1
|
Załuska-Ogryzek K, Wróblewska-Łuczka P, Góralczyk A, Luszczki JJ. Antiproliferative effects of arvanil, olvanil and LY2183240 on human neuroblastoma and glioblastoma cell lines and their in vitro interactions with cisplatin and temozolomide: an isobolographic analysis. Eur J Pharmacol 2025; 1001:177775. [PMID: 40441593 DOI: 10.1016/j.ejphar.2025.177775] [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: 03/24/2025] [Revised: 05/14/2025] [Accepted: 05/26/2025] [Indexed: 06/02/2025]
Abstract
Glioblastoma is the most aggressive malignant brain tumor characterized by rapid development, poor prognosis and high mortality. The purpose of this in vitro study was to assess cytotoxic and anti-proliferative effects of arvanil, olvanil and LY2183240 (three cannabinoid receptor ligands), when used alone and in combination with cisplatin and temozolomide in various neuroblastoma (CHP-134 and KELLY) and glioblastoma (U87MG, C6, and T98G) cell lines in the MTT assay. Results indicate that arvanil, olvanil and LY2183240 considerably affected the viability of neuroblastoma and glioblastoma cell lines with selectivity index values ranging from 0.50 to 17.48 for arvanil; from 0.93 to 24.06 for olvanil; and from 1.68 to 14.52 for LY2183240. With isobolographic analysis we found that the combination of LY2183240 with cisplatin produced three different types of interactions: a synergy in CHP-134; an antagonism in T98G; and additivity in KELLY, C6 and U87MG cell lines in the MTT assay. The combinations of arvanil and olvanil with cisplatin exerted additive interaction in all the tested cell lines in the MTT assay. Similarly, the combinations of temozolomide with arvanil, olvanil and LY 2183240 produced the additive interaction in all the tested glioblastoma and neuroblastoma cell lines. In conclusion, the combinations of temozolomide with the three cannabinoids, due to their additive interactions, can be considered as a promising therapeutic direction in glioblastoma. Favorable combinations of cisplatin with arvanil, olvanil and LY2183240 can also be recommended, except for a combination of cisplatin with LY2183240 in glioblastoma T98G cell line in the MTT assay.
Collapse
Affiliation(s)
- Katarzyna Załuska-Ogryzek
- Department of Occupational Medicine, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090, Lublin, Poland.
| | - Paula Wróblewska-Łuczka
- Department of Occupational Medicine, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090, Lublin, Poland.
| | - Agnieszka Góralczyk
- Department of Occupational Medicine, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090, Lublin, Poland.
| | - Jarogniew J Luszczki
- Department of Occupational Medicine, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090, Lublin, Poland.
| |
Collapse
|
2
|
Qiao Z, Feng X, Sun W, Wang F, Lu C. Independent and synergistic effects of extreme heat and NO 2 pollution on diabetic nephropathy in a type II diabetes mouse model. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 375:126321. [PMID: 40294690 DOI: 10.1016/j.envpol.2025.126321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 04/13/2025] [Accepted: 04/26/2025] [Indexed: 04/30/2025]
Abstract
Extreme heat and traffic-related air pollution (TRAP) have been linked to worsening chronic health disorders, however, their combined effects on diabetic nephropathy (DN) are little understood. Type II diabetic mice were exposed to heat (40 °C) and NO2 (5 ppm) separately for 4 h per day over 6 weeks to investigate the synergistic effects on the progression of DN. We found that exposure to high temperature and NO2 elevated blood glucose levels and exacerbated histopathological changes. Additionally, there were increased oxidation indicators (ROS, MDA, 8-OHdG) and decreased antioxidant indicators (CAT, SOD, GSH-PX), along with elevated inflammation markers (TNF-α, IL-1β, IL-6). The expressions of transient receptor potential (TRP) ion channels (TRPV1, TRPV4, TRPA1, TRPM2) were also upregulated. Our findings suggest that simultaneous exposure to high temperature and NO2 impairs metabolic and autophagy pathways. Exposure to both high temperature and NO2 produces a synergistic effect, leading to more severe damage than exposure to either factor individually. This resulted in increased expression of APOA1, P62, and p-mTOR/mTOR while decreasing the expression of p-AMPKα/AMPKα and LC3-II/I. This disruption promoted the progression of DN. In contrast, capsazepine (CZP) reduced TRP expression, inflammatory markers, oxidative stress, metabolic and autophagy disorders, thereby mitigating renal damage and alleviating the progression of diabetic nephropathy. Our study provides some potential strategies for early prevention and effective reduction of DN.
Collapse
Affiliation(s)
- Zipeng Qiao
- XiangYa School of Public Health, Central South University, Changsha, 410013, China
| | - Xiangling Feng
- XiangYa School of Public Health, Central South University, Changsha, 410013, China
| | - Wenying Sun
- XiangYa School of Public Health, Central South University, Changsha, 410013, China
| | - Faming Wang
- Centre for Molecular Biosciences and Non-communicable Diseases, Xi'an University of Science and Technology, Xi'an, 710054, China
| | - Chan Lu
- XiangYa School of Public Health, Central South University, Changsha, 410013, China; FuRong Laboratory, Changsha, 410078, Hunan, China; Hunan Provincial Key Laboratory of Low Carbon Healthy Building, Central South University, Changsha, 410083, China.
| |
Collapse
|
3
|
da Silva GB, Braga GDC, Simões JLB, Bagatini MD, Kempka AP. Mitochondrial dysfunction and carcinogenesis: The engagement of ion channels in cancer development. Cell Calcium 2025; 128:103010. [PMID: 40043325 DOI: 10.1016/j.ceca.2025.103010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 01/27/2025] [Accepted: 02/20/2025] [Indexed: 05/11/2025]
Abstract
Mitochondria represent a fundamental structure for cellular homeostasis, controlling multiple conditions regarding energetic functions and cellular survival. To maintain these organelles functioning in ideal conditions, their membranes count with ion channels for different inorganic ions, which must be balanced to offer the proper function for both the organelle and the cell. However, studies have shown that other health conditions impair the activities of mitochondrial ion channels, including cancer. In this sense, the altered activities of potassium, calcium, and calcium-activated potassium channels are mainly linked with cancer development and cellular homeostasis alteration, demonstrating their role as pharmacological targets. With that in mind, scientists have found significant mitochondrial and cellular responses related to apoptosis and reduction of cellular survival from cells with modulated ion channels, indicating the potential of this possible therapy in carcinogenic contexts. Nonetheless, few studies still evaluate mitochondrial ion channel modulation as a treatment against cancer. Hence, more research must be conducted on this subject.
Collapse
|
4
|
Ramer R, Hinz B. Effect of cannabinoids on the efficacy and side effects of anticancer therapeutic strategies - Current status of preclinical and clinical research. Pharmacol Ther 2025; 270:108851. [PMID: 40221102 DOI: 10.1016/j.pharmthera.2025.108851] [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: 01/31/2025] [Revised: 03/14/2025] [Accepted: 04/04/2025] [Indexed: 04/14/2025]
Abstract
Cannabinoids have attracted increasing attention in cancer research in recent decades. A major focus of current preclinical and clinical studies is on the interactions and potential risks when combined with chemotherapeutic agents, targeted therapies and other anticancer strategies. Given the extensive preclinical data on additive, synergistic and, in some cases, antagonistic tumor cell killing effects of chemotherapeutic agents and cannabinoids when co-administered, a critical analysis of these data seems essential. The available data mainly relate to combination treatments for glioblastoma, hematological malignancies and breast cancer, but also for other cancer types. Such an analysis also appears necessary because cannabinoids are used as an option to treat nausea and vomiting caused by chemotherapy, as well as tumor-related pain, and cancer patients sometimes take cannabinoids without a medical prescription. In addition, numerous recent preclinical studies also suggest cannabinoid-mediated relief of other chemotherapy-related side effects such as peripheral neuropathy, nephrotoxicity, cardiotoxicity, cystitis, bladder complications and mucositis. To summarize, the data available to date raise the prospect that cannabinoids may increase the efficacy of chemotherapeutic agents while reducing their side effects. However, preclinical studies on anticancer interactions are mostly limited to cytotoxicity analyses. An equally thorough investigation of the effects of such combinations on the immune system and on the tumorigenic levels of angiogenesis, invasion and metastasis is still pending. On this basis, a comprehensive understanding for the evaluation of a targeted additional treatment of various cancers with cannabinoids could be established.
Collapse
Affiliation(s)
- Robert Ramer
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, Schillingallee 70, 18057 Rostock, Germany
| | - Burkhard Hinz
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, Schillingallee 70, 18057 Rostock, Germany.
| |
Collapse
|
5
|
Pu J, Yuan K, Tao J, Qin Y, Li Y, Fu J, Li Z, Zhou H, Tang Z, Li L, Gai X, Qin D. Glioblastoma multiforme: an updated overview of temozolomide resistance mechanisms and strategies to overcome resistance. Discov Oncol 2025; 16:731. [PMID: 40353925 PMCID: PMC12069213 DOI: 10.1007/s12672-025-02567-3] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025] Open
Abstract
Glioblastoma (GBM) is an aggressive primary brain tumor with high lethality. The typical treatment regimen includes post-surgical radiotherapy and temozolomide (TMZ) chemotherapy, which helps extend survival. Nevertheless, TMZ resistance occurs in approximately 50% of patients. This resistance is primarily associated with the expression of O6-methylguanine-DNA methyltransferase (MGMT), which repairs O6-methylguanine lesions generated by TMZ and is thought to be the major mechanism of drug resistance. Additionally, the mismatch repair and base excision repair pathways play crucial roles in TMZ resistance. Emerging studies also point to drug transport mechanisms, glioma stem cells, and the heterogeneous tumor microenvironment as additional influences on TMZ resistance in gliomas. A better understanding of these mechanisms is vital for developing new treatments to improve TMZ effectiveness, such as DNA repair inhibitors, inhibitors of multidrug transporting proteins, TMZ analogs, and combination therapies targeting multiple pathways. This article discusses the main resistance mechanisms and potential strategies to counteract resistance in GBM patients, aiming to broaden the understanding of these mechanisms for future research and to explore the therapeutic effects of traditional Chinese medicines and their active components in overcoming TMZ resistance.
Collapse
Affiliation(s)
- Jianlin Pu
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
- Second Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Kai Yuan
- Second Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Jian Tao
- Department of Rehabilitation Medicine, Mojiang Hani Autonomous Country Hospital of Traditional Chinese Medicine, Mojiang, China
| | - Yuliang Qin
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
| | - Yongxin Li
- Department of Rehabilitation Medicine, Mojiang Hani Autonomous Country Hospital of Traditional Chinese Medicine, Mojiang, China
| | - Jing Fu
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
- Second Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Zhong Li
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
- Second Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Haimei Zhou
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
| | - Zhengxiu Tang
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
| | - Li Li
- Department of Emergency Trauma Surgery, The First People's Hospital of Yunnan Province, Kunming, China
| | - Xuesong Gai
- Department of Rehabilitation Medicine, The First People's Hospital of Yunnan Province, Kunming, China.
| | - Dongdong Qin
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China.
| |
Collapse
|
6
|
An M, Huang J, Zhao J, Wang L, Liu Y. PDZK1 regulated by miR-145-5p protects against endothelial cell apoptosis and diabetic retinopathy by targeting mitochondrial function. Exp Eye Res 2025; 254:110314. [PMID: 40020896 DOI: 10.1016/j.exer.2025.110314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/13/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
Mitochondria are a focus of biomedical research because of their role in apoptosis and diabetic retinopathy (DR) initiation and progression. However, the detailed mechanisms underlying mitochondrial disorders and endothelial dysfunction during DR remain elusive. We identified PDZ domain containing 1 (PDZK1) as a key factor linking endothelial mitochondrial dysfunction and cell apoptosis during DR progression. PDZK1 was downregulated by high concentrations of glucose in human retinal capillary endothelial cells (HRCECs) and decreased in serum from patients with DR. PDZK1 knockout induced endothelial cell apoptosis and an irregular and disordered arrangement of retinal cells, aggravating DR. Moreover, PDZK1 loss impaired endothelial mitochondrial function with accumulated damaged mitochondria, decreased mitochondrial DNA (mtDNA) content, and increased reactive oxygen species (ROS) production. Mechanistically, mRNA sequencing showed that PDZK1 deficiency in endothelial cells interfered with mitochondrial function by increasing ATF4 (Activating Transcription Factor 4) expression. Further studies showed that PDZK1 was inhibited by miR-145-5p. The expression of miR-145-5p was significantly upregulated in the serum of patients with DR and HRCECs with high glucose concentration, leading to endothelial dysfunction and DR progression. Our results suggested that PDZK1 deficiency is crucial in mediating retinal endothelial cell apoptosis and is associated with mitochondrial dysfunction. PDZK1 overexpression by upstream miRNA, or its downstream molecule, ATF4, may represent novel therapeutic approaches for DR treatment.
Collapse
Affiliation(s)
- Meixia An
- Department of Ophthalmology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Jialuo Huang
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Jian Zhao
- Department of Ophthalmology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Lili Wang
- Department of Ophthalmology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Yanli Liu
- Department of Ophthalmology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China.
| |
Collapse
|
7
|
Li Y, Qu S, Zuo J, Long H, Cao F, Jiang F. Progress on the functions and mechanisms of natural products in anti-glioma therapy. Chin J Nat Med 2025; 23:541-559. [PMID: 40383611 DOI: 10.1016/s1875-5364(25)60815-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/12/2024] [Accepted: 01/14/2025] [Indexed: 05/20/2025]
Abstract
Glioma, the most prevalent primary tumor of the central nervous system (CNS), is also the most lethal primary malignant tumor. Currently, there are limited chemotherapeutics available for glioma treatment, necessitating further research to identify and develop new chemotherapeutic agents. A significant approach to discovering anti-glioma drugs involves isolating antitumor active ingredients from natural products (NPs) and optimizing their structures. Additionally, targeted drug delivery systems (TDDSs) are employed to enhance drug solubility and stability and overcome the blood-brain barrier (BBB). TDDSs can penetrate deep into the brain, increase drug concentration and retention time in the CNS, and improve the targeting efficiency of NPs, thereby reducing adverse effects and enhancing anti-glioma efficacy. This paper reviews the research progress of anti-glioma activities of NPs, including alkaloids, polyphenols, flavonoids, terpenoids, saponins, quinones, and their synthetic derivatives over the past decade. The review also summarizes anti-glioma mechanisms, such as suppression of related protein expression, regulation of reactive oxygen species (ROS) levels, control of apoptosis signaling pathways, reduction of matrix metalloproteinases (MMPs) expression, blocking of vascular endothelial growth factor (VEGF), and reversal of immunosuppression. Furthermore, the functions and advantages of NP-based TDDSs in anti-glioma therapy are examined. The key information presented in this review will be valuable for the research and development of NP-based anti-glioma drugs and related TDDSs.
