1
|
Dong PF, Liu TB, Chen K, Li D, Li Y, Lian CY, Wang ZY, Wang L. Cadmium targeting transcription factor EB to inhibit autophagy-lysosome function contributes to acute kidney injury. J Adv Res 2024:S2090-1232(24)00297-2. [PMID: 39033876 DOI: 10.1016/j.jare.2024.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/03/2024] [Accepted: 07/13/2024] [Indexed: 07/23/2024] Open
Abstract
INTRODUCTION Environmental and occupational exposure to cadmium (Cd) has been shown to cause acute kidney injury (AKI). Previous studies have demonstrated that autophagy inhibition and lysosomal dysfunction are important mechanisms of Cd-induced AKI. OBJECTIVES Transcription factor EB (TFEB) is a critical transcription regulator that modulates autophagy-lysosome function, but its role in Cd-induced AKI is yet to be elucidated. Thus, in vivo and in vitro studies were conducted to clarify this issue. METHODS AND RESULTS Data firstly showed that reduced TFEB expression and nuclear translocation were evident in Cd-induced AKI models, accompanied by autophagy-lysosome dysfunction. Pharmacological and genetic activation of TFEB improved Cd-induced AKI via alleviating autophagy inhibition and lysosomal dysfunction, whereas Tfeb knockdown further aggravated this phenomenon, suggesting the key role of TFEB in Cd-induced AKI by regulating autophagy. Mechanistically, Cd activated mechanistic target of rapamycin complex 1 (mTORC1) to enhance TFEB phosphorylation and thereby inhibiting TFEB nuclear translocation. Cd also activated chromosome region maintenance 1 (CRM1) to promote TFEB nuclear export. Meanwhile, Cd activated general control non-repressed protein 5 (GCN5) to enhance nuclear TFEB acetylation, resulting in the decreased TFEB transcriptional activity. Moreover, inhibition of CRM1 or GCN5 alleviated Cd-induced AKI by enhancing TFEB activity, respectively. CONCLUSION In summary, these findings reveal that TFEB phosphorylation, nuclear export and acetylation independently suppress TFEB activity to cause Cd-induced AKI via regulating autophagy-lysosome function, suggesting that TFEB activation might be a promising treatment strategy for Cd-induced AKI.
Collapse
Affiliation(s)
- Peng-Fei Dong
- College of Veterinary Medicine, Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, 271017, Shandong Province, China
| | - Tian-Bin Liu
- New Drug Evaluation Center of Shandong Academy of Pharmaceutical Sciences, Shandong Academy of Pharmaceutical Sciences, 989 Xinluo Street, Ji'nan City, 250101, Shandong Province, China
| | - Kai Chen
- New Drug Evaluation Center of Shandong Academy of Pharmaceutical Sciences, Shandong Academy of Pharmaceutical Sciences, 989 Xinluo Street, Ji'nan City, 250101, Shandong Province, China
| | - Dan Li
- Shandong Medicine Technician College, 999 Fengtian Street, Tai'an City, 271016, Shandong Province, China
| | - Yue Li
- College of Veterinary Medicine, Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, 271017, Shandong Province, China
| | - Cai-Yu Lian
- College of Veterinary Medicine, Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, 271017, Shandong Province, China
| | - Zhen-Yong Wang
- College of Veterinary Medicine, Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, 271017, Shandong Province, China
| | - Lin Wang
- College of Veterinary Medicine, Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, 271017, Shandong Province, China.
| |
Collapse
|
2
|
Wang D, Li Q, Xiao C, Wang H, Dong S. Nanoparticles in Periodontitis Therapy: A Review of the Current Situation. Int J Nanomedicine 2024; 19:6857-6893. [PMID: 39005956 PMCID: PMC11246087 DOI: 10.2147/ijn.s465089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 06/10/2024] [Indexed: 07/16/2024] Open
Abstract
Periodontitis is a disease of inflammation that affects the tissues supporting the periodontium. It is triggered by an immunological reaction of the gums to plaque, which leads to the destruction of periodontal attachment structures. Periodontitis is one of the most commonly recognized dental disorders in the world and a major factor in the loss of adult teeth. Scaling and root planing remain crucial for managing patients with persistent periodontitis. Nevertheless, exclusive reliance on mechanical interventions like periodontal surgery, extractions, and root planning is insufficient to halt the progression of periodontitis. In response to the problem of bacterial resistance, some researchers are committed to finding alternative therapies to antibiotics. In addition, some scholars focus on finding new materials to provide a powerful microenvironment for periodontal tissue regeneration and promote osteogenic repair. Nanoparticles possess distinct therapeutic qualities, including exceptional antibacterial, anti-inflammatory, and antioxidant properties, immunomodulatory capacities, and the promotion of bone regeneration ability, which made them can be used for the treatment of periodontitis. However, there are many problems that limit the clinical translation of nanoparticles, such as toxic accumulation in cells, poor correlation between in vitro and in vivo, and poor animal-to-human transmissibility. In this paper, we review the present researches on nanoparticles in periodontitis treatment from the perspective of three main categories: inorganic nanoparticles, organic nanoparticles, and nanocomposites (including nanofibers, hydrogels, and membranes). The aim of this review is to provide a comprehensive and recent update on nanoparticles-based therapies for periodontitis. The conclusion section summarizes the opportunities and challenges in the design and clinical translation of nanoparticles for the treatment of periodontitis.
