1
|
Wang Y, Gao P, Wu Z, Jiang B, Wang Y, He Z, Zhao B, Tian X, Gao H, Cai L, Li W. Exploring the therapeutic potential of Chinese herbs on comorbid type 2 diabetes mellitus and Parkinson's disease: A mechanistic study. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119095. [PMID: 39537117 DOI: 10.1016/j.jep.2024.119095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/12/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Type 2 diabetes mellitus (T2DM) and Parkinson's disease (PD) are chronic conditions that affect the aging population, with increasing prevalence globally. The rising prevalence of comorbidity between these conditions, driven by demographic shifts, severely impacts the quality of life of patients, posing a significant burden on healthcare resources. Chinese herbal medicine has been used to treat T2DM and PD for millennia. Pharmacological studies have demonstrated that medicinal herbs effectively lower blood glucose levels and exert neuroprotective effects, suggesting their potential as adjunctive therapy for concurrent management of T2DM and PD. AIM OF THE STUDY To elucidate the shared mechanisms underlying T2DM and PD, particularly focusing on the potential mechanisms by which medicinal herbs (including herbal formulas, single herbs, and active compounds) may treat these diseases, to provide valuable insights for developing therapeutics targeting comorbid T2DM and PD. MATERIALS AND METHODS Studies exploring the mechanisms underlying T2DM and PD, as well as the treatment of these conditions with medicinal herbs, were extracted from several electronic databases, including PubMed, Web of Science, Google Scholar, and China National Knowledge Infrastructure (CNKI). RESULTS Numerous studies have shown that inflammation, oxidative stress, insulin resistance, impaired autophagy, gut microbiota dysbiosis, and ferroptosis are shared mechanisms underlying T2DM and PD mediated through the NLRP3 inflammasome, NF-κB, MAPK, Keap1/Nrf2/ARE, PI3K/AKT, AMPK/SIRT1, and System XC--GSH-GPX4 signaling pathways. Thirty-four medicinal herbs, including 2 herbal formulas, 4 single herbs, and 28 active compounds, have been reported to potentially exert anti-T2DM and anti-PD effects by targeting these shared mechanisms. CONCLUSIONS Traditional Chinese medicine effectively combats T2DM and PD through shared pathological mechanisms, highlighting their potential for application in treating these comorbid conditions.
Collapse
Affiliation(s)
- Yan Wang
- Clinical College of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China; Encephalopathy Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Pengpeng Gao
- Department of Preventive Treatment, Ningxia Integrated Chinese and Western Medicine Hospital, Yinchuan, 750004, China
| | - Zicong Wu
- Encephalopathy Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Bing Jiang
- Department of Integrated Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Yanru Wang
- Gansu University Key Laboratory for Molecular Medicine & Chinese Medicine Prevention and Treatment of Major Diseases, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Zhaxicao He
- Clinical College of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Bing Zhao
- Clinical College of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Xinyun Tian
- Encephalopathy Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Han Gao
- Encephalopathy Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Li Cai
- Encephalopathy Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China.
| | - Wentao Li
- Encephalopathy Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China.
| |
Collapse
|
2
|
Zheng H, Li L, Wang D, Zhang S, Li W, Cheng M, Ge C, Chen J, Qiang Y, Chen F, Yu Y. FoxO is required for neoblast differentiation during planarian regeneration. Int J Biol Macromol 2024; 288:138729. [PMID: 39672403 DOI: 10.1016/j.ijbiomac.2024.138729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 12/15/2024]
Abstract
Stem cells are of great importance in the maintenance and regeneration of tissues, with Forkhead box O (FoxO) proteins emerging as pivotal regulators of their functions. However, the precise impact of FoxO proteins on stem cell behavior within regenerative environments remains ambiguous. Planarians, renowned for their abundance of adult stem cells (neoblasts), serve as an excellent model for investigating the dynamics of stem cells during regeneration. In this study, we identified DjfoxO, a conserved foxO gene in the planarian Dugesia japonica, and demonstrated its expression in neoblasts, with elevated levels detected in the regenerative blastema during the regeneration process. Using a FoxO inhibitor (AS1842856) together with RNA interference techniques, we demonstrated that inhibition of FoxO signaling in planarians hinders the regeneration of missing tissues, including the central nervous system, eyespots, anterior intestinal branches, and pharynx. It is noteworthy that the knockdown of DjfoxO does not significantly affect the mitotic activity of neoblasts. Conversely, it impedes the production of lineage-specific progenitors, potentially via modulation of the Erk pathway. These findings elucidate the instructive function of FoxO signaling in regulating stem cell differentiation and provide valuable insights into its potential for improving stem cell-based regenerative therapies.
Collapse
Affiliation(s)
- Hanxue Zheng
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Linfeng Li
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Du Wang
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Shengchao Zhang
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Wenhui Li
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Mengdi Cheng
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Cui Ge
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Jiayi Chen
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Yanmei Qiang
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Fulin Chen
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China; Provincial Key Laboratory of Biotechnology of Shaanxi, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, China
| | - Yuan Yu
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China; Provincial Key Laboratory of Biotechnology of Shaanxi, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, China.
| |
Collapse
|
3
|
Li H, Ye Z, Zheng G, Su Z. Polysaccharides targeting autophagy to alleviate metabolic syndrome. Int J Biol Macromol 2024; 283:137393. [PMID: 39521230 DOI: 10.1016/j.ijbiomac.2024.137393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 10/25/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Metabolic syndrome is a prevalent non-communicable disease characterized by central obesity, insulin resistance, hypertension, hyperglycemia, and hyperlipidemia. Epidemiological statistics indicate that one-third of the world's population is affected by metabolic syndrome. Unfortunately, owing to complicated pathogenesis and limited pharmacological options, the growing prevalence of metabolic syndrome threatens human health worldwide. Autophagy is an intracellular degradation mechanism that involves the degradation of unfolded or aggregated proteins and damaged cellular organelles, thereby maintaining metabolic homeostasis. Increasing evidence indicates that dysfunctional autophagy is closely associated with the development of metabolic syndrome, making it an attractive therapeutic target. Furthermore, a growing number of plant-derived polysaccharides have been shown to regulate autophagy, thereby alleviating metabolic syndrome, such as Astragalus polysaccharides, Laminaria japonica polysaccharides, Ganoderma lucidum polysaccharides and Lycium barbarum polysaccharides. In this review, we summarize recent advances in the discovery of autophagy modulators of plant polysaccharides for the treatment of metabolic syndrome, with the aim of providing precursor compounds for the development of new therapeutic agents. Additionally, we look forward to seeing more diseases being treated with plant polysaccharides by regulating autophagy, as well as the discovery of more intricate mechanisms that govern autophagy.
Collapse
Affiliation(s)
- Hongxia Li
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Pharmacology of Traditional Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Diseases, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zeting Ye
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Pharmacology of Traditional Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Diseases, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guangjuan Zheng
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Pharmacology of Traditional Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Diseases, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Zuqing Su
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Pharmacology of Traditional Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Diseases, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
4
|
Hua W, Xie L, Dong C, Yang G, Chi S, Xu Z, Yang C, Wang H, Wu X. Procyanidin C1 ameliorates acidic pH stress induced nucleus pulposus degeneration through SIRT3/FOXO3-mediated mitochondrial dynamics. J Transl Med 2024; 22:1071. [PMID: 39605029 PMCID: PMC11600718 DOI: 10.1186/s12967-024-05805-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024] Open
Abstract
Intervertebral disc degeneration (IVDD) is a common cause of low back pain. Procyanidin C1 (PCC1) has been demonstrated to exert a protective effect on nucleus pulposus (NP) cells, and therefore, plays a critical role in the prevention and therapy of IVDD. Clarifying the pathophysiological characteristics and molecular mechanisms of IVDD may be helpful in establishing novel preventive and therapeutic strategies. This study aimed to investigate the probable mechanisms underlying the protection against acidic pH stress induced human NP cell injury. In vitro, acidic pH stress induced degeneration, mitochondrial dynamics imbalance, mitophagy, and mitochondria-mediated apoptosis in NP cells, all of which were ameliorated by PCC1. Autophagy inhibition partially eliminated the protective effects of PCC1 on mitochondrial homeostasis in NP cells. Moreover, PCC1 activated the sirtuin 3 (SIRT3)/forkhead box O3 (FOXO3) signaling pathway, a pivotal signaling pathway involved in the regulation of mitochondrial homeostasis in NP cells. In vivo, PCC1 ameliorated IVDD in a rat model and preserved the extracellular matrix of NP cells. Consequently, the protective effects of PCC1 on NP cells may inhibit IVDD progression via regulation of the SIRT3/FOXO3 signaling pathway. Therefore, regulation of the SIRT3/FOXO3 signaling pathway may be a novel preventive and therapeutic strategy for IVDD.
Collapse
Affiliation(s)
- Wenbin Hua
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lin Xie
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chenpeng Dong
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Guoyu Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shouyuan Chi
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhiqiang Xu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Cao Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Huiwen Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Xinghuo Wu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
5
|
Ruan Y, Xue Y, Zhang P, Jia J. Acetylation of FOXO1 is involved in cadmium-induced rat kidney injury via mediating autophagosome-lysosome fusion blockade and autophagy inhibition. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 287:117253. [PMID: 39536554 DOI: 10.1016/j.ecoenv.2024.117253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 10/14/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
Cadmium (Cd), a potentially toxic elements, has the potential to cause harm to the kidneys. Studies has demonstrated that autophagosome-lysosome fusion blockade and consequent autophagy inhibition is related to Cd-induced kidney injury. Studies indicate that acetylation of forkhead box protein O1 (FOXO1) as a transcriptional factor of lysosomal and autophagy genes, but its roles in Cd-exposed kidney tissues remains unclear till now. Therefore, the present study was conducted to elucidate this issue. Data found that Cd enhances the acetylation level of FOXO1 and inhibits the expression level of silent information regulator 1 (Sirt1, deacetylase of FOXO1). Pharmacological activation of Sirt1 (SRT2104 treatment) decreases Cd-increased acetylation level of FOXO1, enhances Cd-inhibited transcription level of Ras-related protein 7 (Rab7), restores Cd-blocked fusion of autophagosome and lysosome, and alleviates Cd-induced autophagy inhibition. Moreover, data corroborated that inhibiting the acetylation level of FOXO1 is conductive to mitigating Cd-induced kidney injury. Collectively, these results demonstrate that acetylation of FOXO1 mediates the autophagosome-lysosome fusion blockade and autophagy inhibition during Cd-induced kidney injury, while regulating the acetylation level of FOXO1 may be a potential mechanism of treating nephrotoxicity after Cd exposure.
Collapse
Affiliation(s)
- Yingxin Ruan
- Department of Nephrology, General Hospital of Tianjin Medical University, Tianjin 300052, PR China
| | - Yang Xue
- Department of Nephrology, General Hospital of Tianjin Medical University, Tianjin 300052, PR China
| | - Pengyu Zhang
- Department of Blood Transfusion, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, PR China.
| | - Junya Jia
- Department of Nephrology, General Hospital of Tianjin Medical University, Tianjin 300052, PR China.
| |
Collapse
|
6
|
Fan C, Chen G, Reiter RJ, Bai Y, Zheng T, Fan L. Glutathione inhibits lung cancer development by reducing interleukin-6 expression and reversing the Warburg effect. Mitochondrion 2024; 79:101953. [PMID: 39214486 DOI: 10.1016/j.mito.2024.101953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Reduced glutathione (GSH) is widely used as an antioxidant in clinical practice, but whether GSH affects the development of early lung cancer remains unclear. Herein, we investigated the mechanism underlying the anticancer effect of GSH in patients with pulmonary nodules. Thirty patients with pulmonary nodules were treated with GSH intravenously for 10 days at a dose of 1.8 g/d, followed by oral administration of the drug at a dose of 0.4 g three times daily for 6 months. The results showed that GSH treatment promoted nodule absorption and reduced the IL-6 level in the peripheral blood of the patients. GSH reduced IL-6 expression in inflammatory BEAS-2B and lung cancer cells and inhibited the proliferation of lung cancer cell lines in vitro. In addition, GSH reduced IL-6 expression by decreasing ROS via down-regulating PI3K/AKT/FoxO pathways. Finally, GSH reversed the Warburg effect, restored mitochondrial function, and reduced the IL-6 expression via PI3K/AKT/FoxO pathways. The in vivo experiment confirmed that GSH inhibited lung cancer growth, improved mitochondrial function, and reduced the IL-6 expression by regulating key enzymes via the PI3K/AKT/FoxO pathway. In conclusion, we uncovered that GSH exerts an unprecedentedly potent anti-cancer effect to prevent the transformation of lung nodules to lung cancer by improving the mitochondrial function and suppressing inflammation via PI3K/AKT/FoxO pathway. This investigation innovatively positions GSH as a potentially safe and efficacious old drug with new uses, inhibiting inflammation and early lung cancer. The use of the drug offers a promising preventive strategy when administered during the early stages of lung cancer.
Collapse
Affiliation(s)
- Chenchen Fan
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Institute of Energy Metabolism and Health, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Guojie Chen
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Institute of Energy Metabolism and Health, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Yidong Bai
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Tiansheng Zheng
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Institute of Energy Metabolism and Health, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Lihong Fan
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Institute of Energy Metabolism and Health, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| |
Collapse
|
7
|
Liu Y, Liu Q, Shang H, Li J, Chai H, Wang K, Guo Z, Luo T, Liu S, Liu Y, Wang X, Zhang H, Wu C, Song SJ, Yang J. Potential application of natural compounds in ischaemic stroke: Focusing on the mechanisms underlying "lysosomocentric" dysfunction of the autophagy-lysosomal pathway. Pharmacol Ther 2024; 263:108721. [PMID: 39284368 DOI: 10.1016/j.pharmthera.2024.108721] [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: 03/27/2024] [Revised: 07/06/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
Ischaemic stroke (IS) is the second leading cause of death and a major cause of disability worldwide. Currently, the clinical management of IS still depends on restoring blood flow via pharmacological thrombolysis or mechanical thrombectomy, with accompanying disadvantages of narrow therapeutic time window and risk of haemorrhagic transformation. Thus, novel pathophysiological mechanisms and targeted therapeutic candidates are urgently needed. The autophagy-lysosomal pathway (ALP), as a dynamic cellular lysosome-based degradative process, has been comprehensively studied in recent decades, including its upstream regulatory mechanisms and its role in mediating neuronal fate after IS. Importantly, increasing evidence has shown that IS can lead to lysosomal dysfunction, such as lysosomal membrane permeabilization, impaired lysosomal acidity, lysosomal storage disorder, and dysfunctional lysosomal ion homeostasis, which are involved in the IS-mediated defects in ALP function. There is tightly regulated crosstalk between transcription factor EB (TFEB), mammalian target of rapamycin (mTOR) and lysosomal function, but their relationship remains to be systematically summarized. Notably, a growing body of evidence emphasizes the benefits of naturally derived compounds in the treatment of IS via modulation of ALP function. However, little is known about the roles of natural compounds as modulators of lysosomes in the treatment of IS. Therefore, in this context, we provide an overview of the current understanding of the mechanisms underlying IS-mediated ALP dysfunction, from a lysosomal perspective. We also provide an update on the effect of natural compounds on IS, according to their chemical structural types, in different experimental stroke models, cerebral regions and cell types, with a primary focus on lysosomes and autophagy initiation. This review aims to highlight the therapeutic potential of natural compounds that target lysosomal and ALP function for IS treatment.