Collapse
Affiliation(s)
- Yanting Li
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Shuhui Qu
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Jiayi Zuo
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Haoping Long
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Feng Cao
- Department of Pharmaceutical, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Feng Jiang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
8
|
El-Hammadi MM, Martín-Navarro L, Berrocoso E, Álvarez-Fuentes J, Crespo-Facorro B, Suárez-Pereira I, Vázquez-Bourgon J, Martín-Banderas L. Enhanced Metabolic Syndrome Management Through Cannabidiol-Loaded PLGA Nanoparticles: Development and In Vitro Evaluation. J Biomed Mater Res A 2025; 113:e37916. [PMID: 40277882 DOI: 10.1002/jbm.a.37916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/07/2025] [Accepted: 04/10/2025] [Indexed: 04/26/2025]
Abstract
Cannabidiol (CBD) holds promise for managing metabolic diseases, yet enhancing its oral bioavailability and efficacy remains challenging. To address this, we developed polymeric nanoparticles (NPs), using poly(lactic-co-glycolic acid) (PLGA), encapsulating CBD using nanoprecipitation, aiming to create an effective CBD-nanoformulation for metabolic disorder treatment. These NPs (135-265 nm) demonstrated high encapsulation efficiency (EE% ≈ 100%) and sustained release kinetics. Their therapeutic potential was evaluated in an in vitro metabolic syndrome model employing sodium palmitate-induced HepG2 cells. Key assessment parameters included cell viability (MTT assay), glucose uptake, lipid accumulation (Oil Red O staining), triglycerides, cholesterol, HDL-c levels, and gene expression of metabolic regulators. Results showed an IC50 of 9.85 μg/mL for free CBD and 11.26 μg/mL for CBD-loaded NPs. CBD-loaded NPs significantly enhanced glucose uptake, reduced lipid content, lowered triglycerides and total cholesterol, and increased HDL-c levels compared to free CBD. Gene analysis indicated reduced gluconeogenesis via downregulation of PPARγ, FOXO-1, PEPCK, and G6Pase and enhanced fatty acid oxidation through CPT-1 upregulation. These findings suggest that CBD-loaded NPs may serve as a novel therapeutic strategy for the management of metabolic disorders, warranting further in vivo studies.
Collapse
Affiliation(s)
- Mazen M El-Hammadi
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidad de Sevilla, Sevilla, Spain
| | - Lucía Martín-Navarro
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidad de Sevilla, Sevilla, Spain
| | - Esther Berrocoso
- Centro de Investigación Biomédica en Red en Salud Mental (CIBERSAM), Madrid, Spain
- Neuropsychopharmacology & Psychobiology Research Group, Department of Psychology, University of Cadiz, Cádiz, Spain
- Biomedical Research and Innovation Institute of Cádiz (INiBICA) Research Unit, Puerta del Mar University Hospital, University of Cádiz, Cádiz, Spain
| | - Josefa Álvarez-Fuentes
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidad de Sevilla, Sevilla, Spain
- Instituto de Biomedicina de Sevilla (IBIS)-Campus Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Benedicto Crespo-Facorro
- Centro de Investigación Biomédica en Red en Salud Mental (CIBERSAM), Madrid, Spain
- Instituto de Biomedicina de Sevilla (IBIS)-Campus Hospital Universitario Virgen del Rocío, Sevilla, Spain
- Department of Psychiatry, School of Medicine, University Hospital Virgen del Rocio, Sevilla, Spain
| | - Irene Suárez-Pereira
- Centro de Investigación Biomédica en Red en Salud Mental (CIBERSAM), Madrid, Spain
- Biomedical Research and Innovation Institute of Cádiz (INiBICA) Research Unit, Puerta del Mar University Hospital, University of Cádiz, Cádiz, Spain
- Neuropsychopharmacology & Psychobiology Research Group, Department of Neuroscience, University of Cadiz, Cádiz, Spain
| | - Javier Vázquez-Bourgon
- Centro de Investigación Biomédica en Red en Salud Mental (CIBERSAM), Madrid, Spain
- Department of Psychiatry, University Hospital Marqués de Valdecilla. Instituto de Investigación Sanitaria Valdecilla (IDIVAL), Santander, Spain
- Departamento de Medicina y Psiquiatría, Universidad de Cantabria, Santander, Spain
| | - Lucía Martín-Banderas
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidad de Sevilla, Sevilla, Spain
- Instituto de Biomedicina de Sevilla (IBIS)-Campus Hospital Universitario Virgen del Rocío, Sevilla, Spain
| |
Collapse
|
9
|
Chang X, Huang Y, Qu Y, Guo Y, Fan W, Zhen H. Integrative analysis of mitochondrial-related gene profiling identifies prognostic clusters and drug resistance mechanisms in low-grade glioma. Discov Oncol 2025; 16:465. [PMID: 40186003 PMCID: PMC11971116 DOI: 10.1007/s12672-025-02201-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 03/21/2025] [Indexed: 04/07/2025] Open
Abstract
Mitochondrial dysfunction has emerged as a critical factor in the progression and prognosis of low-grade glioma (LGG). In this study, we explored the role of mitochondrial-related genes through consensus clustering analysis using multi-omics data from the TCGA, CGGA, and other independent datasets. Patients were categorized into three clusters (Cluster A, B, and C), with Cluster B consistently associated with poorer prognosis. Mutation landscape analysis revealed distinct genetic alterations and copy number variations among clusters, particularly in Cluster B, which exhibited unique genetic signatures. Immune infiltration analysis showed that Cluster B had higher expression levels of immune checkpoint genes, stronger immune evasion activity, and greater immune cell infiltration, suggesting an immunosuppressive tumor microenvironment. Furthermore, we identified mitochondrial-related prognostic markers and developed a MITscore based on gene expression patterns, which stratified patients into high- and low-risk groups. High MITscore groups displayed stronger stemness characteristics, poorer survival outcomes, and differential responses to chemotherapy and immunotherapy. Cross-validation with drug sensitivity and immunotherapy cohorts indicated that high MITscore patients were more sensitive to certain chemotherapeutic agents and responded better to immunotherapy. Finally, using the SRGA method, we identified novel biomarkers (KDR, LRRK2, SQSTM1) closely associated with mitochondrial function, which may serve as potential targets for therapeutic intervention. These findings highlight the critical role of mitochondrial dysfunction in LGG prognosis, tumor microenvironment regulation, and treatment response, providing new avenues for precision oncology.
Collapse
Affiliation(s)
- Xiaozan Chang
- Henan Provincial People's Hospital, Cerebrovascular Disease Hospital, Zhengzhou, 450003, Henan, China
| | - Yingxuan Huang
- Pediatric Intensive Care Unit, The Affiliated Hospital of Youjiang Medical University for Nationalities; Key Laboratory of Research and Development on Clinical Molecular Diagnosis for High-Incidence Diseases of Baise, Baise, China
| | - Ying Qu
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yu Guo
- Nanfang Hospital (ZengCheng Branch), Southern Medical University, No. 28, Innovation Avenue, Ningxi Street, Guangzhou, China.
| | - Wenwen Fan
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17, Nanli, Panjiayuan, Beijing, 100021, China.
| | - Haining Zhen
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, 127 Changle West Road, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
10
|
Liu S, Ma Z. The role of cannabinoid-mediated signaling pathways and mechanisms in brain disorders. Cell Signal 2025; 128:111653. [PMID: 39952540 DOI: 10.1016/j.cellsig.2025.111653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/17/2025] [Accepted: 02/07/2025] [Indexed: 02/17/2025]
Abstract
Cannabinoids play significant roles in the central nervous system (CNS), but cannabinoid-mediated physiopathological functions are not elaborated. Cannabinoid receptors (CBRs) mediate functions that include the regulation of neuroinflammation, oxidative stress, apoptosis, autophagy, and neurogenesis. Microglia are the primary immune cells responsible for mediating neuroinflammation in the CNS. Therefore, this article primarily focuses on microglia to summarize the inflammatory pathways mediated by cannabinoids in the CNS, including nuclear factor-κB (NF-κB), NOD-like receptor protein 3 (NLRP3) inflammasome, mitogen-activated protein kinase (MAPK), protein kinase B (Akt), and cAMP-dependent protein kinase (PKA) signaling pathways. Additionally, we provide a table summarizing the role of cannabinoids in various brain diseases. Medical use of cannabinoids has protective effects in preventing and treating brain diseases; however, excessive and repeated use can be detrimental to the CNS. We propose that cannabinoids hold significant potential for preventing and treating brain diseases, including ferroptosis, lactate metabolism, and mitophagy, providing new insights for further research on cannabinoids.
Collapse
Affiliation(s)
- Shunfeng Liu
- School of Basic Medicine, Qingdao University, Qingdao 266071, China; Institute of Brain Science and Disorders, Qingdao University, Qingdao 266071, China
| | - Zegang Ma
- School of Basic Medicine, Qingdao University, Qingdao 266071, China; Institute of Brain Science and Disorders, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
11
|
Deng K, Zhao W, Dai L, Jing Z, Ma L. Identification and validation of mitochondria-related LncRNA signatures as a novel prognostic model for glioma. Anticancer Drugs 2025; 36:297-305. [PMID: 40053488 DOI: 10.1097/cad.0000000000001677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
A predictive model for long-term survival is needed, and mitochondrial dysfunction is a key feature of cancer metabolism, though its link to glioma is not well understood. The aim of this study was to identify the molecular characteristics associated with glioma prognosis and explore its potential function. We analyzed RNA-seq data from The Cancer Genome Atlas and identified differentially expressed mitochondrial long noncoding RNAs (lncRNAs) using R's 'limma' package. A prognostic model was developed using 10 selected lncRNAs and validated with Cox regression and least absolute shrinkage and selection operator algorithm. The model's efficacy was assessed using Kaplan-Meier and receiver operating characteristic curve analyses, and its correlation with immune cell profiles and drug sensitivity was explored. A 10-mitochondria-related LncRNA signature was generated. The median risk score values are used to classify glioma samples into low-risk and high-risk groups. In breast patients, the signature-based risk score demonstrated a more potent ability to predict survival than conventional clinicopathological features. Furthermore, we noted a substantial disparity in the number of immune cells, including B cells, CD8 + T cells, and macrophages, between the two groups. In addition, the high-risk group exhibited lower half-maximal inhibitory concentration values for specific chemotherapy medications, including bortezomib, luminespib, rapamycin, and 5-fluorouracil. Our study elucidates the diagnostic and prognostic value of mitochondria-related-lncRNAs in the promotion, suppression, and treatment of glioma.
Collapse
Affiliation(s)
- Kaihan Deng
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou
| | - Wei Zhao
- Department of Oncology, Shanghai Pudong New Area Gongli Hospital, Shanghai
| | - Lin Dai
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou
| | - Zixuan Jing
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou
| | - Lixin Ma
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou
- Department of Neurosurgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
12
|
Xi X, Chen S, Zhao X, Zhou Z, Zhu S, Ren X, Wang X, Wu J, Mu S, Li X, Shan E, Cui Y. TUBB4A Inhibits Glioma Development by Regulating ROS-PINK1/Parkin-Mitophagy Pathway. Mol Neurobiol 2025; 62:3125-3142. [PMID: 39230869 DOI: 10.1007/s12035-024-04459-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 08/26/2024] [Indexed: 09/05/2024]
Abstract
Glioma is a refractory malignant tumor with a powerful capacity for invasiveness and a poor prognosis. This study aims to investigate the role and mechanism of tubulin beta class IVA (TUBB4A) in glioma progression. The differential expression of TUBB4A in humans was obtained from databases and analyzed. Glioma cells U251-MG and U87-MG were intervened by pcDNA3.1(+) and TUBB4A overexpression plasmid. MTT, CCK8, LDH, wound healing, transwell, and western blotting were used to explore whether TUBB4A participates in the development of glioma. Reactive oxygen species (ROS) were detected by the DCFH-DA probe. Mitochondrial membrane potential (MMP) was examined by JC-1. It was found that TUBB4A expression level correlated with tumor grade, IDH1 status, 1p/19q status, and poor survival in glioma patients. In addition, TUBB4A overexpression inhibited the proliferation, migration, and invasion of U251-MG and U87-MG, while increasing the degree of apoptosis. Notably, TUBB4A overexpression promotes ROS generation and MMP depolarization, and induces mitophagy through the PINK1/Parkin pathway. Interestingly, mitochondria-targeted ROS scavenger reversed the effect of TUBB4A overexpression on PINK1/Parkin expression and mitophagy, whereas mitophagy inhibitor did not affect ROS production. And the effect of TUBB4A overexpression on mitophagy and glioma progression was consistent with that of PINK1/Parkin agonist. In conclusion, TUBB4A is a molecular marker for predicting the prognosis of glioma patients and an effective target for inhibiting glioma progression by regulating ROS-PINK1/Parkin-mitophagy pathway.
Collapse
Affiliation(s)
- Xueru Xi
- School of Nursing, Nanjing Medical University, Nanjing, China
| | - Suqin Chen
- School of Nursing, Nanjing Medical University, Nanjing, China
| | - Xiaoli Zhao
- School of Nursing, Nanjing Medical University, Nanjing, China
| | - Zimu Zhou
- The Cancer Research Institute, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Shanjie Zhu
- School of Nursing, Nanjing Medical University, Nanjing, China
| | - Xurui Ren
- School of Nursing, Nanjing Medical University, Nanjing, China
| | - Xiaomei Wang
- School of Nursing, Nanjing Medical University, Nanjing, China
| | - Jing Wu
- Department of Anesthesiology, The First Medical Center of Chinese, PLA General Hospital, Beijing, China
| | - Shuai Mu
- Department of Oncology, Senior Department of Oncology, The First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Xianwen Li
- School of Nursing, Nanjing Medical University, Nanjing, China.
| | - Enfang Shan
- School of Nursing, Nanjing Medical University, Nanjing, China.
| | - Yan Cui
- School of Nursing, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
13
|
Ye H, Wan Y, Wang X, Wang S, Zhao X, Wang X, Yu T, Yan C, Tian X, Chen ZP, Liu X. Cannabidiol Protects Against Neurotoxic Reactive Astrocytes-Induced Neuronal Death in Mouse Model of Epilepsy. J Neurochem 2025; 169:e70038. [PMID: 40099400 DOI: 10.1111/jnc.70038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 02/20/2025] [Accepted: 02/25/2025] [Indexed: 03/19/2025]
Abstract
Reactive astrocytes play a critical role in the initiation and progression of epilepsy, but their molecular subtypes and functional characterization are not fully understood. In this study, we report the existence of neurotoxic reactive astrocytes, a recently identified subtype, that contribute to neuronal death in the epileptic brain. In a kainic acid (KA)-induced mouse model of epilepsy, we show that neurotoxic reactive astrocytes are induced by microglia-secreted cytokines, including IL-1α, TNFα, and C1q, and are detectable as early as 7 days post-KA stimulation. These cells exhibit a distinct molecular signature marked by elevated expression of complement 3 and adenosine 2A receptor. Transcriptomics and metabolomics analyses of human brain tissues from temporal lobe epilepsy (TLE) patients and an epileptic mouse model reveal that neurotoxic reactive astrocytes induce neuronal damage through lipid-related mechanisms. Moreover, our results demonstrate that the anti-seizure medication cannabidiol (CBD) and an adenosine 2A receptor antagonist can both suppress the formation of neurotoxic reactive astrocytes, mitigate gliosis, and reduce neuronal loss in a mouse model of epilepsy. Electrophysiological and behavioral studies indicate that cannabidiol attenuates seizure symptoms and enhances memory capabilities in epileptic mice. Our findings suggest that neurotoxic reactive astrocytes are formed at an early stage in both the KA-induced mouse model of epilepsy and TLE patients and can contribute to neuronal loss through releasing toxic lipids. Importantly, cannabidiol emerges as a promising therapeutic drug for targeted intervention against neurotoxic reactive astrocytes in adult epilepsy.
