified key regulatory factors involved in pyroptosis and validated these findings through the establishment of a C3a-induced podocyte injury model and a Sprague-Dawley (SD) rat model of PMN. Our findings reveal that TRAF6 is significantly upregulated in PMN, and its interaction with TAK1 is crucial for the activation of the GSDMD/Caspase-1 axis, ultimately driving podocyte pyroptosis. Further biochemical and molecular analyses confirmed the pivotal role of the TRAF6/TAK1 signaling pathway in the pathogenesis of PMN. These results underscore the importance of TRAF6-mediated signaling in the progression of PMN and suggest that targeting the TRAF6/TAK1/GSDMD/Caspase-1 axis may offer a novel therapeutic strategy for the treatment of this debilitating renal disease.
Collapse
Affiliation(s)
- Yaling Guo
- Department of Nephrology, the First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzihu District, Bengbu, 233000, Anhui Province, China.
| | - Jingliang Min
- Department of Neurosurgery, Second Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, China
| | - Baochao Chang
- Department of Nephrology, the First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzihu District, Bengbu, 233000, Anhui Province, China
| | - Zheng Chen
- Department of Nephrology, the First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzihu District, Bengbu, 233000, Anhui Province, China
| | - Weidong Chen
- Department of Nephrology, the First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzihu District, Bengbu, 233000, Anhui Province, China
| |
Collapse
|
7
|
Zhendong Y, Changjun C, Haocheng H, Qibin L, Dailing C, Linsong T, Xuecheng S, Gong M, Lei Z. Regulation of macrophage polarization and pyroptosis by 4-methylcatechol alleviates collagen-induced arthritis via Nrf2/HO-1 and NF-κB/NLRP3 signaling pathways. Int Immunopharmacol 2025; 146:113855. [PMID: 39709906 DOI: 10.1016/j.intimp.2024.113855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/09/2024] [Accepted: 12/11/2024] [Indexed: 12/24/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease that leads to joint deformity and ultimately disability. The metabolite of quercetin, 4-Methylcatechol (4-MC), has been acknowledged for its anti-inflammatory and antioxidant properties; however, the protective effects of 4-MC on RA and its underlying mechanisms remain incompletely elucidated. In a collagen-induced arthritis (CIA) model, we observed that 4-MC effectively mitigated joint inflammation and bone destruction in CIA mice. Additionally, it significantly suppressed the upregulated expression of inflammatory cytokines in synovial tissues. Mechanistically, upon lipopolysaccharide (LPS) stimulation, 4-MC inhibited M1 polarization of macrophages and induced a phenotypic switch from M1 to M2 phenotype, thereby reducing the release of pro-inflammatory cytokines by M1 macrophages while increasing the release of anti-inflammatory cytokines by M2 macrophages. Furthermore, it attenuated LPS/adenosine triphosphate (ATP)-induced pyroptosis in macrophages by downregulating NLRP3 expression levels along with cleaved caspase-1, cleaved IL-1β, and GSDMD-NT expression levels. Notably, our findings revealed that 4-MC exerted inhibitory effects on the NF-κB signaling pathway through specific modulation of the NF-κB complex as well as phosphorylation of the upstream IKK kinase complex. Collectively, these results highlight significant therapeutic potential for utilizing 4-MC in RA treatment.
Collapse
Affiliation(s)
- Ying Zhendong
- Department of Orthopaedics Surgery, Shandong Provincial Qianfoshan Hospital, Shandong University of Traditional Chinese Medicine, Jinan 250012, PR China.
| | - Chen Changjun
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250012, PR China.
| | - Hou Haocheng
- The First Clinical College, Shandong University, Jinan 250014, PR China.
| | - Liu Qibin
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250012, PR China.
| | - Chen Dailing
- Department of Orthopaedics Surgery, Shandong Provincial Qianfoshan Hospital, Shandong Second Medical University, Jinan 250012, PR China.
| | - Teng Linsong
- Department of Orthopaedics Surgery, Shandong Provincial Qianfoshan Hospital, Shandong University of Traditional Chinese Medicine, Jinan 250012, PR China.
| | - Sun Xuecheng
- Department of Orthopedic Trauma, Weifang People's Hospital, Weifang, Shandong, PR China.
| | - Mouchun Gong
- Department of General Surgery, The First People's Hospital of Hangzhou Lin'an District (Hangzhou Medical College Affiliated Lin'an People's Hospital), Hangzhou 311300, PR China.
| | - Zhang Lei
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250012, PR China.
| |
Collapse
|
8
|
Cao Y, Wen E, Chen Q, Li X, Wang Z. Multifunctional ICG-SB@Lip-ZA Nanosystem Focuses on Remodeling the Inflammatory-Immunosuppressive Microenvironment After Photothermal Therapy to Potentiate Cancer Photothermal Immunotherapy. Adv Healthc Mater 2025; 14:e2402211. [PMID: 39440627 DOI: 10.1002/adhm.202402211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 10/14/2024] [Indexed: 10/25/2024]
Abstract
Achieving full eradication of residual tumors post photothermal therapy (PTT) hinges on the immune system's activation and response. Nevertheless, the resultant local inflammation attracts a significant influx of aberrant immune cells and fibroblasts, such as tumor-associated macrophages (TAMs) and cancer-associated fibroblasts (CAFs), following tumor PTT. This phenomenon exacerbates immune evasion and the persistence of residual tumor cells, culminating in tumor recurrence and advancement. To tackle this challenge, a combined therapeutic approach utilizing multifunctional ICG-SB@Lip-ZA nanosystem has been introduced. Indocyanine green (ICG) as a photothermal-transducer ablated tumor cells, zoledronic acid (ZA) depletes TAMs recruited by the inflammatory tumor microenvironment (mostly M2-like phenotype), SB-505124 affects CAFs proliferation in the tumor microenvironment (TME) by inhibiting the transforming growth factor-β (TGF-β) pathway, thereby removing physical barriers to T cell infiltration. In a breast cancer model, these immunomodulatory nanoliposomes markedly decrease the population of M2-like TAMs in the TME, eliminate physical barriers hindering T cell infiltration, reshape the inflammatory immune-suppressive tumor microenvironment, eventually leading to a rate of tumor eradication of 94%. This multifunctional ICG-SB@Lip-ZA nanosystem (including photothermal conversion, TAM depletion, and TGF-β pathway blockade) offers a promising strategy for mitigating the deteriorating tumor microenvironment following PTT and presents a more efficient approach for clinical photothermal-immune combination therapy.
Collapse
Affiliation(s)
- Yi Cao
- Department of ultrasound, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - E Wen
- Precision Medicine Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Qiaoqi Chen
- Department of ultrasound, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Xingsheng Li
- Department of Gerontology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Zhigang Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| |
Collapse
|
9
|
Guo X, Zuo Z, Wang X, Sun Y, Xu D, Liu G, Tong Y, Zhang Z. Epidemiology, risk factors and mechanism of breast cancer and atrial fibrillation. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:92. [PMID: 39716319 DOI: 10.1186/s40959-024-00298-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/17/2024] [Indexed: 12/25/2024]
Abstract
Cancer and cardiovascular diseases are leading causes of death worldwide. Among them, breast cancer is one of the most common malignancies in women, while atrial fibrillation is one of the most extensively studied arrhythmias, with significant public health implications. As the global population ages and advancements in cancer treatments continue, the survival rates of breast cancer patients have significantly improved, leading to an increasing coexistence of breast cancer and atrial fibrillation. However, the mechanisms underlying this coexistence remain insufficiently studied, and there is no consensus on the optimal treatment strategies for these patients. This review consolidates existing research to systematically explore the epidemiological characteristics, risk factors, and pathophysiological mechanisms of both breast cancer and atrial fibrillation. It focuses on the unique signaling pathways associated with different molecular subtypes of breast cancer and their potential impact on the mechanisms of atrial fibrillation. Additionally, the relationship between atrial fibrillation treatment medications and breast cancer is discussed. These insights not only provide essential evidence for the precise prevention and management of atrial fibrillation in breast cancer patients but also lay a solid theoretical foundation for interdisciplinary clinical management practices.
Collapse
Affiliation(s)
- Xiaoxue Guo
- Department of Cardiology, The First Hospital of Jilin University, No.1 Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Zheng Zuo
- Department of Cardiology, The First Hospital of Jilin University, No.1 Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Xishu Wang
- Department of Cardiology, The First Hospital of Jilin University, No.1 Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Ying Sun
- Department of Cardiology, The First Hospital of Jilin University, No.1 Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Dongyang Xu
- Department of Cardiology, The First Hospital of Jilin University, No.1 Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Guanghui Liu
- Department of Cardiology, The First Hospital of Jilin University, No.1 Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Yi Tong
- Department of Cardiology, The First Hospital of Jilin University, No.1 Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Zhiguo Zhang
- Department of Cardiology, The First Hospital of Jilin University, No.1 Xinmin Street, Changchun, 130021, Jilin Province, China.
| |
Collapse
|
10
|
Jalali AM, Mitchell KJ, Pompoco C, Poludasu S, Tran S, Ramana KV. Therapeutic Significance of NLRP3 Inflammasome in Cancer: Friend or Foe? Int J Mol Sci 2024; 25:13689. [PMID: 39769450 PMCID: PMC11728390 DOI: 10.3390/ijms252413689] [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: 11/15/2024] [Revised: 12/16/2024] [Accepted: 12/19/2024] [Indexed: 01/16/2025] Open
Abstract
Besides various infectious and inflammatory complications, recent studies also indicated the significance of NLRP3 inflammasome in cancer progression and therapy. NLRP3-mediated immune response and pyroptosis could be helpful or harmful in the progression of cancer, and also depend on the nature of the tumor microenvironment. The activation of NLRP3 inflammasome could increase immune surveillance and the efficacy of immunotherapy. It can also lead to the removal of tumor cells by the recruitment of phagocytic macrophages, T-lymphocytes, and other immune cells to the tumor site. On the other hand, NLRP3 activation can also be harmful, as chronic inflammation driven by NLRP3 supports tumor progression by creating an environment that facilitates cancer cell proliferation, migration, invasion, and metastasis. The release of pro-inflammatory cytokines such as IL-1β and IL-18 can promote tumor growth and angiogenesis, while sustained inflammation may lead to immune suppression, hindering effective anti-tumor responses. In this review article, we discuss the role of NLRP3 inflammasome-mediated inflammatory response in the pathophysiology of various cancer types; understanding this role is essential for the development of innovative therapeutic strategies for cancer growth and spread.
Collapse
Affiliation(s)
| | | | | | | | | | - Kota V. Ramana
- Department of Biomedical Sciences, Noorda College of Osteopathic Medicine, Provo, UT 84606, USA
| |
Collapse
|
11
|
Peng W, Guo K, Hu J, Wang Q. Inhibition of Pyroptosis by Hydroxychloroquine as a Neuroprotective Strategy in Ischemic Stroke. eNeuro 2024; 12:ENEURO.0254-24.2024. [PMID: 39694827 PMCID: PMC11728853 DOI: 10.1523/eneuro.0254-24.2024] [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: 06/09/2024] [Revised: 10/25/2024] [Accepted: 12/05/2024] [Indexed: 12/20/2024] Open
Abstract
Hydroxychloroquine (HCQ), a well-known antimalarial and anti-inflammatory drug, has demonstrated potential neuroprotective effects in ischemic stroke by inhibiting pyroptosis, a programmed cell death associated with inflammation. This study investigates the impact of HCQ on ischemic stroke pathology using both in vivo and in vitro models. In vivo, C57BL/6 mice subjected to middle cerebral artery occlusion (MCAO) were treated with HCQ. Neurological deficits, infarct volume, and the expression of pyroptosis markers were evaluated. The results demonstrated that HCQ significantly improved motor function and reduced infarct volume in the MCAO mouse model. In vitro, BV2 microglial cells exposed to lipopolysaccharide (LPS) and oxygen-glucose deprivation (OGD) were treated with HCQ. Western blot and immunofluorescence analyses revealed that HCQ effectively suppressed the expression of pyroptosis markers GSDMD and NLRP3 in both in vivo and in vitro models. These findings suggest that HCQ mitigates ischemic stroke damage by inhibiting pyroptosis, highlighting its potential as a therapeutic agent for ischemic stroke. This study provides novel insights into the molecular mechanisms by which HCQ exerts its neuroprotective effects, offering a promising new avenue for developing safe, cost-effective, and widely applicable stroke treatments. The potential of HCQ to modulate neuroinflammatory pathways presents a significant advancement in ischemic stroke therapy, emphasizing the importance of targeting pyroptosis in stroke management and the broader implications for treating neuroinflammatory conditions.Significance Statement Ischemic stroke remains a leading cause of disability and death globally, with limited effective treatments. This study reveals that HCQ significantly mitigates ischemic stroke damage by inhibiting pyroptosis, a form of programmed cell death. Using in vivo and in vitro models, HCQ was shown to improve motor function and reduce infarct volume, highlighting its potential as a neuroprotective agent. These findings offer a promising new therapeutic approach for ischemic stroke, emphasizing the importance of targeting pyroptosis in stroke treatment.