Collapse
Affiliation(s)
- Yueyang Liu
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Qingbo Liu
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Hanxiao Shang
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Jichong Li
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - He Chai
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Kaixuan Wang
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Zhenkun Guo
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Tianyu Luo
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Shiqi Liu
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Yan Liu
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Xuemei Wang
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Hangyi Zhang
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Chunfu Wu
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Shao-Jiang Song
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China.
| | - Jingyu Yang
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China.
| |
Collapse
|
8
|
Lee DH, Lee HJ, Yang G, Kim DY, Kim JU, Yook TH, Lee JH, Kim HJ. A novel treatment strategy targeting cellular pathways with natural products to alleviate sarcopenia. Phytother Res 2024; 38:5033-5051. [PMID: 39099170 DOI: 10.1002/ptr.8301] [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: 12/26/2023] [Revised: 07/08/2024] [Accepted: 07/12/2024] [Indexed: 08/06/2024]
Abstract
Sarcopenia is a condition marked by a significant reduction in muscle mass and strength, primarily due to the aging process, which critically impacts muscle protein dynamics, metabolic functions, and overall physical functionality. This condition leads to increased body fat and reduced daily activity, contributing to severe health issues and a lower quality of life among the elderly. Recognized in the ICD-10-CM only in 2016, sarcopenia lacks definitive treatment options despite its growing prevalence and substantial social and economic implications. Given the aging global population, addressing sarcopenia has become increasingly relevant and necessary. The primary causes include aging, cachexia, diabetes, and nutritional deficiencies, leading to imbalances in protein synthesis and degradation, mitochondrial dysfunction, and hormonal changes. Exercise remains the most effective intervention, but it is often impractical for individuals with limited mobility, and pharmacological options such as anabolic steroids and myostatin inhibitors are not FDA-approved and are still under investigation. This review is crucial as it examines the potential of natural products as a novel treatment strategy for sarcopenia, targeting multiple mechanisms involved in its pathogenesis. By exploring natural products' multi-targeted effects, this study aims to provide innovative and practical solutions for sarcopenia management. Therefore, this review indicates significant improvements in muscle mass and function with the use of specific natural compounds, suggesting promising alternatives for those unable to engage in regular physical activity.
Collapse
Affiliation(s)
- Da Hee Lee
- College of Korean Medicine, Woosuk University, Jeonju-si, Republic of Korea
| | - Hye Jin Lee
- College of Korean Medicine, Woosuk University, Jeonju-si, Republic of Korea
| | - Gabsik Yang
- College of Korean Medicine, Woosuk University, Jeonju-si, Republic of Korea
| | - Dae Yong Kim
- College of Korean Medicine, Woosuk University, Jeonju-si, Republic of Korea
| | - Jong Uk Kim
- College of Korean Medicine, Woosuk University, Jeonju-si, Republic of Korea
| | - Tae Han Yook
- College of Korean Medicine, Woosuk University, Jeonju-si, Republic of Korea
| | - Jun Ho Lee
- College of Korean Medicine, Woosuk University, Jeonju-si, Republic of Korea
- Da Capo Co., Ltd., Jeonju-si, Republic of Korea
| | - Hong Jun Kim
- College of Korean Medicine, Woosuk University, Jeonju-si, Republic of Korea
| |
Collapse
|
9
|
Lv B, Dion WA, Yang H, Xun J, Kim DH, Zhu B, Tan JX. A TBK1-independent primordial function of STING in lysosomal biogenesis. Mol Cell 2024; 84:3979-3996.e9. [PMID: 39423796 PMCID: PMC11490688 DOI: 10.1016/j.molcel.2024.08.026] [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: 03/26/2024] [Revised: 07/18/2024] [Accepted: 08/22/2024] [Indexed: 10/21/2024]
Abstract
Stimulator of interferon genes (STING) is activated in many pathophysiological conditions, leading to TBK1-dependent interferon production in higher organisms. However, primordial functions of STING independent of TBK1 are poorly understood. Here, through proteomics and bioinformatics approaches, we identify lysosomal biogenesis as an unexpected function of STING. Transcription factor EB (TFEB), an evolutionarily conserved regulator of lysosomal biogenesis and host defense, is activated by STING from multiple species, including humans, mice, and frogs. STING-mediated TFEB activation is independent of TBK1, but it requires STING trafficking and its conserved proton channel. GABARAP lipidation, stimulated by the channel of STING, is key for STING-dependent TFEB activation. STING stimulates global upregulation of TFEB-target genes, mediating lysosomal biogenesis and autophagy. TFEB supports cell survival during chronic sterile STING activation, a common condition in aging and age-related diseases. These results reveal a primordial function of STING in the biogenesis of lysosomes, essential organelles in immunity and cellular stress resistance.
Collapse
Affiliation(s)
- Bo Lv
- Aging Institute, University of Pittsburgh School of Medicine/University of Pittsburgh Medical Center, Pittsburgh, PA 15219, USA; Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - William A Dion
- Aging Institute, University of Pittsburgh School of Medicine/University of Pittsburgh Medical Center, Pittsburgh, PA 15219, USA
| | - Haoxiang Yang
- Aging Institute, University of Pittsburgh School of Medicine/University of Pittsburgh Medical Center, Pittsburgh, PA 15219, USA; Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Jinrui Xun
- Aging Institute, University of Pittsburgh School of Medicine/University of Pittsburgh Medical Center, Pittsburgh, PA 15219, USA; Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Do-Hyung Kim
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Bokai Zhu
- Aging Institute, University of Pittsburgh School of Medicine/University of Pittsburgh Medical Center, Pittsburgh, PA 15219, USA; Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Jay Xiaojun Tan
- Aging Institute, University of Pittsburgh School of Medicine/University of Pittsburgh Medical Center, Pittsburgh, PA 15219, USA; Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.
| |
Collapse
|
10
|
Martin TG, Hunt DR, Langer SJ, Tan Y, Ebmeier CC, Leinwand LA. Regression of postprandial cardiac hypertrophy in burmese pythons is mediated by FoxO1. Proc Natl Acad Sci U S A 2024; 121:e2408719121. [PMID: 39352930 PMCID: PMC11474088 DOI: 10.1073/pnas.2408719121] [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: 05/01/2024] [Accepted: 08/16/2024] [Indexed: 10/04/2024] Open
Abstract
As ambush-hunting predators that consume large prey after long intervals of fasting, Burmese pythons evolved with unique adaptations for modulating organ structure and function. Among these is cardiac hypertrophy that develops within three days following a meal (Andersen et al., 2005, Secor, 2008), which we previously showed was initiated by circulating growth factors (Riquelme et al., 2011). Postprandial cardiac hypertrophy in pythons also rapidly regresses with subsequent fasting (Secor, 2008); however, the molecular mechanisms that regulate the dynamic cardiac remodeling in pythons during digestion are largely unknown. In this study, we employed a multiomics approach coupled with targeted molecular analyses to examine remodeling of the python ventricular transcriptome and proteome throughout digestion. We found that forkhead box protein O1 (FoxO1) signaling was suppressed prior to hypertrophy development and then activated during regression, which coincided with decreased and then increased expression, respectively, of FoxO1 transcriptional targets involved in proteolysis. To define the molecular mechanistic role of FoxO1 in hypertrophy regression, we used cultured mammalian cardiomyocytes treated with postfed python plasma. Hypertrophy regression both in pythons and in vitro coincided with activation of FoxO1-dependent autophagy; however, the introduction of a FoxO1-specific inhibitor prevented both regression of cell size and autophagy activation. Finally, to determine whether FoxO1 activation could induce regression, we generated an adenovirus expressing a constitutively active FoxO1. FoxO1 activation was sufficient to prevent and reverse postfed plasma-induced hypertrophy, which was partially prevented by autophagy inhibition. Our results indicate that modulation of FoxO1 activity contributes to the dynamic ventricular remodeling in postprandial Burmese pythons.
Collapse
Affiliation(s)
- Thomas G. Martin
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO80309
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO80303
| | - Dakota R. Hunt
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO80303
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO80303
| | - Stephen J. Langer
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO80309
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO80303
| | - Yuxiao Tan
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO80309
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO80303
| | | | - Leslie A. Leinwand
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO80309
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO80303
| |
Collapse
|
11
|
Mundo Rivera VM, Tlacuahuac Juárez JR, Murillo Melo NM, Leyva Garcia N, Magaña JJ, Cordero Martínez J, Jiménez Gutierrez GE. Natural Autophagy Activators to Fight Age-Related Diseases. Cells 2024; 13:1611. [PMID: 39404375 PMCID: PMC11476028 DOI: 10.3390/cells13191611] [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: 08/24/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
The constant increase in the elderly population presents significant challenges in addressing new social, economic, and health problems concerning this population. With respect to health, aging is a primary risk factor for age-related diseases, which are driven by interconnected molecular hallmarks that influence the development of these diseases. One of the main mechanisms that has attracted more attention to aging is autophagy, a catabolic process that removes and recycles damaged or dysfunctional cell components to preserve cell viability. The autophagy process can be induced or deregulated in response to a wide range of internal or external stimuli, such as starvation, oxidative stress, hypoxia, damaged organelles, infectious pathogens, and aging. Natural compounds that promote the stimulation of autophagy regulatory pathways, such as mTOR, FoxO1/3, AMPK, and Sirt1, lead to increased levels of essential proteins such as Beclin-1 and LC3, as well as a decrease in p62. These changes indicate the activation of autophagic flux, which is known to be decreased in cardiovascular diseases, neurodegeneration, and cataracts. The regulated administration of natural compounds offers an adjuvant therapeutic alternative in age-related diseases; however, more experimental evidence is needed to support and confirm these health benefits. Hence, this review aims to highlight the potential benefits of natural compounds in regulating autophagy pathways as an alternative approach to combating age-related diseases.
Collapse
Affiliation(s)
- Vianey M. Mundo Rivera
- Departamento de Bioingeniería, Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Campus Ciudad de México, Mexico City 14380, Mexico; (V.M.M.R.); (N.M.M.M.); (J.J.M.)
| | - José Roberto Tlacuahuac Juárez
- Laboratorio de Bioquímica Farmacológica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico;
| | - Nadia Mireya Murillo Melo
- Departamento de Bioingeniería, Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Campus Ciudad de México, Mexico City 14380, Mexico; (V.M.M.R.); (N.M.M.M.); (J.J.M.)
- Laboratorio de Medicina Genómica, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14389, Mexico;
| | - Norberto Leyva Garcia
- Laboratorio de Medicina Genómica, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14389, Mexico;
| | - Jonathan J. Magaña
- Departamento de Bioingeniería, Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Campus Ciudad de México, Mexico City 14380, Mexico; (V.M.M.R.); (N.M.M.M.); (J.J.M.)
- Laboratorio de Medicina Genómica, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14389, Mexico;
| | - Joaquín Cordero Martínez
- Laboratorio de Bioquímica Farmacológica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico;
| | | |
Collapse
|
12
|
Zhu Y, Zhang C, Yin Q, Xu W, Luo Y, Ou J. FOXO4 suppresses cisplatin resistance of triple-negative breast cancer by inhibiting autophagy. Am J Med Sci 2024:S0002-9629(24)01403-4. [PMID: 39154963 DOI: 10.1016/j.amjms.2024.08.012] [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: 01/09/2024] [Revised: 08/12/2024] [Accepted: 08/12/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND Resistance to chemotherapy containing cisplatin (DDP) is a main challenge in the treatment of triple-negative breast cancer (TNBC). Forkhead box O4 (FOXO4) is frequently downregulated in DDP-resistant cells. However, it is unclear whether FOXO4 down-regulation is related to DDP resistance. Here, we investigated the relationship between FOXO4 and DDP resistance in TNBC. METHODS We established the DDP-resistant cell lines MDA-MB-231/DDP and BT-549/DDP through in vitro selection. CCK-8 and colony formation assays analyzed cell growth. The resistance index was calculated. Cell autophagy was evaluated. Western blotting and qRT-PCR measured protein and gene expression. The binding between FOXO4 and TGF-β1 was determined by the dual-luciferase reporter assay. RESULTS FOXO4 expression was significantly lower in MDA-MB-231/DDP and BT-549/DDP cells. FOXO4 overexpression increased the sensitivity of TNBC cells to DDP. The PI3K class Ⅲ and Beclin-1 levels and LC3-II/LC3-I ratio elevated significantly in DDP-resistant cells. Moreover, the autophagic flux was enhanced in DDP-resistant cells. 3-MA enhanced the sensitivity of TNBC cells to DDP by inhibiting autophagy. Overexpression of FOXO4, treatment with 3-MA, and their combination significantly reduced the drug resistance index. FOXO4 directly targeted TGF-β1. Additionally, TGF-β1 knockdown inhibited autophagy and restored the sensitivity of DDP-resistant cells to DDP. Mechanistically, FOXO4 affected TNBC resistance to DDP by regulating autophagy and TGF-β1. CONCLUSION FOXO4 overexpression, in combination with autophagy inhibitors, can significantly improve the sensitivity of TNBC-resistant cells to DDP. These findings reveal the role and mechanism of FOXO4 in DDP sensitivity and may provide evidence for the development of TNBC therapies.
Collapse
Affiliation(s)
- Yating Zhu
- Department of Pharmacy, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Chenguang Zhang
- Department of Breast Surgery, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Qiuyu Yin
- Department of Breast Surgery, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Wenting Xu
- Department of Breast Surgery, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Yulou Luo
- Department of Breast Surgery, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Jianghua Ou
- Department of Breast Surgery, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi 830011, China.
| |
Collapse
|
13
|
Bolado-Carrancio A, Tapia O, Rodríguez-Rey JC. Ubiquitination Insight from Spinal Muscular Atrophy-From Pathogenesis to Therapy: A Muscle Perspective. Int J Mol Sci 2024; 25:8800. [PMID: 39201486 PMCID: PMC11354275 DOI: 10.3390/ijms25168800] [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: 06/27/2024] [Revised: 08/03/2024] [Accepted: 08/07/2024] [Indexed: 09/02/2024] Open
Abstract
Spinal muscular atrophy (SMA) is one of the most frequent causes of death in childhood. The disease's molecular basis is deletion or mutations in the SMN1 gene, which produces reduced survival motor neuron protein (SMN) levels. As a result, there is spinal motor neuron degeneration and a large increase in muscle atrophy, in which the ubiquitin-proteasome system (UPS) plays a significant role. In humans, a paralogue of SMN1, SMN2 encodes the truncated protein SMNΔ7. Structural differences between SMN and SMNΔ7 affect the interaction of the proteins with UPS and decrease the stability of the truncated protein. SMN loss affects the general ubiquitination process by lowering the levels of UBA1, one of the main enzymes in the ubiquitination process. We discuss how SMN loss affects both SMN stability and the general ubiquitination process, and how the proteins involved in ubiquitination could be used as future targets for SMA treatment.