Collapse
Affiliation(s)
- Di Wang
- The First Outpatient Department, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, People’s Republic of China
| | - Qiqi Li
- The First Outpatient Department, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, People’s Republic of China
| | - Chunsheng Xiao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, People’s Republic of China
| | - Hao Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, People’s Republic of China
| | - Shujun Dong
- The First Outpatient Department, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| |
Collapse
|
3
|
Wang Y, Yin D, Sun X, Zhang W, Ma H, Huang J, Yang C, Wang J, Geng Q. Perfluoroalkyl sulfonate induces cardiomyocyte apoptosis via endoplasmic reticulum stress activation and autophagy flux inhibition. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 930:172582. [PMID: 38649052 DOI: 10.1016/j.scitotenv.2024.172582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/14/2024] [Accepted: 04/17/2024] [Indexed: 04/25/2024]
Abstract
Perfluoroalkyl sulfonate (PFOS) is a commonly used chemical compound that often found in materials such as waterproofing agents, food packaging, and fire retardants. Known for its stability and persistence in the environment, PFOS can enter the human body through various pathways, including water and the food chain, raising concerns about its potential harm to human health. Previous studies have suggested a cardiac toxicity of PFOS, but the specific cellular mechanisms remained unclear. Here, by using AC16 cardiomyocyte as a model to investigate the molecular mechanisms potential the cardiac toxicity of PFOS. Our findings revealed that PFOS exposure reduced cell viability and induces apoptosis in human cardiomyocyte. Proteomic analysis and molecular biological techniques showed that the Endoplasmic Reticulum (ER) stress-related pathways were activated, while the cellular autophagy flux was inhibited in PFOS-exposed cells. Subsequently, we employed strategies such as autophagy activation and ER stress inhibition to alleviate the PFOS-induced apoptosis in AC16 cells. These results collectively suggest that PFOS-induced ER stress activation and autophagy flux inhibition contribute to cardiomyocyte apoptosis, providing new insights into the mechanisms of PFOS-induced cardiomyocyte toxicity.
Collapse
Affiliation(s)
- Yuanhao Wang
- Department of Geriatrics, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Da Yin
- Department of Cardiology, Shenzhen Cardiovascular Minimally Invasive Medical Engineering Technology Research and Development Center, Shenzhen People's Hospital, The First Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Xin Sun
- Department of Cardiology, Shenzhen Cardiovascular Minimally Invasive Medical Engineering Technology Research and Development Center, Shenzhen People's Hospital, The First Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Wei Zhang
- Department of Geriatrics, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Huan Ma
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, No.106 Zhongshan Er Road, Guangzhou, Guangdong, China
| | - Jingnan Huang
- Department of Geriatrics, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Chuanbin Yang
- Department of Geriatrics, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China.
| | - Jigang Wang
- Department of Geriatrics, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China; State Key Laboratory for Quality Esurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Qingshan Geng
- Department of Geriatrics, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China.
| |
Collapse
|
4
|
Bonam SR, Mastrippolito D, Georgel P, Muller S. Pharmacological targets at the lysosomal autophagy-NLRP3 inflammasome crossroads. Trends Pharmacol Sci 2024; 45:81-101. [PMID: 38102020 DOI: 10.1016/j.tips.2023.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 11/14/2023] [Accepted: 11/14/2023] [Indexed: 12/17/2023]
Abstract
Many aspects of cell homeostasis and integrity are maintained by the nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) family pyrin domain-containing 3 (NLRP3) inflammasome. The NLRP3 oligomeric protein complex assembles in response to exogenous and endogenous danger signals. This inflammasome has also been implicated in the pathogenesis of a range of disease conditions, particularly chronic inflammatory diseases. Given that NLRP3 modulates autophagy, which is also a key regulator of inflammasome activity, excessive inflammation may be controlled by targeting this intersecting pathway. However, specific niche areas of NLRP3-autophagy interactions and their reciprocal regulatory mechanisms remain underexplored. Consequently, we lack treatment methods specifically targeting this pivotal axis. Here, we discuss the potential of such strategies in the context of autoimmune and metabolic diseases and propose some research avenues.
Collapse
Affiliation(s)
- Srinivasa Reddy Bonam
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Dylan Mastrippolito
- CNRS-University of Strasbourg, Biotechnology and Cell Signaling, Illkirch, France; Strasbourg Institute of Drug Discovery and Development (IMS), Strasbourg, France
| | - Philippe Georgel
- CNRS-University of Strasbourg, Biotechnology and Cell Signaling, Illkirch, France; Strasbourg Institute of Drug Discovery and Development (IMS), Strasbourg, France; Fédération Hospitalo-Universitaire (FHU) OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg University, Strasbourg, France
| | - Sylviane Muller
- CNRS-University of Strasbourg, Biotechnology and Cell Signaling, Illkirch, France; Strasbourg Institute of Drug Discovery and Development (IMS), Strasbourg, France; Fédération Hospitalo-Universitaire (FHU) OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg University, Strasbourg, France; University of Strasbourg Institute for Advanced Study (USIAS), Strasbourg, France.
| |
Collapse
|
5
|
Dwivedi R, Baindara P. Differential Regulation of TFEB-Induced Autophagy during Mtb Infection and Starvation. Microorganisms 2023; 11:2944. [PMID: 38138088 PMCID: PMC10746089 DOI: 10.3390/microorganisms11122944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/01/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Through the promotion of phagolysosome formation, autophagy has emerged as a crucial mechanism to eradicate intracellular Mycobacterium tuberculosis (Mtb). A cell-autonomous host defense mechanism called lysosome biogenesis and autophagy transports cytoplasmic cargos and bacterial phagosomes to lysosomes for destruction during infection. Similar occurrences occurred in stressful or starvation circumstances and led to autophagy, which is harmful to the cell. It is interesting to note that under both hunger and infection states, the transcription factor EB (TFEB) acts as a master regulator of lysosomal activities and autophagy. This review highlighted recent research on the multitier regulation of TFEB-induced autophagy by a variety of host effectors and Mtb sulfolipid during Mtb infection and starvation. In general, the research presented here sheds light on how lysosome biogenesis and autophagy are differentially regulated by the TFEB during Mtb infection and starvation.