Collapse
Affiliation(s)
- Wenshuo Peng
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
| | - Kaiming Guo
- School of Pharmaceutical Sciences, Wenzhou Medical University,Wenzhou 325015, China
| | - Jian Hu
- Department of pharmacy, The Second Affiliated Hospital of Nanjing Medical University, Wenzhou 325015, China
| | - Qianchun Wang
- Department of gastroenterology, The First affiliated hospital of Wenzhou Medical University, Wenzhou 325015, China
| |
Collapse
|
12
|
Jang J, He Z, Huang L, Hwang JY, Kim MY, Cho JY. Upregulation of NK cell activity, cytokine expression, and NF-κB pathway by ginsenoside concentrates from Panax ginseng berries in healthy mice and macrophage cell lines. JOURNAL OF ETHNOPHARMACOLOGY 2024; 335:118681. [PMID: 39121929 DOI: 10.1016/j.jep.2024.118681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 08/01/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Panax ginseng (P. ginseng) C.A. Meyer. Has been studied for decades for its various biological activities, especially in terms of immune-regulatory properties. Traditionally, it has been known that root, leaves, and fruits of P. ginseng were eaten for improving body's Qi and homeostasis. Also, these were used to protect body from various types of infectious diseases. However, molecular mechanisms of immunomodulatory activities of ginseng berries have not been systemically studied as often as other parts of the plant. AIM OF THE STUDY The aim of this research is to discover the regulatory effects of P. ginseng berries, more importantly, their ginsenosides, on innate immune responses and to elucidate the molecular mechanism. MATERIALS AND METHODS Ginseng berry concentrate (GBC) was orally injected into BALB/c mice for 30 days, and spleens were extracted for evaluation of immune-regulatory effects. Murine macrophage RAW264.7 cells were used for detailed molecular mechanism studies. Splenic natural killer (NK) cells were isolated using the magnetic-activated cell sorting (MACS) system, and the cytotoxic activity of isolated NK cells was measured using a lactate dehydrogenase (LDH) release assay. The splenic immune cell population was determined by flow-cytometry. NF-κB promoter activity was assessed by in vitro luciferase assay. Expression of inflammatory proteins and cytokines of the spleen and RAW264.7 cells were evaluated using western blotting and real-time PCR, respectively. RESULTS The GBC enhanced cytotoxic activity of NK cells and the immune-regulation-related splenic cell population. Moreover, GBC promoted NF-κB promoter activity and stimulated the NF-κB signaling cascade. In spleen and RAW264.7 cells, expression of pro-inflammatory cytokines was increased upon GBC application, while expression of anti-inflammatory cytokines decreased. CONCLUSIONS These results suggest that P. ginseng berry can stimulate innate immune responses and help maintain a balanced immune condition, mostly due to the action of its key ginsenoside Re, along with other protopanaxadiol- and protopanaxatriol-type ginsenosides. Such finding will provide a new insight into the field of well-being diet research as well as non-chemical immune modulator, by providing nature-derived and plant-based bioactive materials.
Collapse
Affiliation(s)
- Jiwon Jang
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Ziliang He
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Lei Huang
- Department of Biocosmetics, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Ji Yeon Hwang
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Mi-Yeon Kim
- School of Systems Biomedical Science, Soongsil University, Seoul, 06978, Republic of Korea.
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea; Department of Biocosmetics, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
13
|
Chen X, Yu Z. Insight into the Interaction Mechanism of Pseudorabies Virus Infection. BIOLOGY 2024; 13:1013. [PMID: 39765680 PMCID: PMC11673216 DOI: 10.3390/biology13121013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025]
Abstract
The pseudorabies virus (PRV), also known as suid alphaherpesvirus 1 (SuAHV-1), has garnered significant attention due to its broad host range and the economic losses it incurs in the swine industry. This review aims to provide a comprehensive understanding of the intricate virus-host interactions during PRV infection, focusing on the evasion strategies of the virus against the host responses. We also summarize the mechanisms by which PRV manipulates the host cell machinery to facilitate its replication and spread, while simultaneously evading detection and clearance by the immune system. Furthermore, we discuss the latest advancements, such as metabolic, autophagic, and apoptotic pathways in studying these interactions, highlighting the role of various cellular factors and pathways in elucidating virus-host dynamics. By integrating these insights, the article aims to provide a comprehensive overview of the molecular mechanisms underlying PRV pathogenesis and host response, paving the way for the development of novel therapeutic strategies against this virus.
Collapse
Affiliation(s)
- Xiaoyong Chen
- Xingzhi College, Zhejiang Normal University, Lanxi 321100, China
| | - Ziding Yu
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| |
Collapse
|
14
|
Pallathadka H, Khaleel AQ, Zwamel AH, Malathi H, Sharma S, Rizaev JA, Mustafa YF, Pramanik A, Shuhata Alubiady MH, Jawad MA. Multi-Drug Resistance and Breast Cancer Progression via Toll-Like Receptors (TLRs) Signaling. Cell Biochem Biophys 2024; 82:3015-3030. [PMID: 39110298 DOI: 10.1007/s12013-024-01418-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/10/2024] [Indexed: 11/20/2024]
Abstract
Toll-like receptors (TLRs) are essential receptors involved in inflammation and innate immunity. Various types of cancer cells, as well as innate immune cells, express TLRs. There is mounting proof that TLRs are critical to the development and spread of cancer as well as metabolism. In breast cancer, up-regulated levels of TLRs have been linked to the aggressiveness of the diseases, worse treatment outcomes, and the emergence of therapeutic resistance. Patients with advanced non-resectable, recurring, and metastatic breast cancer currently have few available treatment choices. An intriguing new strategy is an innate immunity-mediated anticancer immunotherapy, either used alone or in conjunction with existing treatments. In fact, several TLR agonists and antagonists have been used in clinical studies for anti-cancer immunotherapy. Consequently, TLRs serve as critical targets for controlling the course of breast cancer and treatment resistance in addition to being implicated in immune responses against pathogen infection and cancer immunology. In this review, we deliver an overview of the most current findings on TLR involvement in the development of breast cancer and treatment resistance.
Collapse
Affiliation(s)
| | - Abdulrahman Qais Khaleel
- Department of Medical Instruments Engineering, Al-Maarif University College, Al Anbar, 31001, Iraq.
| | - Ahmed Hussein Zwamel
- Medical laboratory technique college, the Islamic University, Najaf, Iraq
- Medical laboratory technique college, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical laboratory technique college, the Islamic University of Babylon, Babylon, Iraq
| | - H Malathi
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Snehlata Sharma
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjheri, Mohali, 140307, Punjab, India
| | - Jasur Alimdjanovich Rizaev
- Department of Public health and Healthcare management, Rector, Samarkand State Medical University, 18, Amir Temur Street, Samarkand, Uzbekistan
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | - Atreyi Pramanik
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, Uttarakhand, India
| | | | - Mohammed Abed Jawad
- Department of Medical Laboratories Technology, Al-Nisour University College, Baghdad, Iraq
| |
Collapse
|
15
|
Wang H, Wang T, Yan S, Tang J, Zhang Y, Wang L, Xu H, Tu C. Crosstalk of pyroptosis and cytokine in the tumor microenvironment: from mechanisms to clinical implication. Mol Cancer 2024; 23:268. [PMID: 39614288 PMCID: PMC11607834 DOI: 10.1186/s12943-024-02183-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 11/22/2024] [Indexed: 12/01/2024] Open
Abstract
In the realm of cancer research, the tumor microenvironment (TME) plays a crucial role in tumor initiation and progression, shaped by complex interactions between cancer cells and surrounding non-cancerous cells. Cytokines, as essential immunomodulatory agents, are secreted by various cellular constituents within the TME, including immune cells, cancer-associated fibroblasts, and cancer cells themselves. These cytokines facilitate intricate communication networks that significantly influence tumor initiation, progression, metastasis, and immune suppression. Pyroptosis contributes to TME remodeling by promoting the release of pro-inflammatory cytokines and sustaining chronic inflammation, impacting processes such as immune escape and angiogenesis. However, challenges remain due to the complex interplay among cytokines, pyroptosis, and the TME, along with the dual effects of pyroptosis on cancer progression and therapy-related complications like cytokine release syndrome. Unraveling these complexities could facilitate strategies that balance inflammatory responses while minimizing tissue damage during therapy. This review delves into the complex crosstalk between cytokines, pyroptosis, and the TME, elucidating their contribution to tumor progression and metastasis. By synthesizing emerging therapeutic targets and innovative technologies concerning TME, this review aims to provide novel insights that could enhance treatment outcomes for cancer patients.
Collapse
Affiliation(s)
- Hua Wang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Tao Wang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Shuxiang Yan
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Jinxin Tang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Yibo Zhang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Liming Wang
- School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410011, China.
| | - Haodong Xu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| | - Chao Tu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Shenzhen Research Institute of Central South University, Guangdong, 518063, China.
- Hunan Engineering Research Center of AI Medical Equipment, The Second Xiangya Hospital of Central, South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
16
|
Cheng W, Chen W, Jia R. The Role of Pyroptosis in the Progression and Targeted Therapeutic Approaches for Urological Malignancies. J Inflamm Res 2024; 17:9567-9581. [PMID: 39606639 PMCID: PMC11600921 DOI: 10.2147/jir.s487740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024] Open
Abstract
The prevalence of urological malignancies continues to pose a significant global health challenge, particularly due to the poor prognosis associated with advanced stages of these diseases. Consequently, there is an urgent need to deepen our understanding of the molecular mechanisms governing the development of urological malignancies to facilitate breakthroughs in diagnosis and treatment. Pyroptosis, a novel and specific form of programmed cell death, plays a crucial role in regulating inflammatory responses, cell development, tissue homeostasis, and stress responses. Recent research has revealed a close association between pyroptosis and urological malignancies. In this paper, we review the pathogenesis and recent advancements in the understanding of pyroptosis in urological malignancies, elucidate the molecular mechanisms involved in its regulation, and aim to provide new directions for the clinical management of these diseases.
Collapse
Affiliation(s)
- Wenjie Cheng
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Wei Chen
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Ruipeng Jia
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| |
Collapse
|
17
|
Xu L, Yang X, Liu XT, Li XY, Zhu HZ, Xie YH, Wang SW, Li Y, Zhao Y. Carvacrol alleviates LPS-induced myocardial dysfunction by inhibiting the TLR4/MyD88/NF-κB and NLRP3 inflammasome in cardiomyocytes. J Inflamm (Lond) 2024; 21:47. [PMID: 39548566 PMCID: PMC11568595 DOI: 10.1186/s12950-024-00411-z] [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: 11/15/2023] [Accepted: 09/16/2024] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND Sepsis-induced myocardial dysfunction (SIMD) may contribute to the poor prognosis of septic patients. Carvacrol (2-methyl-5-isopropyl phenol), a phenolic monoterpene compound extracted from various aromatic plants and fragrance essential oils, has multiple beneficial effects such as antibacterial, anti-inflammatory, and antioxidant properties. These attributes make it potentially useful for treating many diseases. This study aims to investigate the effects of CAR on LPS-induced myocardial dysfunction and explore the underlying mechanism. RESULTS H9c2 cells were stimulated with 10 µg/ml LPS for 12 h, and c57BL/6 mice were intraperitoneally injected with 10 mg/kg LPS to establish a septic-myocardial injury model. Our results showed that CAR could improve cardiac function, significantly reduce serum levels of inflammatory cytokines (including TNF-α, IL-1β, and IL-6), decrease oxidative stress, and inhibit cardiomyocyte apoptosis in LPS-injured mice. Additionally, CAR significantly downregulated the expression of TLR4, MyD88, and NF-κB in LPS-injured mice and H9c2 cells. It also inhibited the upregulation of inflammasome components (such as NLRP3, GSDMD, and IL-1β) in H9c2 cells triggered by LPS. CONCLUSION Taken together, CAR exhibited potential cardioprotective effects against sepsis, which may be mainly attributed to the TLR4/MyD88/NF-κB pathway and the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Lu Xu
- The College of Life Science, Northwest University, Xi'an, China
| | - Xu Yang
- The College of Life Science, Northwest University, Xi'an, China
| | - Xiao-Ting Liu
- The College of Life Science, Northwest University, Xi'an, China
| | - Xia-Yun Li
- The College of Life Science, Northwest University, Xi'an, China
| | - Han-Zhao Zhu
- Department of Cardiovascular Surgery, The First Affiliated Hospital, The Air Force Medical University, Xi'an, China
| | - Yan-Hua Xie
- The College of Life Science, Northwest University, Xi'an, China
| | - Si-Wang Wang
- The College of Life Science, Northwest University, Xi'an, China.
| | - Yao Li
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, China.
| | - Ye Zhao
- The College of Life Science, Northwest University, Xi'an, China.
| |
Collapse
|
18
|
Lin Z, Liu Z, Yang X, Pan Z, Feng Y, Zhang Y, Chen H, Lao L, Chen J, Shi F, Gong C, Zeng W. Simeprevir induces ferroptosis through β-TrCP/Nrf2/GPX4 axis in triple-negative breast cancer cells. Biomed Pharmacother 2024; 180:117558. [PMID: 39405915 DOI: 10.1016/j.biopha.2024.117558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/25/2024] [Accepted: 10/08/2024] [Indexed: 11/14/2024] Open
Abstract
The effective treatment regimens of triple-negative breast cancer (TNBC), a specific subtype of breast cancer (BC) with proneness to relapse and poor prognosis, are still lacking. Simeprevir (SIM), approved for hepatitis C infection treatment, has been proved to be a competitive drug for the treatment of various solid tumors recently. However, the anti-tumor mechanisms of SIM and therapeutic effects on TNBC are uncertain. In this study, we suggested that SIM effectively restrained the growth of MDA-MB-231 and BT-549 cells, two cell lines from TNBC. The RNA sequencing revealed that ferroptosis signaling was activated in SIM-treated TNBC cells. SIM induced ferroptosis in TNBC cells through reduced glutathione (GSH) levels, increased iron levels, ROS and lipid peroxidation. Mechanistically, SIM promoted the expression of β-TrCP to inhibit the Nrf2/GPX4 axis in TNBC cells, leading to ferroptosis. Moreover, SIM administration into the xenografts formed by MDA-MB-231 dramatically suppressed the tumor progression by inducing ferroptosis in vivo. Collectively, this finding reveals that SIM may serve as a competitive therapeutic strategy to inhibit TNBC.
