1
|
Sun M, Cai X, Lan Z, Liu M, Zhou M, Tang Y, Liu Y, Zhang X, Zhao X, Zhou Y, Zhang J, Meng Z. The lysosomal-associated membrane protein 2-macroautophagy pathway is involved in the regulatory effects of hippocampal aromatase on Aβ accumulation and AD-like behavior. Life Sci 2025; 366-367:123484. [PMID: 39983826 DOI: 10.1016/j.lfs.2025.123484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 01/19/2025] [Accepted: 02/16/2025] [Indexed: 02/23/2025]
Abstract
AIMS Hippocampal aromatase (AROM) knockdown induces Aβ accumulation and Alzheimer's disease (AD)-like spatial learning and memory impairment, and early hippocampal AROM overexpression in APP/PS1 mice prevents Aβ deposition and memory loss later in life. The aim of this study was to elucidate the underlying mechanism and provide novel prevention and treatment targets for AD. MATERIALS AND METHODS AROM-inhibiting viral vectors were constructed and injected into the hippocampi of adult female mice, after which label-free LC-MS/MS proteomics and bioinformatics analysis were conducted. Additional viral vectors targeting LAMP2 or LC3 were constructed and used to treat HT22 cells. LAMP2 expression was verified, and macroautophagy levels, autophagosome formation and Aβ accumulation were examined. Additionally, ovariectomy combined with the hippocampal injection of LAMP2 inhibition/overexpression viral vectors was applied, and learning and memory abilities and Aβ accumulation were examined. KEY FINDINGS Proteomics revealed the enrichment of CMA and autophagy, and LAMP2 was the most significantly upregulated protein. Higher LAMP2 levels were correlated with lower macroautophagy and autophagosomes levels but were correlated with higher Aβ accumulation, and vice versa. Additionally, hippocampal LAMP2 mediated the effects of ovariectomy on spatial memory and Aβ accumulation. SIGNIFICANCE These results demonstrated the important role of the hippocampal LAMP2-macroautophagy pathway in mediating both hippocampal and ovarian estrogen regulation of Aβ accumulation and AD-like behavior, indicating that LAMP2 might be a novel target for both hippocampal and circulating estrogen deficiency-associated memory impairments, such as AD.
Collapse
Affiliation(s)
- Mingguang Sun
- Beijing Hospital of Integrated Traditional Chinese and Western Medicine, Beijing University of Chinese Medicine, Beijing 100853, China; Department of Neurology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Xiaoxia Cai
- Department of Neurobiology, Army Medical University, Chongqing 400038, China; School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Zhen Lan
- Department of Neurobiology, Army Medical University, Chongqing 400038, China; Department of General Surgery, General Hospital of Central Theater Command, Wuhan 430000, China
| | - Mengying Liu
- Department of Neurobiology, Army Medical University, Chongqing 400038, China; The 305 Hospital of PLA, Beijing 100017, China
| | - Maohu Zhou
- Department of Neurobiology, Army Medical University, Chongqing 400038, China
| | - Yisha Tang
- College of Letters and Science, University of California, Berkeley, CA 94720, United States
| | - Yan Liu
- School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Xuan Zhang
- Department of Neurobiology, Army Medical University, Chongqing 400038, China
| | - Xiao Zhao
- Department of Neurology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Yue Zhou
- Department of Neurology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Jiqiang Zhang
- Department of Neurobiology, Army Medical University, Chongqing 400038, China.
| | - Zhaoyou Meng
- Department of Neurology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China.