Collapse
Affiliation(s)
- Richa Dwivedi
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Piyush Baindara
- Radiation Oncology, NextGen Precision Health, School of Medicine, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
6
|
Pang P, Zhang X, Yuan J, Yan H, Yan D. Acrylamide interferes with autophagy and induces apoptosis in Neuro-2a cells by interfering with TFEB-regulated lysosomal function. Food Chem Toxicol 2023; 177:113818. [PMID: 37172712 DOI: 10.1016/j.fct.2023.113818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/28/2023] [Accepted: 05/08/2023] [Indexed: 05/15/2023]
Abstract
Acrylamide (ACR), a well-documented human neurotoxicant that is widely exists in starchy foods. More than 30% of human daily energy is provided by ACR-containing foods. Evidence indicated that ACR can induce apoptosis and inhibit autophagy, but the mechanisms are limited. Transcription Factor EB (TFEB) is a major transcriptional regulator of the autophagy-lysosomal biogenesis that regulates autophagy processes and cell degradation. Our study aimed to investigated the potential mechanisms of TFEB-regulated lysosomal function in ACR-caused autophagic flux inhibition and apoptosis in Neuro-2a cells. Our results found that ACR exposure inhibited the autophagic flux, as revealed by the elevated LC3-II/LC3-I and p62 levels and a notable increased autophagosomes. ACR exposure reduced the amounts of LAMP1 and mature cathepsin D and caused an accumulation of ubiquitinated proteins, which suggests lysosomal dysfunction. In addition, ACR increased cellular apoptosis via decreasing Bcl-2 expression, increasing Bax and cleaved caspase-3 expression, and raising the apoptotic rate. Interestingly, TFEB overexpression alleviated the ACR-induced lysosomal dysfunction, and then mitigated the autophagy flux inhibition and cellular apoptosis. On the other hand, TFEB knockdown exacerbated the ACR-induced lysosomal dysfunction, autophagy flux inhibition, and cellular apoptosis. These findings strongly suggested that TFEB- regulated lysosomal function is responsible for ACR-caused autophagic flux inhibition and apoptosis in Neuro-2a cells. The present study hopes to explore new sensitive indicators in the mechanism of ACR neurotoxicity and thus provide new targets for the prevention and treatment of ACR intoxication.
Collapse
Affiliation(s)
- Pengcheng Pang
- Department of Pathology, Renmin Hospital of Wuhan University, 238 Jiefang-Road, Wuchang District, Wuhan, 430060, PR China; Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan University, 238 Jiefang-Road, Wuchang District, Wuhan, 430060, PR China
| | - Xing Zhang
- Department of Health Toxicology, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong-Road, Wuhan, 430030, PR China
| | - Jingping Yuan
- Department of Pathology, Renmin Hospital of Wuhan University, 238 Jiefang-Road, Wuchang District, Wuhan, 430060, PR China
| | - Hong Yan
- Department of Health Toxicology, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong-Road, Wuhan, 430030, PR China
| | - Dandan Yan
- Department of Pathology, Renmin Hospital of Wuhan University, 238 Jiefang-Road, Wuchang District, Wuhan, 430060, PR China; Department of Health Toxicology, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong-Road, Wuhan, 430030, PR China.
| |
Collapse
|
7
|
Wang Z, Cui J, Li D, Ran S, Huang J, Chen G. Morin exhibits a neuroprotective effect in MPTP-induced Parkinson's disease model via TFEB/AMPK-mediated mitophagy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 116:154866. [PMID: 37209604 DOI: 10.1016/j.phymed.2023.154866] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 04/26/2023] [Accepted: 05/08/2023] [Indexed: 05/22/2023]
Abstract
BACKGROUND Parkinson's disease (PD) is one of the most common neurodegenerative diseases in the world. Mitophagy has been implicated in PD etiology for decades and its pharmacological activation is recognized as a promising treatment strategy for PD. For mitophagy initiation, low mitochondrial membrane potential (ΔΨm) is essential. We identified a natural compound morin that could induce mitophagy without affecting ΔΨm. Morin is a flavonoid that can be isolated from fruits like mulberry. PURPOSE To reveal the effect of morin on the PD mice model and their potential underlying molecular mechanism. METHODS Mitophagy process induced by morin in N2a cells meditation were measured using flow cytometry and immunofluorescence. JC-1 fluorescence dye used to detect the mitochondrial membrane potential (ΔΨm). The TFEB nuclear translocation were examined by immunofluorescence staining and western blot assay. The PD mice model was induced by MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) intraperitoneal administration. RESULTS We found that morin also promoted nuclear translocation of the mitophagy regulator TFEB and activated the AMPK-ULK1 pathway. In MPTP-induced PD in vivo models, morin protected DA neurons from MPTP neurotoxicity and ameliorated behavioral deficit. CONCLUSION Although morin was previously reported to be neuroprotective in PD, the detailed molecular mechanisms remain to be elucidated. For the first time, we report morin served as a novel and safe mitophagy enhancer underlying AMPK-ULK1 pathway and exhibited anti-Parkinsonian effects indicating its potential as a clinical drug for PD treatment.
Collapse
Affiliation(s)
- Ziying Wang
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China.
| | - Jinshuai Cui
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Dongni Li
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Shuzhen Ran
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Junqing Huang
- Formula-pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Gang Chen
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China.