Collapse
Affiliation(s)
- Haojie Ye
- Department of Neurosurgery, The Affiliated Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
- Epilepsy Center, the Affiliated Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
- Institute for Brain Sciences, Nanjing University, Nanjing, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, China
| | - Yuhui Wan
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xin Wang
- Department of Neurosurgery, The Affiliated Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Suji Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
- Epilepsy Center, the Affiliated Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
- Institute for Brain Sciences, Nanjing University, Nanjing, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, China
| | - Xiansen Zhao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
- Epilepsy Center, the Affiliated Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
- Institute for Brain Sciences, Nanjing University, Nanjing, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, China
| | - Xinshi Wang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Tianfu Yu
- Department of Neurosurgery, The Affiliated Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Chao Yan
- Department of Neurosurgery, The Affiliated Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
- Epilepsy Center, the Affiliated Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
- Institute for Brain Sciences, Nanjing University, Nanjing, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, China
| | - Xin Tian
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
- Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, China
| | - Zhang-Peng Chen
- Epilepsy Center, the Affiliated Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
- Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiangyu Liu
- Department of Neurosurgery, The Affiliated Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
- Epilepsy Center, the Affiliated Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
| |
Collapse
|
14
|
Yin M, Zheng X, Shi L. Targeting p38 MAPK: A potential bridge between ER stress and age-related bone loss. Cell Signal 2025; 127:111549. [PMID: 39638139 DOI: 10.1016/j.cellsig.2024.111549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 11/21/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
The endoplasmic reticulum (ER) is crucial in the development of numerous age-related bone disorders. Notably, ER stress can precipitate bone loss by orchestrating inflammatory responses, apoptosis, and autophagy through the activation of the p38 MAPK pathway. Age-related bone loss diseases pose a significant burden on society and healthcare as the global population ages. This review provides a comprehensive analysis of recent research advancements, delving into the critical role of ER stress-activated p38 MAPK in inflammation, apoptosis, and autophagy, as well as its impact on bone formation and bone resorption. This review elucidates the molecular mechanisms underlying the involvement of ER stress-activated p38 MAPK in osteoporosis, rheumatoid arthritis, periodontitis, and osteoarthritis and discusses the therapeutic potential of targeting p38 MAPK. Furthermore, this review provides a scientific foundation for new therapeutic strategies by highlighting prospective research directions.
Collapse
Affiliation(s)
- Meng Yin
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China; Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xin Zheng
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Liang Shi
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| |
Collapse
|
15
|
Deng Z, Chen X, Zhang R, Kong L, Fang Y, Guo J, Shen B, Zhang L. Delta opioid peptide [D-ala2, D-leu5]-Enkephalin's ability to enhance mitophagy via TRPV4 to relieve ischemia/reperfusion injury in brain microvascular endothelial cells. Stroke Vasc Neurol 2025; 10:32-44. [PMID: 38697767 PMCID: PMC11877439 DOI: 10.1136/svn-2023-003080] [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: 01/03/2024] [Accepted: 04/20/2024] [Indexed: 05/05/2024] Open
Abstract
BACKGROUND Local brain tissue can suffer from ischaemia/reperfusion (I/R) injury, which lead to vascular endothelial damage. The peptide δ opioid receptor (δOR) agonist [D-ala2, D-leu5]-Enkephalin (DADLE) can reduce apoptosis caused by acute I/R injury in brain microvascular endothelial cells (BMECs). OBJECTIVE This study aims to explore the mechanism by which DADLE enhances the level of mitophagy in BMECs by upregulating the expression of transient receptor potential vanilloid subtype 4 (TRPV4). METHODS BMECs were extracted and made to undergo oxygen-glucose deprivation/reoxygenation (OGD/R) accompanied by DADLE. RNA-seq analysis revealed that DADLE induced increased TRPV4 expression. The CCK-8 method was used to assess the cellular viability; quantitative PCR (qPCR) was used to determine the mRNA expression of Drp1; western blot was used to determine the expression of TRPV4 and autophagy-related proteins; and calcium imaging was used to detect the calcium influx. Autophagosomes in in the cells' mitochondria were observed by using transmission electron microscopy. ELISA was used to measure ATP content, and a JC-1 fluorescent probe was used to detect mitochondrial membrane potential. RESULTS When compared with the OGD/R group, OGD/R+DADLE group showed significantly enhanced cellular viability; increased expression of TRPV4, Beclin-1, LC3-II/I, PINK1 and Parkin; decreased p62 expression; a marked rise in calcium influx; further increases in mitophagy, an increase in ATP synthesis and an elevation of mitochondrial membrane potential. These protective effects of DADLE can be blocked by a TRPV4 inhibitor HC067047 or RNAi of TRPV4. CONCLUSION DADLE can promote mitophagy in BMECs through TRPV4, improving mitochondrial function and relieving I/R injury.
Collapse
Affiliation(s)
- Zhongfang Deng
- Department of Physiology, Anhui Medical University, Hefei, Anhui, China
| | - Xiaoyu Chen
- Department of Physiology, Anhui Medical University, Hefei, Anhui, China
| | - Ran Zhang
- Department of Physiology, Anhui Medical University, Hefei, Anhui, China
| | - Lingchao Kong
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yang Fang
- Department of Physiology, Anhui Medical University, Hefei, Anhui, China
| | - Jizheng Guo
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui, China
| | - Bing Shen
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao
| | - Lesha Zhang
- Department of Physiology, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
16
|
Załuska-Ogryzek K, Wróblewska-Łuczka P, Góralczyk A, Łuszczki JJ. Isobolographic interactions of cannabidiol and AM 1172 with cisplatin in human neuroblastoma and glioblastoma cell lines: An in vitro study. Chem Biol Interact 2025; 408:111392. [PMID: 39828184 DOI: 10.1016/j.cbi.2025.111392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/10/2024] [Accepted: 01/17/2025] [Indexed: 01/22/2025]
Abstract
Glioblastoma is the most aggressive brain cancer in humans with very poor prognosis and high mortality rate. Despite advances in treatment, glioblastoma almost always recurs and new therapeutic methods are urgently needed. This study aimed at assessing the cytotoxic and antiproliferative effects of AM 1172 and cannabidiol (two cannabinoid receptor ligands) in vitro, when used alone and in combination with cisplatin (a standard cytotoxic drug), in various human neuroblastoma (CHP-134, KELLY), human glioblastoma (U-87MG and T98G) and rat glioblastoma (C6) cell lines. Our experiments showed that AM 1172 and cannabidiol inhibited cell proliferation with IC50 values in the range of 2.29-17.21 μM (for AM 1172) and 11.61-20.35 μM (for cannabidiol), respectively. The selectivity index for AM 1172 ranged from 0.61 to 4.60 and that for cannabidiol ranged from 1.45 to 2.55 in the studied glioblastoma and neuroblastoma cell lines. With isobolographic analysis, it was found that AM 1172 combined with cisplatin exerted a synergistic interaction in the CHP-134 cell line (p < 0.01). In contrast, AM 1172 when combined with cisplatin produced an antagonistic interaction in the C6 cell line (p < 0.01). The remaining combinations of AM 1172 with cisplatin in the U-87MG, KELLY and T98G cell lines were additive. In case of cannabidiol, its combination with cisplatin produced an antagonistic interaction in the T98G cell line (p < 0.0001), whereas the combinations of cannabidiol with cisplatin in the CHP-134, U-87MG, KELLY, and C6 cell lines were additive in nature. The synergistic and additive interactions for the combination of AM 1172 and cannabidiol with cisplatin seem to be a promising direction in glioblastoma therapy. Unfortunately, the combinations producing antagonistic interactions (AM 1172+cisplatin in C6, and cannabidiol + cisplatin in T98G cell lines) should be avoided due to the antagonistic antiproliferative effect of two-drug mixtures.
Collapse
Affiliation(s)
- Katarzyna Załuska-Ogryzek
- Department of Pathophysiology, Medical University of Lublin, Ul. Jaczewskiego 8b, 20-090, Lublin, Poland
| | - Paula Wróblewska-Łuczka
- Department of Occupational Medicine, Medical University of Lublin, Ul. Jaczewskiego 8b, 20-090, Lublin, Poland
| | - Agnieszka Góralczyk
- Department of Occupational Medicine, Medical University of Lublin, Ul. Jaczewskiego 8b, 20-090, Lublin, Poland
| | - Jarogniew J Łuszczki
- Department of Occupational Medicine, Medical University of Lublin, Ul. Jaczewskiego 8b, 20-090, Lublin, Poland.
| |
Collapse
|
17
|
Ismail J, Shebaby W, Azar Atallah S, Taleb RI, Kawrani S, Faour W, Mroueh M. Combination of Cannabidiol with Cisplatin or Paclitaxel Analysis Using the Chou-Talalay Method and Chemo-Sensitization Evaluation in Platinum-Resistant Ovarian Cancer Cells. Biomedicines 2025; 13:520. [PMID: 40002932 PMCID: PMC11852490 DOI: 10.3390/biomedicines13020520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/20/2025] [Accepted: 01/23/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Cannabidiol (CBD) is known for its anti-cancer properties in preclinical models and is increasingly used alongside conventional chemotherapy in cancer treatment. This study aims to evaluate the anti-cancer activity of CBD from Lebanese Cannabis sativa as a monotherapy and in combination with cisplatin or paclitaxel on human ovarian adenocarcinoma cells. Methods: Cytotoxicity of CBD was tested on OVCAR-3 and SK-OV-3 cell lines using the MTS assay. The Chou-Talalay method and CompuSyn software were used to determine the combination indices (CIs) for predicting interactions between CBD and chemotherapeutic agents. CBD showed dose-dependent tumor growth inhibition at 72 h with comparable IC50 values for both cell lines. Results: The combination of CBD with cisplatin or paclitaxel showed significant antagonistic interaction in SK-OV-3 cells (CI > 1), but mild synergism (CI < 1) at high growth inhibition rates (95% and 97%) was observed in SK-OV-3 cells with CBD/cisplatin. Pure antagonism was found in OVCAR-3 cells with CBD/cisplatin. Priming SK-OV-3 cells with CBD reduced the IC50 values of both drugs significantly, with a similar effect seen when cells were primed with cisplatin or paclitaxel before CBD treatment. Conclusions: Integrating CBD with chemotherapy could improve cancer therapy and address drug resistance. Sequential administration of CBD and chemotherapeutic agents is more beneficial than simultaneous administration. Further in vivo studies are necessary to validate these findings and understand CBD's interactions with other drugs fully.
Collapse
Affiliation(s)
- Jana Ismail
- Pharmaceutical Sciences Department, School of Pharmacy, Lebanese American University, Byblos 1102 2801, Lebanon; (J.I.); (W.S.)
| | - Wassim Shebaby
- Pharmaceutical Sciences Department, School of Pharmacy, Lebanese American University, Byblos 1102 2801, Lebanon; (J.I.); (W.S.)
| | - Shirine Azar Atallah
- Pharmaceutical Sciences Department, School of Pharmacy, Lebanese American University, Byblos 1102 2801, Lebanon; (J.I.); (W.S.)
| | - Robin I. Taleb
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Byblos 1102 2801, Lebanon; (R.I.T.)
| | - Sara Kawrani
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Byblos 1102 2801, Lebanon; (R.I.T.)
| | - Wissam Faour
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos 1102 2801, Lebanon;
| | - Mohamad Mroueh
- Pharmaceutical Sciences Department, School of Pharmacy, Lebanese American University, Byblos 1102 2801, Lebanon; (J.I.); (W.S.)
| |
Collapse
|
18
|
Lamtha T, Jongkon N, Lertvanithphol T, Horprathum M, Seetaha S, Choowongkomon K. Cannabinoids as Promising Inhibitors of HER2-Tyrosine Kinase: A Novel Strategy for Targeting HER2-Positive Ovarian Cancer. ACS OMEGA 2025; 10:6191-6200. [PMID: 39989803 PMCID: PMC11840771 DOI: 10.1021/acsomega.4c11108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/17/2025] [Accepted: 01/30/2025] [Indexed: 02/25/2025]
Abstract
Human epidermal growth factor receptor 2 (HER2) is a transmembrane receptor within the ErbB family that plays a pivotal role in the progression of various aggressive cancers. HER2-positive tumors often develop resistance to standard therapies, necessitating the exploration of innovative treatment options. Cannabinoids, bioactive compounds from Cannabis sativa such as cannabidiol (CBD), cannabigerol (CBG), and cannabinol (CBN), have gained attention for their potential anticancer properties. This study evaluates the efficacy of CBD, CBG, and CBN in targeting HER2-positive ovarian cancer through kinase inhibition assays, surface plasmon resonance (SPR), molecular docking, and cell viability assessments. SPR analysis revealed that cannabinoids bind strongly to HER2-tyrosine kinase (HER2-TK), with CBD showing the highest affinity (K D = 6.16 μM), significantly better than afatinib (K D = 26.30 μM), and CBG demonstrating moderate affinity (K D = 17.07 μM). In kinase inhibition assays, CBG was the most potent inhibitor (IC50 = 24.7 nM), followed by CBD (IC50 = 38 nM), suggesting their ability to disrupt HER2-mediated signaling pathways. Molecular docking studies highlighted critical interactions between cannabinoids and essential HER2 residues (Leu796, Thr862, Asp863). In cell viability assays, CBD and CBG effectively inhibited the growth of HER2-positive SKOV3 cells (IC50 = 13.8 μM and 16.6 μM, respectively), comparable to traditional tyrosine kinase inhibitors. These findings underscore the therapeutic potential of cannabinoids, particularly CBD and CBG, as alternative or adjunct therapies for HER2-positive cancers, with the promise of mitigating resistance and adverse effects associated with existing treatments.
Collapse
Affiliation(s)
- Thomanai Lamtha
- Laboratory
of Protein Engineering and Bioinformatics (PROTEP), Department of
Biochemistry, Faculty of Science, Kasetsart
University, Bangkok 10900, Thailand
- Department
of Clinical Chemistry, Faculty of Medical Technology, Nation University, Lampang 52000, Thailand
| | - Nathjanan Jongkon
- Department
of Social and Applied Science, College of Industrial Technology, King Mongkut’s University of Technology North
Bangkok, Bangkok 10800, Thailand
| | - Tossaporn Lertvanithphol
- The
National Electronics and Computer Technology Center (NECTEC), National
Science and Technology Development Agency (NSTDA), Thailand Science Park, Pathum
Thani 12120, Thailand
| | - Mati Horprathum
- The
National Electronics and Computer Technology Center (NECTEC), National
Science and Technology Development Agency (NSTDA), Thailand Science Park, Pathum
Thani 12120, Thailand
| | - Supaphorn Seetaha
- Laboratory
of Protein Engineering and Bioinformatics (PROTEP), Department of
Biochemistry, Faculty of Science, Kasetsart
University, Bangkok 10900, Thailand
| | - Kiattawee Choowongkomon
- Laboratory
of Protein Engineering and Bioinformatics (PROTEP), Department of
Biochemistry, Faculty of Science, Kasetsart
University, Bangkok 10900, Thailand
| |
Collapse
|
19
|
Melo ESA, Asevedo EA, Duarte-Almeida JM, Nurkolis F, Syahputra RA, Park MN, Kim B, do Couto RO, Ribeiro RIMDA. Mechanisms of Cell Death Induced by Cannabidiol Against Tumor Cells: A Review of Preclinical Studies. PLANTS (BASEL, SWITZERLAND) 2025; 14:585. [PMID: 40006844 PMCID: PMC11859785 DOI: 10.3390/plants14040585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/09/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025]
Abstract
Commonly known as marijuana or hemp, Cannabis sativa L. (Cannabaceae), contains numerous active compounds, particularly cannabinoids, which have been extensively studied for their biological activities. Among these, cannabidiol (CBD) stands out for its therapeutic potential, especially given its non-psychotropic effects. This review evaluates the antitumor properties of CBD, highlighting its various mechanisms of action, including the induction of apoptosis, autophagy, and necrosis. By synthesizing findings from in vitro studies on the cell death mechanisms and signaling pathways activated by CBD in various human tumor cell lines, this literature review emphasizes the therapeutic promise of this natural antineoplastic agent. We conducted a comprehensive search of articles in PubMed, Scopus, Springer, Medline, Lilacs, and Scielo databases from 1984 to February 2022. Of the forty-three articles included, the majority (68.18%) reported that CBD activates apoptosis, while 18.18% observed simultaneous apoptosis and autophagy, 9.09% focused on autophagy alone, and 4.54% indicated necrosis. The antitumor effects of CBD appear to be mediated by transient receptor potential cation channels (TRPVs) in endometrial cancer, glioma, bladder cancer, and myeloma, with TRPV1, TRPV2, and TRPV4 playing key roles in activating apoptosis. This knowledge paves the way for innovative therapeutic strategies that may enhance cancer treatment outcomes while minimizing the toxicity and side effects associated with conventional therapies.