| |
Collapse
|
2
|
Shokr MM, Eladawy RM. HMGB1: Different secretion pathways with pivotal role in epilepsy and major depressive disorder. Neuroscience 2025; 570:55-67. [PMID: 39970982 DOI: 10.1016/j.neuroscience.2025.02.023] [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: 07/30/2024] [Revised: 11/10/2024] [Accepted: 02/12/2025] [Indexed: 02/21/2025]
Abstract
High-mobility group box 1 (HMGB1) protein is a highly prevalent protein that, once it is translocated to an extracellular site, can contribute to the pathogenesis of autoimmune and inflammatory responses, including epilepsy and depression. The conditions needed for release are associated with the production of multiple isoforms, and this translocation may occur in response to both immune cell activation and cell death. HMGB1 has been shown to interact with different mediators, including exportin 1, notch receptors, mitogen-activated protein kinase, STAT, tumor protein 53, and inflammasomes. Furthermore, as a crucial inflammatory mediator, HMGB1 has demonstrated upregulated expression and a higher percentage of translocation from the nucleus to the cytoplasm, acting on downstream receptors such as toll-like receptor 4 and receptor for advanced glycation end products, thereby activating interleukin-1 beta and nuclear factor kappa-B, intensifying inflammatory responses. In this review, we aim to discuss the different molecular interactions for the secretion of HMGB1 along with its pivotal role in epilepsy and major depressive disorder.
Collapse
Affiliation(s)
- Mustafa M Shokr
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University - Arish Branch, 45511 Arish, Egypt.
| | - Reem M Eladawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University - Arish Branch, 45511 Arish, Egypt
| |
Collapse
|
3
|
Hu C, Cui T, Xu Z, Yang K, Wu Y, Cai W, Yu J, Qiu Y. Inhibiting HMGB1/AGER/NF-κB pathway prevents pro-inflammatory microglia polarization and protect photoreceptors in retinitis pigmentosa. Int Immunopharmacol 2025; 149:114192. [PMID: 39904032 DOI: 10.1016/j.intimp.2025.114192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/23/2025] [Accepted: 01/27/2025] [Indexed: 02/06/2025]
Abstract
PURPOSE Retinitis pigmentosa (RP) is an inherited retinal neurodegenerative disease which is a significant contributor to blindness. Microglia-mediated inflammation plays a crucial role in retinitis pigmentosa. However, the activation mechanisms of microglia and the role of polarized microglia in RP remain unclear. High-mobility group box 1 (HMGB1) is a key contributor to aseptic inflammation, and glycyrrhizin exerts anti-inflammatory effects by targeting HMGB1. This study aimed to investigate the role of HMGB1 and microglia in RP and explore the protective effects of glycyrrhizin on photoreceptors. METHODS Male C57BL/6 mice and age-matched rd1 mice were used for in vivo models, while zaprinast-treated 661w cells and HMGB1-treated BV-2 cells were used for in vitro models. In this study, the expression of HMGB1 was analyzed using QPCR and western blot (WB). Immunofluorescence staining and ELISA were performed to assess HMGB1 translocation and secretion. Glycyrrhizin was used to inhibit HMGB1, while FPS-ZM1 served as an inhibitor of the receptor for advanced glycation end products (AGER). Microglial polarization was evaluated by QPCR, and the HMGB1/ AGER/ NF-κB signaling pathway was analyzed through WB. Photoreceptor degeneration and visual function were assessed through H&E staining, electroretinography, and TUNEL staining. RESULTS We observed elevated levels of HMGB1 in the retina of rd1 mice and demonstrated in vitro that photoreceptors may serve as a significant source of HMGB1 in the retina. Additionally, HMGB1 was observed to cause microglial polarization via the HMGB1/AGER/ NF-κB pathway and the polarized microglia secrete inflammatory factors including TNF-α and IL-1β which accelerates the degeneration of photoreceptors. Glycyrrhizin reversed the degeneration of photoreceptors and loss of visual function in rd1 mice through the HMGB1/AGER/ NF-κB pathway. CONCLUSION Our findings showed that HMGB1 secreted by photoreceptors activated the microglia through the HMGB1/AGER/NF-κB pathway and the polarized microglia accelerates the degeneration of photoreceptors. Glycyrrhizin reversed the polarization caused by HMGB1 in vitro and delayed the progression of RP in vivo, presenting a potential novel approach for treating retinitis pigmentosa.
