1
|
Pan T, Peng L, Dong J, Li L. Pterostilbene Induces Pyroptosis in Breast Cancer Cells through Pyruvate Kinase 2/Caspase-8/Gasdermin C Signaling Pathway. Int J Mol Sci 2024; 25:10509. [PMID: 39408842 PMCID: PMC11476961 DOI: 10.3390/ijms251910509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
The incidence and mortality of breast cancer increase year by year, and it is urgent to find high-efficiency and low-toxicity anti-cancer drugs. Pterostilbene (PTE) is a natural product with antitumor activity, but the specific antitumor mechanism is not very clear. Aerobic glycolysis is the main energy supply for cancer cells. Pyroptosis is an inflammatory, programmed cell death. The aim of this study was to investigate the effect of PTE on glycolysis and pyroptosis in EMT6 and 4T1 cells and the specific mechanism, and to elucidate the role of pyruvate kinase 2 (PKM2), a key enzyme in glycolysis, in the antitumor role of PTE. Our study suggested that PTE induced pyroptosis by inhibiting tumor glycolysis. PKM2 played an important role in both the inhibition of glycolysis and the induction of pyroptosis by PTE.
Collapse
Affiliation(s)
| | | | - Jing Dong
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China; (T.P.); (L.P.)
| | - Lin Li
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China; (T.P.); (L.P.)
| |
Collapse
|
2
|
Luo M, Luan X, Yang C, Chen X, Yuan S, Cao Y, Zhang J, Xie J, Luo Q, Chen L, Li S, Xiang W, Zhou J. Revisiting the potential of regulated cell death in glioma treatment: a focus on autophagy-dependent cell death, anoikis, ferroptosis, cuproptosis, pyroptosis, immunogenic cell death, and the crosstalk between them. Front Oncol 2024; 14:1397863. [PMID: 39184045 PMCID: PMC11341384 DOI: 10.3389/fonc.2024.1397863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 07/22/2024] [Indexed: 08/27/2024] Open
Abstract
Gliomas are primary tumors that originate in the central nervous system. The conventional treatment options for gliomas typically encompass surgical resection and temozolomide (TMZ) chemotherapy. However, despite aggressive interventions, the median survival for glioma patients is merely about 14.6 months. Consequently, there is an urgent necessity to explore innovative therapeutic strategies for treating glioma. The foundational study of regulated cell death (RCD) can be traced back to Karl Vogt's seminal observations of cellular demise in toads, which were documented in 1842. In the past decade, the Nomenclature Committee on Cell Death (NCCD) has systematically classified and delineated various forms and mechanisms of cell death, synthesizing morphological, biochemical, and functional characteristics. Cell death primarily manifests in two forms: accidental cell death (ACD), which is caused by external factors such as physical, chemical, or mechanical disruptions; and RCD, a gene-directed intrinsic process that coordinates an orderly cellular demise in response to both physiological and pathological cues. Advancements in our understanding of RCD have shed light on the manipulation of cell death modulation - either through induction or suppression - as a potentially groundbreaking approach in oncology, holding significant promise. However, obstacles persist at the interface of research and clinical application, with significant impediments encountered in translating to therapeutic modalities. It is increasingly apparent that an integrative examination of the molecular underpinnings of cell death is imperative for advancing the field, particularly within the framework of inter-pathway functional synergy. In this review, we provide an overview of various forms of RCD, including autophagy-dependent cell death, anoikis, ferroptosis, cuproptosis, pyroptosis and immunogenic cell death. We summarize the latest advancements in understanding the molecular mechanisms that regulate RCD in glioma and explore the interconnections between different cell death processes. By comprehending these connections and developing targeted strategies, we have the potential to enhance glioma therapy through manipulation of RCD.