Collapse
Affiliation(s)
- Zhirong Lin
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Zifei Liu
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Xinyu Yang
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Zhilong Pan
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Yaxin Feng
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Yunyi Zhang
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Huiping Chen
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Liyan Lao
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Jianing Chen
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Fujun Shi
- Department of Breast Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China.
| | - Chang Gong
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| | - Wenfeng Zeng
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| |
Collapse
|
19
|
Jiu X, Li W, Liu Y, Liu L, Lu H. TREM2, a critical activator of pyroptosis, mediates the anti‑tumor effects of piceatannol in uveal melanoma cells via caspase 3/GSDME pathway. Int J Mol Med 2024; 54:96. [PMID: 39219277 PMCID: PMC11410308 DOI: 10.3892/ijmm.2024.5420] [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: 02/29/2024] [Accepted: 07/19/2024] [Indexed: 09/04/2024] Open
Abstract
Uveal melanoma (UM) is the most prevalent type of primary intraocular malignancy and is prone to metastasize, particularly to the liver. However, due to the poor understanding of the pathogenesis of UM, effective therapeutic approaches are lacking. As a phenolic compound extracted from grapes, piceatannol (PIC) exhibits anti‑cancer properties. To the best of our knowledge, however, the effects of PIC on UM have not been well investigated. Therefore, in the present study, considering the impact of pyroptosis on modulating cell viability, the mechanism underlying the effects of PIC on UM cell proliferation was explored. The inhibitory effect of PIC on proliferation of UM cells was detected by cell counting kit‑8 assay. Wound healing was used to investigate the effects of PIC on the migration of UM cells. Activity detecting assays were performed to test the apoptosis and oxidant level in UM cells. Western blotting and RT‑qPCR were used to detect the inflammatory and pyroptotic levels of UM cell after PIC treatment. PIC‑treated UM cells were screened by high‑throughput sequencing to detect the differential expression of RNA and differential genes. Si‑TREM2 transfection was used to verify the important role of TREM2 in the effects of PIC. Immunohistochemical staining was used to observe the expressions of TREM2 and GSDMR of tumor in nude mice after PIC administration. PIC effectively inhibited proliferation ability of C918 and Mum‑2b UM cell lines via enhancing apoptosis, as evidenced by enhanced activities of caspase 3 and caspase 9. In addition, treatment of UM cells with PIC attenuated cell migration in a dose‑dependent manner. PIC increased reactive oxygen species levels and suppressed the activity of the antioxidant enzymes superoxide dismutase, glutathione‑S‑transferase, glutathione peroxidase and catalase. PIC inhibited inflammatory responses in C918 cells. PIC treatment upregulated IL‑1β, IL‑18 and Nod‑like receptor protein 3 and downregulated gasdermin D (GSDMD). RNA sequencing results revealed the activation of an unconventional pyroptosis‑associated signaling pathway, namely caspase 3/GSDME signaling, following PIC treatment, which was mediated by triggering receptor expressed on myeloid cells 2 (TREM2) upregulation. As an agonist of TREM2, COG1410‑mediated TREM2 upregulation inhibited proliferation of C918 cells, displaying similar effects to PIC. Furthermore, PIC inhibited tumor growth via regulating the TREM2/caspase 3/GSDME pathway in a mouse model. Collectively, the present study revealed a novel mechanism underlying the inhibitory effects of PIC on UM, providing a potential treatment approach for UM in clinic.
Collapse
Affiliation(s)
- Xudong Jiu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
- Department of Ophthalmology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Wenjie Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Yang Liu
- Department of Ophthalmology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Lin Liu
- Department of Ophthalmology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Hong Lu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
20
|
Pedrão LFAT, Medeiros POS, Leandro EC, Falquetto B. Parkinson's disease models and death signaling: what do we know until now? Front Neuroanat 2024; 18:1419108. [PMID: 39533977 PMCID: PMC11555652 DOI: 10.3389/fnana.2024.1419108] [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: 04/17/2024] [Accepted: 09/04/2024] [Indexed: 11/16/2024] Open
Abstract
Parkinson's disease (PD) is the second neurodegenerative disorder most prevalent in the world, characterized by the loss of dopaminergic neurons in the Substantia Nigra (SN). It is well known for its motor and non-motor symptoms including bradykinesia, resting tremor, psychiatric, cardiorespiratory, and other dysfunctions. Pathological apoptosis contributes to a wide variety of diseases including PD. Various insults and/or cellular phenotypes have been shown to trigger distinct signaling events leading to cell death in neurons affected by PD. The intrinsic or mitochondrial pathway, inflammatory or oxidative stress-induced extrinsic pathways are the main events associated with apoptosis in PD-related neuronal loss. Although SN is the main brain area studied so far, other brain nuclei are also affected by the disease leading to non-classical motor symptoms as well as non-motor symptoms. Among these, the respiratory symptoms are often overlooked, yet they can cause discomfort and may contribute to patients shortened lifespan after disease diagnosis. While animal and in vitro models are frequently used to investigate the mechanisms involved in the pathogenesis of PD in both the SN and other brain regions, these models provide only a limited understanding of the disease's actual progression. This review offers a comprehensive overview of some of the most studied forms of cell death, including recent research on potential treatment targets for these pathways. It highlights key findings and milestones in the field, shedding light on the potential role of understanding cell death in the prevention and treatment of the PD. Therefore, unraveling the connection between these pathways and the notable pathological mechanisms observed during PD progression could enhance our comprehension of the disease's origin and provide valuable insights into potential molecular targets for the developing therapeutic interventions.
Collapse
Affiliation(s)
| | | | | | - Barbara Falquetto
- Department of Pharmacology, Instituto de Ciências Biomédica, Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
21
|
Gao K, Liu Y, Sun C, Wang Y, Bao H, Liu G, Ou J, Sun P. TNF-ɑ induces mitochondrial dysfunction to drive NLRP3/Caspase-1/GSDMD-mediated pyroptosis in MCF-7 cells. Sci Rep 2024; 14:25880. [PMID: 39468189 PMCID: PMC11519391 DOI: 10.1038/s41598-024-76997-4] [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: 03/31/2024] [Accepted: 10/18/2024] [Indexed: 10/30/2024] Open
Abstract
Pyroptosis is a gasdermin-mediated pro-inflammatory form of programmed cell death (PCD). Tumor necrosis factor-ɑ (TNF-ɑ) is an inflammatory cytokine, and some studies have shown that TNF-ɑ can cause pyroptosis of cells and exert anti-tumor effects. However, whether TNF-ɑ exerts anti-tumor effects on breast cancer cells by inducing pyroptosis has not been reported. In this study, to explore the impact of TNF-ɑ on pyroptosis in breast cancer cells, we treated MCF-7 cells with TNF-ɑ and found that TNF-ɑ induced cell death. Moreover, we observed that the dead cells were swollen with obvious balloon-like bubbles, which was a typical sign of pyroptosis. Further studies have found that the anti-tumor effect of TNF-ɑ on breast cancer cells in vitro was achieved through the canonical pyroptosis pathway. In addition, TNF-ɑ-induced pyroptosis in MCF-7 cells was associated with mitochondrial dysfunction, in which mitochondrial membrane potential was decreased and mitochondrial ROS production was increased. After inhibiting ROS production, the activation effect of TNF-ɑ on NLRP3/Caspase-1/GSDMD pathway was weakened, and the inhibitory effect of TNF-ɑ on the growth of MCF-7 cells in vitro was also decreased, further confirming the involvement of ROS in TNF-ɑ-induced pyroptosis. Overall, our study revealed a new mechanism by which TNF-ɑ exerts an anti-tumor effect by inducing pyroptosis in MCF-7 cells through the ROS/NLRP3/Caspase-1/GSDMD pathway, which may provide new therapeutic ideas for the treatment of breast cancer.
Collapse
Affiliation(s)
- Kexin Gao
- Department of Anatomy, Mudanjiang Medical University, Mudanjiang City, 157000, Heilongjiang, China
| | - Yancui Liu
- Department of Anatomy, Mudanjiang Medical University, Mudanjiang City, 157000, Heilongjiang, China
| | - Cheng Sun
- Department of Anatomy, Mudanjiang Medical University, Mudanjiang City, 157000, Heilongjiang, China
| | - Ying Wang
- Department of Anatomy, Mudanjiang Medical University, Mudanjiang City, 157000, Heilongjiang, China
| | - Hongrong Bao
- Department of Anatomy, Mudanjiang Medical University, Mudanjiang City, 157000, Heilongjiang, China
| | - Guoyang Liu
- Department of Nuclear Medicine, Hongqi Hospital affiliated to Mudanjiang Medical University, Mudanjiang City, 157000, Heilongjiang, China
| | - Jinrui Ou
- Department of Nuclear Magnetic, the Second People's Hospital of Mudanjiang City, Mudanjiang City, 157000, Heilongjiang, China
| | - Ping Sun
- Department of Anatomy, Mudanjiang Medical University, Mudanjiang City, 157000, Heilongjiang, China.
| |
Collapse
|
22
|
Zhang Q, Guo S, Wang H. The Protective Role of Baicalin in the Regulation of NLRP3 Inflammasome in Different Diseases. Cell Biochem Biophys 2024:10.1007/s12013-024-01597-y. [PMID: 39443419 DOI: 10.1007/s12013-024-01597-y] [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] [Accepted: 10/09/2024] [Indexed: 10/25/2024]
Abstract
The NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome consists of pro-caspase-1, NLRP3 and apoptosis-related speckle-like protein (ASC). It can detect multiple microorganisms, endogenous danger signals and environmental stimulus including adenosine triphosphate (ATP), urate, cholesterol crystals, and so on, thereby forming activated NLRP3 inflammasome. During the course of the activation of NLRP3 inflammasome, pro-caspase-1 is transformed into activated caspase-1 that results in the maturation and secretion of interleukin-1beta (IL-1β) and IL-18. The dysfunction of NLRP3 inflammasome participates in multiple diseases such as liver diseases, renal diseases, nervous system diseases and diabetes. Baicalin is the primary bioactive component of Scutellaria baicalensis, which has been used since ancient times. Baicalin has many types of biological functions, such as anti-bacterial, anti-tumor and antioxidant. More and more evidence suggests that baicalin regulation of NLRP3 inflammasome is involved in different diseases. However, the mechanism is still elusive. Here, we reviewed the progress of baicalin regulation of NLRP3 inflammasome in many kinds of diseases to lay a foundation for future researches.
Collapse
Affiliation(s)
- Qi Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
| | - Shiyun Guo
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
| | - Honggang Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China.
| |
Collapse
|
23
|
Quagliariello V, Canale ML, Bisceglia I, Iovine M, Giordano V, Giacobbe I, Scherillo M, Gabrielli D, Maurea C, Barbato M, Inno A, Berretta M, Tedeschi A, Oliva S, Greco A, Maurea N. Glucagon-like Peptide 1 Receptor Agonists in Cardio-Oncology: Pathophysiology of Cardiometabolic Outcomes in Cancer Patients. Int J Mol Sci 2024; 25:11299. [PMID: 39457081 PMCID: PMC11508560 DOI: 10.3390/ijms252011299] [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: 09/06/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Cancer patients, especially long cancer survivors, are exposed to several cardio-metabolic diseases, including diabetes, heart failure, and atherosclerosis, which increase their risk of cardiovascular mortality. Therapy with glucagon-like peptide 1 (GLP1) receptor agonists demonstrated several beneficial cardiovascular effects, including atherosclerosis and heart failure prevention. Cardiovascular outcome trials (CVOTs) suggest that GLP-1 RA could exert cardiorenal benefits and systemic anti-inflammatory effects in patients with type-2 diabetes through the activation of cAMP and PI3K/AkT pathways and the inhibition of NLRP-3 and MyD88. In this narrative review, we highlight the biochemical properties of GLP-1 RA through a deep analysis of the clinical and preclinical evidence of the primary prevention of cardiomyopathies. The overall picture of this review encourages the study of GLP-1 RA in cancer patients with type-2 diabetes, as a potential primary prevention strategy against heart failure and atherosclerosis.
Collapse
Affiliation(s)
- Vincenzo Quagliariello
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (M.I.); (V.G.); (I.G.); (M.B.); (N.M.)
| | | | - Irma Bisceglia
- Servizi Cardiologici Integrati, Dipartimento Cardio-Toraco-Vascolare, Azienda Ospedaliera San Camillo Forlanini, 00148 Rome, Italy;
| | - Martina Iovine
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (M.I.); (V.G.); (I.G.); (M.B.); (N.M.)
| | - Vienna Giordano
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (M.I.); (V.G.); (I.G.); (M.B.); (N.M.)
| | - Ilaria Giacobbe
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (M.I.); (V.G.); (I.G.); (M.B.); (N.M.)
| | - Marino Scherillo
- Division of Cardiology, Hospital San Pio Benevento (BN), 82100 Benevento, Italy;
| | - Domenico Gabrielli
- U.O.C. Cardiologia, Dipartimento Cardio-Toraco-Vascolare, Azienda Ospedaliera San Camillo Forlanini, 00152 Rome, Italy;
| | - Carlo Maurea
- Department of Medicine, University of Salerno, 84084 Fisciano, Italy;
| | - Matteo Barbato
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (M.I.); (V.G.); (I.G.); (M.B.); (N.M.)
| | - Alessandro Inno
- Medical Oncology, IRCCS Ospedale Sacro Cuore Don Calabria, 37024 Negrar di Valpolicella, Italy;
| | - Massimiliano Berretta
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy;
| | - Andrea Tedeschi
- Cardiology Unit of Emergency Department, Guglielmo da Saliceto Hospital, 29121 Piacenza, Italy;
| | - Stefano Oliva
- UOSD Cardiologia di Interesse Oncologico IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy;
| | - Alessandra Greco
- Divisione di Cardiologia, Fondazione IRCCS San Matteo Hospital, Viale Golgi 19, 27100 Pavia, Italy;
| | - Nicola Maurea
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (M.I.); (V.G.); (I.G.); (M.B.); (N.M.)
| |
Collapse
|
24
|
Balahura Stămat LR, Dinescu S. Inhibition of NLRP3 inflammasome contributes to paclitaxel efficacy in triple negative breast cancer treatment. Sci Rep 2024; 14:24753. [PMID: 39433537 PMCID: PMC11494052 DOI: 10.1038/s41598-024-75805-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 10/08/2024] [Indexed: 10/23/2024] Open
Abstract
Chronic inflammation and NLRP3 inflammasome activation are among the determining factors of breast malignancies. Paclitaxel (PTX) is a drug used in breast cancer treatment which sustains prolonged inflammation, reducing the effectiveness of chemotherapy. Considering the impact of inflammatory processes in cancer progression, there is a strong concern to develop therapeutic strategy targeting NLRP3 inflammasome for triple-negative breast cancer (TNBC) treatment. Therefore, the aim of this study was to evaluate the potential of PTX and NLRP3 inflammasome modulation to counterbalance TNBC by inducing programmed cell death and inhibiting the activity of pro-inflammatory cytokines. The obtained results suggested the strong interaction between NLRP3 inflammasome and TNBC and revealed that pharmacological inhibition, using NLRP3-specific inhibitor MCC950, and genetic silencing of NLRP3 inflammasome using specific small interfering RNA, reduced inflammatory responses and facilitated PTX-determined tumor cell death. Thus, NLRP3 inflammasome manipulation in combination with anti-tumor drugs opens up new therapeutic perspectives for TNBC therapy.