Collapse
Affiliation(s)
- Chengyu Hu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072 China
| | - Tao Cui
- Tianjin Medical Device Evaluation and Inspection Center, Tianjin, China
| | - Zihang Xu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072 China
| | - Kun Yang
- Tianjin Medical Device Evaluation and Inspection Center, Tianjin, China
| | - Yan Wu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072 China
| | - Wenting Cai
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072 China
| | - Jing Yu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072 China; Department of Ophthalmology, The Third People's Hospital of Bengbu, Bengbu, China.
| | - Yaoyan Qiu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072 China.
| |
Collapse
|
4
|
Ren G, Bhatnagar S, Young ME, Lee T, Kim JA. Endothelial autophagy-related gene 7 contributes to high fat diet-induced obesity. Mol Metab 2025; 93:102099. [PMID: 39832563 PMCID: PMC11802379 DOI: 10.1016/j.molmet.2025.102099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/22/2025] Open
Abstract
OBJECTIVE Obesity-associated metabolic dysfunction is a major public health concern worldwide. Endothelial dysfunction is a hallmark of metabolic dysfunction, and endothelial cells affect metabolic functions. Because autophagy-related gene 7 (ATG7) is involved in various cellular physiology, we investigated the roles of endothelial cell-ATG7 (EC-ATG7) on high-fat diet-induced obesity and its related metabolic dysfunction. METHODS We generated an endothelial-specific Atg7 knock-out mouse by breeding Atg7flox/flox mouse with the Chd5-Cre mouse, and investigated the metabolic phenotypes associated with high-fat diet (HFD)-induced obesity. Body weight, food intake, glucose tolerance, insulin sensitivity, and liver fat accumulation were measured in endothelial Atg7 deficient (Atg7ΔEnd) and control mice (Atg7f/f). Adipose tissue inflammation was assessed by measuring the expression of pro-inflammatory genes. Furthermore, we performed indirect calorimetry and examined the insulin signaling pathway molecules. RESULTS We found that deletion of EC-Atg7 ameliorated HFD-induced weight gain, fatty liver, and adipocyte hypertrophy and inflammatory response in adipose tissue, and improved insulin sensitivity without changing glucose tolerance. These metabolic effects seem to be due to the reduced food intake because there were no differences in energy expenditure, energy excretion to feces, and physical activity. Interestingly, the deletion of EC-Atg7 protected from HFD-induced vascular rarefaction, and the knock-down of Atg7 in endothelial cells protected from fatty acid-induced cell death. CONCLUSIONS Our results suggest that EC-Atg7 deletion ameliorates HFD-induced obesity and its related metabolic dysfunction, such as insulin resistance and fatty liver by attenuating appetite and vascular rarefaction. The EC-Atg7 deletion may protect the endothelial cells from lipotoxicity and impaired angiogenesis, which preserves the endothelial function in metabolic tissues. These findings may have implications for developing new therapeutic strategies for preventing and treating obesity and its associated health risks.
Collapse
Affiliation(s)
- Guang Ren
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Sushant Bhatnagar
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL 35294, USA; UAB Comprehensive Diabetes Center, USA
| | - Martin E Young
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL 35294, USA; UAB Comprehensive Diabetes Center, USA
| | - Timmy Lee
- Department of Medicine and Division of Nephrology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jeong-A Kim
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL 35294, USA; UAB Comprehensive Diabetes Center, USA.