Collapse
Affiliation(s)
- Edilene S. A. Melo
- Experimental Pathology Laboratory, Dona Lindu Central-West Campus (CCO), Federal University of São João del-Rei (UFSJ), Sebastião Gonçalves Coelho 400, Chanadour, Divinopolis 35501-296, MG, Brazil; (E.S.A.M.); (E.A.A.)
| | - Estefani A. Asevedo
- Experimental Pathology Laboratory, Dona Lindu Central-West Campus (CCO), Federal University of São João del-Rei (UFSJ), Sebastião Gonçalves Coelho 400, Chanadour, Divinopolis 35501-296, MG, Brazil; (E.S.A.M.); (E.A.A.)
| | - Joaquim Maurício Duarte-Almeida
- Plant Cell Culture Laboratory, Dona Lindu Central-West Campus (CCO), Federal University of São João del-Rei, Sebastião Gonçalves Coelho 400, Chanadour, Divinopolis 35501-296, MG, Brazil;
| | - Fahrul Nurkolis
- Department of Biological Sciences, Faculty of Sciences and Technology, State Islamic University of Sunan Kalijaga (UIN Sunan Kalijaga), Yogyakarta 55281, Indonesia;
| | - Rony Abdi Syahputra
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan 20155, Indonesia;
| | - Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.N.P.); (B.K.)
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.N.P.); (B.K.)
| | - Renê Oliveira do Couto
- Pharmaceutical Development Laboratory, Dona Lindu Central-West Campus (CCO), Federal University of São João del-Rei, Sebastião Gonçalves Coelho 400, Chanadour, Divinopolis 35501-296, MG, Brazil;
| | - Rosy Iara Maciel de A. Ribeiro
- Experimental Pathology Laboratory, Dona Lindu Central-West Campus (CCO), Federal University of São João del-Rei (UFSJ), Sebastião Gonçalves Coelho 400, Chanadour, Divinopolis 35501-296, MG, Brazil; (E.S.A.M.); (E.A.A.)
| |
Collapse
|
20
|
Li Y, Liu H, Fang R, Jin J, Yang F, Chen J, Zhang J. Designing novel Au(III) complexes based on the structure of diazepam: Achieving a multiaction mechanism against glioma. Eur J Med Chem 2025; 283:117171. [PMID: 39705733 DOI: 10.1016/j.ejmech.2024.117171] [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: 10/09/2024] [Revised: 12/01/2024] [Accepted: 12/12/2024] [Indexed: 12/22/2024]
Abstract
Metal-based drugs have been used in the clinical treatment of tumors for over 30 years. However, no metal-based drugs have been clinically approved to treat glioma. Although metal complexes have excellent cytotoxicity, their most critical problem is crossing the blood-brain barrier. Therefore, to enable metal complexes to cross blood-brain barrier and target glioma therapy, herein, we propose to rationally used the basic structure of diazepam (5-chlorobenzophenone) and thiosemicarbazide to synthesize gold (Au) complexes C1, C2 and C3 with antiglioma activity. The C3 complex with two methyl groups attached to the N3 of thiosemicarbazone exhibited excellent cytotoxicity to glioma cells through its multiaction mechanism against glioma, inducing apoptosis, autophagy death, and deoxyribonucleic acid damage. In addition, the synthesized C3 complex can effectively cross the blood-brain barrier and accumulate in glioma, considerably decreasing the untoward reaction in vivo. Our findings provide a novel strategy for designing metal-based complexes for the treatment of glioma.
Collapse
Affiliation(s)
- Yanping Li
- Mental Health Education Center of College Student, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, 541004, PR China
| | - Haoran Liu
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, 541004, PR China
| | - Ronghao Fang
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, 541004, PR China
| | - Jiamin Jin
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, 541004, PR China
| | - Feng Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi, 541004, PR China
| | - Jian Chen
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, 541004, PR China.
| | - Juzheng Zhang
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, 541004, PR China.
| |
Collapse
|
21
|
Ke J, Fan Y, Zhang S. Effects of PARP1 inhibitor PJ-34 on TGFα, IL-6, and IL-1β levels in diabetic nephropathy. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:304-315. [PMID: 40073245 DOI: 10.1093/jimmun/vkae024] [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: 07/10/2024] [Accepted: 11/13/2024] [Indexed: 03/14/2025]
Abstract
Diabetic nephropathy is a severe chronic complication characterized by cytotoxicity, inflammation, and fibrosis, ultimately leading to renal failure. This study systematically investigated the effects of the PARP1 inhibitor PJ-34 on high glucose-induced cytotoxicity, inflammation, and fibrosis in HK-2 cells, as well as its improvement on neuropathic pain response and transforming growth factor β (TGFβ) expression in a type 1 diabetes mellitus diabetic nephropathy mouse model. Through cellular and animal experiments, we observed that PJ-34 significantly enhanced the proliferative capacity of cells damaged by high glucose, reduced apoptosis, and decreased the release of proinflammatory factors TGFα, interleukin-6, and interleukin-1β. In the type 1 diabetes mellitus nephropathy mouse model, the administration of PJ-34 substantially improved parameters of neuropathic pain, alleviated renal tissue damage, reduced indicators of renal functional impairment-inhibited renal fibrosis, and reduced the key protein expression in the epithelial-mesenchymal transition process, acting through the regulation of the TGFβ/Smads signaling pathway. This study elucidated the mechanism of action of the PARP1 inhibitor PJ-34 as a potential therapeutic agent for diabetic nephropathy, offering a novel strategy for its treatment.
Collapse
Affiliation(s)
- Jing Ke
- Department of Endocrinology, Central Hospital of Ezhou, Ezhou, China
| | - Yanan Fan
- Department of Thyroid and Breast Surgery, Central Hospital of Ezhou, Ezhou, China
| | - Shaochun Zhang
- Orthopedics Department, Central Hospital of Ezhou, Ezhou, China
| |
Collapse
|
22
|
Dong X, Nie J, Huang A, Chen L, Zang E, Xiang Z, Hao X, Yan S, Ding X, Zhao Y. A novel small molecule NJH-13 induces pyroptosis via the Ca 2+ driven AKT-FOXO1-GSDME signaling pathway in NSCLC by targeting TRPV5. J Adv Res 2025:S2090-1232(25)00047-5. [PMID: 39832720 DOI: 10.1016/j.jare.2025.01.028] [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: 11/09/2024] [Revised: 01/05/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025] Open
Abstract
INTRODUCTION Pyroptosis represents a mode of programmed necrotic cell death (PCD), mediated by members of gasdermin family (GSDMs), such as GSDME. It is emerging as a promising approach for combating cancer. Notably, GSDME is the key modulator for the switch between apoptosis and pyroptosis in cells. However, GSDME is often downregulated in many malignancies, including lung adenocarcinoma. OBJECTIVE To identify novel pyroptosis inducers in non-small cell lung cancer (NSCLC) and dissect the underlying mechanism. METHODS Pyroptosis was examined by live cell imaging, PI/Hoechst/Annexin V staining, LDH release assay, ELISA, and western blot assays. DARTS, CETSA, molecular docking was used to identify the target of NJH-13. RNA-seq, qPCR, chromatin immunoprecipitation (ChIP), dual luciferase assays were used elucidate the mechanism. RESULTS In this study, NJH-13, an N-containing heterocycle, was screened out and identified to possess the ability to activate GSDME, consequently triggering pyroptosis in NSCLC cells. By using the DARTS strategy, transient receptor potential cation channel subfamily V member 5 (TRPV5) was identified as a potential target of NJH-13. NJH-13 increased intracellular calcium level and triggered oxidative stress, both of which are critical events leading to pyroptosis mediated by GSDME. Mechanistically, NJH-13 enhanced the transcription of GSDME via the protein kinase B (AKT)/forkhead box transcription factor O1 (FOXO1) signaling pathway. ChIP revealed that FOXO1 bound directly to the promoter region of GSDME, thus triggering the GSDME-mediated pyroptosis. Pharmacological and genetic activation of AKT or inhibition of FOXO1 partially rescued NJH-13-induced pyroptotic cell death. Moreover, NJH-13 treatment suppressed tumor growth in vivo. CONCLUSION Taken together, our results revealed that TRPV5 is a distinctive target for manipulating pyroptosis and provided evidence that NJH-13 functions as a potential anti-cancer agent capable of triggering pyroptosis in NSCLC cells.
Collapse
Affiliation(s)
- Xianxiang Dong
- State Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Jiahui Nie
- School of Chemical Science and Technology, Yunnan University, Kunming 650091, PR China
| | - Aiying Huang
- State Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan, PR China; School of Chemical Science and Technology, Yunnan University, Kunming 650091, PR China
| | - Li Chen
- School of Chemical Science and Technology, Yunnan University, Kunming 650091, PR China
| | - Erkang Zang
- State Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan, PR China; School of Life Sciences, Yunnan University, Kunming 650091, PR China
| | - Zhengrui Xiang
- State Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Xiaojiang Hao
- State Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan, PR China; Research Unit of Chemical Biology of Natural Anti-Virus Products, Chinese Academy of Medical Sciences, Beijing 100730, PR China
| | - Shengjiao Yan
- School of Chemical Science and Technology, Yunnan University, Kunming 650091, PR China.
| | - Xiao Ding
- State Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan, PR China; Research Unit of Chemical Biology of Natural Anti-Virus Products, Chinese Academy of Medical Sciences, Beijing 100730, PR China.
| | - Yuhan Zhao
- State Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan, PR China.
| |
Collapse
|
23
|
Schmitt-Ulms G, Wang X, Watts J, Booth S, Wille H, Zhao W. A unified model for the origins of spongiform degeneration and other neuropathological features in prion diseases. ARXIV 2025:arXiv:2412.16678v2. [PMID: 39876936 PMCID: PMC11774453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Decades after their initial observation in prion-infected brain tissues, the identities of virus-like dense particles, varicose tubules, and oval bodies containing parallel bands and fibrils have remained elusive. Our recent work revealed that a phenotype of dilation of the endoplasmic reticulum (ER), most notable for the perinuclear space (PNS), contributes to spongiform degeneration. To assess the significance of this phenotype for the etiology of prion diseases, we explored whether it can be functionally linked to other neuropathological hallmarks observed in these diseases, as this would indicate it to be a central event. Having surveyed the neuropathological record and other distant literature niches, we propose a model in which pathogenic forms of the prion protein poison raft domains, including essential Na+, K+-ATPases (NKAs) embedded within them, thereby triggering an ER-centered cellular rescue program coordinated by the unfolded protein response (UPR). The execution of this program stalls general protein synthesis, causing the deterioration of synaptic spines. As the disease progresses, cells selectively increase sterol biosynthesis, along with ribosome and ER biogenesis. These adaptive rescue attempts cause morphological changes to the ER which manifest as ER dilation or ER hypertrophy in a manner that is influenced by Ca2+ influx into the cell. The nuclear-to-cytoplasmic transport of mRNAs and tRNAs interrupts in late stage disease, thereby depriving ribosomes of supplies and inducing them to aggregate into a paracrystalline form. In support of this model, we share previously reported data, whose features are consistent with the interpretation that 1) the phenotype of ER dilation is observed in major prion diseases, 2) varicose tubules and oval bodies represent ER hypertrophy, and 3) virus-like dense particles are paracrystalline aggregates of inactive ribosomes.
Collapse
Affiliation(s)
- Gerold Schmitt-Ulms
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Canada
| | - Xinzhu Wang
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Canada
| | - Joel Watts
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Stephanie Booth
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Canada
| | - Holger Wille
- Department of Biochemistry, University of Alberta, Edmonton, Canada
- Centre for Prions and Protein Folding Diseases, University of Edmonton, Edmonton, Canada
| | - Wenda Zhao
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Canada
| |
Collapse
|
24
|
Chen Y, Gao Q, Wang D, Zou X, Li X, Ji J, Liu B. An Overview of Research Advances in Oncology Regarding the Transcription Factor ATF4. Curr Drug Targets 2025; 26:59-72. [PMID: 39350552 DOI: 10.2174/0113894501328461240921062056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/09/2024] [Accepted: 09/10/2024] [Indexed: 02/19/2025]
Abstract
This review provides a comprehensive overview of the recent advancements in research on ATF4 (Activating Transcription Factor 4) within the field of oncology. As a crucial transcription factor, ATF4 has garnered increasing attention for its role in cancer research. The review begins with an exploration of the regulatory mechanisms of ATF4, including its transcriptional control, post-translational modifications, and interactions with other transcription factors. It then highlights key research findings on ATF4's involvement in various aspects of tumor biology, such as cell proliferation, differentiation, apoptosis and survival, invasion and metastasis, and the tumor microenvironment. Furthermore, the review discusses the potential of targeting ATF4 as a novel therapeutic strategy for cancer treatment. It also explores how ATF4's interactions with existing anticancer drugs could inform the development of more effective therapeutic agents. By elucidating the role of ATF4 in tumor biology and its potential clinical applications, this review aims to provide new insights and strategies for cancer treatment.
Collapse
Affiliation(s)
- Yulu Chen
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Qi Gao
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Dan Wang
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Xun Zou
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Xiuming Li
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Jing Ji
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Bin Liu
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| |
Collapse
|
25
|
Giannotti L, Di Chiara Stanca B, Spedicato F, Vergara D, Stanca E, Damiano F, Siculella L. Exploring the Therapeutic Potential of Cannabidiol in U87MG Cells: Effects on Autophagy and NRF2 Pathway. Antioxidants (Basel) 2024; 14:18. [PMID: 39857352 PMCID: PMC11761945 DOI: 10.3390/antiox14010018] [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: 11/23/2024] [Revised: 12/14/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025] Open
Abstract
Cannabinoids include both endogenous endocannabinoids and exogenous phytocannabinoids, such as cannabidiol (CBD), and have potential as therapeutic agents in cancer treatment due to their selective anticancer activities. CBD exhibits both antioxidant and pro-oxidant effects depending on its concentration and cell types. These properties allow CBD to influence oxidative stress responses and potentially enhance the efficacy of antitumor therapies. In this study, we treated U87MG glioma cells with low dose (1 μM) CBD and evaluated its molecular effects. Our findings indicate that CBD reduced cell viability by 20% (p < 0.05) through the alteration of mitochondrial membrane potential. The alteration of redox status by CBD caused an attempt to rescue mitochondrial functionality through nuclear localization of the GABP transcription factor involved in mitochondria biogenesis. Moreover, CBD treatment caused an increase in autophagic flux, as supported by the increase in Beclin-1 and the ratio of LC3-II/LC3-I. Due to mitochondria functionality alteration, pro-apoptotic proteins were induced without activating apoptotic effectors Caspase-3 or Caspase-7. The study of the transcription factor NRF2 and the ubiquitin-binding protein p62 expression revealed an increase in their levels in CBD-treated cells. In conclusion, low-dose CBD makes U87MG cells more vulnerable to cytotoxic effects, reducing cell viability and mitochondrial dynamics while increasing autophagic flux and redox systems. This explains the mechanisms by which glioma cells respond to CBD treatment. These findings highlight the therapeutic potential of CBD, suggesting that modulating NRF2 and autophagy pathways could represent a promising strategy for glioblastoma treatment.