Collapse
Affiliation(s)
- Liliana-Roxana Balahura Stămat
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, 050095, Romania
| | - Sorina Dinescu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, 050095, Romania.
- Research Institute of the University of Bucharest, Bucharest, 050663, Romania.
| |
Collapse
|
25
|
Du X, Yang H, Kang X, Fu C, Yang T. Blocking GATA6 Alleviates Pyroptosis and Inhibits Abdominal Wall Endometriosis Lesion Growth Through Inactivating the PI3K/AKT Pathway. Cell Biochem Biophys 2024:10.1007/s12013-024-01583-4. [PMID: 39365516 DOI: 10.1007/s12013-024-01583-4] [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] [Accepted: 09/25/2024] [Indexed: 10/05/2024]
Abstract
Endometriosis is a benign gynecological disorder characterized by the abnormal presence of endometrium-like cells, referred to as ectopic tissue, located outside the uterine cavity. Beyond the abnormal proliferation of endometrium-like tissues within and beyond the pelvic cavity, compelling scientific evidence underscores the crucial involvement of the NOD-like receptor NLRP3 inflammasome and pyroptosis in the pathogenesis of EMS. Our investigation has revealed a striking upregulation of the endogenous protein GATA-binding protein 6 (GATA6) in abdominal wall EMS. Notably, the knockdown of GATA6 significantly impaired the viability and migratory potential of primary ectopic endometrial stromal cells (EESCs) while also inhibiting crucial markers of pyroptosis, such as NLRP3, the gasdermin D N-terminal fragment (GSDMD-N), and reactive oxygen species (ROS) levels within these cells. Delving deeper into the underlying mechanisms, we discovered that suppressing GATA6 mitigated the activation of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway in EESCs. The administration of 740 Y-P, an agonist of the PI3K/AKT pathway, mitigated the inhibitive actions of GATA6 knockdown on EESCs' growth, migration, and pyroptosis, highlighting the intricate crosstalk between GATA6 and this intricate signaling cascade. In vivo experiments corroborated these findings, demonstrating that reduced GATA6 expression effectively restrained the growth of endometrial lesions and concurrently suppressed pyroptosis, accompanied by a dampening of PI3K/AKT signaling within these lesions. In summary, our study underscores the pivotal role of GATA6 in modulating the growth and pyroptosis of abdominal wall EMS through its regulation of the PI3K/AKT signaling pathway. Silencing GATA6 emerges as a promising approach to alleviate pyroptosis and potentially offers a novel therapeutic angle for managing abdominal wall EMS.
Collapse
Affiliation(s)
- Xiufang Du
- Department of Women's Group Health, Shijiazhuang Maternal and Child Health Care Hospital, Shijiazhuang, 050000, Hebei, China.
| | - Hongjie Yang
- Shijiazhuang Maternal and Child Health Care Hospital, Shijiazhuang, 050000, Hebei, China
| | - Xiaobei Kang
- Department of Women's Group Health, Shijiazhuang Maternal and Child Health Care Hospital, Shijiazhuang, 050000, Hebei, China
| | - Changna Fu
- Blood Collection Room, Shijiazhuang Maternal and Child Health Care Hospital, Shijiazhuang, 050000, Hebei, China
| | - Tao Yang
- The Second Department of Obstetrics and Gynecology, Shijiazhuang Maternal and Child Health Hospital, Shijiazhuang, 050000, Hebei, China
| |
Collapse
|
26
|
Al Mamun A, Geng P, Wang S, Shao C. Role of Pyroptosis in Endometrial Cancer and Its Therapeutic Regulation. J Inflamm Res 2024; 17:7037-7056. [PMID: 39377044 PMCID: PMC11457779 DOI: 10.2147/jir.s486878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 09/21/2024] [Indexed: 10/09/2024] Open
Abstract
Pyroptosis is an inflammatory cell death induced by inflammasomes that release several pro-inflammatory mediators such as interleukin-18 (IL-18) and interleukin-1β (IL-1β). Pyroptosis, a type of programmed cell death, has recently received increased interest both as a therapeutic and immunological mechanism. Numerous studies have provided substantial evidence supporting the involvement of inflammasomes and pyroptosis in a variety of pathological conditions including cancers, nerve damage, inflammatory diseases and metabolic conditions. Researchers have demonstrated that dysregulation of pyroptosis and inflammasomes contribute to the progression of endometriosis and gynecological malignancies. Current research also indicates that inflammasome and pyroptosis-dependent signaling pathways may further induce the progression of endometrial cancer (EC). More specifically, dysregulation of NLR family pyrin domain 3 (NLRP3) and caspase-1-dependent pyroptosis play a contributory role in the pathogenesis and development of EC. Therefore, pyroptosis-regulated protein gasdermin D (GSDMD) may be an independent prognostic biomarker for the detection of EC. This review presents the molecular mechanisms of pyroptosis-dependent signaling pathways and their contributory role and function in advancing EC. Moreover, this review offers new insights into potential future applications and innovative approaches in utilizing pyroptosis to develop effective anti-cancer therapies.
Collapse
Affiliation(s)
- Abdullah Al Mamun
- Key Laboratory of Joint Diagnosis and Treatment of Chronic Liver Disease and Liver Cancer of Lishui, The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People’s Hospital, Lishui, Zhejiang, 323000, People’s Republic of China
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People’s Republic of China
| | - Peiwu Geng
- Key Laboratory of Joint Diagnosis and Treatment of Chronic Liver Disease and Liver Cancer of Lishui, The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People’s Hospital, Lishui, Zhejiang, 323000, People’s Republic of China
| | - Shuanghu Wang
- Key Laboratory of Joint Diagnosis and Treatment of Chronic Liver Disease and Liver Cancer of Lishui, The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People’s Hospital, Lishui, Zhejiang, 323000, People’s Republic of China
| | - Chuxiao Shao
- Key Laboratory of Joint Diagnosis and Treatment of Chronic Liver Disease and Liver Cancer of Lishui, The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People’s Hospital, Lishui, Zhejiang, 323000, People’s Republic of China
| |
Collapse
|
27
|
Li LR, Chen L, Sun ZJ. Igniting hope: Harnessing NLRP3 inflammasome-GSDMD-mediated pyroptosis for cancer immunotherapy. Life Sci 2024; 354:122951. [PMID: 39127315 DOI: 10.1016/j.lfs.2024.122951] [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: 05/10/2024] [Revised: 07/19/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
In the contemporary landscape of oncology, immunotherapy, represented by immune checkpoint blockade (ICB) therapy, stands out as a beacon of innovation in cancer treatment. Despite its promise, the therapy's progression is hindered by suboptimal clinical response rates. Addressing this challenge, the modulation of the NLRP3 inflammasome-GSDMD-mediated pyroptosis pathway holds promise as a means to augment the efficacy of immunotherapy. In the pathway, the NLRP3 inflammasome serves as a pivotal molecular sensor that responds to inflammatory stimuli within the organism. Its activation leads to the release of cytokines interleukin 1β and interleukin 18 through the cleavage of GSDMD, thereby forming membrane pores and potentially resulting in pyroptosis. This cascade of processes exerts a profound impact on tumor development and progression, with its function and expression exhibiting variability across different tumor types and developmental stages. Consequently, understanding the specific roles of the NLRP3 inflammasome and GSDMD-mediated pyroptosis in diverse tumors is imperative for comprehending tumorigenesis and crafting precise therapeutic strategies. This review aims to elucidate the structure and activation mechanisms of the NLRP3 inflammasome, as well as the induction mechanisms of GSDMD-mediated pyroptosis. Additionally, we provide a comprehensive overview of the involvement of this pathway in various cancer types and its applications in tumor immunotherapy, nanotherapy, and other fields. Emphasis is placed on the feasibility of leveraging this approach to enhance ICB therapy within the field of immunotherapy. Furthermore, we discuss the potential applications of this pathway in other immunotherapy methods, such as chimeric antigen receptor T-cell (CAR-T) therapy and tumor vaccines.
Collapse
Affiliation(s)
- Ling-Rui Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Lei Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
| | - Zhi-Jun Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
| |
Collapse
|
28
|
Wang H, Wang X, Wang L, Wang H, Zhang Y. Plant‐Derived Phytochemicals and Their Nanoformulations for Inducing Programed Cell Death in Cancer. ADVANCED THERAPEUTICS 2024; 7. [DOI: 10.1002/adtp.202400197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Indexed: 01/05/2025]
Abstract
AbstractPhytochemicals are a diverse class of compounds found in various plant‐based foods and beverages that have displayed the capacity to exert powerful anticancer effects through the induction of programed cell death (PCD) in malignancies. PCD is a sophisticated process that maintains in upholding tissue homeostasis and eliminating injured or neoplastic cells. Phytochemicals have shown the potential to induce PCD in malignant cells through various mechanisms, including modulation of cell signaling pathways, regulation of reactive oxygen species (ROS), and interaction with critical targets in cells such as DNA. Moreover, recent studies have suggested that nanomaterials loaded with phytochemicals may enhance cell death in tumors, which can also stimulate antitumor immunity. In this review, a comprehensive overview of the current understanding of the anticancer effects of phytochemicals and their potential as a promising approach to cancer therapy, is provided. The impacts of phytochemicals such as resveratrol, curcumin, apigenin, quercetin, and some approved plant‐derived drugs, such as taxanes on the regulation of some types of PCD, including apoptosis, pyroptosis, anoikis, autophagic cell death, ferroptosis, and necroptosis, are discussed. The underlying mechanisms and the potential of nanomaterials loaded with phytochemicals to enhance PCD in tumors are also explained.
Collapse
Affiliation(s)
- Haoyu Wang
- Medical College Xijing University Xi'an Shaanxi 710123 China
- Department of Orthopedics The Second Affiliated Hospital Xi'an Jiaotong University Xi'an Shaanxi 710004 China
| | - Xiaoyang Wang
- Department of Orthopedics The Second Affiliated Hospital Xi'an Jiaotong University Xi'an Shaanxi 710004 China
| | - Long Wang
- Medical College Xijing University Xi'an Shaanxi 710123 China
| | - Haifan Wang
- Department of Orthopedics The Second Affiliated Hospital Xi'an Jiaotong University Xi'an Shaanxi 710004 China
| | - Yuxing Zhang
- Medical College Xijing University Xi'an Shaanxi 710123 China
| |
Collapse
|
29
|
Chen Q, Peng B, Lin L, Chen J, Jiang Z, Luo Y, Huang L, Li J, Peng Y, Wu J, Li W, Zhuang K, Liang M. Chondroitin Sulfate-Modified Hydroxyapatite for Caspase-1 Activated Induced Pyroptosis through Ca Overload/ER Stress/STING/IRF3 Pathway in Colorectal Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2403201. [PMID: 39016938 DOI: 10.1002/smll.202403201] [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: 04/21/2024] [Revised: 06/06/2024] [Indexed: 07/18/2024]
Abstract
Immune checkpoint inhibitors, are the fourth most common therapeutic tool after surgery, chemotherapy, and radiotherapy for colorectal cancer (CRC). However, only a small proportion (≈5%) of CRC patients, those with "hot" (immuno-activated) tumors, benefit from the therapy. Pyroptosis, an innovative form of programmed cell death, is a potentially effective means to mediate a "cold" to "hot" transformation of the tumor microenvironment (TME). Calcium-releasing hydroxyapatite (HAP) nanoparticles (NPs) trigger calcium overload and pyroptosis in tumor cells. However, current limitations of these nanomedicines, such as poor tumor-targeting capabilities and insufficient calcium (Ca) ion release, limit their application. In this study, chondroitin sulfate (CS) is used to target tumors via binding to CD44 receptors and kaempferol (KAE) is used as a Ca homeostasis disruptor to construct CS-HAP@KAE NPs that function as pyroptosis inducers in CRC cells. CS-HAP@KAE NPs bind to the tumor cell membrane, HAP released Ca in response to the acidic environment of the TME, and kaempferol (KAE) enhances the influx of extracellular Ca, resulting in intracellular Ca overload and pyroptosis. This is associated with excessive endoplasmic reticulum stress triggered activation of the stimulator of interferon genes/interferon regulatory factor 3 pathway, ultimately transforming the TME from "cold" to "hot".
Collapse
Affiliation(s)
- Qing Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jiaxing University, Jiaxing, 314001, China
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Bin Peng
- Department of Oncology, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510700, China
| | - Lifan Lin
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jiawen Chen
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zhaojun Jiang
- Department of Oncology, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510700, China
| | - Yuanwei Luo
- Department of Oncology, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510700, China
| | - Liyong Huang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jiaxing University, Jiaxing, 314001, China
| | - Jin Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jiaxing University, Jiaxing, 314001, China
| | - Yuping Peng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jiaxing University, Jiaxing, 314001, China
| | - Jiaming Wu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jiaxing University, Jiaxing, 314001, China
| | - Wei Li
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hunan, 421000, China
| | - Kangmin Zhuang
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Min Liang
- Department of Oncology, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510700, China
| |
Collapse
|
30
|
Lin CJ, Tian GA, Zhao WY, Tian Y, Liu YR, Gu DN, Tian L. NLRP1 inhibits lung adenocarcinoma growth through mediating mitochondrial dysregulation in an inflammasome-independent manner. Braz J Med Biol Res 2024; 57:e13885. [PMID: 39258674 PMCID: PMC11379352 DOI: 10.1590/1414-431x2024e13885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 07/21/2024] [Indexed: 09/12/2024] Open
Abstract
NLRP1, the first identified inflammasome-forming sensor, is thought to be involved in cancer, yet its definite function in lung adenocarcinoma (LUAD) remains unclear. Herein, we explored the expression and function of NLRP1 in LUAD. Decreased NLRP1 expression was identified in LUAD, which was associated with a poor prognosis. Overexpression of NLRP1 inhibited tumor growth in vitro and in vivo. Mechanically, this effect was observed regardless of inflammasome activation. Further studies revealed that overexpression of NLRP1 downregulated the phosphorylation of DRP1 and promoted mitochondrial fusion, which was mediated by inhibition of NF-κB activity. NF-κB agonist could neutralize the effect of NLRP1 on mitochondrial dynamics. In addition, LUAD sensitivity to cisplatin was enhanced by decreased mitochondrial fission resulting from up-regulated NLRP1. In conclusion, our findings demonstrated an unexpected role of NLRP1 in LUAD by modulating mitochondrial activities, which provides strong evidence for its potential in LUAD treatment.