Collapse
Affiliation(s)
- Alfonso Bolado-Carrancio
- Departamento de Biología Molecular, Facultad de Medicina, Universidad de Cantabria-and Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011 Santander, Spain;
| | - Olga Tapia
- Departamento de Ciencias Médicas Básicas, Instituto de Tecnologías Biomédicas, Universidad de la Laguna, 38200 La Laguna, Spain
| | - José C. Rodríguez-Rey
- Departamento de Biología Molecular, Facultad de Medicina, Universidad de Cantabria-and Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011 Santander, Spain;
| |
Collapse
|
14
|
Zhang R, Farshadyeganeh P, Ohkawara B, Nakajima K, Takeda JI, Ito M, Zhang S, Miyasaka Y, Ohno T, Mori-Yoshimura M, Masuda A, Ohno K. Muscle-specific lack of Gfpt1 triggers ER stress to alleviate misfolded protein accumulation. Dis Model Mech 2024; 17:dmm050768. [PMID: 38903011 PMCID: PMC11554261 DOI: 10.1242/dmm.050768] [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: 02/27/2024] [Accepted: 06/12/2024] [Indexed: 06/22/2024] Open
Abstract
Pathogenic variants in GFPT1, encoding a key enzyme to synthesize UDP-N-acetylglucosamine (UDP-GlcNAc), cause congenital myasthenic syndrome (CMS). We made a knock-in (KI) mouse model carrying a frameshift variant in Gfpt1 exon 9, simulating that found in a patient with CMS. As Gfpt1 exon 9 is exclusively expressed in striated muscles, Gfpt1-KI mice were deficient for Gfpt1 only in skeletal muscles. In Gfpt1-KI mice, (1) UDP-HexNAc, CMP-NeuAc and protein O-GlcNAcylation were reduced in skeletal muscles; (2) aged Gfpt1-KI mice showed poor exercise performance and abnormal neuromuscular junction structures; and (3) markers of the unfolded protein response (UPR) were elevated in skeletal muscles. Denervation-mediated enhancement of endoplasmic reticulum (ER) stress in Gfpt1-KI mice facilitated protein folding, ubiquitin-proteasome degradation and apoptosis, whereas autophagy was not induced and protein aggregates were markedly increased. Lack of autophagy was accounted for by enhanced degradation of FoxO1 by increased Xbp1-s/u proteins. Similarly, in Gfpt1-silenced C2C12 myotubes, ER stress exacerbated protein aggregates and activated apoptosis, but autophagy was attenuated. In both skeletal muscles in Gfpt1-KI mice and Gfpt1-silenced C2C12 myotubes, maladaptive UPR failed to eliminate protein aggregates and provoked apoptosis.
Collapse
Affiliation(s)
- Ruchen Zhang
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Paniz Farshadyeganeh
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Bisei Ohkawara
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Kazuki Nakajima
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu 501-1193, Japan
| | - Jun-ichi Takeda
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Mikako Ito
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Shaochuan Zhang
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Yuki Miyasaka
- Division of Experimental Animals, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Tamio Ohno
- Division of Experimental Animals, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Madoka Mori-Yoshimura
- Department of Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Kodaira 187-8775, Japan
| | - Akio Masuda
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Kinji Ohno
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
- Graduate School of Nutritional Sciences, Nagoya University of Arts and Sciences, Nisshin 470-0196, Japan
| |
Collapse
|
15
|
Zhao Z, Chu Y, Feng A, Zhang S, Wu H, Li Z, Sun M, Zhang L, Chen T, Xu M. STK3 kinase activation inhibits tumor proliferation through FOXO1-TP53INP1/P21 pathway in esophageal squamous cell carcinoma. Cell Oncol (Dordr) 2024; 47:1295-1314. [PMID: 38436783 PMCID: PMC11322239 DOI: 10.1007/s13402-024-00928-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2024] [Indexed: 03/05/2024] Open
Abstract
PURPOSE Esophageal squamous cell carcinoma (ESCC) is an aggressive disease with a poor prognosis, caused by the inactivation of critical cell growth regulators that lead to uncontrolled proliferation and increased malignancy. Although Serine/Threonine Kinase 3 (STK3), also known as Mammalian STE20-like protein kinase 2 (MST2), is a highly conserved kinase of the Hippo pathway, plays a critical role in immunomodulation, organ development, cellular differentiation, and cancer suppression, its phenotype and function in ESCC require further investigation. In this study, we report for the first time on the role of STK3 kinase and its activation condition in ESCC, as well as the mechanism and mediators of kinase activation. METHODS In this study, we investigated the expression and clinical significance of STK3 in ESCC. We first used bioinformatics databases and immunohistochemistry to analyze STK3 expression in the ESCC patient cohort and conducted survival analysis. In vivo, we conducted a tumorigenicity assay using nude mouse models to demonstrate the phenotypes of STK3 kinase. In vitro, we conducted Western blot analysis, qPCR analysis, CO-IP, and immunofluorescence (IF) staining analysis to detect molecule expression, interaction, and distribution. We measured proliferation, migration, and apoptosis abilities in ESCC cells in the experimental groups using CCK-8 and transwell assays, flow cytometry, and EdU staining. We used RNA-seq to identify genes that were differentially expressed in ESCC cells with silenced STK3 or FOXO1. We demonstrated the regulatory relationship of the TP53INP1/P21 gene medicated by the STK3-FOXO1 axis using Western blotting and ChIP in vitro. RESULTS We demonstrate high STK3 expression in ESCC tissue and cell lines compared to esophageal epithelium. Cellular ROS induces STK3 autophosphorylation in ESCC cells, resulting in upregulated p-STK3/4. STK3 activation inhibits ESCC cell proliferation and migration by triggering apoptosis and suppressing the cell cycle. STK3 kinase activation phosphorylates FOXO1Ser212, promoting nuclear translocation, enhancing transcriptional activity, and upregulating TP53INP1 and P21. We also investigated TP53INP1 and P21's phenotypic effects in ESCC, finding that their knockdown significantly increases tumor proliferation, highlighting their crucial role in ESCC tumorigenesis. CONCLUSION STK3 kinase has a high expression level in ESCC and can be activated by cellular ROS, inhibiting cell proliferation and migration. Additionally, STK3 activation-mediated FOXO1 regulates ESCC cell apoptosis and cell cycle arrest by targeting TP53INP1/P21. Our research underscores the anti-tumor function of STK3 in ESCC and elucidates the mechanism underlying its anti-tumor effect on ESCC.
Collapse
Affiliation(s)
- Ziying Zhao
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Yuan Chu
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Anqi Feng
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Shihan Zhang
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Hao Wu
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Zhaoxing Li
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Mingchuang Sun
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Li Zhang
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Tao Chen
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| | - Meidong Xu
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| |
Collapse
|
16
|
Jin L, Hou P. Yixin-Fumai granules modulate autophagy through the PI3K/AKT/FOXO pathway and lead to amelioration of aging mice with sick sinus syndrome. Immun Ageing 2024; 21:46. [PMID: 38971780 PMCID: PMC11227161 DOI: 10.1186/s12979-024-00439-y] [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: 03/15/2024] [Accepted: 05/23/2024] [Indexed: 07/08/2024]
Abstract
OBJECTIVE By employing network pharmacology alongside molecular docking techniques, we can delve into the intricate workings of Yixin-Fumai granules (YXFMs) and their impact on sick sinus syndrome (SSS) within wrinkles mice. Specifically, we aim to understand how YXFMs enhance autophagy through the PI3K/AKT/FOXO path. METHODS The active ingredients and medicinal uses of Ginseng, ligusticum wallichii, Ophiopogon, Schisandra, salvia, and astragalus were compiled using the BATMAN-TCM database. We also used Genecards, OMIM, and Disgenet files to identify the disease goals. A hierarchical diagram of "disease-drug-key targets" was generated using the Cytoscape programs. In addition, we established a target protein interaction (PPI) network using the STRING database. Then, the Cluster Profiler R package was used to conduct GO functional enrichment evaluation and KEGG pathway enrichment analyses of the targets. Based on the PPI system, we chose the top communicating targets and substances over molecular docking. In vivo studies were performed to validate these selections further. The mouse model was induced to study the damaged sinoatrial node (SAN) in mice with lower heart rates due to age-related changes. Electrocardiogram and Masson staining assessments were performed to obtain the results. The transmission electron microscope was used to assess the autophagy level of SAN cells. Western blot was employed to analyze the impact of YXFMs on protein expression in the PI3K/AKT/FOXO signaling process throughout SSS therapy in aging mice. RESULTS One hundred forty-two active ingredients, 1858 targets, 1226 disease targets, and 266 intersection targets were obtained. The key targets of the PPI network encompassed TP53, AKT1, CTNNB1, INS, and TNF, among others. According to GO functional analysis, the mechanism underlying YXFMs in SSS treatment may primarily be associated with the control of ion transport across membranes, cardiac contraction, regulation of blood circulation, and other biological processes. Based on the results of KEGG pathway enrichment analysis, it was determined that they were mainly enriched in multiple pathways of signaling such as the PI3K-Akt signaling route, MAPK signaling process, AGE-RAGE signaling path, FOXO signaling path, HIF-1 signaling process, and several other paths. Molecular docking demonstrated that five compounds had excellent binding to the key candidate target proteins AKT1 and INS. Through the in vivo studies, we noticed notable effects when administering YXFMs. These effects included the suppression of aging-induced SSS, a decrease in the R-R interval, a rise in heart rate, a reduction in fibrosis, a boost in the autophagy process level, and a spike in the levels of expression of key protein molecules in the PI3K/AKT/FOXO signaling path. CONCLUSION This research has made preliminary predictions about the potential of YXFMs in treating SSS. It suggests that YXFMs may have the ability to target key proteins and critical paths associated with the condition. Further testing has been conducted to discover new findings and evidence of ideas for tackling SSS triggered by aging.
Collapse
Affiliation(s)
- Lianzi Jin
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110000, China
| | - Ping Hou
- Department of Cardiology, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, 110000, China.
| |
Collapse
|
17
|
Lin J, Zhang Y, Guan H, Li S, Sui Y, Hong L, Zheng Z, Huang M. Myricitrin inhibited ferritinophagy-mediated ferroptosis in cisplatin-induced human renal tubular epithelial cell injury. Front Pharmacol 2024; 15:1372094. [PMID: 38910888 PMCID: PMC11190325 DOI: 10.3389/fphar.2024.1372094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 05/14/2024] [Indexed: 06/25/2024] Open
Abstract
Cisplatin-induced acute kidney injury (AKI) increases the patient mortality dramatically and results in an unfavorable prognosis. A strong correlation between AKI and ferroptosis, which is a notable type of programmed cell death, was found in recent studies. Myricitrin is a natural flavonoid compound with diverse pharmacological properties. To investigate the protective effect of myricitrin against cisplatin induced human tubular epithelium (HK-2) cell injury and the underlying anti-ferroptic mechanism by this study. Firstly, a pharmacology network analysis was proposed to explore the myricitrin's effect. HK-2 cells were employed for in vitro experiments. Ferroptosis was detected by cell viability, quantification of iron, malondialdehyde, glutathione, lipid peroxidation fluorescence, and glutathione peroxidase (GPX4) expression. Ferritinophagy was detected by related protein expression (NCOA4, FTH, LC3II/I, and SQSTM1). In our study, GO enrichment presented that myricitrin might be effective in eliminating ferroptosis. The phenomenon of ferroptosis regulated by ferritinophagy was observed in cisplatin-activated HK-2 cells. Meanwhile, pretreatment with myricitrin significantly rescued HK-2 cells from cell death, reduced iron overload and lipid peroxidation biomarkers, and improved GPX4 expression. In addition, myricitrin downregulated the expression of LC3II/LC3I and NCOA4 and elevated the expression of FTH and SQTM. Furthermore, myricitrin inhibited ROS production and preserved mitochondrial function with a lower percentage of green JC-1 monomers. However, the protection could be reserved by the inducer of ferritinophagy rapamycin. Mechanically, the Hub genes analysis reveals that AKT and NF-κB are indispensable mediators in the anti-ferroptic process. In conclusion, myricitrin ameliorates cisplatin induced HK-2 cells damage by attenuating ferritinophagy mediated ferroptosis.
Collapse
Affiliation(s)
- Jiawen Lin
- Department of Nephrology, Kidney and Urology Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yangyang Zhang
- Department of Nephrology, Kidney and Urology Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Hui Guan
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuping Li
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Yuan Sui
- Molecular and Cellular Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, United States
| | - Ling Hong
- Department of Nephrology, Kidney and Urology Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Zhihua Zheng
- Department of Nephrology, Kidney and Urology Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Mingcheng Huang
- Department of Nephrology, Kidney and Urology Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
18
|
Yeh PS, Liu CT, Yu CY, Chang YC, Lin SY, Li YC, Luan YZ, Sung WW. Crebanine, an aporphine alkaloid, induces cancer cell apoptosis through PI3K-Akt pathway in glioblastoma multiforme. Front Pharmacol 2024; 15:1419044. [PMID: 38895635 PMCID: PMC11184677 DOI: 10.3389/fphar.2024.1419044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 05/14/2024] [Indexed: 06/21/2024] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most prevalent and lethal primary central nervous system malignancies. GBM is notorious for its high rates of recurrence and therapy resistance and the PI3K/Akt pathway plays a pivotal role in its malignant behavior. Crebanine (CB), an alkaloid capable of penetrating the blood-brain barrier (BBB), has been shown to have inhibitory effects on proinflammatory molecules and multiple cancer cell lines via pathways such as PI3K/Akt. This study aims to investigate the efficacy and mechanisms of CB treatment on GBM. It is the first study to elucidate the anti-tumor role of CB in GBM, providing new possibilities for GBM therapy. Through a series of experiments, we demonstrate the significant anti-survival, anti-clonogenicity, and proapoptotic effects of CB treatment on GBM cell lines. Next-generation sequencing (NGS) is also conducted and provides a complete list of significant changes in gene expression after treatment, including genes related to apoptosis, the cell cycle, FoxO, and autophagy. The subsequent protein expressions of the upregulation of apoptosis and downregulation of PI3K/Akt are further proved. The clinical applicability of CB to GBM treatment could be high for its BBB-penetrating feature, significant induction of apoptosis, and blockage of the PI3K/Akt pathway. Future research is needed using in vivo experiments and other therapeutic pathways shown in NGS for further clinical or in vivo studies.
Collapse
Affiliation(s)
- Poh-Shiow Yeh
- Department of Neurology, Chi Mei Medical Center, Tainan, Taiwan
| | - Chien-Te Liu
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chia-Ying Yu
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Urology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Ya-Chuan Chang
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Urology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shu-Yu Lin
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yun-Chen Li
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yu-Ze Luan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Wen-Wei Sung
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Urology, Chung Shan Medical University Hospital, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| |
Collapse
|
19
|
Silva RCMC. Mitochondria, Autophagy and Inflammation: Interconnected in Aging. Cell Biochem Biophys 2024; 82:411-426. [PMID: 38381268 DOI: 10.1007/s12013-024-01231-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 02/08/2024] [Indexed: 02/22/2024]
Abstract
In this manuscript, I discuss the direct link between abnormalities in inflammatory responses, mitochondrial metabolism and autophagy during the process of aging. It is focused on the cytosolic receptors nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3) and cyclic GMP-AMP synthase (cGAS); myeloid-derived suppressor cells (MDSCs) expansion and their associated immunosuppressive metabolite, methyl-glyoxal, all of them negatively regulated by mitochondrial autophagy, biogenesis, metabolic pathways and its distinct metabolites.