| |
Collapse
|
8
|
Li Z, Chen L, Zhang D, Huang X, Yang J, Li W, Wang C, Meng X, Huang G. Intranasal 15d-PGJ2 inhibits the growth of rat lactotroph pituitary neuroendocrine tumors by inducing PPARγ-dependent apoptotic and autophagic cell death. Front Neurosci 2023; 17:1109675. [PMID: 37250410 PMCID: PMC10213263 DOI: 10.3389/fnins.2023.1109675] [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: 11/28/2022] [Accepted: 04/21/2023] [Indexed: 05/31/2023] Open
Abstract
PPARγ agonists have been reported to induce cell death in pituitary neuroendocrine tumor (PitNET) cell cultures. However, the therapeutic effects of PPARγ agonists in vivo remain unclear. In the present study, we found that intranasal 15d-PGJ2, an endogenous PPARγ agonist, resulted in growth suppression of Fischer 344 rat lactotroph PitNETs induced by subcutaneous implantation with a mini-osmotic pump containing estradiol. Intranasal 15d-PGJ2 reduced the volume and weight of the pituitary gland and the level of serum prolactin (PRL) in rat lactotroph PitNETs. 15d-PGJ2 treatment attenuated pathological changes and significantly decreased the ratio of PRL/pituitary-specific transcription factor 1 (Pit-1) and estrogen receptor α (ERα)/Pit-1 double-positive cells. Moreover, 15d-PGJ2 treatment induced apoptosis in the pituitary gland characterized by an increased ratio of TUNEL-positive cells, cleavage of caspase-3, and elevated activity of caspase-3. 15d-PGJ2 treatment decreased the levels of cytokines, including TNF-α, IL-1β, and IL-6. Furthermore, 15d-PGJ2 treatment markedly increased the protein expression of PPARγ and blocked autophagic flux, as evidenced by the accumulation of LC3-II and SQSTM1/p62 and the decrease in LAMP-1 expression. Importantly, all these effects mediated by 15d-PGJ2 were abolished by cotreatment with the PPARγ antagonist GW9662. In conclusion, intranasal 15d-PGJ2 suppressed the growth of rat lactotroph PitNETs by inducing PPARγ-dependent apoptotic and autophagic cell death. Therefore, 15d-PGJ2 may be a potential new drug for lactotroph PitNETs.
Collapse
|
9
|
Contreras PS, Tapia PJ, Jeong E, Ghosh S, Altan-Bonnet N, Puertollano R. Beta-coronaviruses exploit cellular stress responses by modulating TFEB and TFE3 activity. iScience 2023; 26:106169. [PMID: 36785787 PMCID: PMC9908431 DOI: 10.1016/j.isci.2023.106169] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 01/09/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Beta-coronaviruses have emerged as a severe threat to global health. Undercovering the interplay between host and beta-coronaviruses is essential for understanding disease pathogenesis and developing efficient treatments. Here we report that the transcription factors TFEB and TFE3 translocate from the cytosol to the nucleus in response to beta-coronavirus infection by a mechanism that requires activation of calcineurin phosphatase. In the nucleus, TFEB and TFE3 bind to the promoter of multiple lysosomal and immune genes. Accordingly, MHV-induced upregulation of immune regulators is significantly decreased in TFEB/TFE3-depleted cells. Conversely, over-expression of either TFEB or TFE3 is sufficient to increase expression of several cytokines and chemokines. The reduced immune response observed in the absence of TFEB and TFE3 results in increased cellular survival of infected cells but also in reduced lysosomal exocytosis and decreased viral infectivity. These results suggest a central role of TFEB and TFE3 in cellular response to beta-coronavirus infection.
Collapse
Affiliation(s)
- Pablo S. Contreras
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Pablo J. Tapia
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Eutteum Jeong
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sourish Ghosh
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nihal Altan-Bonnet
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Rosa Puertollano
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
10
|
Yang J, Hasenbilige, Bao S, Luo S, Jiang L, Li Q, Kong Y, Cao J. Inhibition of ATF4-mediated elevation of both autophagy and AKT/mTOR was involved in antitumorigenic activity of curcumin. Food Chem Toxicol 2023; 173:113609. [PMID: 36640941 DOI: 10.1016/j.fct.2023.113609] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 12/22/2022] [Accepted: 01/09/2023] [Indexed: 01/13/2023]
Abstract
Curcumin, a natural hydrophobic polyphenol, carries significant anticancer activity. The protein kinase B (AKT)/the mammalian target of the rapamycin (mTOR) pathway and autophagy are well known to be involved in carcinogenesis, and usually, inhibition of mTOR is the main reason to promote autophagy. In this study, however, autophagy and mTOR were found to be inhibited simultaneously by curcumin treatments, and both of them played an important role in the effect of curcumin on suppressing the growth of A549 cells. Tunicamycin (TM), the activator of Endoplasmic Reticulum (ER) stress, increased both autophagy and AKT/mTOR, while curcumin could significantly decrease TM-induced autophagy and AKT/mTOR. Furthermore, curcumin could inhibit TM-induced aerobic glycolysis in A549 cells, and decrease the level of cycle-related and migration-related proteins. Blocking activating transcription factor 4 (ATF4) by siRNA strongly reduced both the expression of autophagy-related proteins and AKT/mTOR. ChIP assay illustrated that ATF4 protein could bind to the promotor sequence of either ATG4B or AKT1. The transplantation tumor experiment showed that the weight and volume of the transplanted tumors were reduced significantly in the BALB/c mice subcutaneously injected with A549 cells treated with curcumin. Moreover, intranasal administration of curcumin decreased the protein level of autophagy, AKT/mTOR and ER stress in lung tissues of BALB/c mice. Taken together, our results demonstrated that inhibition of ER stress-dependent ATF4-mediated autophagy and AKT/mTOR pathway plays an important role in anticancer effect of curcumin.
Collapse
Affiliation(s)
- Jie Yang
- Department of Occupational and Environmental Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China
| | - Hasenbilige
- Department of Occupational and Environmental Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China
| | - Shibo Bao
- Department of Occupational and Environmental Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China
| | - Shengxiang Luo
- Department of Occupational and Environmental Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China
| | - Liping Jiang
- Department of Occupational and Environmental Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China
| | - Qiujuan Li
- Department of Occupational and Environmental Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China
| | - Ying Kong
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, 116044, China.
| | - Jun Cao
- Department of Occupational and Environmental Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China.