Collapse
Affiliation(s)
- Chen-jing Lin
- Department of Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang-ang Tian
- Department of Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen-ya Zhao
- Department of Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Tian
- Department of Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi-ru Liu
- Department of Chemotherapy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Dian-na Gu
- Department of Chemotherapy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ling Tian
- Department of Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
31
|
Dai X, Liu Y, Wu Y, Wang S, Guo Q, Feng X, Zhao F, Li Y, Lan L, Li X. DYZY01 alleviates pulmonary hypertension via inhibiting endothelial cell pyroptosis and rescuing endothelial dysfunction. Eur J Pharmacol 2024; 978:176785. [PMID: 38942262 DOI: 10.1016/j.ejphar.2024.176785] [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: 11/07/2023] [Revised: 06/18/2024] [Accepted: 06/26/2024] [Indexed: 06/30/2024]
Abstract
Pulmonary hypertension (PH) is a malignant pulmonary vascular disease with a poor prognosis. Although the development of targeted drugs for this disease has made some breakthroughs in recent decades, PH remains incurable. Therefore, innovative clinical treatment methods and drugs for PH are still urgently needed. DYZY01 is a new drug whose main ingredient is high-purity cannabidiol, a non-psychoactive constituent of cannabinoids that was demonstrated to have anti-inflammatory and anti-pyroptosis properties. Several recent studies have found cannabidiol could improve experimental PH, whereas the mechanistic effect of it warrants further investigation. Thus, this study aimed to investigate whether DYZY01 can treat PH by inhibiting inflammation and pyroptosis and to reveal its underlying mechanism. We established hypoxia and monocrotaline (MCT)-induced PH rat models in vivo and treated them with either DYZY01 (10,50 mg/kg/d) or Riociguat (10 mg/kg/d) by oral administration. The mean pulmonary arterial pressure (mPAP), right ventricular hypertrophy index (RVHI), and extent of vascular remodeling were measured. Meanwhile, the effect of DYZY01 on human pulmonary arterial endothelial cells (HPAECs) was assessed in vitro. The results indicated that DYZY01 significantly reduced mPAP and RVHI in PH rats and reversed the extent of pulmonary vascular remodeling. This improvement may have been achieved by reducing endothelial cell pyroptosis via inhibiting the NF-κB/NLRP3/Caspase-1 pathway. Furthermore, DYZY01 could improve endothelial vascular function, possibly by regulating the secretion of vasodilator factors and inhibiting the proliferation and migration of pulmonary endothelial cells.
Collapse
MESH Headings
- Animals
- Pyroptosis/drug effects
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/physiopathology
- Hypertension, Pulmonary/pathology
- Rats
- Male
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Humans
- Rats, Sprague-Dawley
- NF-kappa B/metabolism
- Vascular Remodeling/drug effects
- Cannabidiol/pharmacology
- Cannabidiol/therapeutic use
- Disease Models, Animal
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Pulmonary Artery/drug effects
- Pulmonary Artery/pathology
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Xuejing Dai
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Yi Liu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Yusi Wu
- School of Medicine, Hunan Normal University, Changsha, 410013, Hunan, China
| | - Shubin Wang
- Deyi Pharmaceutical Company Ltd., 102600, Beijing, China
| | - Qing Guo
- Deyi Pharmaceutical Company Ltd., 102600, Beijing, China
| | - Xuexiang Feng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Feilong Zhao
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Ying Li
- Department of Health Management, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Lan Lan
- Deyi Pharmaceutical Company Ltd., 102600, Beijing, China.
| | - Xiaohui Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China.
| |
Collapse
|
32
|
Shao C, Yan X, Li H, Nian D, Ren L, Pang S, Sun J. Intranuclear Irradiation Inhibits Solid Tumor Growth by Upregulating Caspase8 and Activating Apoptosis. Mol Pharm 2024; 21:4259-4271. [PMID: 39077844 DOI: 10.1021/acs.molpharmaceut.4c00092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/31/2024]
Abstract
Radioimmunotherapy (RIT) is a novel and promising cancer treatment method, with ongoing research focusing on RIT antibody selection, radionuclides, treatment options, and benefited patient groups. As we dive into the mechanisms of tumor biology, a deeper exploration of how RIT affects tumor tissue is needed to provide new ways to improve clinical treatment outcome and patient prognosis. We labeled the anti-PD-L1 monoclonal antibody atezolizumab with iodine-131 (131I), separated and purified the labeled mAb with Sephadex G-25 medium gel filtration resin, and tested product stability. We detected the in vivo activity of 131I-PD-L1 mAb by analyzing its in vivo biodistribution and performing SPECT imaging and then set different treatment groups to study the effect of 131I-atezolizumab on the survival of tumor-bearing mice. Western blot, real-time quantitative PCR, and immunohistochemistry were used to detect the expression level of Caspase8 and Nlrp3 in tumor. TUNEL fluorescence staining was used to detect the apoptosis in the tumor. The radiopharmaceutical molecular probe 131I-atezolizumab showed high stability and in vivo biological activity. The treatment regimen adopted had a positive effect on the survival of tumor-bearing mice. 131I internal irradiation upregulated Caspase8 in tumor and ultimately inhibited solid tumor growth by activating apoptosis pathways. We also found a significant increase in the expression of NLRP3, which plays an important role in the pyroptosis pathway, in tumor. In summary, our data demonstrated that radiopharmaceuticals combined with immunotherapy affected tumor tissue by modulating relevant biological pathways, thereby achieving better antitumor effects compared with single therapy and providing new insights for promoting better patient prognosis and combination treatment strategies.
Collapse
Affiliation(s)
- Chenxu Shao
- Department of Nuclear Medicine, School of Laboratory Medicine, Bengbu Medical University, Bengbu 233030, Anhui Province, P. R. China
| | - Xiaoping Yan
- Department of Radiology, The People's Hospital of Jiangyou, Jiangyou 621799, P. R. China
| | - Hui Li
- Department of Nuclear Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, P. R. China
| | - Di Nian
- Department of Nuclear Medicine, School of Laboratory Medicine, Bengbu Medical University, Bengbu 233030, Anhui Province, P. R. China
| | - Li Ren
- Department of Nuclear Medicine, School of Laboratory Medicine, Bengbu Medical University, Bengbu 233030, Anhui Province, P. R. China
| | - Shangjie Pang
- Department of Nuclear Medicine, School of Laboratory Medicine, Bengbu Medical University, Bengbu 233030, Anhui Province, P. R. China
| | - Junjie Sun
- Department of Nuclear Medicine, School of Laboratory Medicine, Bengbu Medical University, Bengbu 233030, Anhui Province, P. R. China
| |
Collapse
|
33
|
Wang X, Yin QH, Wan LL, Sun RL, Wang G, Gu JF, Tang DC. Research progress on the effect of pyroptosis on the occurrence, development, invasion and metastasis of colorectal cancer. World J Gastrointest Oncol 2024; 16:3410-3427. [PMID: 39171180 PMCID: PMC11334039 DOI: 10.4251/wjgo.v16.i8.3410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/28/2024] [Accepted: 07/04/2024] [Indexed: 08/07/2024] Open
Abstract
Pyroptosis is a type of programmed cell death mediated by gasdermines (GSDMs). The N-terminal domain of GSDMs forms pores in the plasma membrane, causing cell membrane rupture and the release of cell contents, leading to an inflammatory response and mediating pyrodeath. Pyroptosis plays an important role in inflammatory diseases and malignant tumors. With the further study of pyroptosis, an increasing number of studies have shown that the pyroptosis pathway can regulate the tumor microenvironment and antitumor immunity of colorectal cancer and is closely related to the occurrence, development, treatment and prognosis of colorectal cancer. This review aimed to explore the molecular mechanism of pyroptosis and the role of pyroptosis in the occurrence, development, treatment and prognosis of colorectal cancer (CRC) and to provide ideas for the clinical diagnosis and treatment of CRC.
Collapse
Affiliation(s)
- Xu Wang
- School of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Qi-Hang Yin
- School of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Lin-Lu Wan
- School of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Ruo-Lan Sun
- School of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Gang Wang
- Department of Ana and Intestine Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Jun-Fei Gu
- School of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - De-Cai Tang
- School of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| |
Collapse
|
34
|
Zhang K, Lin G, Nie Z, Jin S, Bing X, Li Z, Li M. TRIM38 suppresses migration, invasion, metastasis, and proliferation in non-small cell lung cancer (NSCLC) via regulating the AMPK/NF-κB/NLRP3 pathway. Mol Cell Biochem 2024; 479:2069-2079. [PMID: 37566200 DOI: 10.1007/s11010-023-04823-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 07/25/2023] [Indexed: 08/12/2023]
Abstract
Accumulating data have revealed the pivotal function of tripartite motif protein 38 (TRIM38) in tumors. In view of this, this investigation aims to explore the function and potential mechanism of TRIM38 in non-small cell lung cancer (NSCLC). A xenotypic tumor model was established in vivo by subcutaneously injecting NSCLC cells (2 × 106 cells) in tail vein of each mouse. Relative expression of TRIM38 mRNA was detected via quantitative real-time polymerase chain reaction (qRT-PCR). For exploring the role of TRIM38 in vivo and in vitro, mice or NSCLC cells were divided into two groups: the vector group and the TRIM38 overexpression group. Also, protein expression levels of TRIM38, Vimentin, E-cadherin, and N-cadherin were determined using western blotting and immunohistochemistry staining. Tumor nodules of mouse lung tissues were assessed via performing H&E staining. Moreover, proliferation of NSCLC cells was evaluated through colony formation and CCK-8 assays. Further, migration and invasion of NSCLC cells were assessed through wound healing and transwell assays. Protein levels of pathway-related proteins including p-p65, p65, IκB, p-IκB, p-AMPK, AMPK, and NLRP3 were examined through western blotting analysis. Tumor lung tissues of mice and NSCLC cells showed low protein and mRNA expression of TRIM38. Functionally, up-regulation of TRIM38 reduced the number of tumor nodules and suppressed epithelial-to-mesenchymal transition (EMT) in lung tissues of mice. Furthermore, up-regulation of TRIM38 in NSCLC cells inhibited migration, invasion, EMT, and proliferation. With respect to the mechanism, in vivo experiments, the inhibitory effects of TRIM38 overexpression on tumor nodules, and EMT were reversed by AMPK inhibitor. In vitro experiments, TRIM38 overexpression caused down-regulation of p-IκB and p-p65 as well as up-regulation of p-AMPK. The inhibitory effects of TRIM38 overexpression on migration, proliferation, invasion, and EMT of NSCLC cells were reversed by overexpression of NLRP3. Concurrently, AMPK inhibitor enhanced the TRIM38-overexpressed NSCLC cell's abilities in migration, clone formation, invasion, and proliferation. TRIM38 regulated the AMPK/NF-κB/NLRP3 pathway to suppress the NSCLC's progression and development.
Collapse
Affiliation(s)
- Kaihua Zhang
- Department of Thoracic Surgery, China Aerospace Science & Industry Corporation 731 Hospital, No. 3, Zhen Gang Nan Li, Yun Gang, Feng Tai District, Beijing, 100074, China
| | - Guihu Lin
- Department of Thoracic Surgery, China Aerospace Science & Industry Corporation 731 Hospital, No. 3, Zhen Gang Nan Li, Yun Gang, Feng Tai District, Beijing, 100074, China
| | - Zhenkai Nie
- Department of Thoracic Surgery, China Aerospace Science & Industry Corporation 731 Hospital, No. 3, Zhen Gang Nan Li, Yun Gang, Feng Tai District, Beijing, 100074, China
| | - Shan Jin
- Department of Thoracic Surgery, China Aerospace Science & Industry Corporation 731 Hospital, No. 3, Zhen Gang Nan Li, Yun Gang, Feng Tai District, Beijing, 100074, China
| | - Xiaohan Bing
- Department of Thoracic Surgery, China Aerospace Science & Industry Corporation 731 Hospital, No. 3, Zhen Gang Nan Li, Yun Gang, Feng Tai District, Beijing, 100074, China
| | - Zhantao Li
- Department of Thoracic Surgery, China Aerospace Science & Industry Corporation 731 Hospital, No. 3, Zhen Gang Nan Li, Yun Gang, Feng Tai District, Beijing, 100074, China
| | - Mingru Li
- Department of Thoracic Surgery, China Aerospace Science & Industry Corporation 731 Hospital, No. 3, Zhen Gang Nan Li, Yun Gang, Feng Tai District, Beijing, 100074, China.
| |
Collapse
|
35
|
Brint A, Greene S, Fennig-Victor AR, Wang S. Multiple sclerosis: the NLRP3 inflammasome, gasdermin D, and therapeutics. ANNALS OF TRANSLATIONAL MEDICINE 2024; 12:62. [PMID: 39118955 PMCID: PMC11304424 DOI: 10.21037/atm-23-1960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 04/26/2024] [Indexed: 08/10/2024]
Abstract
Multiple sclerosis (MS) stands as a chronic inflammatory disease characterized by its neurodegenerative impacts on the central nervous system. The complexity of MS and the significant challenges it poses to patients have made the exploration of effective treatments a crucial area of research. Among the various mechanisms under investigation, the role of inflammation in MS progression is of particular interest. Inflammatory responses within the body are regulated by various cellular mechanisms, one of which involves the nucleotide-binding oligomerization domain (NOD)-, leucine-rich repeat (LRR)-, and pyrin domains (PYD)-containing protein 3 (NLRP3). NLRP3 acts as a sensor within cells, playing a pivotal role in controlling the inflammatory response. Its activation is a critical step leading to the assembly of the NLRP3 inflammasome complex, a process that has profound implications for inflammatory diseases like MS. The NLRP3 inflammasome's activation is intricately linked to the subsequent activation of caspase 1 and gasdermin D (GsdmD), signaling pathways that are central to the inflammatory process. GsdmD, a prominent member of the Gasdermin protein family, is particularly noteworthy for its role in pyroptotic cell death, a form of programmed cell death that is distinct from apoptosis and is characterized by its inflammatory nature. This pathway's activation contributes significantly to the pathology of MS by exacerbating inflammatory responses within the nervous system. Given the detrimental effects of unregulated inflammation in MS, therapeutics targeting these inflammatory processes offer a promising avenue for alleviating the symptoms experienced by patients. This review delves into the intricacies of the pyroptotic pathways, highlighting how the formation of the NLRP3 inflammasome induces such pathways and the potential intervention points for therapeutic agents. By inhibiting key steps within these pathways, it is possible to mitigate the inflammatory response, thereby offering relief to those suffering from MS. Understanding these mechanisms not only sheds light on the pathophysiology of MS but also paves the way for the development of novel therapeutic strategies aimed at controlling the disease's progression through the modulation of the body's inflammatory response.