Collapse
Affiliation(s)
- Maowen Luo
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Xingzhao Luan
- Department of Neurosurgery, the Affiliated Hospital of Panzhihua University, Panzhihua, Sichuan, China
- School of Clinical Medicine, the Affiliated Hospital of Panzhihua University, Panzhihua, Sichuan, China
| | - Chaoge Yang
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, China
| | - Xiaofan Chen
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Suxin Yuan
- School of Clinical Medicine, the Affiliated Hospital of Panzhihua University, Panzhihua, Sichuan, China
| | - Youlin Cao
- Department of Neurosurgery, the Affiliated Hospital of Panzhihua University, Panzhihua, Sichuan, China
- School of Clinical Medicine, the Affiliated Hospital of Panzhihua University, Panzhihua, Sichuan, China
| | - Jing Zhang
- School of Clinical Medicine, the Affiliated Hospital of Panzhihua University, Panzhihua, Sichuan, China
| | - Jiaying Xie
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Qinglian Luo
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, China
| | - Ligang Chen
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, China
| | - Shenjie Li
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, China
| | - Wei Xiang
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, China
| | - Jie Zhou
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, China
| |
Collapse
|
3
|
Nishiguch Y, Fujiwara-Tani R, Nukaga S, Nishida R, Ikemoto A, Sasaki R, Mori S, Ogata R, Kishi S, Hojo Y, Shinohara H, Sho M, Kuniyasu H. Pterostilbene Induces Apoptosis from Endoplasmic Reticulum Stress Synergistically with Anticancer Drugs That Deposit Iron in Mitochondria. Int J Mol Sci 2024; 25:2611. [PMID: 38473857 DOI: 10.3390/ijms25052611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Anticancer agents are playing an increasing role in the treatment of gastric cancer (GC); however, novel anticancer agents have not been fully developed. Therefore, it is important to investigate compounds that improve sensitivity to the existing anticancer drugs. We have reported that pterostilbene (PTE), a plant stilbene, enhances the antitumor effect of low doses of sunitinib in gastric cancer cells accumulating mitochondrial iron (II) (mtFe) at low doses. In this study, we investigated the relationship between the mtFe deposition and the synergistic effect of PTE and different anticancer drugs. For this study, we used 5-fluorouracil (5FU), cisplatin (CPPD), and lapatinib (LAP), which are frequently used in the treatment of GC, and doxorubicin (DOX), which is known to deposit mtFe. A combination of low-dose PTE and these drugs suppressed the expression of PDZ domain-containing 8 (PDZD8) and increased mtFe accumulation and mitochondrial H2O2. Consequently, reactive oxygen species-associated hypoxia inducible factor-1α activation induced endoplasmic reticulum stress and led to apoptosis, but not ferroptosis. In contrast, 5FU and CDDP did not show the same changes as those observed with PTE and DOX or LAP, and there was no synergistic effect with PTE. These results indicate that the combination of PTE with iron-accumulating anticancer drugs exhibits a strong synergistic effect. These findings would help in developing novel therapeutic strategies for GC. However, further clinical investigations are required.
Collapse
Grants
- 22K16497 Ministry of Education, Culture, Sports, Science and Technology
- 19K16564 Ministry of Education, Culture, Sports, Science and Technology
- 23K19900 Ministry of Education, Culture, Sports, Science and Technology
- 20K21659 Ministry of Education, Culture, Sports, Science and Technology
Collapse
Affiliation(s)
- Yukiko Nishiguch
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Rina Fujiwara-Tani
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Shota Nukaga
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Ryoichi Nishida
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Ayaka Ikemoto
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Rika Sasaki
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Shiori Mori
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Ruiko Ogata
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Shingo Kishi
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
- Pathology Laboratory, Research Institute, Tokushukai Nozaki Hospital, 2-10-50 Tanigawa, Daito 574-0074, Osaka, Japan
| | - Yudai Hojo
- Department of Surgery, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya 663-8501, Hyogo, Japan
| | - Hisashi Shinohara
- Department of Surgery, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya 663-8501, Hyogo, Japan
| | - Masayuki Sho
- Department of Surgery, Nara Medical University, Kashihara 634-8522, Nara, Japan
| | - Hiroki Kuniyasu
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| |
Collapse
|
4
|
Qu X, Zhang L, Wang L. Pterostilbene as a Therapeutic Alternative for Central Nervous System Disorders: A Review of the Current Status and Perspectives. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:14432-14457. [PMID: 37786984 DOI: 10.1021/acs.jafc.3c06238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
Neurological disorders are diverse, have complex causes, and often result in disability; yet, effective treatments remain scarce. The resveratrol derivative pterostilbene possesses numerous physiological activities that hold promise as a novel therapy for the central nervous system (CNS) disorders. This review aimed to summarize the protective mechanisms of pterostilbene in in vitro and in vivo models of CNS disorders and the pharmacokinetics and safety to assess its possible effects on CNS disorders. Available evidence supports the protective effects of pterostilbene in CNS disorders involving mechanisms such as antioxidant and anti-inflammatory activity, regulation of lipid metabolism and vascular smooth muscle cell proliferation, improvement of synaptic function and neurogenesis, induction of glioma cell cycle arrest, and inhibition of glioma cell migration and invasion. Studies have identified possible molecular targets and pathways for the protective actions of pterostilbene in CNS disorders including the AMPK/STAT3, Akt, NF-κB, MAPK, and ERK signaling pathways. The possible pharmacological effects and molecular pathways of pterostilbene in CNS disorders are critically discussed in this review. Future studies should aim to increase our understanding of pterostilbene in animal models and humans to further evaluate its role in CNS disorders and the detailed mechanisms.