| |
Collapse
|
5
|
Zhang Q, Liu X, Wei Q, Xiong S, Luo W, Zhou Y, Cao J, Xu X, Liu R, Tang X, Zhang W, Luo B. Apoptotic breast cancer cells after chemotherapy induce pro-tumour extracellular vesicles via LAP-competent macrophages. Redox Biol 2025; 80:103485. [PMID: 39756316 PMCID: PMC11758215 DOI: 10.1016/j.redox.2024.103485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 12/27/2024] [Indexed: 01/07/2025] Open
Abstract
Chemotherapy is important in the systemic therapy for breast cancer. However, after chemotherapy, the left living tumour cells are more progressive. There is an urgent need to study the underlying mechanism which is still unclear to further improve the therapeutic efficacy of chemotherapy in breast cancer. Here we find a pro-tumour effect of the apoptotic cells induced by the chemotherapy, which is mediated by a new subset of macrophages undergoing LC3-associated phagocytosis (LAP). By transferring exosomal S100A11 into the living tumour cells after chemotherapy, the macrophage exhibits a more pro-tumour phenotype than classic M2-type macrophages. Moreover, S100A11 binds to IFITM3, inducing Akt phosphorylation of living tumour cells after chemotherapy, which promotes tumour progression. Of note, Akt inhibitor can enhance the therapeutic effcicay of chemotherapy in breast cancer. This study provides a novel mechanistic link between tumour-associated macrophages and breast cancer, uncovering Akt as a potential therapeutic target to improve chemotherapy efficacy.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Ultrasound, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China; Department of Ultrasound, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Xiaodi Liu
- Department of Ultrasound, Laboratory of Ultrasound Imaging and Drug, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiuxia Wei
- Department of Ultrasound, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Shiyu Xiong
- Department of Ultrasound, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Wanrong Luo
- Department of Ultrasound, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yingshi Zhou
- Department of Ultrasound, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Jincheng Cao
- Department of Ultrasound, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Xiaolin Xu
- Department of Ultrasound, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Rongbin Liu
- Department of Ultrasound, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Xinyu Tang
- Department of Breast Surgery, Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Wenyue Zhang
- Department of Ultrasound, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| | - Baoming Luo
- Department of Ultrasound, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| |
Collapse
|
6
|
Cune D, Pitasi CL, Rubiola A, Jamma T, Simula L, Boucher C, Fortun A, Adoux L, Letourneur F, Saintpierre B, Donnadieu E, Terris B, Bossard P, Chassaing B, Romagnolo B. Inhibition of Atg7 in intestinal epithelial cells drives resistance against Citrobacter rodentium. Cell Death Dis 2025; 16:112. [PMID: 39971913 PMCID: PMC11840101 DOI: 10.1038/s41419-025-07422-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 01/14/2025] [Accepted: 02/03/2025] [Indexed: 02/21/2025]
Abstract
Autophagy, a cytoprotective mechanism in intestinal epithelial cells, plays a crucial role in maintaining intestinal homeostasis. Beyond its cell-autonomous effects, the significance of autophagy in these cells is increasingly acknowledged in the dynamic interplay between the microbiota and the immune response. In the context of colon cancer, intestinal epithelium disruption of autophagy has been identified as a critical factor influencing tumor development. This disruption modulates the composition of the gut microbiota, eliciting an anti-tumoral immune response. Here, we report that Atg7 deficiency in intestinal epithelial cells shapes the intestinal microbiota leading to an associated limitation of colitis induced by Citrobacter rodentium infection. Mice with an inducible, intestinal epithelial-cell-specific deletion of the autophagy gene, Atg7, exhibited enhanced clearance of C. rodentium, mitigated hyperplasia, and reduced pathogen-induced goblet cell loss. This protective effect is linked to a higher proportion of neutrophils and phagocytic cells in the early phase of infection. At later stages, it is associated with the downregulation of pro-inflammatory pathways and an increase in Th17 and Treg responses-immune responses known for their protective roles against C. rodentium infection, modulated by specific gut microbiota. Fecal microbiota transplantation and antibiotic treatment approaches revealed that the Atg7-deficiency-shapped microbiota, especially Gram-positive bacteria, playing a central role in driving resistance to C. rodentium infection. In summary, our findings highlight that inhibiting autophagy in intestinal epithelial cells contributes to maintaining homeostasis and preventing detrimental intestinal inflammation through microbiota-mediated colonization resistance against C. rodentium. This underscores the central role played by autophagy in shaping the microbiota in promoting immune-mediated resistance against enteropathogens.