Collapse
Affiliation(s)
- Amie Brint
- Chemistry Department, University of Arkansas at Little Rock, Little Rock, AR, USA
- College of Medicine and Graduate School, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Seth Greene
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, USA
| | - Alyssa R. Fennig-Victor
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, USA
| | - Shanzhi Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, USA
| |
Collapse
|
36
|
Bayat M, Nahand JS. Let's make it personal: CRISPR tools in manipulating cell death pathways for cancer treatment. Cell Biol Toxicol 2024; 40:61. [PMID: 39075259 PMCID: PMC11286699 DOI: 10.1007/s10565-024-09907-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 07/18/2024] [Indexed: 07/31/2024]
Abstract
Advancements in the CRISPR technology, a game-changer in experimental research, have revolutionized various fields of life sciences and more profoundly, cancer research. Cell death pathways are among the most deregulated in cancer cells and are considered as critical aspects in cancer development. Through decades, our knowledge of the mechanisms orchestrating programmed cellular death has increased substantially, attributed to the revolution of cutting-edge technologies. The heroic appearance of CRISPR systems have expanded the available screening platform and genome engineering toolbox to detect mutations and create precise genome edits. In that context, the precise ability of this system for identification and targeting of mutations in cell death signaling pathways that result in cancer development and therapy resistance is an auspicious choice to transform and accelerate the individualized cancer therapy. The concept of personalized cancer therapy stands on the identification of molecular characterization of the individual tumor and its microenvironment in order to provide a precise treatment with the highest possible outcome and minimum toxicity. This study explored the potential of CRISPR technology in precision cancer treatment by identifying and targeting specific cell death pathways. It showed the promise of CRISPR in finding key components and mutations involved in programmed cell death, making it a potential tool for targeted cancer therapy. However, this study also highlighted the challenges and limitations that need to be addressed in future research to fully realize the potential of CRISPR in cancer treatment.
Collapse
Affiliation(s)
- Mobina Bayat
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, 15731, Iran
| | - Javid Sadri Nahand
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, 15731, Iran.
| |
Collapse
|
37
|
Alipour S, Mardi A, Shajari N, Kazemi T, Sadeghi MR, Ahmadian Heris J, Masoumi J, Baradaran B. Unmasking the NLRP3 inflammasome in dendritic cells as a potential therapeutic target for autoimmunity, cancer, and infectious conditions. Life Sci 2024; 348:122686. [PMID: 38710282 DOI: 10.1016/j.lfs.2024.122686] [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: 01/17/2024] [Revised: 04/13/2024] [Accepted: 05/03/2024] [Indexed: 05/08/2024]
Abstract
Proper and functional immune response requires a complex interaction between innate and adaptive immune cells, which dendritic cells (DCs) are the primary actors in this coordination as professional antigen-presenting cells. DCs are armed with numerous pattern recognition receptors (PRRs) such as nucleotide-binding and oligomerization domain-like receptors (NLRs) like NLRP3, which influence the development of their activation state upon sensation of ligands. NLRP3 is a crucial component of the immune system for protection against tumors and infectious agents, because its activation leads to the assembly of inflammasomes that cause the formation of active caspase-1 and stimulate the maturation and release of proinflammatory cytokines. But, when NLRP3 becomes overactivated, it plays a pathogenic role in the progression of several autoimmune disorders. So, NLRP3 activation is strictly regulated by diverse signaling pathways that are mentioned in detail in this review. Furthermore, the role of NLRP3 in all of the diverse immune cells' subsets is briefly mentioned in this study because NLRP3 plays a pivotal role in modulating other immune cells which are accompanied by DCs' responses and subsequently influence differentiation of T cells to diverse T helper subsets and even impact on cytotoxic CD8+ T cells' responses. This review sheds light on the functional and therapeutic role of NLRP3 in DCs and its contribution to the occurrence and progression of autoimmune disorders, prevention of diverse tumors' development, and recognition and annihilation of various infectious agents. Furthermore, we highlight NLRP3 targeting potential for improving DC-based immunotherapeutic approaches, to be used for the benefit of patients suffering from these disorders.
Collapse
Affiliation(s)
- Shiva Alipour
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amirhossein Mardi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Neda Shajari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Kazemi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Sadeghi
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Javad Masoumi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
38
|
Xia S, Ma R. Tributyltin chloride induces chondrocyte damage through the activation of NLRP3‑mediated inflammation and pyroptosis. Mol Med Rep 2024; 30:122. [PMID: 38785157 PMCID: PMC11130746 DOI: 10.3892/mmr.2024.13247] [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: 02/06/2024] [Accepted: 04/11/2024] [Indexed: 05/25/2024] Open
Abstract
Tributyltin chloride (TBTC) is known to have effects and mechanisms in various diseases; however, whether TBTC is detrimental to joints and causes osteoarthritis (OA), as well as its underlying mechanism, has not yet been fully elucidated. The present study explored the effects of TBTC on rat chondrocytes, as well as on mouse OA. The toxicity of TBTC toward rat chondrocytes was detected using a lactate dehydrogenase (LDH) leakage assay and cell viability was evaluated using the Cell Counting Kit‑8 assay. The results showed that TBTC decreased the viability of rat chondrocytes and increased the LDH leakage rate in a concentration‑dependent manner. Moreover, compared with in the control group, TBTC increased the expression levels of interleukin (IL)‑1β, IL‑18, matrix metalloproteinase (MMP)‑1, MMP‑13, NLR family pyrin domain containing 3 (NLRP3), caspase‑1, PYD and CARD domain containing, and gasdermin D in chondrocytes. Furthermore, knockdown of NLRP3 reversed the TBTC‑induced increases in LDH leakage and NLRP3 inflammasome‑associated protein levels. In vivo, TBTC exacerbated cartilage tissue damage in mice from the OA group, as evidenced by the attenuation of safranin O staining. In conclusion, TBTC may aggravate OA in mice by promoting chondrocyte damage and inducing pyroptosis through the activation of NLRP3 and caspase‑1 signaling. The present study demonstrated that TBTC can cause significant damage to the articular cartilage; therefore, TBTC contamination should be strictly monitored.
Collapse
Affiliation(s)
- Silong Xia
- Department of Orthopedics, The Affiliated Jianhu Hospital of Yangzhou University, Yancheng, Jiangsu 224700, P.R. China
| | - Rong Ma
- Department of Orthopedics, The Affiliated Jianhu Hospital of Yangzhou University, Yancheng, Jiangsu 224700, P.R. China
| |
Collapse
|
39
|
Al Mamun A, Shao C, Geng P, Wang S, Xiao J. Pyroptosis in Diabetic Peripheral Neuropathy and its Therapeutic Regulation. J Inflamm Res 2024; 17:3839-3864. [PMID: 38895141 PMCID: PMC11185259 DOI: 10.2147/jir.s465203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 06/05/2024] [Indexed: 06/21/2024] Open
Abstract
Pyroptosis is a pro-inflammatory form of cell death resulting from the activation of gasdermins (GSDMs) pore-forming proteins and the release of several pro-inflammatory factors. However, inflammasomes are the intracellular protein complexes that cleave gasdermin D (GSDMD), leading to the formation of robust cell membrane pores and the initiation of pyroptosis. Inflammasome activation and gasdermin-mediated membrane pore formation are the important intrinsic processes in the classical pyroptotic signaling pathway. Overactivation of the NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome triggers pyroptosis and amplifies inflammation. Current evidence suggests that the overactivation of inflammasomes and pyroptosis may further induce the progression of cancers, nerve injury, inflammatory disorders and metabolic dysfunctions. Current evidence also indicates that pyroptosis-dependent cell death accelerates the progression of diabetes and its frequent consequences including diabetic peripheral neuropathy (DPN). Pyroptosis-mediated inflammatory reaction further exacerbates DPN-mediated CNS injury. Accumulating evidence shows that several molecular signaling mechanisms trigger pyroptosis in insulin-producing cells, further leading to the development of DPN. Numerous studies have suggested that certain natural compounds or drugs may possess promising pharmacological properties by modulating inflammasomes and pyroptosis, thereby offering potential preventive and practical therapeutic approaches for the treatment and management of DPN. This review elaborates on the underlying molecular mechanisms of pyroptosis and explores possible therapeutic strategies for regulating pyroptosis-regulated cell death in the pharmacological treatment of DPN.
Collapse
Affiliation(s)
- Abdullah Al Mamun
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, 323000, People’s Republic of China
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People’s Republic of China
| | - Chuxiao Shao
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, 323000, People’s Republic of China
| | - Peiwu Geng
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, 323000, People’s Republic of China
| | - Shuanghu Wang
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, 323000, People’s Republic of China
| | - Jian Xiao
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, 323000, People’s Republic of China
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People’s Republic of China
- Department of Wound Healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People’s Republic of China
| |
Collapse
|
40
|
Lin CN, Liang YL, Tsai HF, Wu PY, Huang LY, Lin YH, Kang CY, Yao CL, Shen MR, Hsu KF. Adipocyte pyroptosis occurs in omental tumor microenvironment and is associated with chemoresistance of ovarian cancer. J Biomed Sci 2024; 31:62. [PMID: 38862973 PMCID: PMC11167873 DOI: 10.1186/s12929-024-01051-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 05/27/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND Ovarian carcinoma (OC) is a fatal malignancy, with most patients experiencing recurrence and resistance to chemotherapy. In contrast to hematogenous metastasizing tumors, ovarian cancer cells disseminate within the peritoneal cavity, especially the omentum. Previously, we reported omental crown-like structure (CLS) number is associated with poor prognosis of advanced-stage OC. CLS that have pathologic features of a dead or dying adipocyte was surrounded by several macrophages is well known a histologic hallmark for inflammatory adipose tissue. In this study, we attempted to clarify the interaction between metastatic ovarian cancer cells and omental CLS, and to formulate a therapeutic strategy for advanced-stage ovarian cancer. METHODS A three-cell (including OC cells, adipocytes and macrophages) coculture model was established to mimic the omental tumor microenvironment (TME) of ovarian cancer. Caspase-1 activity, ATP and free fatty acids (FFA) levels were detected by commercial kits. An adipocyte organoid model was established to assess macrophages migration and infiltration. In vitro and in vivo experiments were performed for functional assays and therapeutic effect evaluations. Clinical OC tissue samples were collected for immunochemistry stain and statistics analysis. RESULTS In three-cell coculture model, OC cells-derived IL-6 and IL-8 could induce the occurrence of pyroptosis in omental adipocytes. The pyroptotic adipocytes release ATP to increase macrophage infiltration, release FFA into TME, uptake by OC cells to increase chemoresistance. From OC tumor samples study, we demonstrated patients with high gasdermin D (GSDMD) expression in omental adipocytes is highly correlated with chemoresistance and poor outcome in advanced-stage OC. In animal model, by pyroptosis inhibitor, DSF, effectively retarded tumor growth and prolonged mice survival. CONCLUSIONS Omental adipocyte pyroptosis may contribute the chemoresistance in advanced stage OC. Omental adipocytes could release FFA and ATP through the GSDMD-mediate pyroptosis to induce chemoresistance and macrophages infiltration resulting the poor prognosis in advanced-stage OC. Inhibition of adipocyte pyroptosis may be a potential therapeutic modality in advanced-stage OC with omentum metastasis.
Collapse
Affiliation(s)
- Chang-Ni Lin
- Department of Obstetrics and Gynecology, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, 138, Sheng-Li Road, Tainan, 70428, Taiwan
| | - Yu-Ling Liang
- Department of Obstetrics and Gynecology, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, 138, Sheng-Li Road, Tainan, 70428, Taiwan
| | - Hsing-Fen Tsai
- Department of Obstetrics and Gynecology, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, 138, Sheng-Li Road, Tainan, 70428, Taiwan
| | - Pei-Ying Wu
- Department of Obstetrics and Gynecology, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, 138, Sheng-Li Road, Tainan, 70428, Taiwan
| | - Lan-Yin Huang
- Department of Obstetrics and Gynecology, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, 138, Sheng-Li Road, Tainan, 70428, Taiwan
| | - Yu-Han Lin
- Department of Obstetrics and Gynecology, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, 138, Sheng-Li Road, Tainan, 70428, Taiwan
| | - Chieh-Yi Kang
- Department of Obstetrics and Gynecology, Chi-Mei Medical Center, Tainan, Taiwan
| | - Chao-Ling Yao
- Department of Chemical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Meng-Ru Shen
- Department of Obstetrics and Gynecology, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, 138, Sheng-Li Road, Tainan, 70428, Taiwan
| | - Keng-Fu Hsu
- Department of Obstetrics and Gynecology, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, 138, Sheng-Li Road, Tainan, 70428, Taiwan.