Collapse
Affiliation(s)
- Xin Qu
- Department of Orthopedics, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, P.R. China
| | - Lijuan Zhang
- Departments of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 39 Huaxiang Road, Tiexi District, Shenyang 110000, Liaoning, P.R. China
| | - Lin Wang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, No. 39 Huaxiang Road, Tiexi District, Shenyang 110000, Liaoning, P.R. China
| |
Collapse
|
5
|
Khalil MI, Agamy AF, Elshewemi SS, Sultan AS, Abdelmeguid NE. Pterostilbene induces apoptosis in hepatocellular carcinoma cells: Biochemical, pathological, and molecular markers. Saudi J Biol Sci 2023; 30:103717. [PMID: 37483838 PMCID: PMC10359945 DOI: 10.1016/j.sjbs.2023.103717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/13/2023] [Accepted: 06/23/2023] [Indexed: 07/25/2023] Open
Abstract
Worldwide, hepatocellular carcinoma (HCC) is considered the sixth most prevalent cancer and ranked third in causes leading to death. Pterostilbene (PTE), a dimethylated analog of resveratrol, is a phytochemical found in fruits such as blueberries and grapes, and is known for its anticancer effect. The current study intended to investigate the effect of PTE on HepG2 cells. Cell viability, colony-forming potential, lipid peroxidation, catalase enzyme (CAT), superoxide dismutase (SOD), and caspase 3 activities, histone release, and expression levels of mTOR, S6K1, p53, and STAT3 proteins were assessed in PTE-treated HepG2 cells. In addition, the cellular and ultrastructural alterations were evaluated by light and transmission electron microscopy. PTE induced a significant reduction in HepG2 viability in a dose-dependent manner (IC50 of PTE = 74 ± 6 μM), accompanied by a decrease in colony formation potential. PTE-treated cancer cells exhibited a decrease in lipid peroxidation and CAT activity, and an increase in histone release, caspase-3, and SOD activities. Ultrastructurally, PTE-treated cells exhibited notable cell shrinkage, reduced number of filopodia, increased vacuolization, apoptotic bodies, accumulation of lipid droplets, enlarged mitochondria, dilated endoplasmic reticulum, pyknotic nuclei, and cellular fragmentation. mTOR, S6K1, and STAT3 levels were downregulated, however p53 level was modulated in PTE-treated cells. The anticancer potential of PTE might be related to its ability to alter the ultrastructure morphology, reduce mitotic activity, and modulate some key protein required for cell proliferation, suggesting its potential to trigger cancer cells towards apoptosis.
Collapse
Affiliation(s)
- Mahmoud I. Khalil
- Department of Biological Sciences, Faculty of Science, Beirut Arab University, Lebanon
- Molecular Biology Unit, Zoology Department, Faculty of Science, Alexandria University, Egypt
| | - Alaa F. Agamy
- Molecular Biology Unit, Zoology Department, Faculty of Science, Alexandria University, Egypt
| | | | - Ahmed S. Sultan
- Biochemistry Department, Faculty of Science, Alexandria University, Egypt
| | | |
Collapse
|
6
|
Li W, Xu X. Advances in mitophagy and mitochondrial apoptosis pathway-related drugs in glioblastoma treatment. Front Pharmacol 2023; 14:1211719. [PMID: 37456742 PMCID: PMC10347406 DOI: 10.3389/fphar.2023.1211719] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/23/2023] [Indexed: 07/18/2023] Open
Abstract
Glioblastoma (GBM) is the most common malignant tumor of the central nervous system (CNS). It is a leading cause of death among patients with intracranial malignant tumors. GBM exhibits intra- and inter-tumor heterogeneity, leading to drug resistance and eventual tumor recurrence. Conventional treatments for GBM include maximum surgical resection of glioma tissue, temozolomide administration, and radiotherapy, but these methods do not effectively halt cancer progression. Therefore, development of novel methods for the treatment of GBM and identification of new therapeutic targets are urgently required. In recent years, studies have shown that drugs related to mitophagy and mitochondrial apoptosis pathways can promote the death of glioblastoma cells by inducing mitochondrial damage, impairing adenosine triphosphate (ATP) synthesis, and depleting large amounts of ATP. Some studies have also shown that modern nano-drug delivery technology targeting mitochondria can achieve better drug release and deeper tissue penetration, suggesting that mitochondria could be a new target for intervention and therapy. The combination of drugs targeting mitochondrial apoptosis and autophagy pathways with nanotechnology is a promising novel approach for treating GBM.This article reviews the current status of drug therapy for GBM, drugs targeting mitophagy and mitochondrial apoptosis pathways, the potential of mitochondria as a new target for GBM treatment, the latest developments pertaining to GBM treatment, and promising directions for future research.