Collapse
Affiliation(s)
- Laura Giannotti
- Department of Experimental Medicine, University of Salento, 73100 Lecce, Italy; (B.D.C.S.); (E.S.); (L.S.)
| | - Benedetta Di Chiara Stanca
- Department of Experimental Medicine, University of Salento, 73100 Lecce, Italy; (B.D.C.S.); (E.S.); (L.S.)
- Institute of Polymers, Composites and Biomaterials, National Research Council (IPCB-CNR), 80125 Naples, Italy
| | - Francesco Spedicato
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (F.S.); (D.V.)
| | - Daniele Vergara
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (F.S.); (D.V.)
| | - Eleonora Stanca
- Department of Experimental Medicine, University of Salento, 73100 Lecce, Italy; (B.D.C.S.); (E.S.); (L.S.)
| | - Fabrizio Damiano
- Department of Experimental Medicine, University of Salento, 73100 Lecce, Italy; (B.D.C.S.); (E.S.); (L.S.)
| | - Luisa Siculella
- Department of Experimental Medicine, University of Salento, 73100 Lecce, Italy; (B.D.C.S.); (E.S.); (L.S.)
| |
Collapse
|
26
|
Moya-Utrera F, Fuentes-Ríos D, Romero-Carrasco A, Doña-Flores M, Cheng-Sánchez I, Díaz-Morilla A, Soledad Pino-González M, Martínez-Ferez A, Moreno J, Mesas C, Melguizo C, Prados J, Sarabia F, López-Romero JM. Synthesis of (-)-Cannabidiol (CBD), (-)-Δ 9- and (-)-Δ 8-Tetrahydrocannabinols, Encapsulation of CBD with Nanoparticles for Controlled Delivery and Biological Evaluation. Chemistry 2024; 30:e202402496. [PMID: 39307687 DOI: 10.1002/chem.202402496] [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: 07/01/2024] [Indexed: 11/01/2024]
Abstract
Cannabidiol (CBD) is garnering increasing interest due to its significant biological activity. This natural compound is one of the major cannabinoids in Cannabis sativa L. In this work, we describe the encapsulation of CBD in solid and hollow pH-sensitive poly(4-vinylpyridine) (solid@p4VP and hollow@p4VP) nanoparticles, and temperature-sensitive poly(N-isopropylacrylamide) (solid@pNIPAM and hollow@pNIPAM) nanoparticles for transport and release CBD in a controlled manner. The CBD loading into these smart polymeric systems was effective and their release profiles, solubility and resistance to stomach and intestinal conditions were evaluated, showing satisfactory properties and improved bioavailability with respect to free CBD. Finally, the A549 human lung cancer cell line was used as lung adenocarcinoma epithelial cellular model to carry out preliminary assays of the in vitro activity of the vehiculized CBD. For all these studies, synthetic CBD was employed, for which a new efficient and scalable synthesis of cannabinoids has been developed.
Collapse
Affiliation(s)
- Federico Moya-Utrera
- Department of Organic Chemistry, Faculty of Sciences, University of Málaga, Campus de Teatinos, s/n, 29071, Málaga, Spain
| | - David Fuentes-Ríos
- Department of Organic Chemistry, Faculty of Sciences, University of Málaga, Campus de Teatinos, s/n, 29071, Málaga, Spain
| | - Antonio Romero-Carrasco
- Department of Organic Chemistry, Faculty of Sciences, University of Málaga, Campus de Teatinos, s/n, 29071, Málaga, Spain
| | - Manuel Doña-Flores
- Department of Organic Chemistry, Faculty of Sciences, University of Málaga, Campus de Teatinos, s/n, 29071, Málaga, Spain
| | - Iván Cheng-Sánchez
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Amelia Díaz-Morilla
- Department of Organic Chemistry, Faculty of Sciences, University of Málaga, Campus de Teatinos, s/n, 29071, Málaga, Spain
| | - María Soledad Pino-González
- Department of Organic Chemistry, Faculty of Sciences, University of Málaga, Campus de Teatinos, s/n, 29071, Málaga, Spain
| | | | - Javier Moreno
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100, Granada, Spain
| | - Cristina Mesas
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100, Granada, Spain
- Instituto de Investigación Biosanitaria ibs. Granada, 18012, Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071, Granada, Spain
| | - Consolación Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100, Granada, Spain
- Instituto de Investigación Biosanitaria ibs. Granada, 18012, Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071, Granada, Spain
| | - José Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100, Granada, Spain
- Instituto de Investigación Biosanitaria ibs. Granada, 18012, Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071, Granada, Spain
| | - Francisco Sarabia
- Department of Organic Chemistry, Faculty of Sciences, University of Málaga, Campus de Teatinos, s/n, 29071, Málaga, Spain
| | - J Manuel López-Romero
- Department of Organic Chemistry, Faculty of Sciences, University of Málaga, Campus de Teatinos, s/n, 29071, Málaga, Spain
| |
Collapse
|
27
|
Chen L, Zhang H, Shang C, Hong Y. The Role and Applied Value of Mitochondria in Glioma-Related Research. CNS Neurosci Ther 2024; 30:e70121. [PMID: 39639571 PMCID: PMC11621238 DOI: 10.1111/cns.70121] [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: 07/18/2024] [Revised: 10/06/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024] Open
Abstract
Mitochondria, known as the "energy factory" of cells, are essential organelles with a double membrane structure and genetic material found in most eukaryotic cells. They play a crucial role in tumorigenesis and development, with alterations in mitochondrial structure and function in tumor cells leading to characteristics such as rapid proliferation, invasion, and drug resistance. Glioma, the most common brain tumor with a high recurrence rate and limited treatment options, has been linked to changes in mitochondrial structure and function. This review focuses on the bioenergetics, dynamics, metastasis, and autophagy of mitochondria in relation to glioma proliferation, as well as the potential use of mitochondria-targeting drugs in glioma treatment.
Collapse
Affiliation(s)
- Liwen Chen
- Department of Neurobiology, School of Life SciencesChina Medical UniversityShenyangLiaoningChina
- Department of Neurosurgery, Shengjing HospitalChina Medical UniversityShenyangLiaoningChina
| | - Hui Zhang
- Department of Urology, Shengjing HospitalChina Medical UniversityShenyangLiaoningChina
| | - Chao Shang
- Department of Neurobiology, School of Life SciencesChina Medical UniversityShenyangLiaoningChina
| | - Yang Hong
- Department of Neurosurgery, Shengjing HospitalChina Medical UniversityShenyangLiaoningChina
| |
Collapse
|
28
|
Tang Q, Ren T, Bai P, Wang X, Zhao L, Zhong R, Sun G. Novel strategies to overcome chemoresistance in human glioblastoma. Biochem Pharmacol 2024; 230:116588. [PMID: 39461382 DOI: 10.1016/j.bcp.2024.116588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024]
Abstract
Temozolomide (TMZ) is currently the first-line chemotherapeutic agent for the treatment of glioblastoma multiforme (GBM). However, the inherent heterogeneity of GBM often results in suboptimal outcomes, particularly due to varying degrees of resistance to TMZ. Over the past several decades, O6-methylguanine-DNA methyltransferase (MGMT)-mediated DNA repair pathway has been extensively investigated as a target to overcome TMZ resistance. Nonetheless, the combination of small molecule covalent MGMT inhibitors with TMZ and other chemotherapeutic agents has frequently led to adverse clinical effects. Recently, additional mechanisms contributing to TMZ resistance have been identified, including epidermal growth factor receptor (EGFR) mutations, overactivation of intracellular signalling pathways, energy metabolism reprogramming or survival autophagy, and changes in tumor microenvironment (TME). These findings suggest that novel therapeutic strategies targeting these mechanisms hold promise for overcoming TMZ resistance in GBM patients. In this review, we summarize the latest advancements in understanding the mechanisms underlying intrinsic and acquired TMZ resistance. Additionally, we compile various small-molecule compounds with potential to mitigate chemoresistance in GBM. These mechanism-based compounds may enhance the sensitivity of GBM to TMZ and related chemotherapeutic agents, thereby improving overall survival rates in clinical practice.
Collapse
Affiliation(s)
- Qing Tang
- Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Ting Ren
- Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Peiying Bai
- Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Xin Wang
- Department of Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100029, China
| | - Lijiao Zhao
- Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Rugang Zhong
- Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Guohui Sun
- Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China.
| |
Collapse
|
29
|
Cham PS, Singh A, Jamwal A, Singh R, Anand R, Manhas D, Sharma S, Singh VP, Nandi U, Singh SK, Singh PP. Discovery of Ring-Annulated Analogues of Cannabidiol as Potential Anticancer Agents: Synthesis and Biological Evaluation. ACS Med Chem Lett 2024; 15:1832-1842. [PMID: 39563806 PMCID: PMC11571011 DOI: 10.1021/acsmedchemlett.4c00233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 11/21/2024] Open
Abstract
Cannabidiol (CBD) is a nonpsychoactive cannabinoid derived from Cannabis sativa and its potential therapeutic effects extend beyond its well-known antiepileptic properties. Exploring CBD and its analogues as anticancer agents has gained significant attention in recent years. In this study, a series of novel ring-annulated analogues of CBD with oxazinyl moiety were synthesized and evaluated for their antiproliferative effect. The analogues 4d and 4h demonstrate promising activity against breast and colorectal cancer. Furthermore, mechanistic insights revealed that the identified candidates arrest the G1 phase of the cell cycle and induce apoptosis via the mitochondrial pathway in breast cancer cell lines. Notably, CBD ring-annulated analogues 4d or 4h exhibit enhanced solubility, better metabolic stability, and lowered cytochrome P450 (CYP) inhibition liability compared to CBD. These multifaceted attributes highlight the potential of cannabinoid-based candidates for further preclinical development.
Collapse
Affiliation(s)
- Pankaj Singh Cham
- Natural Product & Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine (CSIR-IIIM), Canal Road, Jammu 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Ajeet Singh
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine (CSIR-IIIM), Canal Road, Jammu 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Ashiya Jamwal
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine (CSIR-IIIM), Canal Road, Jammu 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Rattandeep Singh
- Natural Product & Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine (CSIR-IIIM), Canal Road, Jammu 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Radhika Anand
- Natural Product & Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine (CSIR-IIIM), Canal Road, Jammu 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Diksha Manhas
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine (CSIR-IIIM), Canal Road, Jammu 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sucheta Sharma
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine (CSIR-IIIM), Canal Road, Jammu 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Varun Pratap Singh
- Natural Product & Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine (CSIR-IIIM), Canal Road, Jammu 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Utpal Nandi
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine (CSIR-IIIM), Canal Road, Jammu 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Shashank K Singh
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine (CSIR-IIIM), Canal Road, Jammu 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Parvinder Pal Singh
- Natural Product & Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine (CSIR-IIIM), Canal Road, Jammu 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
30
|
Ma W, Lu Y, Jin X, Lin N, Zhang L, Song Y. Targeting selective autophagy and beyond: From underlying mechanisms to potential therapies. J Adv Res 2024; 65:297-327. [PMID: 38750694 PMCID: PMC11518956 DOI: 10.1016/j.jare.2024.05.009] [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: 03/07/2024] [Revised: 04/26/2024] [Accepted: 05/08/2024] [Indexed: 05/21/2024] Open
Abstract
BACKGROUND Autophagy is an evolutionarily conserved turnover process for intracellular substances in eukaryotes, relying on lysosomal (in animals) or vacuolar (in yeast and plants) mechanisms. In the past two decades, emerging evidence suggests that, under specific conditions, autophagy can target particular macromolecules or organelles for degradation, a process termed selective autophagy. Recently, accumulating studies have demonstrated that the abnormality of selective autophagy is closely associated with the occurrence and progression of many human diseases, including neurodegenerative diseases, cancers, metabolic diseases, and cardiovascular diseases. AIM OF REVIEW This review aims at systematically and comprehensively introducing selective autophagy and its role in various diseases, while unravelling the molecular mechanisms of selective autophagy. By providing a theoretical basis for the development of related small-molecule drugs as well as treating related human diseases, this review seeks to contribute to the understanding of selective autophagy and its therapeutic potential. KEY SCIENTIFIC CONCEPTS OF REVIEW In this review, we systematically introduce and dissect the major categories of selective autophagy that have been discovered. We also focus on recent advances in understanding the molecular mechanisms underlying both classical and non-classical selective autophagy. Moreover, the current situation of small-molecule drugs targeting different types of selective autophagy is further summarized, providing valuable insights into the discovery of more candidate small-molecule drugs targeting selective autophagy in the future. On the other hand, we also reveal clinically relevant implementations that are potentially related to selective autophagy, such as predictive approaches and treatments tailored to individual patients.
Collapse
Affiliation(s)
- Wei Ma
- Department of Breast Surgery, Department of Ultrasound, Department of Hematology and Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yingying Lu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Xin Jin
- Department of Breast Surgery, Department of Ultrasound, Department of Hematology and Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Na Lin
- Department of Breast Surgery, Department of Ultrasound, Department of Hematology and Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang 110001, China.
| | - Lan Zhang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China.
| | - Yaowen Song
- Department of Breast Surgery, Department of Ultrasound, Department of Hematology and Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang 110001, China.
| |
Collapse
|
31
|
Tabatabaei K, Moazzezi S, Emamgholizadeh M, Vaez H, Baradaran B, Shokouhi B. Improved Therapeutic Efficacy of Doxorubicin Chemotherapy With Cannabidiol in 4T1 Mice Breast Cancer Model. Cancer Med 2024; 13:e70395. [PMID: 39503169 PMCID: PMC11538943 DOI: 10.1002/cam4.70395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 10/02/2024] [Accepted: 10/20/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND High dose chemotherapy is one of the therapeutic strategies for breast cancer and doxorubicin (DOX) as a chemotherapy agent is widely used. DOX indication is limited due to its dose-depended cardiotoxicity. Recently, cannabidiol (CBD) shows antitumoral and cardioprotective effects, so we hypothesized that CBD administration with high-dose DOX chemotherapy can improve anticancer activity and reduce cardiotoxic side effects. METHOD Mice breast cancer model established by injecting 4T1 cell lines. One group was not injected by 4T1 cells as a not cancerous group and received normal saline (NS, 0.1 mL). In cancerous groups, first group was considered as cancerous control and received NS (0.1 mL); the second group received CBD (5 mg/kg, IP) on Days 1,7, and 14; in the third group DOX (5 mg/kg, IV) as CBD schedule was administrated; the fourth group treated with CBD 1 day before DOX injection as pretreatment, and the last group was treated with CBD and DOX at same time with previous doses and schedules. On Day 21, all mice were sacrificed, heart and lungs tissues were obtained and histological sections were isolated. SOD2, iNOS, MMP2, MMP9 were evaluated through western blot and TUNEL test preformed for breast tumor. RESULTS Tumor size and weight significantly decreased in DOX, pretreatment CBD + DOX and CBD + DOX groups. Administration of CBD with DOX could not prevent weight loss. TUNEL test demonstrated the highest tumor cell apoptosis in pretreatment CBD + DOX and CBD + DOX. In lungs belonged to CBD + DOX, there was not any sign of metastasis. Cardiac histopathological examination of pretreatment CBD + DOX and CBD + DOX did not show any sign of congestion or inflammation. In CBD + DOX SOD2 increased, also iNOS, MMP2, and MMP9 decreased compared to DOX. CONCLUSIONS This study demonstrated that simultaneous administration of CBD and DOX can increase antitumoral effect and reduce DOX cardiotoxicity. Nevertheless, CBD can induce cardiotoxicity as administrated alone.