| |
Collapse
|
11
|
Zeng P, Liu YC, Wang XM, Ye CY, Sun YW, Su HF, Qiu SW, Li YN, Wang Y, Wang YC, Ma J, Li M, Tian Q. Targets and mechanisms of Alpinia oxyphylla Miquel fruits in treating neurodegenerative dementia. Front Aging Neurosci 2022; 14:1013891. [PMID: 36533181 PMCID: PMC9749063 DOI: 10.3389/fnagi.2022.1013891] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 11/10/2022] [Indexed: 11/04/2023] Open
Abstract
The dried and ripe fruits of Alpinia oxyphylla and ripe fruits of Alpinia oxyphylla Miquel (AO) have the effects of tonifying kidney-essence and nourishing intelligence and thus have been widely used in treating dementia. Alzheimer's disease (AD) is a typical form of neurodegenerative dementia with kidney-essence deficiency in Traditional Chinese Medicine (TCM). So far, there is a lack of systematic studies on the biological basis of tonifying kidney-essence and nourishing intelligence and the corresponding phytochemicals. In this study, we investigated the targets of AO in tonifying kidney-essence and nourishing intelligence based on the key pathophysiological processes of neurodegenerative dementia. According to ultra-high-performance liquid chromatography with triple quadrupole mass spectrometry data and Lipinski's rule of five, 49 bioactive phytochemicals from AO were identified, and 26 of them were found to target 168 key molecules in the treatment of neurodegenerative dementia. Nine phytochemicals of AO were shown to target acetylcholinesterase (ACHE), and 19 phytochemicals were shown to target butyrylcholinesterase (BCHE). A database of neurodegenerative dementia with kidney-essence deficiency involving 731 genes was constructed. Furthermore, yakuchinone B, 5-hydroxy-1,7-bis (4-hydroxy-3-methoxyphenyl) heptan-3-one (5-HYD), oxyhylladiketone, oxyphyllacinol, butyl-β-D-fructopyranoside, dibutyl phthalate, chrysin, yakuchinone A, rhamnetin, and rhamnocitrin were identified as the key phytochemicals from AO that regulate the pathogenesis of neurodegenerative dementia in a multitargeted manner. The approach of studying the pharmacological mechanism underlying the effects of medicinal plants and the biological basis of TCM syndrome may be helpful in studying the translation of TCM.
Collapse
Affiliation(s)
- Peng Zeng
- Key Laboratory of Neurological Disease of National Education Ministry, School of Basic Medicine of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Histology and Embryology, School of Basic Medicine, Hengyang Medical College, University of South China, Hengyang, China
| | - Yuan-Cheng Liu
- Key Laboratory of Neurological Disease of National Education Ministry, School of Basic Medicine of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Ming Wang
- Key Laboratory of Neurological Disease of National Education Ministry, School of Basic Medicine of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chao-Yuan Ye
- Key Laboratory of Neurological Disease of National Education Ministry, School of Basic Medicine of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi-Wen Sun
- Key Laboratory of Neurological Disease of National Education Ministry, School of Basic Medicine of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong-Fei Su
- Key Laboratory of Neurological Disease of National Education Ministry, School of Basic Medicine of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuo-Wen Qiu
- Key Laboratory of Neurological Disease of National Education Ministry, School of Basic Medicine of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ya-Nan Li
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Yao Wang
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Yan-Chun Wang
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Jun Ma
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Man Li
- Key Laboratory of Neurological Disease of National Education Ministry, School of Basic Medicine of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Tian
- Key Laboratory of Neurological Disease of National Education Ministry, School of Basic Medicine of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
12
|
Ahsan MJ, Ali A, Ali A, Thiriveedhi A, Bakht MA, Yusuf M, Salahuddin, Afzal O, Altamimi AS. Pyrazoline Containing Compounds as Therapeutic Targets for Neurodegenerative Disorders. ACS OMEGA 2022; 7:38207-38245. [PMID: 36340076 PMCID: PMC9631758 DOI: 10.1021/acsomega.2c05339] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 10/03/2022] [Indexed: 09/21/2023]
Abstract
Pyrazolines are a significant class of heterocyclic compounds with essential biological activities. They are quite stable, which has inspired medicinal chemists to experiment with the ring's structure in many different ways to create a variety of pharmacological activities. The structures of numerous commercially available therapeutic agents contain a pyrazoline ring. Pyrazolines are well-known for their ability to treat neurodegenerative diseases. The neurodegenerative diseases that affect huge populations globally include Alzheimer's disease (AD), Parkinson's disease (PD), and psychiatric disorders. The neuroprotective properties of pyrazolines published since 2003 are covered in the current review. Structure-activity relationships (SARs), molecular docking simulation, anticholinesterase (anti-AChE), and monoamine oxidase (MAO A/B) inhibitory actions are all covered in this article. Pyrazolines were discovered to have beneficial effects in the management of AD and were revealed to be inhibitors of acetylcholine esterase (AChE) and beta-amyloid (Aβ) plaques. They were discovered to be efficient against PD and also targeted MAO B and COMT. It was discovered that the pyrazolines block MAO A to treat psychiatric diseases. Pyrazolines are significant heteroaromatic scaffolds with a variety of biological functions. They were discovered to be remarkably stable and serve as an indispensable anchor for the development of new drugs. By blocking AChE and MAOs, they may be used to treat neurodegenerative diseases. The discussion outlined here is an essential and helpful resource for medicinal chemists who are investigating and applying pyrazolines in neurodegenerative research initiatives as well as to expedite future research programs on neurodegenerative disorders.