Collapse
|
7
|
Alzahrani B, Elderdery AY, Alzerwi NAN, Alsrhani A, Alsultan A, Rayzah M, Idrees B, Rayzah F, Baksh Y, Alzahrani AM, Subbiah SK, Mok PL. Pluronic-F-127-Passivated SnO 2 Nanoparticles Derived by Using Polygonum cuspidatum Root Extract: Synthesis, Characterization, and Anticancer Properties. PLANTS (BASEL, SWITZERLAND) 2023; 12:plants12091760. [PMID: 37176818 PMCID: PMC10181209 DOI: 10.3390/plants12091760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/01/2023] [Accepted: 04/07/2023] [Indexed: 05/15/2023]
Abstract
Nanotechnology has emerged as the most popular research topic with revolutionary applications across all scientific disciplines. Tin oxide (SnO2) has been gaining considerable attention lately owing to its intriguing features, which can be enhanced by its synthesis in the nanoscale range. The establishment of a cost-efficient and ecologically friendly procedure for its production is the result of growing concerns about human well-being. The novelty and significance of this study lie in the fact that the synthesized SnO2 nanoparticles have been tailored to have specific properties, such as size and morphology. These properties are crucial for their applications. Moreover, this study provides insights into the synthesis process of SnO2 nanoparticles, which can be useful for developing efficient and cost-effective methods for large-scale production. In the current study, green Pluronic-coated SnO2 nanoparticles (NPs) utilizing the root extracts of Polygonum cuspidatum have been formulated and characterized by several methods such as UV-visible, Fourier transform infrared spectroscopy (FTIR), energy dispersive X-ray (EDAX), transmission electron microscope (TEM), field emission-scanning electron microscope (FE-SEM), X-ray diffraction (XRD), photoluminescence (PL), and dynamic light scattering (DLS) studies. The crystallite size of SnO2 NPs was estimated to be 45 nm, and a tetragonal rutile-type crystalline structure was observed. FESEM analysis validated the NPs' spherical structure. The cytotoxic potential of the NPs against HepG2 cells was assessed using the in vitro MTT assay. The apoptotic efficiency of the NPs was evaluated using a dual-staining approach. The NPs revealed substantial cytotoxic effects against HepG2 cells but failed to exhibit cytotoxicity in different liver cell lines. Furthermore, dual staining and flow cytometry studies revealed higher apoptosis in NP-treated HepG2 cells. Nanoparticle treatment also inhibited the cell cycle at G0/G1 stage. It increased oxidative stress and promoted apoptosis by encouraging pro-apoptotic protein expression in HepG2 cells. NP treatment effectively blocked the PI3K/Akt/mTOR axis in HepG2 cells. Thus, green Pluronic-F-127-coated SnO2 NPs exhibits enormous efficiency to be utilized as an talented anticancer agent.