Collapse
Affiliation(s)
- Rafael Cardoso Maciel Costa Silva
- Laboratory of Immunoreceptors and signaling, Instituto de Biofísica Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
20
|
Jang EH, Kim SA. Long-Term Epigenetic Regulation of Foxo3 Expression in Neonatal Valproate-Exposed Rat Hippocampus with Sex-Related Differences. Int J Mol Sci 2024; 25:5287. [PMID: 38791325 PMCID: PMC11121443 DOI: 10.3390/ijms25105287] [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: 03/14/2024] [Revised: 05/06/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Perinatal exposure to valproic acid is commonly used for autism spectrum disorder (ASD) animal model development. The inhibition of histone deacetylases by VPA has been proposed to induce epigenetic changes during neurodevelopment, but the specific alterations in genetic expression underlying ASD-like behavioral changes remain unclear. We used qPCR-based gene expression and epigenetics tools and Western blotting in the hippocampi of neonatal valproic acid-exposed animals at 4 weeks of age and conducted the social interaction test to detect behavioral changes. Significant alterations in gene expression were observed in males, particularly concerning mRNA expression of Foxo3, which was significantly associated with behavioral changes. Moreover, notable differences were observed in H3K27ac chromatin immunoprecipitation, quantitative PCR (ChIP-qPCR), and methylation-sensitive restriction enzyme-based qPCR targeting the Foxo3 gene promoter region. These findings provide evidence that epigenetically regulated hippocampal Foxo3 expression may influence social interaction-related behavioral changes. Furthermore, identifying sex-specific gene expression and epigenetic changes in this model may elucidate the sex disparity observed in autism spectrum disorder prevalence.
Collapse
Affiliation(s)
| | - Soon-Ae Kim
- Department of Pharmacology, School of Medicine, Eulji University, Daejeon 34824, Republic of Korea;
| |
Collapse
|
21
|
Pierzynowska K, Podlacha M, Gaffke L, Rintz E, Wiśniewska K, Cyske Z, Węgrzyn G. Correction of symptoms of Huntington disease by genistein through FOXO3-mediated autophagy stimulation. Autophagy 2024; 20:1159-1182. [PMID: 37992314 PMCID: PMC11135876 DOI: 10.1080/15548627.2023.2286116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 11/03/2023] [Accepted: 11/15/2023] [Indexed: 11/24/2023] Open
Abstract
Huntington disease (HD) is a neurodegenerative disorder caused by a mutation in the HTT gene. The expansion of CAG triplets leads to the appearance of misfolded HTT (huntingtin) forming aggregates and leading to impairment of neuronal functions. Here we demonstrate that stimulation of macroautophagy/autophagy by genistein (4',5,7-trihydroxyisoflavone or 5,7-dihydroxy-3-(4-hydroxyphenyl)-4 H-1-benzopyran-4-one) caused a reduction of levels of mutated HTT in brains of HD mice and correction of their behavior as assessed in a battery of cognitive, anxiety and motor tests, even if the compound was administered after symptoms had developed in the animals. Biochemical and immunological parameters were also improved in HD mice. Studies on molecular mechanisms of genistein-mediated stimulation of autophagy in HD cells indicated the involvement of the FOXO3-related pathway. In conclusion, treatment with genistein stimulates the autophagy process in the brains of HD mice, leading to correction of symptoms of HD, suggesting that it might be considered as a potential drug for this disease. Combined with a very recently published report indicating that impaired autophagy may be a major cause of neurodegenerative changes, these results may indicate the way to the development of effective therapeutic approaches for different neurodegenerative diseases by testing compounds (or possibly combinations of compounds) capable of stimulating autophagy and/or unblocking this process.Abbreviations: CNS: central nervous system; EPM: elevated plus-maze; GOT1/ASPAT: glutamic-oxaloacetic transaminase 1, soluble; GPT/ALAT/ALT: glutamic pyruvic transaminase, soluble; HD: Huntington disease; HTT: huntingtin; IL: interleukin; mHTT: mutant huntingtin; NOR: novel object recognition; MWM: Morris water maze; OF: open field; ROS: reactive oxygen species; TNF: tumor necrosis factor.
Collapse
Affiliation(s)
- Karolina Pierzynowska
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Gdańsk, Poland
| | - Magdalena Podlacha
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Gdańsk, Poland
| | - Lidia Gaffke
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Gdańsk, Poland
| | - Estera Rintz
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Gdańsk, Poland
| | - Karolina Wiśniewska
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Gdańsk, Poland
| | - Zuzanna Cyske
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Gdańsk, Poland
| | - Grzegorz Węgrzyn
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
22
|
Liu F, Zhao L, Wu T, Yu W, Li J, Wang W, Huang C, Diao Z, Xu Y. Targeting autophagy with natural products as a potential therapeutic approach for diabetic microangiopathy. Front Pharmacol 2024; 15:1364616. [PMID: 38659578 PMCID: PMC11039818 DOI: 10.3389/fphar.2024.1364616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 03/26/2024] [Indexed: 04/26/2024] Open
Abstract
As the quality of life improves, the incidence of diabetes mellitus and its microvascular complications (DMC) continues to increase, posing a threat to people's health and wellbeing. Given the limitations of existing treatment, there is an urgent need for novel approaches to prevent and treat DMC. Autophagy, a pivotal mechanism governing metabolic regulation in organisms, facilitates the removal of dysfunctional proteins and organelles, thereby sustaining cellular homeostasis and energy generation. Anomalous states in pancreatic β-cells, podocytes, Müller cells, cardiomyocytes, and Schwann cells in DMC are closely linked to autophagic dysregulation. Natural products have the property of being multi-targeted and can affect autophagy and hence DMC progression in terms of nutrient perception, oxidative stress, endoplasmic reticulum stress, inflammation, and apoptosis. This review consolidates recent advancements in understanding DMC pathogenesis via autophagy and proposes novel perspectives on treating DMC by either stimulating or inhibiting autophagy using natural products.
Collapse
Affiliation(s)
- Fengzhao Liu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lijuan Zhao
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Tao Wu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenfei Yu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jixin Li
- Xi yuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wenru Wang
- Xi yuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chengcheng Huang
- Department of Endocrinology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
| | - Zhihao Diao
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yunsheng Xu
- Department of Endocrinology, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
23
|
Chen RX, Xu SD, Deng MH, Hao SH, Chen JW, Ma XD, Zhuang WT, Cao JH, Lv YR, Lin JL, Li SY, Qiao GB, Xie D, Wang FW. Mex-3 RNA binding family member A (MEX3A)/circMPP6 complex promotes colorectal cancer progression by inhibiting autophagy. Signal Transduct Target Ther 2024; 9:80. [PMID: 38565536 PMCID: PMC10987644 DOI: 10.1038/s41392-024-01787-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 01/30/2024] [Accepted: 03/01/2024] [Indexed: 04/04/2024] Open
Abstract
RNA-binding proteins (RBPs)-RNA networks have contributed to cancer development. Circular RNAs (circRNAs) are considered as protein recruiters; nevertheless, the patterns of circRNA-protein interactions in colorectal cancer (CRC) are still lacking. Processing bodies (PBs) formed through liquid-liquid phase separation (LLPS) are membrane-less organelles (MLOs) consisting of RBPs and RNA. Previous evidence suggests a connection between PBs dynamics and cancer progression. Despite the increasingly acknowledged crucial role of RBPs and RNA in the accumulation and maintenance of MLOs, there remains a lack of specific research on the interactions between PBs-related RBPs and circRNAs in CRC. Herein, we identify that MEX-3 RNA binding family member A (MEX3A), frequently upregulated in CRC tissues, predicts poorer patient survival. Elevated MEX3A accelerates malignance and inhibits autophagy of CRC cells. Importantly, MEX3A undergoes intrinsically disordered regions (IDRs)-dependent LLPS in the cytoplasm. Specifically, circMPP6 acts as a scaffold to facilitate the interaction between MEX3A and PBs proteins. The MEX3A/circMPP6 complex modulates PBs dynamic and promotes UPF-mediated phosphodiesterase 5A (PDE5A) mRNA degradation, consequently leading to the aggressive properties of CRC cells. Clinically, CRC patients exhibiting high MEX3A expression and low PDE5A expression have the poorest overall survival. Our findings reveal a collaboration between MEX3A and circMPP6 in the regulation of mRNA decay through triggering the PBs aggregation, which provides prognostic markers and/or therapeutic targets for CRC.
Collapse
Affiliation(s)
- Ri-Xin Chen
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Shui-Dan Xu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Min-Hua Deng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Shi-Hui Hao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Jie-Wei Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xiao-Dan Ma
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Wei-Tao Zhuang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Jing-Hua Cao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yong-Rui Lv
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Jin-Long Lin
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Si-Yu Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Gui-Bin Qiao
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| | - Dan Xie
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| | - Feng-Wei Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
24
|
Zuo Z, Li Q, Zhou S, Yu R, Wu C, Chen J, Xiao Y, Chen H, Song J, Pan Y, Wang W. Berberine ameliorates contrast-induced acute kidney injury by regulating HDAC4-FoxO3a axis-induced autophagy: In vivo and in vitro. Phytother Res 2024; 38:1761-1780. [PMID: 37922559 DOI: 10.1002/ptr.8059] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 09/30/2023] [Accepted: 10/15/2023] [Indexed: 11/07/2023]
Abstract
In hospitals, contrast-induced acute kidney injury (CI-AKI) is a major cause of renal failure. This study evaluates berberine's (BBR) renal protection and its potential HDAC4 mechanism. CI-AKI in rats was induced with 10 mL kg-1 ioversol. Rats were divided into five groups: Ctrl, BBR, CI-AKI, CI-AKI + BBR, and CI-AKI + Tasq. The renal function of CI-AKI rats was determined by measuring serum creatinine and blood urea nitrogen. Histopathological changes and apoptosis of renal tubular epithelial cells were observed by HE and terminal deoxynucleotidyl transferase (TdTase)-mediated dUTP-biotin nick end labeling (TUNEL) staining. Transmission electron microscopy was used to observe autophagic structures. In vitro, a CI-AKI cell model was created with ioversol-treated HK-2 cells. Treatments included BBR, Rapa, HCQ, and Tasq. Analyses focused on proteins and genes associated with kidney injury, apoptosis, autophagy, and the HDAC4-FoxO3a axis. BBR showed significant protective effects against CI-AKI both in vivo and in vitro. It inhibited apoptosis by increasing Bcl-2 protein levels and decreasing Bax levels. BBR also activated autophagy, as indicated by changes in autophagy-related proteins and autophagic flux. The study further revealed that the contrast agent ioversol increased the expression of HDAC4, which led to elevated levels of phosphorylated FoxO3a (p-FoxO3a) and acetylated FoxO3a (Ac-FoxO3a). However, BBR inhibited HDAC4 expression, resulting in decreased levels of p-FoxO3a and Ac-FoxO3a. This activation of autophagy-related genes, regulated by the transcription factor FoxO3a, played a role in BBR's protective effects. BBR, a traditional Chinese medicine, shows promise against CI-AKI. It may counteract CI-AKI by modulating HDAC4 and FoxO3a, enhancing autophagy, and limiting apoptosis.
Collapse
Affiliation(s)
- Zhi Zuo
- Department of Cardiology, The First Affiliated Hospital with Nanjing Medical University/Jiangsu Province Hospital, Nanjing, China
| | - Qingju Li
- Lianshui People's Hospital, Affiliated Kangda College of Nanjing Medical University, Huai'an, China
- School of Clinical Medicine, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College of Yangzhou University, Yangzhou, China
- Jiangsu College of Nursing, Huai'an, China
| | - Suqin Zhou
- Lianshui People's Hospital, Affiliated Kangda College of Nanjing Medical University, Huai'an, China
| | - Ran Yu
- Lianshui People's Hospital, Affiliated Kangda College of Nanjing Medical University, Huai'an, China
- School of Clinical Medicine, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College of Yangzhou University, Yangzhou, China
- Jiangsu College of Nursing, Huai'an, China
| | - Caixia Wu
- Lianshui People's Hospital, Affiliated Kangda College of Nanjing Medical University, Huai'an, China
| | - Jiajia Chen
- Lianshui People's Hospital, Affiliated Kangda College of Nanjing Medical University, Huai'an, China
| | - Yao Xiao
- Lianshui People's Hospital, Affiliated Kangda College of Nanjing Medical University, Huai'an, China
- Jiangsu College of Nursing, Huai'an, China
| | - Haoyu Chen
- Lianshui People's Hospital, Affiliated Kangda College of Nanjing Medical University, Huai'an, China
| | - Jian Song
- Lianshui People's Hospital, Affiliated Kangda College of Nanjing Medical University, Huai'an, China
| | - Yan Pan
- Lianshui People's Hospital, Affiliated Kangda College of Nanjing Medical University, Huai'an, China
| | - Wanpeng Wang
- Lianshui People's Hospital, Affiliated Kangda College of Nanjing Medical University, Huai'an, China
- School of Clinical Medicine, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College of Yangzhou University, Yangzhou, China
- Jiangsu College of Nursing, Huai'an, China
| |
Collapse
|
25
|
Shi L, Yang J, Tao Z, Zheng L, Bui T, Alonso R, Yue F, Cheng Z. Loss of FoxO1 activates an alternate mechanism of mitochondrial quality control for healthy adipose browning. Clin Sci (Lond) 2024; 138:371-385. [PMID: 38469619 PMCID: PMC10932742 DOI: 10.1042/cs20230973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/13/2024]
Abstract
Browning of white adipose tissue is hallmarked by increased mitochondrial density and metabolic improvements. However, it remains largely unknown how mitochondrial turnover and quality control are regulated during adipose browning. In the present study, we found that mice lacking adipocyte FoxO1, a transcription factor that regulates autophagy, adopted an alternate mechanism of mitophagy to maintain mitochondrial turnover and quality control during adipose browning. Post-developmental deletion of adipocyte FoxO1 (adO1KO) suppressed Bnip3 but activated Fundc1/Drp1/OPA1 cascade, concurrent with up-regulation of Atg7 and CTSL. In addition, mitochondrial biogenesis was stimulated via the Pgc1α/Tfam pathway in adO1KO mice. These changes were associated with enhanced mitochondrial homeostasis and metabolic health (e.g., improved glucose tolerance and insulin sensitivity). By contrast, silencing Fundc1 or Pgc1α reversed the changes induced by silencing FoxO1, which impaired mitochondrial quality control and function. Ablation of Atg7 suppressed mitochondrial turnover and function, causing metabolic disorder (e.g., impaired glucose tolerance and insulin sensitivity), regardless of elevated markers of adipose browning. Consistently, suppression of autophagy via CTSL by high-fat diet was associated with a reversal of adO1KO-induced benefits. Our data reveal a unique role of FoxO1 in coordinating mitophagy receptors (Bnip3 and Fundc1) for a fine-tuned mitochondrial turnover and quality control, underscoring autophagic clearance of mitochondria as a prerequisite for healthy browning of adipose tissue.