Collapse
Affiliation(s)
- Mohamed Jawed Ahsan
- Department
of Pharmaceutical Chemistry, Maharishi Arvind
College of Pharmacy, Jaipur, Rajasthan 302 039, India
| | - Amena Ali
- Department
of Pharmaceutical Chemistry, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Abuzer Ali
- Department
of Pharmacognosy, College of Pharmacy, Taif
University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Arunkumar Thiriveedhi
- Vignan’s
Foundation for Science, Technology & Research Deemed to be University
Guntur, Vadlamudi, Andhra Pradesh 522213, India
| | - Mohammed A. Bakht
- Department
of Chemistry, College of Science and Humanity Studies, Prince Sattam Bin Abdulaziz University, P.O. Box 83, Al-Kharj 11942, Saudi Arabia
| | - Mohammad Yusuf
- Department
of Clinical Pharmacy, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Salahuddin
- Department
of Pharmaceutical Chemistry, Noida Institute
of Technology (Pharmacy Institute), Knowledge Park-2, Greater Noida, Uttar
Pradesh 201 306, India
| | - Obaid Afzal
- Department
of Pharmaceutical Chemistry, College of
Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box- 173, Al-Kharj 11942, Saudi Arabia
| | - Abdulmalik Saleh
Alfawaz Altamimi
- Department
of Pharmaceutical Chemistry, College of
Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box- 173, Al-Kharj 11942, Saudi Arabia
| |
Collapse
|
13
|
Inhibited transcription factor EB function induces reactive oxygen species overproduction to promote pyroptosis in cadmium-exposed renal tubular epithelial cells. Chem Biol Interact 2022; 368:110249. [DOI: 10.1016/j.cbi.2022.110249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/12/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022]
|
14
|
Jeong YH, Kim TI, Oh YC, Ma JY. Selaginella tamariscina Inhibits Glutamate-Induced Autophagic Cell Death by Activating the PI3K/AKT/mTOR Signaling Pathways. Int J Mol Sci 2022; 23:ijms231911445. [PMID: 36232743 PMCID: PMC9569781 DOI: 10.3390/ijms231911445] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/20/2022] [Accepted: 09/23/2022] [Indexed: 11/21/2022] Open
Abstract
Glutamate-induced neural toxicity in autophagic neuron death is partially mediated by increased oxidative stress. Therefore, reducing oxidative stress in the brain is critical for treating or preventing neurodegenerative diseases. Selaginella tamariscina is a traditional medicinal plant for treating gastrointestinal bleeding, hematuria, leucorrhea, inflammation, chronic hepatitis, gout, and hyperuricemia. We investigate the inhibitory effects of Selaginella tamariscina ethanol extract (STE) on neurotoxicity and autophagic cell death in glutamate-exposed HT22 mouse hippocampal cells. STE significantly increased cell viability and mitochondrial membrane potential and decreased the expression of reactive oxygen species, lactate dehydrogenase release, and cell apoptosis in glutamate-exposed HT22 cells. In addition, while glutamate induced the excessive activation of mitophagy, STE attenuated glutamate-induced light chain (LC) 3 II and Beclin-1 expression and increased p62 expression. Furthermore, STE strongly enhanced the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) phosphorylation activation. STE strongly inhibited glutamate-induced autophagy by activating the PI3K/Akt/mTOR signaling pathway. In contrast, the addition of LY294002, a PI3K/Akt inhibitor, remarkably suppressed cell viability and p-Akt and p62 expression, while markedly increasing the expression of LC3 II and Beclin-1. Our findings indicate that autophagy inhibition by activating PI3K/Akt/mTOR phosphorylation levels could be responsible for the neuroprotective effects of STE on glutamate neuronal damage.
Collapse
Affiliation(s)
| | | | - You-Chang Oh
- Correspondence: (Y.-C.O.); (J.Y.M.); Tel.: +82-53-940-3882 (Y.-C.O.); +82-53-940-3812 (J.Y.M.)
| | - Jin Yeul Ma
- Correspondence: (Y.-C.O.); (J.Y.M.); Tel.: +82-53-940-3882 (Y.-C.O.); +82-53-940-3812 (J.Y.M.)
| |
Collapse
|
15
|
Li M, Liu G, Yuan LX, Yang J, Liu J, Li Z, Yang C, Wang J. Triphenyl phosphate (TPP) promotes hepatocyte toxicity via induction of endoplasmic reticulum stress and inhibition of autophagy flux. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 840:156461. [PMID: 35660595 DOI: 10.1016/j.scitotenv.2022.156461] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/30/2022] [Accepted: 05/31/2022] [Indexed: 06/15/2023]
Abstract
Triphenyl phosphate (TPP), a commonly used organophosphate flame retardant, is frequently found in environmental and biota samples, indicating widespread human exposure. Recent studies have shown that TPP causes hepatotoxicity, but the underlying cellular mechanisms are not fully elucidated. Here, by using normal hepatocyte AML12 cells as a model, we showed that TPP induced apoptotic cell death. RNA sequencing analyses revealed that differentially expressed genes induced by TPP were related to endoplasmic reticulum (ER) stress and autophagy. Immunostaining and western blot results further confirmed that TPP activated ER stress. Interestingly, though TPP increased LC3-II, a canonical marker for autophagy, TPP inhibited autophagy flux rather than induced autophagy. Interestingly, TPP-induced ER stress facilitated autophagy flux inhibition and apoptosis. Furthermore, inhibition of autophagy aggravated, and activation of autophagy attenuated apoptosis induced by TPP. Collectively, these results uncovered that ER stress and autophagy flux inhibition were responsible for TPP-induced apoptosis in mouse hepatocytes. Thus, our foundlings provided novel insight into the potential mechanisms of TPP-induced hepatocyte toxicity.
Collapse
Affiliation(s)
- Miaoran Li
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Geriatric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China
| | - Gang Liu
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Li-Xia Yuan
- School of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou 510515, China
| | - Jing Yang
- Department of Geriatric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China
| | - Jing Liu
- Department of Geriatric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China
| | - Zhijie Li
- Department of Geriatric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China
| | - Chuanbin Yang
- Department of Geriatric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China.
| | - Jigang Wang
- Department of Geriatric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China; School of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou 510515, China; Artemisinin Research Center, Institute of Chinese Materia Medica, Chinese Academy of Chinese Medical Sciences, Beijing 100700, China.