Collapse
Affiliation(s)
- David Cune
- Université Paris Cité, Institut Cochin, Inserm, CNRS, Paris, France
- Equipe Labellisée Ligue Nationale Contre Le Cancer, Paris, France
| | - Caterina Luana Pitasi
- Université Paris Cité, Institut Cochin, Inserm, CNRS, Paris, France
- Equipe Labellisée Ligue Nationale Contre Le Cancer, Paris, France
| | - Alessia Rubiola
- Université Paris Cité, Institut Cochin, Inserm, CNRS, Paris, France
- Equipe Labellisée Ligue Nationale Contre Le Cancer, Paris, France
| | - Trinath Jamma
- Department of Biological Sciences, Birla Institute of Technology and Science-Pilani, Hyderabad, India
| | - Luca Simula
- Université Paris Cité, Institut Cochin, Inserm, CNRS, Paris, France
| | - Camille Boucher
- Université Paris Cité, Institut Cochin, Inserm, CNRS, Paris, France
- Equipe Labellisée Ligue Nationale Contre Le Cancer, Paris, France
| | - Apolline Fortun
- Université Paris Cité, Institut Cochin, Inserm, CNRS, Paris, France
- Equipe Labellisée Ligue Nationale Contre Le Cancer, Paris, France
| | - Lucie Adoux
- Genomic Facility, Université de Paris Cité, Institut Cochin, INSERM, CNRS, Paris, France
| | - Franck Letourneur
- Genomic Facility, Université de Paris Cité, Institut Cochin, INSERM, CNRS, Paris, France
| | - Benjamin Saintpierre
- Genomic Facility, Université de Paris Cité, Institut Cochin, INSERM, CNRS, Paris, France
| | | | - Benoît Terris
- Université Paris Cité, Institut Cochin, Inserm, CNRS, Paris, France
- Equipe Labellisée Ligue Nationale Contre Le Cancer, Paris, France
- Pathology Department, AP-HP, Hôpital Cochin, Paris, France
| | - Pascale Bossard
- Université Paris Cité, Institut Cochin, Inserm, CNRS, Paris, France
- Equipe Labellisée Ligue Nationale Contre Le Cancer, Paris, France
| | - Benoît Chassaing
- Université Paris Cité, Institut Cochin, Inserm, CNRS, Paris, France
- Microbiome-Host Interactions, Institut Pasteur, Université Paris Cité, INSERM, Paris, France
| | - Béatrice Romagnolo
- Université Paris Cité, Institut Cochin, Inserm, CNRS, Paris, France.
- Equipe Labellisée Ligue Nationale Contre Le Cancer, Paris, France.
| |
Collapse
|
7
|
Sakurai M, Kuwahara T. Canonical and noncanonical autophagy: involvement in Parkinson's disease. Front Cell Dev Biol 2025; 13:1518991. [PMID: 39949604 PMCID: PMC11821624 DOI: 10.3389/fcell.2025.1518991] [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: 10/29/2024] [Accepted: 01/15/2025] [Indexed: 02/16/2025] Open
Abstract
Autophagy is the major degradation process in cells and is involved in a variety of physiological and pathological functions. While macroautophagy, which employs a series of molecular cascades to form ATG8-coated double membrane autophagosomes for degradation, remains the well-known type of canonical autophagy, microautophagy and chaperon-mediated autophagy have also been characterized. On the other hand, recent studies have focused on the functions of autophagy proteins beyond intracellular degradation, including noncanonical autophagy, also known as the conjugation of ATG8 to single membranes (CASM), and autophagy-related extracellular secretion. In particular, CASM is unique in that it does not require autophagy upstream mechanisms, while the ATG8 conjugation system is involved in a manner different from canonical autophagy. There have been many reports on the involvement of these autophagy-related mechanisms in neurodegenerative diseases, with Parkinson's disease (PD) receiving particular attention because of the important roles of several causative and risk genes, including LRRK2. In this review, we will summarize and discuss the contributions of canonical and noncanonical autophagy to cellular functions, with a special focus on the pathogenesis of PD.