Collapse
Affiliation(s)
- Limin Shi
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, FL 32611, U.S.A
- Interdisciplinary Nutritional Sciences Doctoral Program, Center for Nutritional Sciences, University of Florida, Gainesville, FL 32611, U.S.A
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL 32610, U.S.A
| | - Jinying Yang
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, FL 32611, U.S.A
- Interdisciplinary Nutritional Sciences Doctoral Program, Center for Nutritional Sciences, University of Florida, Gainesville, FL 32611, U.S.A
| | - Zhipeng Tao
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA 24061, U.S.A
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, U.S.A
| | - Louise Zheng
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA 24061, U.S.A
| | - Tyler F. Bui
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, FL 32611, U.S.A
| | - Ramon L. Alonso
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, FL 32611, U.S.A
| | - Feng Yue
- Department of Animal Sciences, University of Florida, Gainesville, FL 32611, U.S.A
| | - Zhiyong Cheng
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, FL 32611, U.S.A
- Interdisciplinary Nutritional Sciences Doctoral Program, Center for Nutritional Sciences, University of Florida, Gainesville, FL 32611, U.S.A
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL 32610, U.S.A
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA 24061, U.S.A
| |
Collapse
|
26
|
Gao Y, Zhang L, Zhang F, Liu R, Liu L, Li X, Zhu X, Liang Y. Traditional Chinese medicine and its active substances reduce vascular injury in diabetes via regulating autophagic activity. Front Pharmacol 2024; 15:1355246. [PMID: 38505420 PMCID: PMC10949535 DOI: 10.3389/fphar.2024.1355246] [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: 12/13/2023] [Accepted: 02/26/2024] [Indexed: 03/21/2024] Open
Abstract
Due to its high prevalence, poor prognosis, and heavy burden on healthcare costs, diabetic vascular complications have become a significant public health issue. Currently, the molecular and pathophysiological mechanisms underlying diabetes-induced vascular complications remain incompletely understood. Autophagy, a highly conserved process of lysosomal degradation, maintains intracellular homeostasis and energy balance via removing protein aggregates, damaged organelles, and exogenous pathogens. Increasing evidence suggests that dysregulated autophagy may contribute to vascular abnormalities in various types of blood vessels, including both microvessels and large vessels, under diabetic conditions. Traditional Chinese medicine (TCM) possesses the characteristics of "multiple components, multiple targets and multiple pathways," and its safety has been demonstrated, particularly with minimal toxicity in liver and kidney. Thus, TCM has gained increasing attention from researchers. Moreover, recent studies have indicated that Chinese herbal medicine and its active compounds can improve vascular damage in diabetes by regulating autophagy. Based on this background, this review summarizes the classification, occurrence process, and related molecular mechanisms of autophagy, with a focus on discussing the role of autophagy in diabetic vascular damage and the protective effects of TCM and its active compounds through the regulation of autophagy in diabetes. Moreover, we systematically elucidate the autophagic mechanisms by which TCM formulations, individual herbal extracts, and active compounds regulate diabetic vascular damage, thereby providing new candidate drugs for clinical treatment of vascular complications in diabetes. Therefore, further exploration of TCM and its active compounds with autophagy-regulating effects holds significant research value for achieving targeted therapeutic approaches for diabetic vascular complications.
Collapse
Affiliation(s)
- Yankui Gao
- Department of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Lei Zhang
- Department of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Fei Zhang
- Department of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Lanzhou, China
| | - Rong Liu
- Department of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Lei Liu
- Department of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Xiaoyan Li
- Department of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Xiangdong Zhu
- Department of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, China
| | - Yonglin Liang
- Department of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| |
Collapse
|
27
|
Guo X, Peng K, He Y, Xue L. Mechanistic regulation of FOXO transcription factors in the nucleus. Biochim Biophys Acta Rev Cancer 2024; 1879:189083. [PMID: 38309444 DOI: 10.1016/j.bbcan.2024.189083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/28/2024] [Accepted: 01/31/2024] [Indexed: 02/05/2024]
Abstract
FOXO proteins represent evolutionarily conserved transcription factors (TFs) that play critical roles in responding to various physiological signals or pathological stimuli, either through transcription-dependent or -independent mechanisms. Dysfunction of these proteins have been implicated in numerous diseases, including cancer. Although the regulation of FOXO TFs shuttling between the cytoplasm and the nucleus has been extensively studied and reviewed, there's still a lack of a comprehensive review focusing on the intricate interactions between FOXO, DNA, and cofactors in the regulation of gene expression. In this review, we aim to summarize recent advances and provide a detailed understanding of the mechanism underlying FOXO proteins binding to target DNA. Additionally, we will discuss the challenges associated with pharmacological approaches in modulating FOXO function, and explore the dynamic association between TF, DNA, and RNA on chromatin. This review will contribute to a better understanding of mechanistic regulations of eukaryotic TFs within the nucleus.
Collapse
Affiliation(s)
- Xiaowei Guo
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha, China; The Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Changsha, China.
| | - Kai Peng
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Yanwen He
- Changsha Stomatological Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Lei Xue
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China.
| |
Collapse
|
28
|
Li YY, Qin ZH, Sheng R. The Multiple Roles of Autophagy in Neural Function and Diseases. Neurosci Bull 2024; 40:363-382. [PMID: 37856037 PMCID: PMC10912456 DOI: 10.1007/s12264-023-01120-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 07/11/2023] [Indexed: 10/20/2023] Open
Abstract
Autophagy involves the sequestration and delivery of cytoplasmic materials to lysosomes, where proteins, lipids, and organelles are degraded and recycled. According to the way the cytoplasmic components are engulfed, autophagy can be divided into macroautophagy, microautophagy, and chaperone-mediated autophagy. Recently, many studies have found that autophagy plays an important role in neurological diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease, neuronal excitotoxicity, and cerebral ischemia. Autophagy maintains cell homeostasis in the nervous system via degradation of misfolded proteins, elimination of damaged organelles, and regulation of apoptosis and inflammation. AMPK-mTOR, Beclin 1, TP53, endoplasmic reticulum stress, and other signal pathways are involved in the regulation of autophagy and can be used as potential therapeutic targets for neurological diseases. Here, we discuss the role, functions, and signal pathways of autophagy in neurological diseases, which will shed light on the pathogenic mechanisms of neurological diseases and suggest novel targets for therapies.
Collapse
Affiliation(s)
- Yan-Yan Li
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China
| | - Zheng-Hong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China.
| | - Rui Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China.
| |
Collapse
|
29
|
Liu TW, Zhao YM, Jin KY, Wang JX, Zhao XF. KAT8 is upregulated and recruited to the promoter of Atg8 by FOXO to induce H4 acetylation for autophagy under 20-hydroxyecdysone regulation. J Biol Chem 2024; 300:105704. [PMID: 38309506 PMCID: PMC10904276 DOI: 10.1016/j.jbc.2024.105704] [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: 10/16/2023] [Revised: 01/11/2024] [Accepted: 01/14/2024] [Indexed: 02/05/2024] Open
Abstract
Selective gene expression in cells in physiological or pathological conditions is important for the growth and development of organisms. Acetylation of histone H4 at K16 (H4K16ac) catalyzed by histone acetyltransferase 8 (KAT8) is known to promote gene transcription; however, the regulation of KAT8 transcription and the mechanism by which KAT8 acetylates H4K16ac to promote specific gene expression are unclear. Using the lepidopteran insect Helicoverpa armigera as a model, we reveal that the transcription factor FOXO promotes KAT8 expression and recruits KAT8 to the promoter region of autophagy-related gene 8 (Atg8) to increase H4 acetylation at that location, enabling Atg8 transcription under the steroid hormone 20-hydroxyecdysone (20E) regulation. H4K16ac levels are increased in the midgut during metamorphosis, which is consistent with the expression profiles of KAT8 and ATG8. Knockdown of Kat8 using RNA interference results in delayed pupation and repression of midgut autophagy and decreases H4K16ac levels. Overexpression of KAT8-GFP promotes autophagy and increases H4K16ac levels. FOXO, KAT8, and H4K16ac colocalized at the FOXO-binding region to promote Atg8 transcription under 20E regulation. Acetylated FOXO at K180 and K183 catalyzed by KAT8 promotes gene transcription for autophagy. 20E via FOXO promotes Kat8 transcription. Knockdown or overexpression of FOXO appeared to give similar results as knockdown or overexpression of KAT8. Therefore, FOXO upregulates KAT8 expression and recruits KAT8 to the promoter region of Atg8, where the KAT8 induces H4 acetylation to promote Atg8 transcription for autophagy under 20E regulation. This study reveals the mechanism that KAT8 promotes transcription of a specific gene.
Collapse
Affiliation(s)
- Tian-Wen Liu
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Yu-Meng Zhao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Ke-Yan Jin
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Jin-Xing Wang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China.
| | - Xiao-Fan Zhao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China.
| |
Collapse
|
30
|
Zhou Y, Zhao Q, Zhang Y, Di L, Xue F, Xu W, Gao W, Guo Y, He Y, Kou J, Qin Y, Xie X, Du L, Han G, Pang X. A new andrographolide derivative ADA targeting SIRT3-FOXO3a signaling mitigates cognitive impairment by activating mitophagy and inhibiting neuroinflammation in Apoe4 mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 124:155298. [PMID: 38185066 DOI: 10.1016/j.phymed.2023.155298] [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: 07/29/2023] [Revised: 12/04/2023] [Accepted: 12/16/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is one of the most common neurodegenerative diseases and mitophagy deficit was identified as the typical abnormality in early stage of AD. The neuroprotective effect of andrographolide (AGA) has been confirmed, anda acetylated derivative of AGA (3,14,19-triacetylandrographolide, ADA) was considered to have stronger efficacy. PURPOSE The current study aims to investigate the impact of ADA on cognitive ability in a sporadic AD model and explore its potential mechanism. STUDY DESIGN/ METHODS Apoe4 mouse was adopted for evaluating the impact of AGA on cognitive impairment through a serious of behavioral tests. The molecular mechanism of ADA involved in mitophagy and neuroinflammation was investigated in detailby Western blot, ELISA, immunofluorescence and transmission electron microscopy in Apoe4 mice, as well as Apoe4-transfected BV2 cells and HT22 cells. RESULTS ADA application significantly improved cognitive impairment of Apoe4 mice, and lessened Aβ load and neuronal damage, which has stronger activity than its prototype AGA. Accumulated mitophagy markers LC3II, P62, TOM20, PINK1 and Parkin, and decreased mitophagy receptor BNIP3 in hippocampus of Apoe4 mice were greatly reversed after ADA treatment. Meanwhile, ADA promoted the recruitment of BNIP3 to mitochondria, and the transport of damaged mitochondria to lysosome, indicating that disturbed mitophagy in AD mice was restored by ADA. Inhibited SIRT3 and FOXO3a in Apoe4 mice brains were elevated after ADA treatment. ADA also lightened the neuroinflammation caused by NLRP3 inflammasome activation. Additionally, damaged mitophagy and/or activated NLRP3 inflammasome were also observed in BV2 cells and HT22 cells transfected with Apoe4, all of which were rescued by ADA incubation. Noteworthily, SIRT3 inhibitor 3-TYP could abolish the impact of ADA on mitophagy and NLRP3 inflammasome in vitro. CONCLUSION ADA exerted stronger cognition-enhancing ability in relative to AGA, and ADA could repaire mitophagy deficiency via SIRT3-FOXO3a pathway, and subsequently inhibite NLRP3 inflammasome to mitigate AD pathology.
Collapse
Affiliation(s)
- Yunfeng Zhou
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng 475004, China; State Key Laboratroy of Antiviral Drugs, Henan University, Kaifeng 475004, China
| | - Qian Zhao
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Yixuan Zhang
- Huaihe Hosptial of Henan University, Kaifeng 475000, China
| | - Lulu Di
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Feng Xue
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Wangjun Xu
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Weiping Gao
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Yukun Guo
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Yangyang He
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng 475004, China; Institutes of Traditional Chinese Medicine, Henan University, Kaifeng 475004, China; State Key Laboratroy of Antiviral Drugs, Henan University, Kaifeng 475004, China
| | - Jiejian Kou
- Huaihe Hosptial of Henan University, Kaifeng 475000, China
| | - Ying Qin
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Xinmei Xie
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng 475004, China; State Key Laboratroy of Antiviral Drugs, Henan University, Kaifeng 475004, China.
| | - Lida Du
- Institute of Molecular Medicine & Innovative Pharmaceutics, Qingdao University, Qingdao 266071, China.
| | - Guang Han
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng 475004, China; State Key Laboratroy of Antiviral Drugs, Henan University, Kaifeng 475004, China.
| | - Xiaobin Pang
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng 475004, China; Institutes of Traditional Chinese Medicine, Henan University, Kaifeng 475004, China; State Key Laboratroy of Antiviral Drugs, Henan University, Kaifeng 475004, China.
| |
Collapse
|
31
|
Yang X, Weng Q, Li X, Lu K, Wang L, Song K, Zhang C, Rahimnejad S. High water temperature raised the requirements of methionine for spotted seabass (Lateolabrax maculatus). FISH PHYSIOLOGY AND BIOCHEMISTRY 2024; 50:23-40. [PMID: 36322361 DOI: 10.1007/s10695-022-01136-7] [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: 07/09/2022] [Accepted: 10/17/2022] [Indexed: 06/16/2023]
Abstract
This study evaluated the effects of dietary methionine level and rearing water temperature on growth, antioxidant capacity, methionine metabolism, and hepatocyte autophagy in spotted seabass (Lateolabrax maculatus). A factorial design was used with six methionine levels [0.64, 0.85, 1.11, 1.33, 1.58, and 1.76%] and two temperatures [moderate temperature (MT): 27 ℃, and high temperature (HT): 33 ℃]. The results revealed the significant effects of both dietary methionine level and water temperature on weight gain (WG) and feed efficiency (FE), and their interaction effect was found on WG (P < 0.05). In both water temperatures tested, fish WG increased with increasing methionine level up to 1.11% and decreased thereafter. The groups of fish reared at MT exhibited dramatically higher WG and FE than those kept at HT while an opposite trend was observed for feed intake. Liver antioxidant indices including reduced glutathione and malondialdehyde (MDA) concentrations, and catalase and superoxide dismutase (SOD) activities remarkably increased in the HT group compared to the MT group. Moreover, the lowest MDA concentration and the highest SOD activity were recorded at methionine levels between 1.11% and 0.85%, respectively, regardless of water temperatures. Expression of methionine metabolism-related key enzyme genes (mat2b, cbs, ms, and bhmt) in the liver was increased at moderate methionine levels, and higher expression levels were detected at MT compared to HT with the exception of ms gene relative expression. Relative expression of hepatocyte autophagy-related genes (pink1, atg5, mul1, foxo3) and hsp70 was upregulated by increasing methionine level up to a certain level and decreased thereafter and increasing water temperature led to significantly enhanced expression of hsp70. In summary, HT induced heat stress and reduced fish growth, and an appropriate dietary methionine level improved the antioxidant capacity and stress resistance of fish. A second-order polynomial regression analysis based on the WG suggested that the optimal dietary methionine level for maximum growth of spotted seabass is 1.22% of the diet at 27 ℃ and 1.26% of the diet at 33 ℃, then 1.37 g and 1.68 g dietary methionine intake is required for 100 g weight gain at 27 ℃ or 33 ℃, respectively.