Collapse
Affiliation(s)
- Koorosh Tabatabaei
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
- Faculty of Veterinary MedicineTabriz Islamic Azad UniversityTabrizIran
| | - Sara Moazzezi
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
- Faculty of Veterinary MedicineTabriz Islamic Azad UniversityTabrizIran
| | | | - Haleh Vaez
- Department of Pharmacology and Toxicology, Faculty of PharmacyTabriz University of Medical SciencesTabrizIran
| | - Behzad Baradaran
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
| | - Behrooz Shokouhi
- Department of PathologyTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
32
|
Kong L, Kong L, Li P, Gao L, Ma H, Shi B. Tribbles pseudokinase 3 promoted renal fibrosis by regulating the expression of DNA damage-inducible transcript 3 in diabetic nephropathy. BIOMOLECULES & BIOMEDICINE 2024; 24:1559-1570. [PMID: 38733632 PMCID: PMC11496876 DOI: 10.17305/bb.2024.10419] [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: 02/27/2024] [Revised: 05/03/2024] [Accepted: 05/03/2024] [Indexed: 05/13/2024]
Abstract
Diabetic nephropathy (DN) is a severe complication of prolonged diabetes, impacting millions worldwide with an increasing incidence. This study investigates the role of tribbles pseudokinase 3 (TRIB3), a protein implicated in the progression of DN, focusing on its mechanisms underlying glomerular damage. Through analysis of the Gene Expression Omnibus (GEO) database, we identified TRIB, among differentially expressed genes (DEGs) in streptozotocin (STZ)-treated C57BL/6J mice. Both in vitro and in vivo experiments were conducted to examine the effects of TRIB3 inhibition on high glucose (HG)-induced damage in podocytes and DN mouse models. The results demonstrated that TRIB3 inhibition reduced inflammatory responses and extracellular matrix (ECM) production inMPC5 cells, mediated by the downregulation of DNA damage-inducible transcript 3 (DDIT3) - a critical regulator of proinflammatory cytokine secretion and ECM synthesis. Inhibiting TRIB3 decreased inflammatory factors and ECM deposition in diabetic mice in vivo, confirming its pivotal role in DN pathogenesis. These findings indicate that TRIB3 and its interaction with DDIT3 contribute significantly to DN by promoting inflammatory cascades and ECM accumulation, presenting potential therapeutic targets for managing the disease.
Collapse
Affiliation(s)
- Lulu Kong
- Department of Endocrinology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Endocrinology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Liusha Kong
- Department of Nephrology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Peipei Li
- Department of Endocrinology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Li Gao
- Department of Endocrinology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Hongqin Ma
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Bimin Shi
- Department of Endocrinology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
33
|
Calheiros LGRDM, Pedro G, Oliveira da Silva T, Amorim RM, Alves CEF, Laufer-Amorim R. In Vitro Antitumor Effect of Oils Rich in CBD and THC Cannabis Extract in Canine Prostate Carcinoma Cell Lines. Vet Sci 2024; 11:501. [PMID: 39453093 PMCID: PMC11512242 DOI: 10.3390/vetsci11100501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024] Open
Abstract
Prostate cancer is one of the leading causes of cancer-related deaths worldwide, even when diagnosed at an early stage in humans and dogs. Dogs have a significant incidence of spontaneous prostate cancer, which is highly similar to human androgen-independent prostate cancer and represents a valuable model for comparative studies. Cannabidiol (CBD) and Δ9-tetrahydrocannabinol (THC) are the two main cannabinoids extracted from Cannabis sativa and have demonstrated antiproliferative and anti-invasive properties in different tumor types. In this study, CBD or THC-rich extracts inhibited the proliferation of two canine prostatic carcinoma cell lines, PC1 and PC2, showing an IC50 of 3.43 and 3.57 μM for CBD rich extracts, and 4.90 and 4.48 μM THC rich extracts, respectively. Cell death was also observed with both Annexin V and Propidium iodide staining for the canine cell lines. These results provide new information concerning the use of rich oil in canine PC and open a promising opportunity for further in vitro and in vivo studies to establish the mechanisms of action of these compounds using dogs as a natural model for prostatic carcinoma.
Collapse
Affiliation(s)
| | | | | | | | | | - Renée Laufer-Amorim
- School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu 18618-681, Brazil; (L.G.R.d.M.C.); (G.P.); (T.O.d.S.); (R.M.A.); (C.E.F.A.)
| |
Collapse
|
34
|
Wang Q, Zhang C, Pang Y, Cheng M, Wang R, Chen X, Ji T, Yang Y, Zhang J, Zhong C. Comprehensive analysis of bulk, single-cell RNA sequencing, and spatial transcriptomics revealed IER3 for predicting malignant progression and immunotherapy efficacy in glioma. Cancer Cell Int 2024; 24:332. [PMID: 39354533 PMCID: PMC11443732 DOI: 10.1186/s12935-024-03511-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/18/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND As part of stress-triggered molecules, immediate early response 3 (IER3) dysregulation has been reported to sustain pro-oncogenic pathways and precede malignant transformation. However, the role of IER3 in glioma pathology is ill-defined. METHODS Immunohistochemistry (IHC) assay and publicly available glioma datasets were used to calculate the IER3 expression level in glioma. Wound healing, invasion and cell counting kit-8 (CCK8) assays were applied to measure the cell viability and capacities of migration and invasion of glioma cells in vitro. The immunofluorescence (IF) assay was used to assess the expression associations of IER3 with CCL2 and TGFBI. Cox regression analysis and Kaplan-Meier (K-M) curve were introduced to compute the prognosis-predicting value of IER3. Variations in copy number (CNVs), single nucleotide (SNVs), and methylation profiles were analyzed to illustrate the epigenetic modifications of IER3. Gliomas were divided into two subgroups using the restricted cubic spline (RCS) method. RESULTS IER3: was overexpressed and hypomethylated in gliomas and significantly associated with the dismal prognosis of glioma samples. Samples in the high IER3 subgroup were characterized by increased infiltration of tumor-associated monocytes/macrophages (TAMMs), as well as the elevated sensitivity to Dabrafenib, an inhibitor of BRAF. In addition, this subgroup demonstrated a low mutation rate of IDH, high gain rates of BRAF, ELTD1, and PDGFA. Gliomas with relatively low IER3 expression demonstrated a less invasive subtype and were featured by favorable prognosis, increased response to immunotherapy, and adjuvant chemotherapy plus radiotherapy. The IF assay revealed that IER3 was co-localized and co-expressed with TGFBI. The glioma cells with small interfering RNA (siRNA)-silenced IER3 displayed lower migration, invasion, proliferation, and cell viability than the control group. CONCLUSIONS In this study, we identified IER3 upregulation as an essential biomarker that could assist in adjuvant therapy and prognosis prediction for gliomas.
Collapse
Affiliation(s)
- Qi Wang
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chunyu Zhang
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ying Pang
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Meng Cheng
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Rui Wang
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xu Chen
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Tongjie Ji
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuntong Yang
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jing Zhang
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute for Advanced Study, Tongji University, Shanghai, China
| | - Chunlong Zhong
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
35
|
Brogyanyi T, Kejík Z, Veselá K, Dytrych P, Hoskovec D, Masařik M, Babula P, Kaplánek R, Přibyl T, Zelenka J, Ruml T, Vokurka M, Martásek P, Jakubek M. Iron chelators as mitophagy agents: Potential and limitations. Biomed Pharmacother 2024; 179:117407. [PMID: 39265234 DOI: 10.1016/j.biopha.2024.117407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 09/14/2024] Open
Abstract
Mitochondrial autophagy (mitophagy) is very important process for the maintenance of cellular homeostasis, functionality and survival. Its dysregulation is associated with high risk and progression numerous serious diseases (e.g., oncological, neurodegenerative and cardiovascular ones). Therefore, targeting mitophagy mechanisms is very hot topic in the biological and medicinal research. The interrelationships between the regulation of mitophagy and iron homeostasis are now becoming apparent. In short, mitochondria are central point for the regulation of iron homeostasis, but change in intracellular cheatable iron level can induce/repress mitophagy. In this review, relationships between iron homeostasis and mitophagy are thoroughly discussed and described. Also, therapeutic applicability of mitophagy chelators in the context of individual diseases is comprehensively and critically evaluated.
Collapse
Affiliation(s)
- Tereza Brogyanyi
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic; Institute of Pathological Physiology, First Faculty of Medicine, Charles University in Prague, U Nemocnice 5, 1, Prague 28 53, Czech Republic
| | - Zdeněk Kejík
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic
| | - Kateřina Veselá
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic
| | - Petr Dytrych
- 1st Department of Surgery-Department of Abdominal, Thoracic Surgery and Traumatology, First Faculty of Medicine, Charles University and General University Hospital, U Nemocnice 2, Prague 121 08, Czech Republic
| | - David Hoskovec
- 1st Department of Surgery-Department of Abdominal, Thoracic Surgery and Traumatology, First Faculty of Medicine, Charles University and General University Hospital, U Nemocnice 2, Prague 121 08, Czech Republic
| | - Michal Masařik
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic; Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno CZ-625 00, Czech Republic; Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
| | - Petr Babula
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno CZ-625 00, Czech Republic
| | - Robert Kaplánek
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic
| | - Tomáš Přibyl
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, Prague 166 28, Czech Republic
| | - Jaroslav Zelenka
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, Prague 166 28, Czech Republic
| | - Tomáš Ruml
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, Prague 166 28, Czech Republic
| | - Martin Vokurka
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University in Prague, U Nemocnice 5, 1, Prague 28 53, Czech Republic
| | - Pavel Martásek
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic
| | - Milan Jakubek
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic.
| |
Collapse
|
36
|
Li X, Tie J, Sun Y, Gong C, Deng S, Chen X, Li S, Wang Y, Wang Z, Wu F, Liu H, Wu Y, Zhang G, Guo Q, Yang Y, Wang Y. Targeting DNM1L/DRP1-FIS1 axis inhibits high-grade glioma progression by impeding mitochondrial respiratory cristae remodeling. J Exp Clin Cancer Res 2024; 43:273. [PMID: 39350223 PMCID: PMC11440692 DOI: 10.1186/s13046-024-03194-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/17/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND The dynamics of mitochondrial respiratory cristae (MRC) and its impact on oxidative phosphorylation (OXPHOS) play a crucial role in driving the progression of high-grade glioma (HGG). However, the underlying mechanism remains unclear. METHODS In the present study, we employed machine learning-based transmission electron microscopy analysis of 7141 mitochondria from 54 resected glioma patients. Additionally, we conducted bioinformatics analysis and multiplex immunohistochemical (mIHC) staining of clinical glioma microarrays to identify key molecules involved in glioma. Subsequently, we modulated the expression levels of mitochondrial dynamic-1-like protein (DNM1L/DRP1), and its two receptors, mitochondrial fission protein 1 (FIS1) and mitochondrial fission factor (MFF), via lentiviral transfection to further investigate the central role of these molecules in the dynamics of glioblastoma (GBM) cells and glioma stem cells (GSCs). We then evaluated the potential impact of DNM1L/DRP1, FIS1, and MFF on the proliferation and progression of GBM cells and GSCs using a combination of CCK-8 assay, Transwell assay, Wound Healing assay, tumor spheroid formation assay and cell derived xenograft assay employing NOD/ShiLtJGpt-Prkdcem26Cd52Il2rgem26Cd22/Gpt (NCG) mouse model. Subsequently, we validated the ability of the DNM1L/DRP1-FIS1 axis to remodel MRC structure through mitophagy by utilizing Seahorse XF analysis technology, mitochondrial function detection, MRC abundance detection and monitoring dynamic changes in mitophagy. RESULTS Our findings revealed that compared to low-grade glioma (LGG), HGG exhibited more integrated MRC structures. Further research revealed that DNM1L/DRP1, FIS1, and MFF played pivotal roles in governing mitochondrial fission and remodeling MRC in HGG. The subsequent validation demonstrated that DNM1L/DRP1 exerts a positive regulatory effect on FIS1, whereas the interaction between MFF and FIS1 demonstrates a competitive inhibition relationship. The down-regulation of the DNM1L/DRP1-FIS1 axis significantly impaired mitophagy, thereby hindering the remodeling of MRC and inhibiting OXPHOS function in glioma, ultimately leading to the inhibition of its aggressive progression. In contrast, MFF exerts a contrasting effect on MRC integrity, OXPHOS activity, and glioma progression. CONCLUSIONS This study highlights that the DNM1L/DRP1-FIS1 axis stabilizes MRC structures through mitophagy in HGG cells while driving their OXPHOS activity ultimately leading to robust disease progression. The inhibition of the DNM1L/DRP1-FIS1 axis hinders MRC remodeling and suppresses GBM progression. We propose that down-regulation of the DNM1L/DRP1-FIS1 axis could be a potential therapeutic strategy for treating HGG.
Collapse
Affiliation(s)
- Xiaodong Li
- Specific Lab for Mitochondrial Plasticity Underlying Nervous System Diseases, National Demonstration Center for Experimental Preclinical Medicine Education, The Fourth Military Medical University, Xi'an, 710032, China
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Jingjing Tie
- Specific Lab for Mitochondrial Plasticity Underlying Nervous System Diseases, National Demonstration Center for Experimental Preclinical Medicine Education, The Fourth Military Medical University, Xi'an, 710032, China
- Department of Human Anatomy, Histology and Embryology, Medical School of Yan'an University, Yan'an, China
| | - Yuze Sun
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Chengrong Gong
- Department of Computer Fundamentals, The Fourth Military Medical University, Xi'an, 710032, China
| | - Shizhou Deng
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Xiyu Chen
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Shujiao Li
- Specific Lab for Mitochondrial Plasticity Underlying Nervous System Diseases, National Demonstration Center for Experimental Preclinical Medicine Education, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yaoliang Wang
- Department of Neurosurgery, Xi-Jing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Zhenhua Wang
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Feifei Wu
- Specific Lab for Mitochondrial Plasticity Underlying Nervous System Diseases, National Demonstration Center for Experimental Preclinical Medicine Education, The Fourth Military Medical University, Xi'an, 710032, China
| | - Hui Liu
- Specific Lab for Mitochondrial Plasticity Underlying Nervous System Diseases, National Demonstration Center for Experimental Preclinical Medicine Education, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yousheng Wu
- Specific Lab for Mitochondrial Plasticity Underlying Nervous System Diseases, National Demonstration Center for Experimental Preclinical Medicine Education, The Fourth Military Medical University, Xi'an, 710032, China
| | - Guopeng Zhang
- Department of Computer Fundamentals, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Qingdong Guo
- Department of Neurosurgery, Xi-Jing Hospital, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Yanling Yang
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Yayun Wang
- Specific Lab for Mitochondrial Plasticity Underlying Nervous System Diseases, National Demonstration Center for Experimental Preclinical Medicine Education, The Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
37
|
Meena D, Jha S. Autophagy in glioblastoma: A mechanistic perspective. Int J Cancer 2024; 155:605-617. [PMID: 38716809 DOI: 10.1002/ijc.34991] [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: 01/29/2024] [Revised: 03/28/2024] [Accepted: 04/12/2024] [Indexed: 06/20/2024]
Abstract
Glioblastoma (GBM) is one of the most lethal malignancies in humans. Even after surgical resection and aggressive radio- or chemotherapies, patients with GBM can survive for less than 14 months. Extreme inter-tumor and intra-tumor heterogeneity of GBM poses a challenge for resolving recalcitrant GBM pathophysiology. GBM tumor microenvironment (TME) exhibits diverse heterogeneity in cellular composition and processes contributing to tumor progression and therapeutic resistance. Autophagy is such a cellular process; that demonstrates a cell-specific and TME context-dependent role in GBM progression, leading to either the promotion or suppression of GBM progression. Autophagy can regulate GBM cell function directly via regulation of survival, migration, and invasion, or indirectly by affecting GBM TME composition such as immune cell population, tumor metabolism, and glioma stem cells. This review comprehensively investigates the role of autophagy in GBM pathophysiology.