Collapse
Affiliation(s)
| | - Tomoki Kuwahara
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
8
|
Lee SK, Park SW, Jang DJ, Lee JA. Mechanisms and roles of membrane-anchored ATG8s. Front Cell Dev Biol 2025; 13:1532050. [PMID: 39936034 PMCID: PMC11810923 DOI: 10.3389/fcell.2025.1532050] [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: 11/21/2024] [Accepted: 01/09/2025] [Indexed: 02/13/2025] Open
Abstract
Autophagy-related protein 8 (ATG8) family proteins, including LC3 and GABARAP subfamilies, are pivotal in canonical autophagy, driving autophagosome formation, cargo selection, and lysosomal fusion. However, recent studies have identified non-canonical roles for lipidated ATG8 in processes such as LC3-associated phagocytosis (LAP), LC3-associated endocytosis (LANDO), and lipidated ATG8-mediated secretory autophagy. These pathways expand ATG8's functional repertoire in immune regulation, membrane repair, and pathogen clearance, as ATG8 becomes conjugated to single-membrane structures (e.g., phagosomes and lysosomes). This review examines the molecular mechanisms of ATG8 lipidation, focusing on its selective conjugation to phosphatidylethanolamine (PE) in autophagy and phosphatidylserine (PS) in CASM. We highlight LIR-based probes and LC3/GABARAP-specific deconjugases as critical tools that allow precise tracking and manipulation of ATG8 in autophagic and non-autophagic contexts. These advancements hold therapeutic promise for treating autophagy-related diseases, including cancer and neurodegenerative disorders, by targeting ATG8-driven pathways that maintain cellular homeostasis.
Collapse
Affiliation(s)
- Soo-Kyeong Lee
- Department of Biological Sciences and Biotechnology, College of Life Sciences and Nanotechnology, Hannam University, Daejeon, Republic of Korea
| | - Sang-Won Park
- Research Institute of Invertebrate Vector, Kyungpook National University, Sangju, Republic of Korea
| | - Deok-Jin Jang
- Department of Ecological Science, College of Ecology and Environment, Kyungpook National University, Sangju, Republic of Korea
| | - Jin-A. Lee
- Department of Biological Sciences and Biotechnology, College of Life Sciences and Nanotechnology, Hannam University, Daejeon, Republic of Korea
| |
Collapse
|
9
|
Shirani N, Abdi N, Chehelgerdi M, Yaghoobi H, Chehelgerdi M. Investigating the role of exosomal long non-coding RNAs in drug resistance within female reproductive system cancers. Front Cell Dev Biol 2025; 13:1485422. [PMID: 39925739 PMCID: PMC11802832 DOI: 10.3389/fcell.2025.1485422] [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: 08/23/2024] [Accepted: 01/02/2025] [Indexed: 02/11/2025] Open
Abstract
Exosomes, as key mediators of intercellular communication, have been increasingly recognized for their role in the oncogenic processes, particularly in facilitating drug resistance. This article delves into the emerging evidence linking exosomal lncRNAs to the modulation of drug resistance mechanisms in cancers such as ovarian, cervical, and endometrial cancer. It synthesizes current research findings on how these lncRNAs influence cancer cell survival, tumor microenvironment, and chemotherapy efficacy. Additionally, the review highlights potential therapeutic strategies targeting exosomal lncRNAs, proposing a new frontier in overcoming drug resistance. By mapping the interface of exosomal lncRNAs and drug resistance, this article aims to provide a comprehensive understanding that could pave the way for innovative treatments and improved patient outcomes in female reproductive system cancers.
Collapse
Affiliation(s)
- Nooshafarin Shirani
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Neda Abdi
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Matin Chehelgerdi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Hajar Yaghoobi
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mohammad Chehelgerdi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| |
Collapse
|
10
|
Moresco P, Kastan JP, Yang JI, Prabakar R, Minicozzi F, Adams DW, Cifani P, Tuveson DA, Fearon DT. Signal peptide-independent secretion of keratin-19 by pancreatic cancer cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.18.633717. [PMID: 39896665 PMCID: PMC11785074 DOI: 10.1101/2025.01.18.633717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
The exclusion of T cells causes immune escape of pancreatic ductal adenocarcinoma (PDA). T cell exclusion is mediated by the interaction between CXCR4 on T cells and its ligand, CXCL12, which is complexed to keratin-19 (KRT19) on the surface of PDA cells. KRT19 secretion by PDA cells is essential to this process but is unusual because KRT19 lacks an endoplasmic reticulum (ER)-directing signal peptide (SP). By using biotinylation by an ER-restricted TurboID system and a split-GFP assay in PDA cells, we demonstrate that KRT19 enters the ER via its "head" domain. Additionally, KRT19 is shown to interact with the signal recognition particle and its secretion is sensitive to canonical protein secretion inhibitors. In vivo, mouse tumors formed with ER-TurboID-expressing PDA cells contain biotinylated KRT19. In contrast, keratin-8 (KRT8), which colocalizes with KRT19 on the surface of PDA cells, does not enter the ER. Rather, KRT8 is externalized via secretory autophagy possibly in a complex with KRT19. Thus, despite lacking a classical SP, PDA cells secrete KRT19 to capture CXCL12 and protect against immune attack.