Collapse
Affiliation(s)
- Xin Yang
- Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College, Jimei University, Xiamen, 361021, People's Republic of China
| | - Qinjiang Weng
- Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College, Jimei University, Xiamen, 361021, People's Republic of China
| | - Xueshan Li
- Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College, Jimei University, Xiamen, 361021, People's Republic of China
| | - Kangle Lu
- Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College, Jimei University, Xiamen, 361021, People's Republic of China
| | - Ling Wang
- Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College, Jimei University, Xiamen, 361021, People's Republic of China
| | - Kai Song
- Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College, Jimei University, Xiamen, 361021, People's Republic of China.
| | - Chunxiao Zhang
- Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College, Jimei University, Xiamen, 361021, People's Republic of China.
| | - Samad Rahimnejad
- Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, University of South Bohemia in Ceske Budejovice, Zatisi 728/II, 389 25, Vodnany, Czech Republic
| |
Collapse
|
32
|
Martin TG, Hunt DR, Langer SJ, Tan Y, Ebmeier CC, Crocini C, Chung E, Leinwand LA. A Conserved Mechanism of Cardiac Hypertrophy Regression through FoxO1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.27.577585. [PMID: 38328143 PMCID: PMC10849654 DOI: 10.1101/2024.01.27.577585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
The heart is a highly plastic organ that responds to diverse stimuli to modify form and function. The molecular mechanisms of adaptive physiological cardiac hypertrophy are well-established; however, the regulation of hypertrophy regression is poorly understood. To identify molecular features of regression, we studied Burmese pythons which experience reversible cardiac hypertrophy following large, infrequent meals. Using multi-omics screens followed by targeted analyses, we found forkhead box protein O1 (FoxO1) transcription factor signaling, and downstream autophagy activity, were downregulated during hypertrophy, but re-activated with regression. To determine whether these events were mechanistically related to regression, we established an in vitro platform of cardiomyocyte hypertrophy and regression from treatment with fed python plasma. FoxO1 inhibition prevented regression in this system, while FoxO1 activation reversed fed python plasma-induced hypertrophy in an autophagy-dependent manner. We next examined whether FoxO1 was implicated in mammalian models of reversible hypertrophy from exercise and pregnancy and found that in both cases FoxO1 was activated during regression. In these models, as in pythons, activation of FoxO1 was associated with increased expression FoxO1 target genes involved in autophagy. Taken together, our findings suggest FoxO1-dependent autophagy is a conserved mechanism for regression of physiological cardiac hypertrophy across species.
Collapse
Affiliation(s)
- Thomas G. Martin
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder CO
- BioFrontiers Institute, University of Colorado Boulder, Boulder CO
| | - Dakota R. Hunt
- Department of Biochemistry, University of Colorado Boulder, Boulder CO
- BioFrontiers Institute, University of Colorado Boulder, Boulder CO
| | - Stephen J. Langer
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder CO
- BioFrontiers Institute, University of Colorado Boulder, Boulder CO
| | - Yuxiao Tan
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder CO
- BioFrontiers Institute, University of Colorado Boulder, Boulder CO
| | - Christopher C. Ebmeier
- Department of Biochemistry, University of Colorado Boulder, Boulder CO
- BioFrontiers Institute, University of Colorado Boulder, Boulder CO
| | - Claudia Crocini
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder CO
- BioFrontiers Institute, University of Colorado Boulder, Boulder CO
| | - Eunhee Chung
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder CO
- BioFrontiers Institute, University of Colorado Boulder, Boulder CO
- Department of Kinesiology, University of Texas at San Antonio, San Antonio, TX
| | - Leslie A. Leinwand
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder CO
- BioFrontiers Institute, University of Colorado Boulder, Boulder CO
| |
Collapse
|
33
|
de Lima Junior EA, Teixeira AADS, Silveira LS, Jové Q, Ladrón NÁ, Pereira MG, López-Soriano FJ, Argilés JM, Brum PC, Busquets S, Neto JCR. Formoterol reduces muscle wasting in mice undergoing doxorubicin chemotherapy. Front Oncol 2024; 13:1237709. [PMID: 38234397 PMCID: PMC10791811 DOI: 10.3389/fonc.2023.1237709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 11/27/2023] [Indexed: 01/19/2024] Open
Abstract
Background Even though doxorubicin (DOX) chemotherapy promotes intense muscle wasting, this drug is still widely used in clinical practice due to its remarkable efficiency in managing cancer. On the other hand, intense muscle loss during the oncological treatment is considered a bad prognosis for the disease's evolution and the patient's quality of life. In this sense, strategies that can counteract the muscle wasting induced by DOX are essential. In this study, we evaluated the effectiveness of formoterol (FOR), a β2-adrenoceptor agonist, in managing muscle wasting caused by DOX. Methods and results To evaluate the effect of FOR on DOX-induced muscle wasting, mice were treated with DOX (2.5 mg/kg b.w., i.p. administration, twice a week), associated or not to FOR treatment (1 mg/kg b.w., s.c. administration, daily). Control mice received vehicle solution. A combination of FOR treatment with DOX protected against the loss of body weight (p<0.05), muscle mass (p<0.001), and grip force (p<0.001) promoted by chemotherapy. FOR also attenuated muscle wasting (p<0.01) in tumor-bearing mice on chemotherapy. The potential mechanism by which FOR prevented further DOX-induced muscle wasting occurred by regulating Akt/FoxO3a signaling and gene expression of atrogenes in skeletal muscle. Conclusions Collectively, our results suggest that FOR can be used as a pharmacological strategy for managing muscle wasting induced by DOX. This study provides new insights into the potential therapeutic use of FOR to improve the overall wellbeing of cancer patients undergoing DOX chemotherapy.
Collapse
Affiliation(s)
- Edson Alves de Lima Junior
- Immunometabolism Research Group, Department of Cell and Developmental Biology, University of São Paulo, São Paulo, Brazil
| | | | - Loreana Sanches Silveira
- Immunometabolism Research Group, Department of Cell and Developmental Biology, University of São Paulo, São Paulo, Brazil
| | - Queralt Jové
- Cancer Research Group, Departament de Bioquímica i Molecular Biomedicine, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain and Institut de Biomedicina de la Barcelona (IBUB), Barcelona, Spain
| | - Natalia Álvarez Ladrón
- Cancer Research Group, Departament de Bioquímica i Molecular Biomedicine, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain and Institut de Biomedicina de la Barcelona (IBUB), Barcelona, Spain
| | - Marcelo G. Pereira
- Leeds School of Biomedical Sciences, Faculty of Biological Sciences University of Leeds, Leeds, United Kingdom
| | - Francisco Javier López-Soriano
- Cancer Research Group, Departament de Bioquímica i Molecular Biomedicine, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain and Institut de Biomedicina de la Barcelona (IBUB), Barcelona, Spain
| | - Josep M. Argilés
- Cancer Research Group, Departament de Bioquímica i Molecular Biomedicine, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain and Institut de Biomedicina de la Barcelona (IBUB), Barcelona, Spain
| | - Patrícia Chakur Brum
- School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil
| | - Silvia Busquets
- Cancer Research Group, Departament de Bioquímica i Molecular Biomedicine, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain and Institut de Biomedicina de la Barcelona (IBUB), Barcelona, Spain
| | - José Cesar Rosa Neto
- Immunometabolism Research Group, Department of Cell and Developmental Biology, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
34
|
Sengupta S, Levy DL. Organelle Communication with the Nucleus. Results Probl Cell Differ 2024; 73:3-23. [PMID: 39242372 PMCID: PMC11409190 DOI: 10.1007/978-3-031-62036-2_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2024]
Abstract
Compartmentalization of cellular components is critical to the spatiotemporal and environmental regulation of biochemical activities inside a cell, ensures the proper division of cellular labor and resources, and increases the efficiency of metabolic processes. However, compartmentalization also poses a challenge as organelles often need to communicate across these compartments to complete reaction pathways. These communication signals are often critical aspects of the cellular response to changing environmental conditions. A central signaling hub in the cell, the nucleus communicates with mitochondria, lysosomes, the endoplasmic reticulum, and the Golgi body to ensure optimal organellar and cellular performance. Here we review different mechanisms by which these organelles communicate with the nucleus, focusing on anterograde and retrograde signaling of mitochondria, localization-based signaling of lysosomes, the unfolded protein response of the endoplasmic reticulum, and evidence for nucleus-Golgi signaling. We also include a brief overview of some less well-characterized mechanisms of communication between non-nuclear organelles.
Collapse
Affiliation(s)
- Sourabh Sengupta
- Department of Molecular Biology, University of Wyoming, Laramie, WY 82071, USA
| | - Daniel L. Levy
- Department of Molecular Biology, University of Wyoming, Laramie, WY 82071, USA
| |
Collapse
|
35
|
Cui D, Zhang Y, Chen L, Du H, Zheng B, Huang M, Li X, Wei J, Chen Q. CD30 plays a role in T-dependent immune response and T cell proliferation. FASEB J 2024; 38:e23365. [PMID: 38069862 DOI: 10.1096/fj.202301747rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/22/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023]
Abstract
CD30 is a member of the tumor necrosis factor receptor (TNFR) superfamily and expressed in both normal and malignant lymphoid cells. However, the role of CD30 in lymphopoiesis is not known. In this study, we showed CD30 was expressed both in T and B cells, but its deficiency in mice had no effect on T- and B-cell development. In fact, CD30 deficiency attenuated B-cell response to T-cell-dependent antigens. The impaired B cell response in CD30-deficient mice is caused by the reduction of activation-induced cytidine deaminase (AID) expression. Moreover, CD30-deficient mice exhibited decreased TCR-mediated T cell proliferation and slightly impaired TCR signaling. High-throughput RNA sequencing analysis revealed that CD30 deficiency led to a decrease of FOXO-autophagy axis in T cells upon TCR stimulation. Thus, CD30 positively regulates T-cell-dependent immune response and T cell proliferation.
Collapse
Affiliation(s)
- Dongya Cui
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China
| | - Yongguang Zhang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China
| | - Liling Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China
| | - Hekang Du
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China
| | - Baijiao Zheng
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China
| | - Miaohui Huang
- Department of Reproductive Medicine, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
| | - Xinxin Li
- The Cancer Center, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Jianhui Wei
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China
| | - Qi Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China
| |
Collapse
|
36
|
Le TV, Truong NH, Holterman AXL. Autophagy modulates physiologic and adaptive response in the liver. LIVER RESEARCH 2023; 7:304-320. [DOI: 10.1016/j.livres.2023.12.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
37
|
Feng W, Ma C, Rao H, Zhang W, Liu C, Xu Y, Aji R, Wang Z, Xu J, Gao WQ, Li L. Setd2 deficiency promotes gastric tumorigenesis through inhibiting the SIRT1/FOXO pathway. Cancer Lett 2023; 579:216470. [PMID: 37914019 DOI: 10.1016/j.canlet.2023.216470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/19/2023] [Accepted: 10/29/2023] [Indexed: 11/03/2023]
Abstract
Gastric cancer (GC) is the fifth most common cancer and the second leading cause of cancer death globally. SETD2 is a histone methyltransferase catalyzing tri-methylation of H3K36 (H3K36me3) and has been shown to participate in diverse biological processes and human tumors. However, the mechanism of SETD2 in GC remains unclear. Here, we reported that Setd2 deficiency predicts poor prognosis of gastric cancer. SETD2 loss facilitated H. felis/MNU and c-Myc-induced gastric tumorigenesis, respectively. The mouse model of stomach-specific Setd2 depletion together with c-MYC overexpression (AMS) developed high-grade epithelial defects, intestinal metaplasia and dysplasia at only 10-12 weeks of age. Mechanistically, Setd2 depletion resulted in impaired epigenetic regulation of Sirt1, thus inhibiting the SIRT1/FOXO pathway. Moreover, the agonists of FOXO signaling or overexpression of SIRT1 significantly rescued the enhanced cell proliferation and migration caused by Setd2 deficiency in SGC7901 cells. Together, our findings highlight an epigenetic mechanism by which SETD2 regulates gastric tumorigenesis through SIRT1/FOXO pathway. It may also pave the way for the development of targeted, patient-tailored therapies for GC patients with Setd2 deficiency.
Collapse
Affiliation(s)
- Wenxin Feng
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, China; School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Chunxiao Ma
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, China; School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Hanyu Rao
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, China; School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Zhang
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, China; School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Changwei Liu
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, China; School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Yue Xu
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, China; School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Rebiguli Aji
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, China; School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Ziyi Wang
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, China; School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Jin Xu
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Wei-Qiang Gao
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, China; School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Li Li
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, China; School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
38
|
Chen Z, Wang R, Zhu Y, Huang Z, Yang X, Li Q, Zhong M, Zhang W, Chen L, Wu W, Feng L, An N, Yan Y. A novel circular RNA, circSQSTM1, protects the endothelial function in atherosclerosis. Free Radic Biol Med 2023; 209:301-319. [PMID: 37865306 DOI: 10.1016/j.freeradbiomed.2023.10.398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/11/2023] [Accepted: 10/18/2023] [Indexed: 10/23/2023]
Abstract
A novel circRNA named circSQSTM1 (hsa_circRNA_075320) was screened out in atorvastatin (ATV) stimulated endothelial cells (ECs) by our group. Considering the anti-atherosclerotic function of ATV, we hypothesized the circSQSTM1 could protect ECs functions in AS progression. The effects of circSQSTM1 on ECs inflammation, oxidative stress and autophagy were measured by qRT-PCR, Western blotting, monocyte-endothelial adhesion assay, dichloro-dihydro-fluorescein diacetate and mCherry-GFP-LC3 labeling. A luciferase reporter assay, RNA immunoprecipitation, MS2-tagging system and fluorescence in situ hybridization were performed to identify the biological functions of circSQSTM1. The partial left carotid artery ligation model and atherosclerosis model were established to analyze the effects of circSQSTM1 on atherosclerosis progression in vivo. Our results revealed that ATV induced the accumulation of circSQSTM1 in ECs via suppressing m6A modified degradation. In the cytoplasm, circSQSTM1 could relieve Sirt1 by competitively sponging miR-23b-3p. In the nucleus, circSQSTM1 directly interacts with eIF4A3 and promoting the efficient nuclear export of FOXO1 mRNA, which encodes FOXO1 transcription factor to directly activate Sirt1 promoter activity. Hence, circSQSTM1 reduced inflammation, inhibited oxidative stress and promoted autophagy by upregulating Sirt1 in ECs. Moreover, circSQSTM1 overexpression in ECs attenuated the progression of atherosclerosis in ApoE-/- mice. Taken together, the unique noncoding RNA known as circSQSTM1 took a protective role to the ECs in atherosclerosis.