Collapse
Affiliation(s)
- Durgesh Meena
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India
| | - Sushmita Jha
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India
| |
Collapse
|
38
|
Chen S, Li X, Wu Q, Li Y, Puig M, Moulin F, Choudhuri S, Gingrich J, Guo L. Investigation of cannabidiol-induced cytotoxicity in human hepatic cells. Toxicology 2024; 506:153884. [PMID: 39004336 PMCID: PMC11648445 DOI: 10.1016/j.tox.2024.153884] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/23/2024] [Accepted: 07/08/2024] [Indexed: 07/16/2024]
Abstract
Cannabidiol (CBD) is one of the primary cannabinoids present in extracts of the plant Cannabis sativa L. A CBD-based drug, Epidiolex, has been approved by the U.S. FDA for the treatment of seizures in childhood-onset epileptic disorders. Although CBD-associated liver toxicity has been reported in clinical studies, the underlying mechanisms remain unclear. In this study, we demonstrated that CBD causes cytotoxicity in primary human hepatocytes and hepatic HepG2 cells. A 24-h CBD treatment induced cell cycle disturbances, cellular apoptosis, and endoplasmic reticulum (ER) stress in HepG2 cells. A potent ER stress inhibitor, 4-phenylbutyrate, markedly attenuated CBD-induced apoptosis and cell death. Additionally, we investigated the role of cytochrome P450 (CYP)-mediated metabolism in CBD-induced cytotoxicity using HepG2 cell lines engineered to express 14 individual CYPs. We identified CYP2C9, 2C19, 2D6, 2C18, and 3A5 as participants in CBD metabolism. Notably, cells overexpressing CYP2C9, 2C19, and 2C18 produced 7-hydroxy-CBD, while cells overexpressing CYP2C9, 2C19, 2D6, and 2C18 generated 7-carboxy-CBD. Furthermore, CBD-induced cytotoxicity was significantly attenuated in the cells expressing CYP2D6. Taken together, these data suggest that cell cycle disturbances, apoptosis, and ER stress are associated with CBD-induced cytotoxicity, and CYP2D6-mediated metabolism plays a critical role in decreasing the cytotoxicity of CBD.
Collapse
Affiliation(s)
- Si Chen
- Division of Biochemical Toxicology, National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA), Jefferson, AR 72079, USA.
| | - Xilin Li
- Division of Genetic and Molecular Toxicology, NCTR, U.S. FDA, Jefferson, AR 72079, USA
| | - Qiangen Wu
- Division of Biochemical Toxicology, National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA), Jefferson, AR 72079, USA
| | - Yuxi Li
- Division of Biochemical Toxicology, National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA), Jefferson, AR 72079, USA
| | - Montserrat Puig
- Division of Biotechnology Review and Research III, Office of Biotechnology Products, Center for Drug Evaluation and Research, U.S. FDA, Silver Spring, MD 20993, USA
| | - Frederic Moulin
- Division of Hepatology and Nutrition, Office of New Drugs, Center for Drug Evaluation and Research, U.S. FDA, Silver Spring, MD 20993, USA
| | - Supratim Choudhuri
- Division of Food Ingredients, Office of Food Additive Safety, Center for Food Safety and Applied Nutrition, U.S. FDA, College Park, MD 20740, USA
| | - Jeremy Gingrich
- Division of Food Ingredients, Office of Food Additive Safety, Center for Food Safety and Applied Nutrition, U.S. FDA, College Park, MD 20740, USA
| | - Lei Guo
- Division of Biochemical Toxicology, National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA), Jefferson, AR 72079, USA.
| |
Collapse
|
39
|
Freire NF, Cordani M, Aparicio-Blanco J, Fraguas Sanchez AI, Dutra L, Pinto MC, Zarrabi A, Pinto JC, Velasco G, Fialho R. Preparation and characterization of PBS (Polybutylene Succinate) nanoparticles containing cannabidiol (CBD) for anticancer application. J Drug Deliv Sci Technol 2024; 97:105833. [DOI: 10.1016/j.jddst.2024.105833] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
40
|
Yang M, Wei X, Yi X, Jiang DS. Mitophagy-related regulated cell death: molecular mechanisms and disease implications. Cell Death Dis 2024; 15:505. [PMID: 39013891 PMCID: PMC11252137 DOI: 10.1038/s41419-024-06804-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 05/26/2024] [Accepted: 06/03/2024] [Indexed: 07/18/2024]
Abstract
During oxidative phosphorylation, mitochondria continuously produce reactive oxygen species (ROS), and untimely ROS clearance can subject mitochondria to oxidative stress, ultimately resulting in mitochondrial damage. Mitophagy is essential for maintaining cellular mitochondrial quality control and homeostasis, with activation involving both ubiquitin-dependent and ubiquitin-independent pathways. Over the past decade, numerous studies have indicated that different forms of regulated cell death (RCD) are connected with mitophagy. These diverse forms of RCD have been shown to be regulated by mitophagy and are implicated in the pathogenesis of a variety of diseases, such as tumors, degenerative diseases, and ischemia‒reperfusion injury (IRI). Importantly, targeting mitophagy to regulate RCD has shown excellent therapeutic potential in preclinical trials, and is expected to be an effective strategy for the treatment of related diseases. Here, we present a summary of the role of mitophagy in different forms of RCD, with a focus on potential molecular mechanisms by which mitophagy regulates RCD. We also discuss the implications of mitophagy-related RCD in the context of various diseases.
Collapse
Affiliation(s)
- Molin Yang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiang Wei
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China
| | - Xin Yi
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Ding-Sheng Jiang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China.
| |
Collapse
|
41
|
Feng S, Pan Y, Lu P, Li N, Zhu W, Hao Z. From bench to bedside: the application of cannabidiol in glioma. J Transl Med 2024; 22:648. [PMID: 38987805 PMCID: PMC11238413 DOI: 10.1186/s12967-024-05477-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 07/03/2024] [Indexed: 07/12/2024] Open
Abstract
Glioma is the most common malignant tumor in central nervous system, with significant health burdens to patients. Due to the intrinsic characteristics of glioma and the lack of breakthroughs in treatment modalities, the prognosis for most patients remains poor. This results in a heavy psychological and financial load worldwide. In recent years, cannabidiol (CBD) has garnered widespread attention and research due to its anti-tumoral, anti-inflammatory, and neuroprotective properties. This review comprehensively summarizes the preclinical and clinical research on the use of CBD in glioma therapy, as well as the current status of nanomedicine formulations of CBD, and discusses the potential and challenges of CBD in glioma therapy in the future.
Collapse
Affiliation(s)
- Shiying Feng
- Department of Oncology, Baotou City Central Hospital, Baotou, 014040, China
- Central Clinical Medical School, Baotou Medical College, Baotou, 014040, China
| | - Yuanming Pan
- Cancer Research Center, Beijing Tuberculosis & Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
| | - Pu Lu
- Department of Oncology, Baotou City Central Hospital, Baotou, 014040, China
| | - Na Li
- Department of Gynecology, Baotou City Central Hospital, Baotou, 014040, China.
| | - Wei Zhu
- Department of Oncology, Baotou City Central Hospital, Baotou, 014040, China.
| | - Zhiqiang Hao
- Department of Oncology, Baotou City Central Hospital, Baotou, 014040, China
| |
Collapse
|
42
|
Nafe R, Hattingen E. Forms of Non-Apoptotic Cell Death and Their Role in Gliomas-Presentation of the Current State of Knowledge. Biomedicines 2024; 12:1546. [PMID: 39062119 PMCID: PMC11274595 DOI: 10.3390/biomedicines12071546] [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: 06/10/2024] [Revised: 07/05/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
In addition to necrosis and apoptosis, the two forms of cell death that have been known for many decades, other non-apoptotic forms of cell death have been discovered, many of which also play a role in tumors. Starting with the description of autophagy more than 60 years ago, newer forms of cell death have become important for the biology of tumors, such as ferroptosis, pyroptosis, necroptosis, and paraptosis. In this review, all non-apoptotic and oncologically relevant forms of programmed cell death are presented, starting with their first descriptions, their molecular characteristics, and their role and their interactions in cell physiology and pathophysiology. Based on these descriptions, the current state of knowledge about their alterations and their role in gliomas will be presented. In addition, current efforts to therapeutically influence the molecular components of these forms of cell death will be discussed. Although research into their exact role in gliomas is still at a rather early stage, our review clarifies that all these non-apoptotic forms of cell death show significant alterations in gliomas and that important insight into understanding them has already been gained.
Collapse
Affiliation(s)
- Reinhold Nafe
- Department of Neuroradiology, Clinics of Johann Wolfgang Goethe-University, Schleusenweg 2-16, D-60528 Frankfurt am Main, Germany;
| | | |
Collapse
|
43
|
Bellone ML, Syed AA, Vitale RM, Sigismondo G, Mensitieri F, Pollastro F, Amodeo P, Appendino G, De Tommasi N, Krijgsveld J, Dal Piaz F. Eukaryotic Initiation Translation Factor 2A activation by cannabidiolic acid alters the protein homeostasis balance in glioblastoma cells. Int J Biol Macromol 2024; 273:132968. [PMID: 38871097 DOI: 10.1016/j.ijbiomac.2024.132968] [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: 03/04/2024] [Revised: 06/01/2024] [Accepted: 06/05/2024] [Indexed: 06/15/2024]
Abstract
Eukaryotic Initiation Translation Factor 2A (EIF2A) is considered to be primarily responsible for the initiation of translation when a cell is subjected to stressful conditions. However, information regarding this protein is still incomplete. Using a combination of proteomic approaches, we demonstrated that EIF2A is the molecular target of the naturally occurring bioactive compound cannabidiolic acid (CBDA) within human glioblastoma cells. This finding allowed us to undertake a study aimed at obtaining further information on the functions that EIF2A plays in tumor cells. Indeed, our data showed that CBDA is able to activate EIF2A when the cells are in no-stress conditions. It induces conformational changes in the protein structure, thus increasing EIF2A affinity towards the proteins participating in the Eukaryotic Translation Machinery. Consequently, following glioblastoma cells incubation with CBDA we observed an enhanced neosynthesis of proteins involved in the stress response, nucleic acid translation and organization, and protein catabolism. These changes in gene expression resulted in increased levels of ubiquitinated proteins and accumulation of the autophagosome. Our results, in addition to shedding light on the molecular mechanism underlying the biological effect of a phytocannabinoid in cancer cells, demonstrated that EIF2A plays a critical role in regulation of protein homeostasis.
Collapse
Affiliation(s)
| | - Azmal Ali Syed
- Division of Proteomics of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Gianluca Sigismondo
- Division of Proteomics of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Federica Pollastro
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, Vercelli, Italy
| | - Pietro Amodeo
- Institute of Biomolecular Chemistry (ICB-CNR), Pozzuoli, Italy
| | - Giovanni Appendino
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, Vercelli, Italy
| | | | - Jeroen Krijgsveld
- Division of Proteomics of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Fabrizio Dal Piaz
- Department of Medicine and Surgery, University of Salerno, Baronissi, Italy.
| |
Collapse
|
44
|
Zhang N, Zhang S, Dong X. Plant-derived bioactive compounds and their novel role in central nervous system disorder treatment via ATF4 targeting: A systematic literature review. Biomed Pharmacother 2024; 176:116811. [PMID: 38795641 DOI: 10.1016/j.biopha.2024.116811] [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: 03/30/2024] [Revised: 05/18/2024] [Accepted: 05/20/2024] [Indexed: 05/28/2024] Open
Abstract
Central nervous system (CNS) disorders exhibit exceedingly intricate pathogenic mechanisms. Pragmatic and effective solutions remain elusive, significantly compromising human life and health. Activating transcription factor 4 (ATF4) participates in the regulation of multiple pathophysiological processes, including CNS disorders. Considering the widespread involvement of ATF4 in the pathological process of CNS disorders, the targeted regulation of ATF4 by plant-derived bioactive compounds (PDBCs) may become a viable strategy for the treatment of CNS disorders. However, the regulatory relationship between PDBCs and ATF4 remains incompletely understood. Here, we aimed to comprehensively review the studies on PDBCs targeting ATF4 to ameliorate CNS disorders, thereby offering novel directions and insights for the treatment of CNS disorders. A computerized search was conducted on PubMed, Embase, Web of Science, and Google Scholar databases to identify preclinical experiments related to PDBCs targeting ATF4 for the treatment of CNS disorders. The search timeframe was from the inception of the databases to December 2023. Two assessors conducted searches using the keywords "ATF4," "Central Nervous System," "Neurological," "Alzheimer's disease," "Parkinson's Disease," "Stroke," "Spinal Cord Injury," "Glioblastoma," "Traumatic Brain Injury," and "Spinal Cord Injury." Overall, 31 studies were included, encompassing assessments of 27 PDBCs. Combining results from in vivo and in vitro studies, we observed that these PDBCs, via ATF4 modulation, prevent the deposition of amyloid-like fibers such as Aβ, tau, and α-synuclein. They regulate ERS, reduce the release of inflammatory factors, restore mitochondrial membrane integrity to prevent oxidative stress, regulate synaptic plasticity, modulate autophagy, and engage anti-apoptotic mechanisms. Consequently, they exert neuroprotective effects in CNS disorders. Numerous PDBCs targeting ATF4 have shown potential in facilitating the restoration of CNS functionality, thereby presenting expansive prospects for the treatment of such disorders. However, future endeavors necessitate high-quality, large-scale, and comprehensive preclinical and clinical studies to further validate this therapeutic potential.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Neurology, the Seventh Clinical College of China Medical University, No. 24 Central Street, Xinfu District, Fushun, Liaoning 113000, China
| | - Shun Zhang
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao street, Heping District, Shenyang, Liaoning 110000, China
| | - Xiaoyu Dong
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao street, Heping District, Shenyang, Liaoning 110000, China.
| |
Collapse
|
45
|
Marquez AB, Vicente J, Castro E, Vota D, Rodríguez-Varela MS, Lanza Castronuovo PA, Fuentes GM, Parise AR, Romorini L, Alvarez DE, Bueno CA, Ramirez CL, Alaimo A, García CC. Broad-Spectrum Antiviral Effect of Cannabidiol Against Enveloped and Nonenveloped Viruses. Cannabis Cannabinoid Res 2024; 9:751-765. [PMID: 37682578 DOI: 10.1089/can.2023.0103] [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: 09/09/2023] Open
Abstract
Introduction: Cannabidiol (CBD), the main non-psychoactive cannabinoid of the Cannabis sativa plant, is a powerful antioxidant compound that in recent years has increased interest due to causes effects in a wide range of biological functions. Zika virus (ZIKV) is a virus transmitted mainly by the Aedes aegypti mosquitoes, which causes neurological diseases, such as microcephaly and Guillain-Barre syndrome. Although the frequency of viral outbreaks has increased recently, no vaccinations or particular chemotherapeutic treatments are available for ZIKV infection. Objectives: The major aim of this study was to explore the in vitro antiviral activity of CBD against ZIKV, expanding also to other dissimilar viruses. Materials and Methods: Cell cultures were infected with enveloped and nonenveloped viruses and treated with non-cytotoxic concentrations of CBD and then, viral titers were determined. Additionally, the mechanism of action of the compound during ZIKV in vitro infections was studied. To study the possible immunomodulatory role of CBD, infected and uninfected Huh-7 cells were exposed to 10 μM CBD during 48 h and levels of interleukins 6 and 8 and interferon-beta (IFN-β) expression levels were measured. On the other hand, the effect of CBD on cellular membranes was studied. For this, an immunofluorescence assay was performed, in which cell membranes were labeled with wheat germ agglutinin. Finally, intracellular cholesterol levels were measured. Results: CBD exhibited a potent antiviral activity against all the tested viruses in different cell lines with half maximal effective concentration values (CE50) ranging from 0.87 to 8.55 μM. Regarding the immunomodulatory effect of CBD during ZIKV in vitro infections, CBD-treated cells exhibited significantly IFN-β increased levels, meanwhile, interleukins 6 and 8 were not induced. Furthermore, it was determined that CBD affects cellular membranes due to the higher fluorescence intensity that was observed in CBD-treated cells and lowers intracellular cholesterol levels, thus affecting the multiplication of ZIKV and other viruses. Conclusions: It was demonstrated that CBD inhibits structurally dissimilar viruses, suggesting that this phytochemical has broad-spectrum antiviral effect, representing a valuable alternative in emergency situations during viral outbreaks, like the one caused by severe acute respiratory syndrome coronavirus 2 in 2020.