| |
Collapse
|
16
|
Wang S, Wang G, Wu W, Xu Z, Yang J, Cao M, Wang Q, Wang J, Yang C, Zhang W. Autophagy activation by dietary piceatannol enhances the efficacy of immunogenic chemotherapy. Front Immunol 2022; 13:968686. [PMID: 35979349 PMCID: PMC9376326 DOI: 10.3389/fimmu.2022.968686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Immunogenic cell death (ICD) promotes the immune antitumor response via releasing damage-associated molecular patterns (DAMPs) from dying tumor cells. The induction of autophagy improves the efficacy of multiple immunogenic chemotherapies. Here, we show that piceatannol, a dietary phenolic compound that is widely distributed in multiple fruits and vegetables such as grapes, blueberries, and mushrooms, induces autophagy and enhances oxaliplatin (OXA)-induced anticancer immune response. Specifically, piceatannol enhanced OXA-induced release of DAMPs, several key hallmarks of ICD including ATP release, cell surface exposure of calreticulin, and high-mobility group box 1 (HMGB1) release. Mechanistically, piceatannol promoted autophagy via activating TFEB/TFE3, two key transcription factors of the autophagy-lysosome pathway, and inhibiting autophagy attenuated piceatannol plus OXA-induced ATP release. Furthermore, piceatannol induced endoplasmic reticulum stress, which is critical for its role in enhancing OXA-induced cell surface exposure of calreticulin, another key hallmark of ICD. Consistently, the combination of piceatannol with OXA promoted the anticancer effects in immunocompetent mice. Taken together, our results indicate the importance and great potential of dietary piceatannol in cancer immunotherapy. Therefore, piceatannol may be used as an ICD enhancer that improves the efficacy of chemotherapeutics such as OXA in cancer treatment with minimized toxicity.
Collapse
Affiliation(s)
- Shuang Wang
- Department of Geriatrics, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Guangsuo Wang
- Department of Thoracic Surgery, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Weiqing Wu
- Department of Health Management, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Zhenglei Xu
- Department of Gastroenterology, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Jing Yang
- Department of Geriatrics, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Min Cao
- Department of Geriatrics, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Qi Wang
- Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
- *Correspondence: Wei Zhang, ; Chuanbin Yang, ; Jigang Wang, ; Qi Wang,
| | - Jigang Wang
- Department of Geriatrics, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
- Artemisinin Research Center, Institute of Chinese Materia Medica, Chinese Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Wei Zhang, ; Chuanbin Yang, ; Jigang Wang, ; Qi Wang,
| | - Chuanbin Yang
- Department of Geriatrics, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
- *Correspondence: Wei Zhang, ; Chuanbin Yang, ; Jigang Wang, ; Qi Wang,
| | - Wei Zhang
- Department of Geriatrics, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, China
- *Correspondence: Wei Zhang, ; Chuanbin Yang, ; Jigang Wang, ; Qi Wang,
| |
Collapse
|
17
|
Zhang W, Xia S, Zhong X, Gao G, Yang J, Wang S, Cao M, Liang Z, Yang C, Wang J. Characterization of 2,2'4,4'-Tetrabromodiphenyl ether (BDE47)-induced testicular toxicity via single-cell RNA-sequencing. PRECISION CLINICAL MEDICINE 2022; 5:pbac016. [PMID: 35875604 PMCID: PMC9306015 DOI: 10.1093/pcmedi/pbac016] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 06/12/2022] [Indexed: 11/30/2022] Open
Abstract
Background The growing male reproductive diseases have been linked to higher exposure to certain environmental compounds such as 2,2′,4,4′-tetrabromodiphenyl ether (BDE47) that are widely distributed in the food chain. However, the specific underlying molecular mechanisms for BDE47-induced male reproductive toxicity are not completely understood. Methods Here, for the first time, advanced single-cell RNA sequencing (ScRNA-seq) was employed to dissect BDE47-induced prepubertal testicular toxicity in mice from a pool of 76 859 cells. Results Our ScRNA-seq results revealed shared and heterogeneous information of differentially expressed genes, signaling pathways, transcription factors, and ligands-receptors in major testicular cell types in mice upon BDE47 treatment. Apart from disruption of hormone homeostasis, BDE47 was discovered to downregulate multiple previously unappreciated pathways such as double-strand break repair and cytokinesis pathways, indicative of their potential roles involved in BDE47-induced testicular injury. Interestingly, transcription factors analysis of ScRNA-seq results revealed that Kdm5b (lysine-specific demethylase 5B), a key transcription factor required for spermatogenesis, was downregulated in all germ cells as well as in Sertoli and telocyte cells in BDE47-treated testes of mice, suggesting its contribution to BDE47-induced impairment of spermatogenesis. Conclusions Overall, for the first time, we established the molecular cell atlas of mice testes to define BDE47-induced prepubertal testicular toxicity using the ScRNA-seq approach, providing novel insight into our understanding of the underlying mechanisms and pathways involved in BDE47-associated testicular injury at a single-cell resolution. Our results can serve as an important resource to further dissect the potential roles of BDE47, and other relevant endocrine-disrupting chemicals, in inducing male reproductive toxicity.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology) , Shenzhen 518020 , China
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University , Guangzhou 510632 , China
| | - Siyu Xia
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology) , Shenzhen 518020 , China
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University , Guangzhou 510632 , China
| | - Xiaoru Zhong
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology) , Shenzhen 518020 , China
| | - Guoyong Gao
- Department of Spine Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology) , Shenzhen 518020 , China
| | - Jing Yang
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology) , Shenzhen 518020 , China
| | - Shuang Wang
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology) , Shenzhen 518020 , China
| | - Min Cao
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology) , Shenzhen 518020 , China
| | - Zhen Liang
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology) , Shenzhen 518020 , China
| | - Chuanbin Yang
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology) , Shenzhen 518020 , China
| | - Jigang Wang
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology) , Shenzhen 518020 , China
- Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences , Beijing 100700 , China
- Center for Reproductive Medicine, Dongguan Maternal and Child Health Care Hospital, Southern Medical University , Dongguan, 523125, Guangdong , China
| |
Collapse
|
18
|
Fung TY, Iyaswamy A, Sreenivasmurthy SG, Krishnamoorthi S, Guan XJ, Zhu Z, Su CF, Liu J, Kan Y, Zhang Y, Wong HLX, Li M. Klotho an Autophagy Stimulator as a Potential Therapeutic Target for Alzheimer’s Disease: A Review. Biomedicines 2022; 10:biomedicines10030705. [PMID: 35327507 PMCID: PMC8945569 DOI: 10.3390/biomedicines10030705] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/17/2022] [Accepted: 03/17/2022] [Indexed: 02/04/2023] Open
Abstract
Alzheimer’s disease (AD) is an age-associated neurodegenerative disease; it is the most common cause of senile dementia. Klotho, a single-pass transmembrane protein primarily generated in the brain and kidney, is active in a variety of metabolic pathways involved in controlling neurodegeneration and ageing. Recently, many studies have found that the upregulation of Klotho can improve pathological cognitive deficits in an AD mice model and have demonstrated that Klotho plays a role in the induction of autophagy, a major contributing factor for AD. Despite the close association between Klotho and neurodegenerative diseases, such as AD, the underlying mechanism by which Klotho contributes to AD remains poorly understood. In this paper, we will introduce the expression, location and structure of Klotho and its biological functions. Specifically, this review is devoted to the correlation of Klotho protein and the AD phenotype, such as the effect of Klotho in upregulating the amyloid-beta clearance and in inducing autophagy for the clearance of toxic proteins, by regulating the autophagy lysosomal pathway (ALP). In summary, the results of multiple studies point out that targeting Klotho would be a potential therapeutic strategy in AD treatment.