Collapse
Affiliation(s)
- Philip Moresco
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
- Graduate Program in Genetics, Stony Brook University, Stony Brook, NY 11794, USA
- Medical Scientist Training Program, Stony Brook University Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | | | - Jung-in Yang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | | | | | - Dexter W. Adams
- Graduate Program in Genetics, Stony Brook University, Stony Brook, NY 11794, USA
- W. M. Keck Structural Biology Laboratory, Howard Hughes Medical Institute, Cold Spring Harbor, NY 11724, USA
| | - Paolo Cifani
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - David A. Tuveson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Douglas T. Fearon
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
- Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
11
|
Le Nihouannen D, Boiziau C, Rey S, Agadzhanian N, Dusserre N, Cordelières F, Priault M, Boeuf H. Inhibiting Autophagy by Chemicals During SCAPs Osteodifferentiation Elicits Disorganized Mineralization, While the Knock-Out of Atg5/7 Genes Leads to Cell Adaptation. Cells 2025; 14:146. [PMID: 39851574 PMCID: PMC11840282 DOI: 10.3390/cells14020146] [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: 11/07/2024] [Revised: 01/07/2025] [Accepted: 01/15/2025] [Indexed: 01/26/2025] Open
Abstract
SCAPs (Stem Cells from Apical Papilla), derived from the apex of forming wisdom teeth, extracted from teenagers for orthodontic reasons, belong to the MSCs (Mesenchymal Stromal Cells) family. They have multipotent differentiation capabilities and are a potentially powerful model for investigating strategies of clinical cell therapies. Since autophagy-a regulated self-eating process-was proposed to be essential in osteogenesis, we investigated its involvement in the SCAP model. By using a combination of chemical and genetic approaches to inhibit autophagy, we studied early and late events of osteoblastic differentiation. We showed that blocking the formation of autophagosomes with verteporfin did not induce a dramatic alteration in early osteoblastic differentiation monitored by ALP (alkaline phosphatase) activity. However, blocking the autophagy flux with bafilomycin A1 led to ALP repression. Strikingly, the mineralization process was observed with both compounds, with calcium phosphate (CaP) nodules that remained inside cells under bafilomycin A1 treatment and numerous but smaller CaP nodules after verteporfin treatment. In contrast, deletion of Atg5 or Atg7, two genes involved in the formation of autophagosomes and essential to trigger canonical autophagy, indicated that both genes could be involved differently in the mineralization process with a modification of the ALP activity while final mineralization was not altered.
Collapse
Affiliation(s)
- Damien Le Nihouannen
- Univ. Bordeaux, INSERM, BIOTIS, U1026, F-33000 Bordeaux, France; (D.L.N.); (S.R.); (N.A.); (N.D.)
| | - Claudine Boiziau
- Univ. Bordeaux, INSERM, BIOTIS, U1026, F-33000 Bordeaux, France; (D.L.N.); (S.R.); (N.A.); (N.D.)
| | - Sylvie Rey
- Univ. Bordeaux, INSERM, BIOTIS, U1026, F-33000 Bordeaux, France; (D.L.N.); (S.R.); (N.A.); (N.D.)
| | - Nicole Agadzhanian
- Univ. Bordeaux, INSERM, BIOTIS, U1026, F-33000 Bordeaux, France; (D.L.N.); (S.R.); (N.A.); (N.D.)
| | - Nathalie Dusserre
- Univ. Bordeaux, INSERM, BIOTIS, U1026, F-33000 Bordeaux, France; (D.L.N.); (S.R.); (N.A.); (N.D.)