Collapse
Affiliation(s)
- Agostina B Marquez
- Laboratorio de Virología, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Josefina Vicente
- Laboratorio de Virología, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Eliana Castro
- Instituto de Investigaciones Biotecnológicas (IIBIO), Universidad Nacional de San Martín (UNSAM)-(CONICET), Buenos Aires, Argentina
| | - Daiana Vota
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Laboratorio de Inmunofarmacología, IQUIBICEN, UBA-CONICET, Buenos Aires, Argentina
| | - María S Rodríguez-Varela
- Laboratorio de Investigación Aplicada a Neurociencias (LIAN), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (Fleni)-CONICET, Instituto de Neurociencias (INEU), Buenos Aires, Argentina
| | - Priscila A Lanza Castronuovo
- Instituto de Investigaciones en Biodiversidad y Biotecnología (INBIOTEC), Química Analítica y Modelado Molecular (QUIAMM), Universidad Nacional de Mar del Plata-CONICET, Mar del Plata, Argentina
| | - Giselle M Fuentes
- Instituto de Investigaciones en Producción Sanidad y Ambiente (IIPROSAM), Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata, Centro Científico Tecnológico Mar del Plata, CONICET, Mar del Plata, Argentina
- Centro de Asociación Simple CIC PBA, Mar del Plata, Argentina
- Centro de Investigaciones en Abejas Sociales, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata, Mar del Plata, Argentina
| | - Alejandro R Parise
- Instituto de Investigaciones en Biodiversidad y Biotecnología (INBIOTEC), Química Analítica y Modelado Molecular (QUIAMM), Universidad Nacional de Mar del Plata-CONICET, Mar del Plata, Argentina
- Departamento de Química Biológica y Bioquímica, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata, Mar del Plata, Argentina
| | - Leonardo Romorini
- Laboratorio de Investigación Aplicada a Neurociencias (LIAN), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (Fleni)-CONICET, Instituto de Neurociencias (INEU), Buenos Aires, Argentina
| | - Diego E Alvarez
- Instituto de Investigaciones Biotecnológicas (IIBIO), Universidad Nacional de San Martín (UNSAM)-(CONICET), Buenos Aires, Argentina
- Escuela de Bio y Nanotecnologías (EByN), Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Carlos A Bueno
- Laboratorio de Virología, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Cristina L Ramirez
- Departamento de Química Biológica y Bioquímica, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata, Mar del Plata, Argentina
- Asociación Civil CBG2000, Mar del Plata, Argentina
| | - Agustina Alaimo
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Cybele C García
- Laboratorio de Virología, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
46
|
Zhu SL, Qi M, Chen MT, Lin JP, Huang HF, Deng LJ, Zhou XW. A novel DDIT3 activator dehydroevodiamine effectively inhibits tumor growth and tumor cell stemness in pancreatic cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155377. [PMID: 38503154 DOI: 10.1016/j.phymed.2024.155377] [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: 04/11/2023] [Revised: 12/19/2023] [Accepted: 01/17/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND The existence of pancreatic cancer stem cells (PCSCs) results in limited survival benefits from current treatment options. There is a scarcity of effective agents for treating pancreatic cancer patients. Dehydroevodiamine (DeHE), a quinazoline alkaloid isolated from the traditional Chinese herb Evodiae fructus, exhibited potent inhibition of pancreatic ductal adenocarcinoma (PDAC) cell proliferation and tumor growth both in vitro and in vivo. METHODS The cytotoxic effect of DeHE on PDAC cells was assessed using CCK-8 and colony formation assays. The antitumor efficacy of DeHE were appraised in human PANC-1 xenograft mouse model. Sphere formation assay and flow cytometry were employed to quantify the tumor stemness. RNA-Seq analysis, drug affinity responsive target stability assay (DARTS), and RNA interference transfection were conducted to elucidate potential signaling pathways. Western blotting and immunohistochemistry were utilized to assess protein expression levels. RESULTS DeHE effectively inhibited PDAC cell proliferation and tumor growth in vitro and in vivo, and exhibited a better safety profile compared to the clinical drug gemcitabine (GEM). DeHE inhibited PCSCs, as evidenced by its suppression of self-renewal capabilities of PCSCs, reduced the proportion of ALDH+ cells and downregulated stemness-associated proteins (Nanog, Sox-2, and Oct-4) both in vitro and in vivo. Furthermore, there is potential involvement of DDIT3 and its downstream DDIT3/TRIB3/AKT/mTOR pathway in the suppression of stemness characteristics within DeHE-treated PDAC cells. Additionally, results from the DARTS assay indicated that DeHE interacts with DDIT3, safeguarding it against degradation mediated by pronase. Notably, the inhibitory capabilities of DeHE on PDAC cell proliferation and tumor stemness were partially restored by siDDIT3 or the AKT activator SC-79. CONCLUSION In summary, our study has identified DeHE, a novel antitumor natural product, as an activator of DDIT3 with the ability to suppress the AKT/mTOR pathway. This pathway is intricately linked to tumor cell proliferation and stemness characteristics in PDAC. These findings suggest that DeHE holds potential as a promising candidate for the development of innovative anticancer therapeutics.
Collapse
Affiliation(s)
- Su-Li Zhu
- Department of Biochemistry and Pharmacology, Sun Yat-Sen University Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, PR China
| | - Ming Qi
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Mei-Ting Chen
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, PR China
| | - Jia-Peng Lin
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Hai-Fu Huang
- Internal Medicine-Oncology, Shenzhen Hospital of Guangzhou University of Traditional Chinese Medicine, PR China
| | - Li-Juan Deng
- Guangzhou Key Laboratory of Formula-pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, PR China.
| | - Xing-Wang Zhou
- Department of Biochemistry and Pharmacology, Sun Yat-Sen University Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, PR China.
| |
Collapse
|
47
|
Li W, Huang C, Qiu L, Tang Y, Zhang X, Zhang L, Zhao H, Miyagishi M, Kasim V, Wu S. p52-ZER6/IGF1R axis maintains cancer stem cell population to promote cancer progression by enhancing pro-survival mitophagy. Oncogene 2024; 43:2115-2131. [PMID: 38773262 DOI: 10.1038/s41388-024-03058-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/27/2024] [Accepted: 05/01/2024] [Indexed: 05/23/2024]
Abstract
Cancer stem cells (CSCs), which are distinct subpopulations of tumor cells, have a substantially higher tumor-initiating capacity and are closely related to poor clinical outcomes. Damage to organelles can trigger CSC pool exhaustion; however, the underlying mechanisms are poorly understood. ZER6 is a zinc-finger protein with two isoforms possessing different amino termini: p52-ZER6 and p71-ZER6. Since their discovery, almost no study reported on their biological and pathological functions. Herein, we found that p52-ZER6 was crucial for CSC population maintenance; p52-ZER6-knocking down almost abolished the tumor initiation capability. Through transcriptomic analyses together with in vitro and in vivo studies, we identified insulin like growth factor 1 receptor (IGF1R) as the transcriptional target of p52-ZER6 that mediated p52-ZER6 regulation of CSC by promoting pro-survival mitophagy. Moreover, this regulation of mitophagy-mediated CSC population maintenance is specific to p52-ZER6, as p71-ZER6 failed to exert the same effect, most possibly due to the presence of the HUB1 domain at its N-terminus. These results provide a new perspective on the regulatory pathway of pro-survival mitophagy in tumor cells and the molecular mechanism underlying p52-ZER6 oncogenic activity, suggesting that targeting p52-ZER6/IGF1R axis to induce CSC pool exhaustion may be a promising anti-tumor therapeutic strategy.
Collapse
Affiliation(s)
- Wenfang Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
- School of Pharmaceutical Sciences and Institute of Materia Medica, Xinjiang University, Urumqi, 830017, China
| | - Can Huang
- Metabolic Disease Research Center, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, 230032, China.
| | - Li Qiu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Yu Tang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Xia Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Lei Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Hezhao Zhao
- Department of Gastrointestinal Surgery, Chongqing University Cancer Hospital, Chongqing University, Chongqing, 400030, China
| | - Makoto Miyagishi
- Life Science Innovation, School of Integrative and Global Majors, University of Tsukuba, Tsukuba, Ibaraki, 305-0006, Japan
| | - Vivi Kasim
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing University, Chongqing, 400030, China.
| | - Shourong Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing University, Chongqing, 400030, China.
| |
Collapse
|
48
|
Kim NY, Shivanne Gowda SG, Lee SG, Sethi G, Ahn KS. Cannabidiol induces ERK activation and ROS production to promote autophagy and ferroptosis in glioblastoma cells. Chem Biol Interact 2024; 394:110995. [PMID: 38583854 DOI: 10.1016/j.cbi.2024.110995] [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: 02/26/2024] [Revised: 03/27/2024] [Accepted: 04/04/2024] [Indexed: 04/09/2024]
Abstract
Small molecule-driven ERK activation is known to induce autophagy and ferroptosis in cancer cells. Herein the effect of cannabidiol (CBD), a phytochemical derived from Cannabis sativa, on ERK-driven autophagy and ferroptosis has been demonstrated in glioblastoma (GBM) cells (U87 and U373 cells). CBD imparted significant cytotoxicity in GBM cells, induced activation of ERK (not JNK and p38), and increased intracellular reactive oxygen species (ROS) levels. It increased the autophagy-related proteins such as LC3 II, Atg7, and Beclin-1 and modulated the expression of ferroptosis-related proteins such as glutathione peroxidase 4 (GPX4), SLC7A11, and TFRC. CBD significantly elevated the endoplasmic reticulum stress, ROS, and iron load, and decreased GSH levels. Inhibitors of autophagy (3-MA) and ferroptosis (Fer-1) had a marginal effect on CBD-induced autophagy/ferroptosis. Treatment with N-acetyl-cysteine (antioxidant) or PD98059 (ERK inhibitor) partly reverted the CBD-induced autophagy/ferroptosis by decreasing the activation of ERK and the production of ROS. Overall, CBD induced autophagy and ferroptosis through the activation of ERK and generation of ROS in GBM cells.
Collapse
Affiliation(s)
- Na Young Kim
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
| | | | - Seok-Geun Lee
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
| |
Collapse
|
49
|
Ma L, Liu M, Liu C, Zhang H, Yang S, An J, Qu G, Song S, Cao Q. Research Progress on the Mechanism of the Antitumor Effects of Cannabidiol. Molecules 2024; 29:1943. [PMID: 38731434 PMCID: PMC11085351 DOI: 10.3390/molecules29091943] [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: 03/19/2024] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 05/13/2024] Open
Abstract
Cannabidiol (CBD), a non-psychoactive ingredient extracted from the hemp plant, has shown therapeutic effects in a variety of diseases, including anxiety, nervous system disorders, inflammation, and tumors. CBD can exert its antitumor effect by regulating the cell cycle, inducing tumor cell apoptosis and autophagy, and inhibiting tumor cell invasion, migration, and angiogenesis. This article reviews the proposed antitumor mechanisms of CBD, aiming to provide references for the clinical treatment of tumor diseases and the rational use of CBD.
Collapse
Affiliation(s)
- Li Ma
- Department of Immunology, School of Basic Medical Sciences, Binzhou Medical University, Yantai 264003, China; (L.M.); (M.L.); (C.L.); (H.Z.)
| | - Mengke Liu
- Department of Immunology, School of Basic Medical Sciences, Binzhou Medical University, Yantai 264003, China; (L.M.); (M.L.); (C.L.); (H.Z.)
| | - Chuntong Liu
- Department of Immunology, School of Basic Medical Sciences, Binzhou Medical University, Yantai 264003, China; (L.M.); (M.L.); (C.L.); (H.Z.)
| | - Huachang Zhang
- Department of Immunology, School of Basic Medical Sciences, Binzhou Medical University, Yantai 264003, China; (L.M.); (M.L.); (C.L.); (H.Z.)
| | - Shude Yang
- Department of Edible Mushrooms, School of Agriculture, Ludong University, Yantai 264025, China;
| | - Jing An
- Division of Infectious Diseases and Global Health, School of Medicine, University of California San Diego (UCSD), La Jolla, CA 92037, USA;
| | - Guiwu Qu
- Department of Traditional Chinese Medicine, School of Pharmacy, Binzhou Medical University, Yantai 264003, China;
| | - Shuling Song
- Department of Immunology, School of Basic Medical Sciences, Binzhou Medical University, Yantai 264003, China; (L.M.); (M.L.); (C.L.); (H.Z.)
| | - Qizhi Cao
- Department of Immunology, School of Basic Medical Sciences, Binzhou Medical University, Yantai 264003, China; (L.M.); (M.L.); (C.L.); (H.Z.)
| |
Collapse
|
50
|
Majchrzak-Celińska A, Studzińska-Sroka E. New Avenues and Major Achievements in Phytocompounds Research for Glioblastoma Therapy. Molecules 2024; 29:1682. [PMID: 38611962 PMCID: PMC11013944 DOI: 10.3390/molecules29071682] [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: 02/23/2024] [Revised: 03/22/2024] [Accepted: 04/04/2024] [Indexed: 04/14/2024] Open
Abstract
Phytocompounds have been evaluated for their anti-glioblastoma actions for decades, with promising results from preclinical studies but only limited translation into clinics. Indeed, by targeting multiple signaling pathways deregulated in cancer, they often show high efficacy in the in vitro studies, but their poor bioavailability, low tumor accumulation, and rapid clearance compromise their efficacy in vivo. Here, we present the new avenues in phytocompound research for the improvement of glioblastoma therapy, including the ways to enhance the response to temozolomide using phytochemicals, the current focus on phytocompound-based immunotherapy, or the use of phytocompounds as photosensitizers in photodynamic therapy. Moreover, we present new, intensively evaluated approaches, such as chemical modifications of phytochemicals or encapsulation into numerous types of nanoformulations, to improve their bioavailability and delivery to the brain. Finally, we present the clinical trials evaluating the role of phytocompounds or phytocompound-derived drugs in glioblastoma therapy and the less studied phytocompounds or plant extracts that have only recently been found to possess promising anti-glioblastoma properties. Overall, recent advancements in phytocompound research are encouraging; however, only with more 3D glioblastoma models, in vivo studies, and clinical trials it is possible to upgrade the role of phytocompounds in glioblastoma treatment to a satisfactory level.
Collapse
Affiliation(s)
- Aleksandra Majchrzak-Celińska
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 3 Str., 60-806 Poznan, Poland
| | - Elżbieta Studzińska-Sroka
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, Rokietnicka 3 Str., 60-806 Poznan, Poland;
| |
Collapse
|