Collapse
Affiliation(s)
- Tsz Yan Fung
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (T.Y.F.); (S.G.S.); (S.K.); (X.-J.G.); (Z.Z.); (C.-F.S.); (J.L.); (Y.K.)
| | - Ashok Iyaswamy
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (T.Y.F.); (S.G.S.); (S.K.); (X.-J.G.); (Z.Z.); (C.-F.S.); (J.L.); (Y.K.)
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen 518057, China
- Correspondence: or (A.I.); (H.L.X.W.); (M.L.); Tel.: +852-3411-2919 (M.L.); Fax: +852-3411-2461 (M.L.)
| | - Sravan G. Sreenivasmurthy
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (T.Y.F.); (S.G.S.); (S.K.); (X.-J.G.); (Z.Z.); (C.-F.S.); (J.L.); (Y.K.)
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen 518057, China
| | - Senthilkumar Krishnamoorthi
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (T.Y.F.); (S.G.S.); (S.K.); (X.-J.G.); (Z.Z.); (C.-F.S.); (J.L.); (Y.K.)
- Centre for Trans-Disciplinary Research, Department of Pharmacology, Saveetha Dental College and Hospitals, Chennai 600077, Tamil Nadu, India
| | - Xin-Jie Guan
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (T.Y.F.); (S.G.S.); (S.K.); (X.-J.G.); (Z.Z.); (C.-F.S.); (J.L.); (Y.K.)
| | - Zhou Zhu
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (T.Y.F.); (S.G.S.); (S.K.); (X.-J.G.); (Z.Z.); (C.-F.S.); (J.L.); (Y.K.)
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen 518057, China
| | - Cheng-Fu Su
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (T.Y.F.); (S.G.S.); (S.K.); (X.-J.G.); (Z.Z.); (C.-F.S.); (J.L.); (Y.K.)
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen 518057, China
| | - Jia Liu
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (T.Y.F.); (S.G.S.); (S.K.); (X.-J.G.); (Z.Z.); (C.-F.S.); (J.L.); (Y.K.)
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen 518057, China
| | - Yuxuan Kan
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (T.Y.F.); (S.G.S.); (S.K.); (X.-J.G.); (Z.Z.); (C.-F.S.); (J.L.); (Y.K.)
| | - Yuan Zhang
- Shenzhen Key Laboratory of Neurosurgery, Department of Neurosurgery, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518025, China;
| | - Hoi Leong Xavier Wong
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
- Correspondence: or (A.I.); (H.L.X.W.); (M.L.); Tel.: +852-3411-2919 (M.L.); Fax: +852-3411-2461 (M.L.)
| | - Min Li
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (T.Y.F.); (S.G.S.); (S.K.); (X.-J.G.); (Z.Z.); (C.-F.S.); (J.L.); (Y.K.)
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen 518057, China
- Correspondence: or (A.I.); (H.L.X.W.); (M.L.); Tel.: +852-3411-2919 (M.L.); Fax: +852-3411-2461 (M.L.)
| |
Collapse
|
19
|
Yang C, Su C, Iyaswamy A, Krishnamoorthi SK, Zhu Z, Yang S, Tong BC, Liu J, Sreenivasmurthy SG, Guan X, Kan Y, Wu AJ, Huang AS, Tan J, Cheung K, Song J, Li M. Celastrol enhances transcription factor EB (TFEB)-mediated autophagy and mitigates Tau pathology: Implications for Alzheimer’s disease therapy. Acta Pharm Sin B 2022; 12:1707-1722. [PMID: 35847498 PMCID: PMC9279716 DOI: 10.1016/j.apsb.2022.01.017] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 12/11/2021] [Accepted: 12/16/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD), characterized by the accumulation of protein aggregates including phosphorylated Tau aggregates, is the most common neurodegenerative disorder with limited therapeutic agents. Autophagy plays a critical role in the degradation of phosphorylated Tau aggregates, and transcription factor EB (TFEB) is a master regulator of autophagy and lysosomal biogenesis. Thus, small-molecule autophagy enhancers targeting TFEB hold promise for AD therapy. Here, we found that celastrol, an active ingredient isolated from the root extracts of Tripterygium wilfordii (Lei Gong Teng in Chinese) enhanced TFEB-mediated autophagy and lysosomal biogenesis in vitro and in mouse brains. Importantly, celastrol reduced phosphorylated Tau aggregates and attenuated memory dysfunction and cognitive deficits in P301S Tau and 3xTg mice, two commonly used AD animal models. Mechanistical studies suggest that TFEB-mediated autophagy-lysosomal pathway is responsible for phosphorylated Tau degradation in response to celastrol. Overall, our findings indicate that Celastrol is a novel TFEB activator that promotes the degradation of phosphorylated Tau aggregates and improves memory in AD animal models. Therefore, Celastrol shows potential as a novel agent for the treatment and/or prevention of AD and other tauopathies.
Collapse
|