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
41
|
Zhang R, Guan S, Meng Z, Deng X, Lu J. 3-MCPD Induces Renal Cell Pyroptosis and Inflammation by Inhibiting ESCRT-III-Mediated Cell Repair and Mitophagy. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024. [PMID: 38857427 DOI: 10.1021/acs.jafc.4c01994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
3-Monochloropropane-1,2-diol (3-MCPD) is a chloropropyl alcohol contaminant mainly from the thermal processing of food and could affect kidneys. Pyroptosis is programmed cell death mediated by inflammasomes and gasdermins, and excessive cellular pyroptosis and inflammation can lead to tissue injury. In the present study, we found that 3-MCPD increased lactate dehydrogenase (LDH) levels in vitro and in vivo, increased the protein expression of NOD-like receptor family pyrin domain containing 3 (NLRP3), N-terminal domain of GSDMD (GSDMD-N), and cleaved caspase-1 and promoted the release of interleukin-1β (IL-1β) and interleukin-18 (IL-18), which induced renal cell pyroptosis and inflammation. Mechanistic studies indicated that the addition of N-acetylcysteine (NAC), a ROS scavenger, inhibited NLRP3 activation and attenuated pyroptosis. Furthermore, we revealed that 3-MCPD induced ROS accumulation by inhibiting ESCRT-III-mediated mitophagy. These results were further validated by the overexpression of charged multivesicular body protein 4B (CHMP4B), a key subunit of ESCRT-III, and the addition of the mitophagy activator carbonyl cyanide m-chlorophenylhydrazone (CCCP) and rapamycin (Rapa). Thus, our results showed that 3-MCPD could induce mitochondrial damage and produce ROS. 3-MCPD suppressed mitophagy, leading to the accumulation of damaged mitochondria and ROS, thereby activating NLRP3 and pyroptosis. Meanwhile, 3-MCPD-mediated suppression of ESCRT-III hindered the repair of GSDMD-induced cell membrane rupture, which further caused the occurrence of pyroptosis. Our findings provide new perspectives for studying the mechanisms underlying 3-MCPD-induced renal injury.
Collapse
Affiliation(s)
- Ranran Zhang
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - Shuang Guan
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Zhuoqun Meng
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - Xuming Deng
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Jing Lu
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| |
Collapse
|
42
|
Wu X, Wang S, Lin L, Jia X, Hu C, Qi H, Lin H, Zheng R, Li M, Xu Y, Xu M, Chen L, Zeng T, Hu R, Ye Z, Shi L, Su Q, Yu X, Yan L, Wang T, Zhao Z, Zheng J, Qin G, Wan Q, Chen G, Dai M, Tang X, Gao Z, Shen F, Gu X, Luo Z, Qin Y, Chen L, Hou X, Huo Y, Li Q, Wang G, Zhang Y, Liu C, Wang Y, Wu S, Yang T, Deng H, Zhao J, Mu Y, Ning G, Wang W, Bi Y, Chen Y, Lu J. Association between triglyceride glucose index and breast cancer in 142,184 Chinese adults: findings from the REACTION study. Front Endocrinol (Lausanne) 2024; 15:1321622. [PMID: 38904041 PMCID: PMC11186986 DOI: 10.3389/fendo.2024.1321622] [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: 10/14/2023] [Accepted: 05/21/2024] [Indexed: 06/22/2024] Open
Abstract
Background The triglyceride glucose (TyG) index has been associated with an increased risk in breast cancer. However, this association remains unclear among the Chinese population. This study aimed to investigate whether the TyG index is associated with the risk of prevalent breast cancer in Chinese women. Methods This cross-sectional study included 142,184 women from the REACTION (Risk Evaluation of Cancers in Chinese Diabetic Individuals: A Longitudinal) Study, which recruited adults aged 40 years or older from 25 centers across mainland China between 2011 and 2012. The TyG index was calculated according to the formula: Ln (fasting triglycerides [mg/dL] × fasting glucose [mg/dL]/2). Multivariable-adjusted logistic regression models were used to evaluate odds ratios (ORs) and 95% confidence intervals (CIs) regarding the associations between the TyG index and breast cancer. Results Multivariable-adjusted logistic regression analysis showed that compared with the lowest quartile of the TyG index, the highest quartile of the TyG index was significantly associated with an increased risk of prevalent breast cancer, with an OR (95% CI) of 1.61 (1.19-2.17). In the stratified analysis, the association of each 1 SD increase in the TyG index with risk of prevalent breast cancer was more dominant in individuals with menarche at age 13-17, those who were postmenopausal, those with a history of breastfeeding, and those who had two to four children, with the ORs (95% CIs) of 1.35 (1.09-1.68), 1.27 (1.05-1.54), 1.26 (1.05-1.52), and 1.32 (1.08-1.62), respectively. Moreover, among those without discernible insulin resistance (homeostatic model assessment-insulin resistance [HOMA-IR] ≥2.5), hyperglycemia and dyslipidemia, each 1 SD increase in the TyG index was associated with a 1.36-fold increase in breast cancer risk, with an OR (95% CI) of 2.36 (1.44-3.87). Conclusion The TyG index is significantly associated with the prevalent breast cancer risk among middle-aged and elderly Chinese women.
Collapse
Affiliation(s)
- Xueyan Wu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuangyuan Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin Lin
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaojing Jia
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chunyan Hu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongyan Qi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Lin
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruizhi Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mian Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lulu Chen
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tianshu Zeng
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruying Hu
- Institute of Chronic Diseases, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Zhen Ye
- Institute of Chronic Diseases, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Lixin Shi
- Department of Endocrinology, Affiliated Hospital of Guiyang Medical College, Guiyang, China
| | - Qing Su
- Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xuefeng Yu
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Yan
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tiange Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiyun Zhao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guijun Qin
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qin Wan
- Department of Endocrinology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Gang Chen
- Fujian Provincial Hospital, Fujian Medical University, Fuzhou, China
| | - Meng Dai
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xulei Tang
- Department of Endocrinology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Zhengnan Gao
- Department of Endocrinology, Dalian Municipal Central Hospital, Dalian, China
| | - Feixia Shen
- Department of Endocrinology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xuejiang Gu
- Department of Endocrinology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zuojie Luo
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yingfen Qin
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Li Chen
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China
| | - Xinguo Hou
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China
| | - Yanan Huo
- Department of Endocrinology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| | - Qiang Li
- Department of Endocrinology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guixia Wang
- Department of Endocrinology, The First Hospital of Jilin University, Changchun, China
| | - Yinfei Zhang
- Department of Endocrinology, Central Hospital of Shanghai Jiading District, Shanghai, China
| | - Chao Liu
- Department of Endocrinology, Jiangsu Province Hospital on Integration of Chinese and Western Medicine, Nanjing, China
| | - Youmin Wang
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shengli Wu
- Department of Endocrinology, Karamay Municipal People's Hospital, Xinjiang, China
| | - Tao Yang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Huacong Deng
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiajun Zhao
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Yiming Mu
- Department of Endocrinology, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yufang Bi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhong Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jieli Lu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
43
|
Stępień S, Smycz-Kubańska M, Kruszniewska-Rajs C, Gola JM, Kabut J, Olczyk P, Mielczarek-Palacz A. Clinical significance of the CXCL8/CXCR1/R2 signalling axis in patients with invasive breast cancer. Oncol Lett 2024; 27:260. [PMID: 38646491 PMCID: PMC11027109 DOI: 10.3892/ol.2024.14393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 02/08/2024] [Indexed: 04/23/2024] Open
Abstract
The C-X-C motif chemokine ligand 8 (CXCL8)-C-X-C chemokine receptor (CXCR)1/2 signalling axis is among numerous mechanisms which stimulate the immune system to defend against tumour growth and influence the tumour microenvironment to promote tumour growth. This pathway plays an important role in the development of a number of cancers including breast cancer (BC). The aim of the present study was to analyse the levels of the chemokine CXCL8 and its receptors, CXCR1 and CXCR2, in the serum of female patients with invasive BC and to assess the expression of these parameters at the mRNA level, considering molecular subtypes and degrees of cancer malignancy. The study group consisted of 62 patients with histopathologically confirmed invasive BC. The control group consisted of 18 patients with histopathologically confirmed fibroadenoma, a benign breast tumour. The levels of CXCL8, CXCR1 and CXCR2 were determined by sandwich ELISA using the CLOUD-CLONE ELISA kit. CXCL8, CXCR1 and CXCR2 transcript levels were analysed using reverse transcription-quantitative PCR. Results showed that serum CXCL8 levels in female patients with invasive BC were significantly higher compared with those in the control group (P<0.05). In addition, significantly elevated CXCR1 levels were observed in luminal B human epidermal growth factor receptor 2+ carcinoma compared with those in the control group. Analysis of CXCL8 in the serum of female patients with BC showed a statistically significant difference between clinical stage G1 and G2 (P<0.05), G2 and G3 (P<0.01), and G1 and G3 (P<0.0001). On the other hand, the analysis of CXCR1 and CXCR2 levels in the serum of the patients revealed a statistically significant difference between G2 and G3 (P<0.05). The current study showed that abnormalities in the immune response involving the CXCL8-CXCR1/2 signalling axis in patients with invasive BC are involved in the development of these tumours. Moreover, the demonstrated severity of changes occurring at protein level may suggest the potential usefulness of their determination as potential diagnostic markers in the clinic.
Collapse
Affiliation(s)
- Sebastian Stępień
- Department of Immunology and Serology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Marta Smycz-Kubańska
- Department of Immunology and Serology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Celina Kruszniewska-Rajs
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Joanna Magdalena Gola
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Jacek Kabut
- Department of Immunology and Serology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Paweł Olczyk
- Department of Community Pharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Aleksandra Mielczarek-Palacz
- Department of Immunology and Serology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| |
Collapse
|
44
|
Liu S, Hu H, Zhang M, Zhang Y, Geng R, Jin Y, Cao Y, Guo W, Liu J, Fu S. Puerarin Delays Mammary Gland Aging by Regulating Gut Microbiota and Inhibiting the p38MAPK Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:10879-10896. [PMID: 38686994 DOI: 10.1021/acs.jafc.3c09444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
Mammary gland aging is one of the most important problems faced by humans and animals. How to delay mammary gland aging is particularly important. Puerarin is a kind of isoflavone substance extracted from Pueraria lobata, which has anti-inflammatory, antioxidant, and other pharmacological effects. However, the role of puerarin in delaying lipopolysaccharide (LPS)-induced mammary gland aging and its underlying mechanism remains unclear. On the one hand, we found that puerarin could significantly downregulate the expression of senescence-associated secretory phenotype (SASP) and age-related indicators (SA-β-gal, p53, p21, p16) in mammary glands of mice. In addition, puerarin mainly inhibited the p38MAPK signaling pathway to repair mitochondrial damage and delay mammary gland aging. On the other hand, puerarin could also delay the cellular senescence of mice mammary epithelial cells (mMECs) by targeting gut microbiota and promoting the secretion of gut microbiota metabolites. In conclusion, puerarin could not only directly act on the mMECs but also regulate the gut microbiota, thus, playing a role in delaying the aging of the mammary gland. Based on the above findings, we have discovered a new pathway for puerarin to delay mammary gland aging.
Collapse
Affiliation(s)
- Shu Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Huijie Hu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Meng Zhang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yufei Zhang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Ruiqi Geng
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yuhang Jin
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yu Cao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Wenjin Guo
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
- Chongqing Research Institute, Jilin University, Chongqing 401120, China
| | - Juxiong Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Shoupeng Fu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| |
Collapse
|
45
|
Ai L, Yi N, Qiu C, Huang W, Zhang K, Hou Q, Jia L, Li H, Liu L. Revolutionizing breast cancer treatment: Harnessing the related mechanisms and drugs for regulated cell death (Review). Int J Oncol 2024; 64:46. [PMID: 38456493 PMCID: PMC11000534 DOI: 10.3892/ijo.2024.5634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 02/22/2024] [Indexed: 03/09/2024] Open
Abstract
Breast cancer arises from the malignant transformation of mammary epithelial cells under the influence of various carcinogenic factors, leading to a gradual increase in its prevalence. This disease has become the leading cause of mortality among female malignancies, posing a significant threat to the health of women. The timely identification of breast cancer remains challenging, often resulting in diagnosis at the advanced stages of the disease. Conventional therapeutic approaches, such as surgical excision, chemotherapy and radiotherapy, exhibit limited efficacy in controlling the progression and metastasis of the disease. Regulated cell death (RCD), a process essential for physiological tissue cell renewal, occurs within the body independently of external influences. In the context of cancer, research on RCD primarily focuses on cuproptosis, ferroptosis and pyroptosis. Mounting evidence suggests a marked association between these specific forms of RCD, and the onset and progression of breast cancer. For example, a cuproptosis vector can effectively bind copper ions to induce cuproptosis in breast cancer cells, thereby hindering their proliferation. Additionally, the expression of ferroptosis‑related genes can enhance the sensitivity of breast cancer cells to chemotherapy. Likewise, pyroptosis‑related proteins not only participate in pyroptosis, but also regulate the tumor microenvironment, ultimately leading to the death of breast cancer cells. The present review discusses the unique regulatory mechanisms of cuproptosis, ferroptosis and pyroptosis in breast cancer, and the mechanisms through which they are affected by conventional cancer drugs. Furthermore, it provides a comprehensive overview of the significance of these forms of RCD in modulating the efficacy of chemotherapy and highlights their shared characteristics. This knowledge may provide novel avenues for both clinical interventions and fundamental research in the context of breast cancer.