Collapse
Affiliation(s)
- Badr Alzahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Saudi Arabia
| | - Abozer Y Elderdery
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Saudi Arabia
| | - Nasser A N Alzerwi
- Department of Surgery, College of Medicine, Majmaah University, P.O. Box 66, Al-Majmaah 11952, Saudi Arabia
| | - Abdullah Alsrhani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Saudi Arabia
| | - Afnan Alsultan
- Department of Surgery, King Saud Medical City, Riyadh 12746, Saudi Arabia
| | - Musaed Rayzah
- Department of Surgery, College of Medicine, Majmaah University, P.O. Box 66, Al-Majmaah 11952, Saudi Arabia
| | - Bandar Idrees
- Department of Surgery, Prince Sultan Military Medical City, P.O. Box 7897, Riyadh 11159, Saudi Arabia
| | - Fares Rayzah
- Aseer Central Hospital, Abha 62523, Saudi Arabia
| | - Yaser Baksh
- Iman General Hospital, Riyadh 12684, Saudi Arabia
| | - Ahmed M Alzahrani
- Department of Surgery, College of Medicine, Majmaah University, P.O. Box 66, Al-Majmaah 11952, Saudi Arabia
| | - Suresh K Subbiah
- Centre for Materials Engineering and Regenerative Medicine, Bharath Institute of Higher Education and Research, Chennai 600073, India
| | - Pooi Ling Mok
- Department of Biomedical Science, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| |
Collapse
|
8
|
Tian L, Jia Z, Yan Y, Jia Q, Shi W, Cui S, Chen H, Han Y, Zhao X, He K. Low-dose of caffeine alleviates high altitude pulmonary edema via regulating mitochondrial quality control process in AT1 cells. Front Pharmacol 2023; 14:1155414. [PMID: 37081967 PMCID: PMC10110878 DOI: 10.3389/fphar.2023.1155414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/23/2023] [Indexed: 04/07/2023] Open
Abstract
Backgrounds: High-altitude pulmonary edema (HAPE) is a life-threatening disease without effective drugs. Caffeine is a small molecule compound with antioxidant biological activity used to treat respiratory distress syndrome. However, it is unclear whether caffeine plays a role in alleviating HAPE.Methods: We combined a series of biological experiments and label-free quantitative proteomics analysis to detect the effect of caffeine on treating HAPE and explore its mechanism in vivo and in vitro.Results: Dry and wet weight ratio and HE staining of pulmonary tissues showed that the HAPE model was constructed successfully, and caffeine relieved pulmonary edema. The proteomic results of mice lungs indicated that regulating mitochondria might be the mechanism by which caffeine reduced HAPE. We found that caffeine blocked the reduction of ATP production and oxygen consumption rate, decreased ROS accumulation, and stabilized mitochondrial membrane potential to protect AT1 cells from oxidative stress damage under hypoxia. Caffeine promoted the PINK1/parkin-dependent mitophagy and enhanced mitochondrial fission to maintain the mitochondria quality control process.Conclusion: Low-dose of caffeine alleviated HAPE by promoting PINK1/parkin-dependent mitophagy and mitochondrial fission to control the mitochondria quality. Therefore, caffeine could be a potential treatment for HAPE.
Collapse
Affiliation(s)
- Liuyang Tian
- School of Medicine, Nankai University, Tianjin, China
- Medical Big Data Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
- National Engineering Research Center for Medical Big Data Application Technology, Chinese PLA General Hospital, Beijing, China
| | - Zhilong Jia
- National Engineering Research Center for Medical Big Data Application Technology, Chinese PLA General Hospital, Beijing, China
- Center for Artificial Intelligence in Medicine, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
- *Correspondence: Zhilong Jia, ; Xiaojing Zhao, ; Kunlun He,
| | - Yan Yan
- Research Center for Translational Medicine, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| | - Qian Jia
- National Engineering Research Center for Medical Big Data Application Technology, Chinese PLA General Hospital, Beijing, China
- Research Center for Translational Medicine, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| | - Wenjie Shi
- Technical Research Centre for Prevention and Control of Birth Defects, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| | - Saijia Cui
- Research Center for Translational Medicine, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| | - Huining Chen
- Research Center for Translational Medicine, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| | - Yang Han
- Medical Big Data Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
- National Engineering Research Center for Medical Big Data Application Technology, Chinese PLA General Hospital, Beijing, China
- Research Center for Translational Medicine, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| | - Xiaojing Zhao
- National Engineering Research Center for Medical Big Data Application Technology, Chinese PLA General Hospital, Beijing, China
- Research Center for Translational Medicine, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
- *Correspondence: Zhilong Jia, ; Xiaojing Zhao, ; Kunlun He,
| | - Kunlun He
- School of Medicine, Nankai University, Tianjin, China
- Medical Big Data Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
- National Engineering Research Center for Medical Big Data Application Technology, Chinese PLA General Hospital, Beijing, China
- *Correspondence: Zhilong Jia, ; Xiaojing Zhao, ; Kunlun He,
| |
Collapse
|
9
|
Cheng B, Wang Y, Ayanlaja AA, Zhu J, Kambey PA, Qiu Z, Zhang C, Hu W. Glutathione S-Transferases S1, Z1 and A1 Serve as Prognostic Factors in Glioblastoma and Promote Drug Resistance through Antioxidant Pathways. Cells 2022; 11:3232. [PMID: 36291099 PMCID: PMC9600210 DOI: 10.3390/cells11203232] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 09/29/2022] [Accepted: 10/10/2022] [Indexed: 11/17/2022] Open
Abstract
The glutathione S-transferase (GST) family of detoxification enzymes can regulate the malignant progression and drug resistance of various tumors. Hematopoietic prostaglandin D synthase (HPGDS, also referred to as GSTS1), GSTZ1, and GSTA1 are abnormally expressed in multiple cancers, but their roles in tumorigenesis and development remain unclear. In this study, we used bioinformatics tools to analyze the connections of HPGDS, GSTZ1, and GSTA1 to a variety of tumors in genetic databases. Then, we performed biochemical assays in GBM cell lines to investigate the involvement of HPGDS in proliferation and drug resistance. We found that HPGDS, GSTZ1, and GSTA1 are abnormally expressed in a variety of tumors and are associated with prognoses. The expression level of HPGDS was significantly positively correlated with the grade of glioma, and high levels of HPGDS predicted a poor prognosis. Inhibiting HPGDS significantly downregulated GBM proliferation and reduced resistance to temozolomide by disrupting the cellular redox balance and inhibiting the activation of JNK signaling. In conclusion, this study suggested that HPGDS, GSTZ1, and GSTA1 are related to the progression of multiple tumors, and HPGDS is expected to be a prognostic factor in GBM.