Collapse
Affiliation(s)
- Ziqi Chen
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Ruoyu Wang
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, China
| | - Yinghong Zhu
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Ziyao Huang
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Xuewen Yang
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Qiushi Li
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Meijun Zhong
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Wei Zhang
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Lin Chen
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Wei Wu
- Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Li Feng
- Department of Cardiology, Zhongshan Hospital of Sun Yat-sen University, Zhongshan, 528400, China.
| | - Ningbo An
- Dermatology Hospital of Southern Medical University, Southern Medical University, Guangzhou, 510515, China.
| | - Yi Yan
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| |
Collapse
|
39
|
Morris BJ, Donlon TA. Genes That Extend Lifespan May Do So by Mitigating the Increased Risk of Death Posed by Having Hypertension. Am J Hypertens 2023; 36:631-640. [PMID: 37561089 PMCID: PMC10647014 DOI: 10.1093/ajh/hpad070] [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: 05/12/2023] [Revised: 07/22/2023] [Accepted: 08/09/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Genetic factors influence lifespan. In humans, there appears to be a particularly strong genetic effect in those aged ≥ 90 years. An important contribution is nutrient sensing genes which confer cell resilience. METHODS Our research has been investigating the genetic factors by longitudinal studies of American men of Japanese descent living on the island of Oahu in Hawaii. This cohort began as the Honolulu Heart Program in the mid-1960s and most subjects are now deceased. RESULTS We previously discovered various genes containing polymorphisms associated with longevity. In recent investigations of the mechanism involved we found that the longevity genotypes ameliorated the risk of mortality posed by having a cardiometabolic disease (CMD)-most prominently hypertension. For the gene FOXO3 the protective alleles mitigated the risk of hypertension, coronary heart disease (CHD) and diabetes. For the kinase MAP3K5 it was hypertension, CHD and diabetes, for the kinase receptor PIK3R1 hypertension, CHD and stroke, and for the growth hormone receptor gene (GHR) and vascular endothelial growth factor receptor 1 gene (FLT1), it was nullifying the higher mortality risk posed by hypertension. Subjects with a CMD who had a longevity genotype had similar survival as men without CMD. No variant protected against risk of death from cancer. We have postulated that the longevity-associated genotypes reduced mortality risk by effects on intracellular resilience mechanisms. In a proteomics study, 43 "stress" proteins and associated biological pathways were found to influence the association of FOXO3 genotype with reduced mortality. CONCLUSIONS Our landmark findings indicate how heritable genetic components affect longevity.
Collapse
Affiliation(s)
- Brian J Morris
- Department of Research, NIH Center of Biomedical Research Excellence on Aging, Kuakini Medical Center, Honolulu, Hawaii 96817, USA
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii 96813, USA
- School of Medical Sciences, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Timothy A Donlon
- Department of Research, NIH Center of Biomedical Research Excellence on Aging, Kuakini Medical Center, Honolulu, Hawaii 96817, USA
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii 96813, USA
| |
Collapse
|
40
|
Sadeghi A, Niknam M, Momeni-Moghaddam MA, Shabani M, Aria H, Bastin A, Teimouri M, Meshkani R, Akbari H. Crosstalk between autophagy and insulin resistance: evidence from different tissues. Eur J Med Res 2023; 28:456. [PMID: 37876013 PMCID: PMC10599071 DOI: 10.1186/s40001-023-01424-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 10/03/2023] [Indexed: 10/26/2023] Open
Abstract
Insulin is a critical hormone that promotes energy storage in various tissues, as well as anabolic functions. Insulin resistance significantly reduces these responses, resulting in pathological conditions, such as obesity and type 2 diabetes mellitus (T2DM). The management of insulin resistance requires better knowledge of its pathophysiological mechanisms to prevent secondary complications, such as cardiovascular diseases (CVDs). Recent evidence regarding the etiological mechanisms behind insulin resistance emphasizes the role of energy imbalance and neurohormonal dysregulation, both of which are closely regulated by autophagy. Autophagy is a conserved process that maintains homeostasis in cells. Accordingly, autophagy abnormalities have been linked to a variety of metabolic disorders, including insulin resistance, T2DM, obesity, and CVDs. Thus, there may be a link between autophagy and insulin resistance. Therefore, the interaction between autophagy and insulin function will be examined in this review, particularly in insulin-responsive tissues, such as adipose tissue, liver, and skeletal muscle.
Collapse
Affiliation(s)
- Asie Sadeghi
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Maryam Niknam
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Maryam Shabani
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Aria
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Alireza Bastin
- Clinical Research Development Center "The Persian Gulf Martyrs" Hospital, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Maryam Teimouri
- Department of Biochemistry, School of Allied Medical Sciences, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Reza Meshkani
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Akbari
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran.
- Department of Clinical Biochemistry, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
41
|
Kim Y, Li C, Gu C, Fang Y, Tycksen E, Puri A, Pietka TA, Sivapackiam J, Kidd K, Park SJ, Johnson BG, Kmoch S, Duffield JS, Bleyer AJ, Jackrel ME, Urano F, Sharma V, Lindahl M, Chen YM. MANF stimulates autophagy and restores mitochondrial homeostasis to treat autosomal dominant tubulointerstitial kidney disease in mice. Nat Commun 2023; 14:6493. [PMID: 37838725 PMCID: PMC10576802 DOI: 10.1038/s41467-023-42154-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 09/25/2023] [Indexed: 10/16/2023] Open
Abstract
Misfolded protein aggregates may cause toxic proteinopathy, including autosomal dominant tubulointerstitial kidney disease due to uromodulin mutations (ADTKD-UMOD), a leading hereditary kidney disease. There are no targeted therapies. In our generated mouse model recapitulating human ADTKD-UMOD carrying a leading UMOD mutation, we show that autophagy/mitophagy and mitochondrial biogenesis are impaired, leading to cGAS-STING activation and tubular injury. Moreover, we demonstrate that inducible tubular overexpression of mesencephalic astrocyte-derived neurotrophic factor (MANF), a secreted endoplasmic reticulum protein, after the onset of disease stimulates autophagy/mitophagy, clears mutant UMOD, and promotes mitochondrial biogenesis through p-AMPK enhancement, thus protecting kidney function in our ADTKD mouse model. Conversely, genetic ablation of MANF in the mutant thick ascending limb tubular cells worsens autophagy suppression and kidney fibrosis. Together, we have discovered MANF as a biotherapeutic protein and elucidated previously unknown mechanisms of MANF in the regulation of organelle homeostasis, which may have broad therapeutic applications to treat various proteinopathies.
Collapse
Affiliation(s)
- Yeawon Kim
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Chuang Li
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Chenjian Gu
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Yili Fang
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Eric Tycksen
- Genome Technology Access Center, McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Anuradhika Puri
- Department of Chemistry, Washington University, St. Louis, MO, USA
| | - Terri A Pietka
- Nutrition and Geriatrics Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Jothilingam Sivapackiam
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kendrah Kidd
- Section of Nephrology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Research Unit of Rare Diseases, Department of Pediatric and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Sun-Ji Park
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Bryce G Johnson
- Pfizer Worldwide Research and Development, Inflammation & Immunology, Cambridge, MA, USA
| | - Stanislav Kmoch
- Section of Nephrology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Research Unit of Rare Diseases, Department of Pediatric and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | | | - Anthony J Bleyer
- Section of Nephrology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Research Unit of Rare Diseases, Department of Pediatric and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | | | - Fumihiko Urano
- Division of Endocrinology, Metabolism, and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Vijay Sharma
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Engineering, School of Engineering & Applied Science, Washington University, St. Louis, MO, USA
| | - Maria Lindahl
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Ying Maggie Chen
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
42
|
Huang HJ, Zhang JL, Zhang CX. Insight into phenotypic plasticity in planthoppers. CURRENT OPINION IN INSECT SCIENCE 2023; 59:101106. [PMID: 37625640 DOI: 10.1016/j.cois.2023.101106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/08/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023]
Abstract
Planthoppers possess an impressive ability to exhibit phenotypic plasticity, which allows them to adjust their morphology for migration, overwintering, and adaptation to different environmental conditions. The wing and color polyphenism are the two most outward morphologies. Wing polyphenism serves as a classic illustration of a life history trade-off between reproduction and migration, while color polyphenism is potentially correlated with the insect development and immunity. In this review, we present the important contributions that link environment cues to wing and color polyphenism, and highlight recent advances in insulin/insulin-like growth factor signaling-forkhead transcription factor subgroup O (FoxO) pathway-mediated wing development and tyrosine-melanin pathway-mediated coloration. Further work, particularly in the identification of the genes that FoxO regulates and in the elucidation of the intracellular signals that link the stimuli to the tyrosine-melanin pathway, is required.
Collapse
Affiliation(s)
- Hai-Jian Huang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of Ministry of Agriculture and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo 315211, China
| | - Jin-Li Zhang
- Institute of Insect Science, Zhejiang University, Hangzhou, China
| | - Chuan-Xi Zhang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of Ministry of Agriculture and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo 315211, China; Institute of Insect Science, Zhejiang University, Hangzhou, China.
| |
Collapse
|
43
|
Mu M, Zhang Q, Zhao C, Li X, Chen Z, Sun X, Yu J. 3-Bromopyruvate overcomes cetuximab resistance in human colorectal cancer cells by inducing autophagy-dependent ferroptosis. Cancer Gene Ther 2023; 30:1414-1425. [PMID: 37558749 PMCID: PMC10581902 DOI: 10.1038/s41417-023-00648-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 06/15/2023] [Accepted: 07/12/2023] [Indexed: 08/11/2023]
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer-related death worldwide. Cetuximab, in combination with chemotherapy, is effective for treating patients with wild-type KRAS/BRAF metastatic CRC (mCRC). However, intrinsic or acquired drug resistance often limits the use of cetuximab. In this study, we investigated the potential of co-treatment with 3-Bromopyruvate (3-BP) and cetuximab to overcome cetuximab resistance in CRC, both in vitro and in vivo. Our results demonstrated that the co-treatment of 3-BP and cetuximab synergistically induced an antiproliferative effect in both CRC cell lines with intrinsic cetuximab resistance (DLD-1 (KRASG13D/-) and HT29 (BRAFV600E)) and in a cetuximab-resistant cell line derived from Caco-2 with acquired resistance (Caco-2-CR). Further analysis revealed that co-treatment induced ferroptosis, autophagy, and apoptosis. Mechanistically, co-treatment inhibited FOXO3a phosphorylation and degradation and activated the FOXO3a/AMPKα/pBeclin1 and FOXO3a/PUMA pathways, leading to the promotion of ferroptosis, autophagy, and apoptosis in DLD-1 (KRASG13D/-), HT29 (BRAFV600E), and Caco-2-CR cells. In conclusion, our findings suggest that co-treatment with 3-BP and cetuximab could be a promising strategy to overcome cetuximab resistance in human CRC.
Collapse
Affiliation(s)
- Mingchao Mu
- Department of General Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Qin Zhang
- Department of Dermatology, Northwest Hospital, the Second Affiliated Hospital of Xi'an Jiaotong University, 710004, Xi'an, Shaanxi, China
| | - Chenye Zhao
- Department of General Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Xiaopeng Li
- Department of General Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Zilu Chen
- Department of General Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Xuejun Sun
- Department of General Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China.
| | - Junhui Yu
- Department of General Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China.
| |
Collapse
|
44
|
Hassan MA, Elmageed GMA, El-Qazaz IG, El-Sayed DS, El-Samad LM, Abdou HM. The Synergistic Influence of Polyflavonoids from Citrus aurantifolia on Diabetes Treatment and Their Modulation of the PI3K/AKT/FOXO1 Signaling Pathways: Molecular Docking Analyses and In Vivo Investigations. Pharmaceutics 2023; 15:2306. [PMID: 37765275 PMCID: PMC10535482 DOI: 10.3390/pharmaceutics15092306] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/05/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
This study was aimed at probing the modulatory influence of polyflavonoids extracted from Citrus aurantifolia, lemon peel extract (LPE-polyflavonoids), on attenuating diabetes mellitus (DM) and its complications. HPLC investigations of the LPE exhibited the incidence of five flavonoids, including diosmin, biochanin A, hesperidin, quercetin, and hesperetin. The in silico impact on ligand-phosphatidylinositol 3-kinase (PI3K) interaction was investigated in terms of polyflavonoid class to explore the non-covalent intakes and binding affinity to the known protein active site. The drug likeness properties and pharmacokinetic parameters of the LPE-polyflavonoids were investigated to assess their bioavailability in relation to Myricetin as a control. Remarkably, the molecular docking studies demonstrated a prominent affinity score of all these agents together with PI3K, implying the potency of the extract to orchestrate PI3K, which is the predominant signal for lessening the level of blood glucose. To verify these findings, in vivo studies were conducted, utilizing diabetic male albino rats treated with LPE-polyflavonoids and other groups treated with hesperidin and diosmin as single flavonoids. Our findings demonstrated that the LPE-polyflavonoids significantly ameliorated the levels of glucose, insulin, glycogen, liver function, carbohydrate metabolizing enzymes, G6Pd, and AGEs compared to the diabetic rats and those exposed to hesperidin and diosmin. Furthermore, the LPE-polyflavonoids regulated the TBARS, GSH, CAT, TNF-α, IL-1β, IL-6, and AFP levels in the pancreatic and hepatic tissues, suggesting their antioxidant and anti-inflammatory properties. In addition, the pancreatic and hepatic GLUT4 and GLUT2 were noticeably increased in addition to the pancreatic p-AKT in the rats administered with the LPE-polyflavonoids compared to the other diabetic rats. Remarkably, the administration of LPE-polyflavonoids upregulated the expression of the pancreatic and hepatic PI3K, AMPK, and FOXO1 genes, emphasizing the efficiency of the LPE in orchestrating all the signaling pathways necessitated to reduce the diabetes mellitus. Notably, the histopathological examinations of the pancreatic and hepatic tissues corroborated the biochemical results. Altogether, our findings accentuated the potential therapeutic role of LPE-polyflavonoids in controlling diabetes mellitus.