Collapse
Affiliation(s)
- Leyu Ai
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830017, P.R. China
- Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Urumqi, Xinjiang Uygur Autonomous Region 830017, P.R. China
- Department of Clinical Medicine, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830017, P.R. China
| | - Na Yi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830017, P.R. China
- Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Urumqi, Xinjiang Uygur Autonomous Region 830017, P.R. China
| | - Chunhan Qiu
- Department of Clinical Medicine, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830017, P.R. China
| | - Wanyi Huang
- Medical College, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| | - Keke Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830017, P.R. China
- Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Urumqi, Xinjiang Uygur Autonomous Region 830017, P.R. China
| | - Qiulian Hou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830017, P.R. China
- Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Urumqi, Xinjiang Uygur Autonomous Region 830017, P.R. China
| | - Long Jia
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830017, P.R. China
- Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Urumqi, Xinjiang Uygur Autonomous Region 830017, P.R. China
| | - Hui Li
- Central Laboratory of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830017, P.R. China
| | - Ling Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830017, P.R. China
- Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Urumqi, Xinjiang Uygur Autonomous Region 830017, P.R. China
| |
Collapse
|
46
|
Parveen R, Kashif M, Srinivasan H, Khan J, Yousif A, Ghataty DS, Ali N, Attia SM, Waseem M. An In Silico Investigation of Pharmacological Modulators and Inflammasomes in Glioblastoma Multiforme. Appl Biochem Biotechnol 2024; 196:2771-2797. [PMID: 37466884 DOI: 10.1007/s12010-023-04655-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2023] [Indexed: 07/20/2023]
Abstract
For the past decades, inflammatory signals have been considered a possible key for pharmacological interventions. There are several compounds and/or molecules that have been known as most promising medication against inflammation and its mediated chronic disorders. Inflammasomes could be recognized as a trigger by detrimental stimuli as pathogenic attack and endogenous signals mediated injury inside the cells. In addition, there has been an inflammatory key mechanism involved in cancers including glioblastoma multiforme (GBM). GBM has been considered the foremost aggressive primary brain tumors in adult stage. There is a scattered beam of light on both cellular and molecular links in inflammation and GBM. However, the immune response of GBM has been characterized extensively by macrophages and lymphocytes related to tumors, and some recent investigations have pinpointed the focus of inflammasomes on the progression of GBM. Nevertheless, risk factors linked with GBM are still debatable. In our study, the most considerable compounds and their bonded and/or targeted proteins have depicted the most promising highlights under in silico condition. Our in silico investigations have revealed a powerful pharmacological agents/compound against inflammasome-mediated GBM.
Collapse
Affiliation(s)
- Roohi Parveen
- School of Life Sciences, B.S. Abdur Rahman Crescent Institute of Science and Technology, Chennai, 600048, India
| | - Mohd Kashif
- School of Life Sciences, B.S. Abdur Rahman Crescent Institute of Science and Technology, Chennai, 600048, India
| | - Hemalatha Srinivasan
- School of Life Sciences, B.S. Abdur Rahman Crescent Institute of Science and Technology, Chennai, 600048, India
| | - Jasim Khan
- UAB Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Amar Yousif
- School of Pharmacy and Health Professions, University of Maryland Eastern Shore, Princess Anne, MD, USA
| | - Dina Saeed Ghataty
- Department of Pharmaceutics, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Giza, Egypt
| | - Nemat Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Mohammad Waseem
- School of Life Sciences, B.S. Abdur Rahman Crescent Institute of Science and Technology, Chennai, 600048, India.
| |
Collapse
|
47
|
Meybodi SM, Ejlalidiz M, Manshadi MR, Raeisi M, Zarin M, Kalhor Z, Saberiyan M, Hamblin MR. Crosstalk between hypoxia-induced pyroptosis and immune escape in cancer: From mechanisms to therapy. Crit Rev Oncol Hematol 2024; 197:104340. [PMID: 38570176 DOI: 10.1016/j.critrevonc.2024.104340] [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: 02/08/2024] [Revised: 03/12/2024] [Accepted: 03/28/2024] [Indexed: 04/05/2024] Open
Abstract
Pyroptosis can be triggered through both canonical and non-canonical inflammasome pathways, involving the cleavage of gasdermin (GSDM) protein family members, like GSDMD and GSDME. The impact of pyroptosis on tumors is nuanced, because its role in regulating cancer progression and anti-tumor immunity may vary depending on the tumor type, stage, location, and immune status. However, pyroptosis cannot be simply categorized as promoting or inhibiting tumors based solely on whether it is acute or chronic in nature. The interplay between pyroptosis and cancer is intricate, with some evidence suggesting that chronic pyroptosis may facilitate tumor growth, while the acute induction of pyroptosis could stimulate anti-cancer immune responses. Tumor hypoxia activates hypoxia inducible factor (HIF) signaling to modulate pyroptosis and immune checkpoint expression. Targeting this hypoxia-pyroptosis-immune escape axis could be a promising therapeutic strategy. This review highlights the complex crosstalk between hypoxia, pyroptosis, and immune evasion in the TME.
Collapse
Affiliation(s)
| | - Mahsa Ejlalidiz
- Medical Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadsadegh Rezaeian Manshadi
- Clinical Research Development Center, Imam Hossein Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Raeisi
- Clinical Research Developmental Unit, Hajar Hospital, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Maryam Zarin
- Department of Medical Genetics, Semnan University of Medical Sciences, Semnan, Iran
| | - Zahra Kalhor
- Department of Anatomical Sciences, Factulty of Medicine, Kurdistan University of Medical Scidnces, Sanandaj, Iran
| | - Mohammadreza Saberiyan
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran; Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| | - Michael R Hamblin
- Laser Research Centre, University of Johannesburg, Doornfontein, South Africa.
| |
Collapse
|
48
|
Magni L, Yu H, Christensen NM, Poulsen MH, Frueh A, Deshar G, Johansen AZ, Johansen JS, Pless SA, Jørgensen NR, Novak I. Human P2X7 receptor variants Gly150Arg and Arg276His polymorphisms have differential effects on risk association and cellular functions in pancreatic cancer. Cancer Cell Int 2024; 24:148. [PMID: 38664691 PMCID: PMC11044319 DOI: 10.1186/s12935-024-03339-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 04/18/2024] [Indexed: 04/29/2024] Open
Abstract
BACKGROUND The purinergic P2X7 receptor (P2X7R) plays an important role in the crosstalk between pancreatic stellate cells (PSCs) and cancer cells, thus promoting progression of pancreatic ductal adenocarcinoma (PDAC). Single nucleotide polymorphisms (SNPs) in the P2X7R have been reported for several cancers, but have not been explored in PDAC. MATERIALS AND METHODS Blood samples from PDAC patients and controls were genotyped for 11 non-synonymous SNPs in P2X7R and a risk analysis was performed. Relevant P2X7R-SNP GFP variants were expressed in PSCs and cancer cells and their function was assayed in the following tests. Responses in Ca2+ were studied with Fura-2 and dye uptake with YO-PRO-1. Cell migration was monitored by fluorescence microscopy. Released cytokines were measured with MSD assay. RESULTS Risk analysis showed that two SNPs 474G>A and 853G>A (rs28360447, rs7958316), that lead to the Gly150Arg and Arg276His variants, had a significant but opposite risk association with PDAC development, protecting against and predisposing to the disease, respectively. In vitro experiments performed on cancer cells and PSCs expressing the Gly150Arg variant showed reduced intracellular Ca2+ response, fluorescent dye uptake, and cell migration, while the Arg276His variant reduced dye uptake but displayed WT-like Ca2+ responses. As predicted, P2X7R was involved in cytokine release (IL-6, IL-1β, IL-8, TNF-α), but the P2X7R inhibitors displayed varied effects. CONCLUSION In conclusion, we provide evidence for the P2X7R SNPs association with PDAC and propose that they could be considered as potential biomarkers.
Collapse
Affiliation(s)
- Lara Magni
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Universitetsparken 13, 2100, Copenhagen Ø, Denmark
| | - Haoran Yu
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Universitetsparken 13, 2100, Copenhagen Ø, Denmark
| | - Nynne M Christensen
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Universitetsparken 13, 2100, Copenhagen Ø, Denmark
| | - Mette H Poulsen
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Alexander Frueh
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Universitetsparken 13, 2100, Copenhagen Ø, Denmark
| | - Ganga Deshar
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Universitetsparken 13, 2100, Copenhagen Ø, Denmark
| | - Astrid Z Johansen
- Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte Hospital, Herlev, Denmark
| | - Julia S Johansen
- Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte Hospital, Herlev, Denmark
- Department of Medicine, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
| | - Stephan A Pless
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Niklas R Jørgensen
- Department of Clinical Biochemistry, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ivana Novak
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Universitetsparken 13, 2100, Copenhagen Ø, Denmark.
| |
Collapse
|
49
|
Yang NY, Zheng HH, Yu C, Ye Y, Xie GH. Diagnosis of Canine Tumours and the Value of Combined Detection of VEGF, P53, SF and NLRP3 for the Early Diagnosis of Canine Mammary Carcinoma. Animals (Basel) 2024; 14:1272. [PMID: 38731276 PMCID: PMC11083559 DOI: 10.3390/ani14091272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 04/21/2024] [Accepted: 04/21/2024] [Indexed: 05/13/2024] Open
Abstract
The average life of a dog is generally maintained at ten to fifteen years, and tumours are the predominant reason that leads to the death of dogs, especially canine mammary carcinoma. Therefore, early diagnosis of tumours is very important. In this study, tumor size, morphology, and texture could be seen through general clinical examination, tumor metastasis could be seen through imaging examination, inflammatory reactions could be seen through hematological examination, and abnormal cell morphology could be seen through cytological and histopathological examination. In the 269 malignant cases and 179 benign cases, we randomly selected 30 cases each, and an additional 30 healthy dogs were selected for the experiment (healthy dogs: dogs in good physical condition without any tumor or other diseases). We used RT-qPCR and ELISA to determine the relative expression of vascular endothelial growth factor (VEGF), tumor protein P53 (P53), serum ferritin (SF), and NOD-like receptor protein 3 (NLRP3) in 30 healthy dogs, 30 dogs with benign mammary tumours, and 30 dogs with malignant mammary tumours. In the results, the same expression trend was obtained both in serum and tissues, and the expression of the four markers was the highest in malignant mammary tumours, with highly significant differences compared with the benign and healthy/paracancerous groups. By plotting the ROC curves, it was found that the results of combined tests were better than a single test and the combination of the four markers was the best for the early diagnosis. In conclusion, this can assist the clinical early diagnosis to a certain extent, and also provides some references and assistance for the development of tumor detection kits in clinical practice.
Collapse
Affiliation(s)
- Ning-Yu Yang
- College of Veterinary Medicine, Jilin University, No. 5333 Xi’an Road, Changchun 130062, China; (N.-Y.Y.); (H.-H.Z.); (C.Y.); (Y.Y.)
| | - Hui-Hua Zheng
- College of Veterinary Medicine, Jilin University, No. 5333 Xi’an Road, Changchun 130062, China; (N.-Y.Y.); (H.-H.Z.); (C.Y.); (Y.Y.)
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Hangzhou 311300, China
| | - Chao Yu
- College of Veterinary Medicine, Jilin University, No. 5333 Xi’an Road, Changchun 130062, China; (N.-Y.Y.); (H.-H.Z.); (C.Y.); (Y.Y.)
| | - Yan Ye
- College of Veterinary Medicine, Jilin University, No. 5333 Xi’an Road, Changchun 130062, China; (N.-Y.Y.); (H.-H.Z.); (C.Y.); (Y.Y.)
| | - Guang-Hong Xie
- College of Veterinary Medicine, Jilin University, No. 5333 Xi’an Road, Changchun 130062, China; (N.-Y.Y.); (H.-H.Z.); (C.Y.); (Y.Y.)
| |
Collapse
|
50
|
Lee Y, Yoon B, Son S, Cho E, Kim KB, Choi EY, Kim DE. Inhibition of Immunoproteasome Attenuates NLRP3 Inflammasome Response by Regulating E3 Ubiquitin Ligase TRIM31. Cells 2024; 13:675. [PMID: 38667290 PMCID: PMC11048918 DOI: 10.3390/cells13080675] [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: 02/24/2024] [Revised: 04/08/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Excessive secretion of pro-inflammatory cytokines leads to the disruption of intestinal barrier in inflammatory bowel disease (IBD). The inflammatory cytokine tumor necrosis factor alpha (TNFα) induces the assembly of the NLRP3 inflammasome, resulting in the augmented secretion of inflammatory cytokines implicated in the pathogenesis of inflammatory bowel disease (IBD). TNFα has also been known to induce the formation of immunoproteasome (IP), which incorporates immunosubunits LMP2, LMP7, and MECL-1. Inhibition of IP activity using the IP subunit LMP2-specific inhibitor YU102, a peptide epoxyketone, decreased the protein levels of NLRP3 and increased the K48-linked polyubiquitination levels of NLRP3 in TNFα-stimulated intestinal epithelial cells. We observed that inhibition of IP activity caused an increase in the protein level of the ubiquitin E3 ligase, tripartite motif-containing protein 31 (TRIM31). TRIM31 facilitated K48-linked polyubiquitination and proteasomal degradation of NLRP3 with an enhanced interaction between NLRP3 and TRIM31 in intestinal epithelial cells. In addition, IP inhibition using YU102 ameliorated the symptoms of colitis in the model mice inflicted with dextran sodium sulfate (DSS). Administration of YU102 in the DSS-treated colitis model mice caused suppression of the NLRP3 protein levels and accompanied inflammatory cytokine release in the intestinal epithelium. Taken together, we demonstrated that inhibiting IP under inflammatory conditions induces E3 ligase TRIM31-mediated NLRP3 degradation, leading to attenuation of the NLRP3 inflammatory response that triggers disruption of intestinal barrier.
Collapse
Affiliation(s)
- Yubin Lee
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (Y.L.); (B.Y.); (S.S.); (E.C.)
| | - Boran Yoon
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (Y.L.); (B.Y.); (S.S.); (E.C.)
| | - Sumin Son
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (Y.L.); (B.Y.); (S.S.); (E.C.)
| | - Eunbin Cho
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (Y.L.); (B.Y.); (S.S.); (E.C.)
| | - Kyung Bo Kim
- Department of Cellular & Molecular Medicine, Herbert Wertheim College of Medicine, Center for Translational Science at Port St. Lucie, Florida International University, 11350 SW Village Pkwy, Port St. Lucie, FL 34987, USA;
| | - Eun Young Choi
- Department of Cellular & Molecular Medicine, Herbert Wertheim College of Medicine, Center for Translational Science at Port St. Lucie, Florida International University, 11350 SW Village Pkwy, Port St. Lucie, FL 34987, USA;
| | - Dong-Eun Kim
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (Y.L.); (B.Y.); (S.S.); (E.C.)
| |
Collapse
|