Collapse
Affiliation(s)
- Bo Cheng
- Department of Psychiatry, The Affiliated Xuzhou Eastern Hospital of Xuzhou Medical University, Tongshan Road 379, Xuzhou 221000, China
- The Key Lab of Psychiatry, Xuzhou Medical University, Tongshan Road 209, Xuzhou 221000, China
| | - Yu Wang
- Department of Geriatric Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Guangzhou Road 264, Nanjing 220029, China
| | - Abiola Abdulrahman Ayanlaja
- Department of Neurology, Johns Hopkins University School of Medicine, 201 N Broadway, Baltimore, MD 21287, USA
| | - Jing Zhu
- Department of Psychiatry, The Affiliated Xuzhou Eastern Hospital of Xuzhou Medical University, Tongshan Road 379, Xuzhou 221000, China
| | - Piniel Alphayo Kambey
- Department of Neurobiology and Cell Biology, Xuzhou Medical University, Tongshan Road 209, Xuzhou 221000, China
| | - Ziqiang Qiu
- Department of Psychiatry, The Affiliated Xuzhou Eastern Hospital of Xuzhou Medical University, Tongshan Road 379, Xuzhou 221000, China
| | - Caiyi Zhang
- Department of Psychiatry, The Affiliated Xuzhou Eastern Hospital of Xuzhou Medical University, Tongshan Road 379, Xuzhou 221000, China
- The Key Lab of Psychiatry, Xuzhou Medical University, Tongshan Road 209, Xuzhou 221000, China
| | - Wei Hu
- Department of Psychiatry, The Affiliated Xuzhou Eastern Hospital of Xuzhou Medical University, Tongshan Road 379, Xuzhou 221000, China
| |
Collapse
|
10
|
GPX8 as a Novel Prognostic Factor and Potential Therapeutic Target in Primary Glioma. J Immunol Res 2022; 2022:8025055. [PMID: 36052280 PMCID: PMC9427289 DOI: 10.1155/2022/8025055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/26/2022] [Indexed: 11/17/2022] Open
Abstract
One of the most prevalent malignant primary brain tumors is primary glioma. Although glutathione peroxidase 8 (GPX8) is intimately associated with carcinogenesis, its function in primary gliomas has not yet been thoroughly understood. Here, we leveraged Chinese Glioma Genome Atlas (CGGA), The Cancer Genome Atlas (TCGA), and Genotype-Tissue Expression (GTEx) database to investigate the association between GPX8 and overall survival (OS) of patients with primary gliomas, and our results showed that GPX8 expression was negatively correlated with OS. Moreover, the expression of GPX8 is significantly lower in normal tissue when compared to glioma tissue. According to results of univariate and multivariate analysis from CGGA using R studio, GPX8 is a valuable primary glioma prognostic indicator. Interestingly, high GPX8 expression is correlated positively with the hedgehog and kras signaling pathways and negatively with G2 checkpoint, apoptosis, reactive oxygen species (ROS) pathway, and interferon gamma pathway, which could be beneficial for the proliferation of glioma cells. Furthermore, GPX8 knockdown caused G1 cell cycle arrest, increased cell death, and reduced colony formation in U87MG and U118MG cells. In conclusion, GPX8 is a promising therapeutic target and meaningful prognostic biomarker of primary glioma.
Collapse
|