Collapse
Affiliation(s)
- Mohamed A. Hassan
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab City 21934, Egypt
| | - Ghada M. Abd Elmageed
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria 21321, Egypt; (G.M.A.E.); (I.G.E.-Q.); (L.M.E.-S.)
| | - Ibtehal G. El-Qazaz
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria 21321, Egypt; (G.M.A.E.); (I.G.E.-Q.); (L.M.E.-S.)
| | - Doaa S. El-Sayed
- Chemistry Department, Faculty of Science, Alexandria University, Alexandria 21321, Egypt;
| | - Lamia M. El-Samad
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria 21321, Egypt; (G.M.A.E.); (I.G.E.-Q.); (L.M.E.-S.)
| | - Heba M. Abdou
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria 21321, Egypt; (G.M.A.E.); (I.G.E.-Q.); (L.M.E.-S.)
| |
Collapse
|
45
|
Oh HJ, Jin H, Lee JY, Lee BY. Silk Peptide Ameliorates Sarcopenia through the Regulation of Akt/mTOR/FoxO3a Signaling Pathways and the Inhibition of Low-Grade Chronic Inflammation in Aged Mice. Cells 2023; 12:2257. [PMID: 37759480 PMCID: PMC10527450 DOI: 10.3390/cells12182257] [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: 08/07/2023] [Revised: 08/30/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
As populations around the world age, interest in healthy aging is growing. One of the first physical changes that occurs with aging is the loss of muscle mass and strength, termed sarcopenia. Sarcopenia limits the activity of older people, reduces their quality of life, and increases the likelihood of their developing disease. In the present study, we aimed to evaluate the effects of the ingestion of acid-hydrolyzed silk peptide (SP) on the muscle mass and strength of mice of >22 months of age with naturally occurring sarcopenia, and to identify the mechanisms involved. The daily administration of SP for 8 weeks increased the activation of the Akt/mTOR/FoxO3a signaling pathways and increased the muscle mass and strength of the old mice. In addition, SP inhibited oxidative stress and inflammation in muscle, which are direct causes of sarcopenia. Therefore, SP represents a promising potential treatment for sarcopenia that may improve the healthy lifespan and quality of life of older people.
Collapse
Affiliation(s)
- Hyun-Ji Oh
- Department of Food Science and Biotechnology, College of Life Science, CHA University, Seongnam 13488, Republic of Korea; (H.-J.O.); (H.J.)
| | - Heegu Jin
- Department of Food Science and Biotechnology, College of Life Science, CHA University, Seongnam 13488, Republic of Korea; (H.-J.O.); (H.J.)
| | | | - Boo-Yong Lee
- Department of Food Science and Biotechnology, College of Life Science, CHA University, Seongnam 13488, Republic of Korea; (H.-J.O.); (H.J.)
| |
Collapse
|
46
|
Yu F, Wang M, Luo K, Sun L, Yu S, Zuo J, Wang Y. Expression Profiles of Long Non-Coding RNAs in the Articular Cartilage of Rats Exposed to T-2 Toxin. Int J Mol Sci 2023; 24:13703. [PMID: 37762015 PMCID: PMC10530968 DOI: 10.3390/ijms241813703] [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: 07/15/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
T-2 toxin could induce bone damage. But there is no specific mechanism about the long non-coding RNAs (lncRNAs) involved in T-2 toxin-induced articular cartilage injury. In this study, 24 SD rats were randomly divided into a control group and a T-2 group, which were administered 4% absolute ethanol and 100 ng/g · bw/day of T-2 toxin, respectively. After treatment for 4 weeks, safranin O/fast green staining identified the pathological changes in the articular cartilage of rats, and immunofluorescence verified the autophagy level increase in the T-2 group. Total RNA was isolated, and high-throughput sequencing was performed. A total of 620 differentially expressed lncRNAs (DE-lncRNAs) were identified, and 326 target genes were predicted. Enrichment analyses showed that the target genes of DE-lncRNAs were enriched in the autophagy-related biological processes and pathways. According to the autophagy database, a total of 23 autophagy-related genes were identified, and five hub genes (Foxo3, Foxo1, Stk11, Hdac4, and Rela) were screened using the Maximal Clique Centrality algorithm. The Human Protein Atlas database indicated that Rela and Hdac4 proteins were highly expressed in the bone marrow tissue, while Foxo3, Foxo1, and Stk11 proteins were reduced. According to Enrichr, etoposide and diatrizoic acid were identified as the key drugs. The real-time quantitative PCR results were consistent with the RNA sequencing (RNA-Seq) results. These results suggested that autophagy was involved in the rat articular cartilage lesions induced by T-2 toxin. The lncRNAs of NONRATG014223.2, NONRATG012484.2, NONRATG021591.2, NONRATG024691.2, and NONRATG002808.2, and their target genes of Foxo3, Foxo1, Stk11, Hdac4, and Rela, respectively, were the key regulator factors of autophagy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yanjie Wang
- School of Public Health, Zhengzhou University, Zhengzhou 450001, China; (F.Y.); (M.W.); (K.L.); (L.S.); (S.Y.); (J.Z.)
| |
Collapse
|
47
|
Ko SH, Choi JH, Kim JM. Bacteroides fragilis Enterotoxin Induces Autophagy through an AMPK and FoxO3-Pathway, Leading to the Inhibition of Apoptosis in Intestinal Epithelial Cells. Toxins (Basel) 2023; 15:544. [PMID: 37755970 PMCID: PMC10535581 DOI: 10.3390/toxins15090544] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 09/28/2023] Open
Abstract
Macroautophagy/autophagy is essential for preserving cellular homeostasis by recycling nutrients and removing spoiled or aged proteins and organelles. It also has an essential role in defense mechanisms against microbial infections. However, the role of autophagy in enterotoxigenic Bacteroides fragilis infection remains largely unknown. In this study, we explored the role of B. fragilis enterotoxin (BFT) in the autophagic process of intestinal epithelial cells (IECs). The LC3-I of human HCT-116 IECs was converted to LC3-II by BFT stimulation. In addition, BFT-exposed cells showed the decreased expression of p62 in a time-dependent manner and increased levels of ATG5 and ATG12 gradually. Evidence of an enhanced autophagic process was supported by autophagosomes co-localized with LC3-lysosome-associated protein 2 in BFT-stimulated cells. The AMP-activated protein kinase (AMPK) and Forkhead box O3 (FoxO3a) axis were required for BFT-induced autophagy activation. In contrast with the activation of autophagy at 3-6 h after BFT exposure, IECs induced apoptosis-related signals at 12-48 h. HCT-116 IECs suppressing the formation of autophagosomes significantly activated apoptosis signals instead of autophagy early after BFT exposure. These data suggest that BFT can activate autophagy through the AMPK-FoxO3a pathway and the autophagy may suppress apoptosis during early exposure of IECs to BFT.
Collapse
Affiliation(s)
- Su Hyuk Ko
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center, San Antonio, TX 78229, USA;
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Jun Ho Choi
- Department of Microbiology, Hanyang University College of Medicine, Seoul 04763, Republic of Korea
| | - Jung Mogg Kim
- Department of Microbiology, Hanyang University College of Medicine, Seoul 04763, Republic of Korea
| |
Collapse
|
48
|
Diez AF, Leroux LP, Chagneau S, Plouffe A, Gold M, Chaparro V, Jaramillo M. Toxoplasma gondii inhibits the expression of autophagy-related genes through AKT-dependent inactivation of the transcription factor FOXO3a. mBio 2023; 14:e0079523. [PMID: 37387601 PMCID: PMC10470550 DOI: 10.1128/mbio.00795-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/15/2023] [Indexed: 07/01/2023] Open
Abstract
The intracellular parasite Toxoplasma gondii induces host AKT activation to prevent autophagy-mediated clearance; however, the molecular underpinnings are not fully understood. Autophagy can be negatively regulated through AKT-sensitive phosphorylation and nuclear export of the transcription factor Forkhead box O3a (FOXO3a). Using a combination of pharmacological and genetic approaches, herein we investigated whether T. gondii hinders host autophagy through AKT-dependent inactivation of FOXO3a. We found that infection by type I and II strains of T. gondii promotes gradual and sustained AKT-dependent phosphorylation of FOXO3a at residues S253 and T32 in human foreskin fibroblasts (HFF) and murine 3T3 fibroblasts. Mechanistically, AKT-sensitive phosphorylation of FOXO3a by T. gondii required live infection and the activity of PI3K but was independent of the plasma membrane receptor EGFR and the kinase PKCα. Phosphorylation of FOXO3a at AKT-sensitive residues was paralleled by its nuclear exclusion in T. gondii-infected HFF. Importantly, the parasite was unable to drive cytoplasmic localization of FOXO3a upon pharmacological blockade of AKT or overexpression of an AKT-insensitive mutant form of FOXO3a. Transcription of a subset of bona fide autophagy-related targets of FOXO3a was reduced during T. gondii infection in an AKT-dependent fashion. However, parasite-directed repression of autophagy-related genes was AKT-resistant in cells deficient in FOXO3a. Consistent with this, T. gondii failed to inhibit the recruitment of acidic organelles and LC3, an autophagy marker, to the parasitophorous vacuole upon chemically or genetically induced nuclear retention of FOXO3a. In all, we provide evidence that T. gondii suppresses FOXO3a-regulated transcriptional programs to prevent autophagy-mediated killing. IMPORTANCE The parasite Toxoplasma gondii is the etiological agent of toxoplasmosis, an opportunistic infection commonly transmitted by ingestion of contaminated food or water. To date, no effective vaccines in humans have been developed and no promising drugs are available to treat chronic infection or prevent congenital infection. T. gondii targets numerous host cell processes to establish a favorable replicative niche. Of note, T. gondii activates the host AKT signaling pathway to prevent autophagy-mediated killing. Herein, we report that T. gondii inhibits FOXO3a, a transcription factor that regulates the expression of autophagy-related genes, through AKT-dependent phosphorylation. The parasite's ability to block the recruitment of the autophagy machinery to the parasitophorous vacuole is impeded upon pharmacological inhibition of AKT or overexpression of an AKT-insensitive form of FOXO3a. Thus, our study provides greater granularity in the role of FOXO3a during infection and reinforces the potential of targeting autophagy as a therapeutic strategy against T. gondii.
Collapse
Affiliation(s)
- Andres Felipe Diez
- Institut National de la Recherche Scientifique (INRS)—Centre Armand-Frappier Santé Biotechnologie (AFSB), Laval, Québec, Canada
| | - Louis-Philippe Leroux
- Institut National de la Recherche Scientifique (INRS)—Centre Armand-Frappier Santé Biotechnologie (AFSB), Laval, Québec, Canada
| | - Sophie Chagneau
- Institut National de la Recherche Scientifique (INRS)—Centre Armand-Frappier Santé Biotechnologie (AFSB), Laval, Québec, Canada
| | - Alexandra Plouffe
- Institut National de la Recherche Scientifique (INRS)—Centre Armand-Frappier Santé Biotechnologie (AFSB), Laval, Québec, Canada
| | - Mackenzie Gold
- Institut National de la Recherche Scientifique (INRS)—Centre Armand-Frappier Santé Biotechnologie (AFSB), Laval, Québec, Canada
| | - Visnu Chaparro
- Institut National de la Recherche Scientifique (INRS)—Centre Armand-Frappier Santé Biotechnologie (AFSB), Laval, Québec, Canada
| | - Maritza Jaramillo
- Institut National de la Recherche Scientifique (INRS)—Centre Armand-Frappier Santé Biotechnologie (AFSB), Laval, Québec, Canada
| |
Collapse
|
49
|
Oh HJ, Jin H, Lee BY. Hesperidin Ameliorates Sarcopenia through the Regulation of Inflammaging and the AKT/mTOR/FoxO3a Signaling Pathway in 22-26-Month-Old Mice. Cells 2023; 12:2015. [PMID: 37566094 PMCID: PMC10417333 DOI: 10.3390/cells12152015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/12/2023] Open
Abstract
Faced with a globally aging society, the maintenance of health and quality of life in older people is very important. The age-related loss of muscle mass and strength, known as sarcopenia, severely reduces quality of life and increases the risks of various diseases. In this study, we investigated the inhibitory effect of hesperidin (HES) on inflammaging, with the intention of evaluating its potential use as a treatment for sarcopenia. We studied 22-26-month-old mice, corresponding to humans aged ≥70 years, with aging-related sarcopenia, and young mice aged 3-6 months. The daily administration of HES for 8 weeks resulted in greater muscle mass and strength and increased the fiber size of the old mice. HES also restored the immune homeostasis that had been disrupted by aging, such as the imbalance in M1/M2 macrophage ratio. In addition, we found that HES ameliorated the sarcopenia by regulating AKT/mammalian target of rapamycin/forkhead box 3a signaling through an increase in insulin-like growth factor (IGF)-1 expression in the old mice. Therefore, HES represents a promising candidate inhibitor of sarcopenia in older people, and its effects are achieved through the maintenance of immune homeostasis.
Collapse
Affiliation(s)
| | | | - Boo-Yong Lee
- Department of Food Science and Biotechnology, College of Life Science, CHA University, Seongnam 13488, Gyeonggi, Republic of Korea; (H.-J.O.); (H.J.)
| |
Collapse
|
50
|
Qiao S, Wang X, Li H, Zhang C, Wang A, Zhang S. Atherosclerosis-associated endothelial dysfunction is promoted by miR-199a-5p/SIRT1 axis regulated by circHIF1ɑ. Nutr Metab Cardiovasc Dis 2023; 33:1619-1631. [PMID: 37336718 DOI: 10.1016/j.numecd.2023.05.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 04/29/2023] [Accepted: 05/03/2023] [Indexed: 06/21/2023]
Abstract
BACKGROUND AND AIMS Atherosclerosis (AS) is a chronic inflammatory disease that damages the arterial wall as a result of hyperlipidemia and causes endothelial cell dysfunction, which increases the risk of atherothrombotic events. Multiple pathological conditions have shown ectopic miR-199a-5p levels to cause endothelial injury, but its role in the AS competitive endogenous RNA (CeRNA) network is still unknown. METHODS AND RESULTS The high-fat diet (HFD) apoE-/- mouse model was constructed in vivo, and ECs were cultured under ox-LDL treatment to induce EC injury in vitro. Immunohistochemistry and immunofluorescence staining were used to assess the effect of miR-199a-5p on the macrophage, SMC, collagen content, and endothelial coverage in the artery wall of mouse model. miR-199a-5p level was validated to be overexpression in the aorta tissue of HFD apoE-/- mice and in the ox-LDL-treated ECs, and even in the plasma EVs of the patients with cerebral AS. Silencing of miR-199a-5p significantly attenuated atherosclerotic progress in HFD apoE-/- mice, and the gain/loss-of-function assay indicated that miR-199a-5p overexpression aggravated ox-LDL-induced disabilities of endothelial proliferation, motility, and neovascularization based on cell counting kit-8 assay, transwell assay and matrigel assay. Mechanistically, miR-199a-5p prevented EC activation by activating the FOXO signaling pathway by targeting SIRT1. Additionally, circular RNA (circRNA) circHIF1ɑ was identified as having a low expression in the ox-LDL-treated EC and mediated SIRT1 expression via sponging miR-199a-5p to rescue ox-LDL-induced EC injury. CONCLUSIONS Our study demonstrated the vital role of miR-199a-5p/SIRT1 axis regulated by circHIF1ɑ in AS pathogenesis and provided novel effective targets for AS treatment.
Collapse
Affiliation(s)
- Shan Qiao
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong First Medical University, Jinan, China; Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China; Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China; Shandong Laibo Biotechnology Co., Ltd, China
| | - Xing Wang
- Department of Neurology, Tianyou Affiliated Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Haiyun Li
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Canling Zhang
- Nursing Department, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong First Medical University, Jinan, China
| | - Aihua Wang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong First Medical University, Jinan, China; Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Shanchao Zhang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong First Medical University, Jinan, China; Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China; School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|