| | - Fabrice Cordelières
- Bordeaux Imaging Center (BIC), US4, UAR 3420, National Center for Scientific Research (CNRS), National Institute of Health and Medical Research (INSERM), Université de Bordeaux, F-33000 Bordeaux, France;
| | - Muriel Priault
- National Center for Scientific Research (CNRS), Institut de Biochimie et Génétique Cellulaires (IBGC), UMR 5095, Université de Bordeaux, F-33000 Bordeaux, France;
| | - Helene Boeuf
- Univ. Bordeaux, INSERM, BIOTIS, U1026, F-33000 Bordeaux, France; (D.L.N.); (S.R.); (N.A.); (N.D.)
| |
Collapse
|
12
|
Wang W, Hu Y, Ding N, Wei J, Li C. The role of SIRT1 in kidney diseases. Int Urol Nephrol 2025; 57:147-158. [PMID: 39030438 DOI: 10.1007/s11255-024-04162-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: 06/10/2024] [Accepted: 07/15/2024] [Indexed: 07/21/2024]
Abstract
SIRT1, a nicotinamide adenine dinucleotide (NAD +)-dependent class III histone deacetylase, exhibits a high level of expression within renal tissues. It has garnered considerable recognition for its pivotal role in modulating signaling pathways intricately linked with the aging process; however, it extends beyond this in the organism. The literature reports that SIRT1 regulates biological processes such as glucose metabolism, lipid metabolism, oxidative stress, inflammation, autophagy, endoplasmic reticulum stress, and apoptosis. Therefore, our study reviews the primary mechanisms by which SIRT1 induces kidney disease and the regulation of related signaling pathways in different models of renal disease. We also discuss commonly studied SIRT1-targeted interventional drugs reported in the literature, including inhibitors (e.g., Ex-527) and activators (e.g., resveratrol). This study aims to provide theoretical foundations and clinical insights for the development and screening of clinical drugs targeting SIRT1, aiming at enhanced scientific approaches for the prevention and treatment of kidney diseases.
Collapse
Affiliation(s)
- Wei Wang
- School of Pharmacy, School of Stomatology and Ophthalmology, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, China
| | - Yuanyuan Hu
- School of Pharmacy, School of Stomatology and Ophthalmology, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, China
| | - Ning Ding
- School of Pharmacy, School of Stomatology and Ophthalmology, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, China
| | - Jiping Wei
- School of Pharmacy, School of Stomatology and Ophthalmology, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, China
| | - Cairong Li
- Second Affiliated Hospital, Clinical Medical School, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, China.
| |
Collapse
|
13
|
Blandino G, Satchi-Fainaro R, Tinhofer I, Tonon G, Heilshorn SC, Kwon YJ, Pestana A, Frascolla C, Pompili L, Puce A, Iachettini S, Tocci A, Karkampouna S, Kruithof-de Julio M, Tocci P, Porciello N, Maccaroni K, Rutigliano D, Shen X, Ciliberto G. Cancer Organoids as reliable disease models to drive clinical development of novel therapies. J Exp Clin Cancer Res 2024; 43:334. [PMID: 39731178 DOI: 10.1186/s13046-024-03258-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 12/29/2024] Open
Abstract
On September 23-24 (2024) the 6th Workshop IRE on Translational Oncology, titled "Cancer Organoids as Reliable Disease Models to Drive Clinical Development of Novel Therapies," took place at the IRCCS Regina Elena Cancer Institute in Rome. This prominent international conference focused on tumor organoids, bringing together leading experts from around the world.A central challenge in precision oncology is modeling the dynamic tumor ecosystem, which encompasses numerous elements that evolve spatially and temporally. Patient-derived 3D culture models, including organoids, explants, and engineered or bioprinted systems, have recently emerged as sophisticated tools capable of capturing the complexity and diversity of cancer cells interacting within their microenvironments. These models address critical unmet needs in precision medicine, particularly in aiding clinical decision-making. The rapid development of these human tissue avatars has enabled advanced modeling of cellular alterations in disease states and the screening of compounds to uncover novel therapeutic pathways.Throughout the event, distinguished speakers shared their expertise and research findings, illustrating how organoids are transforming our understanding of treatment resistance, metastatic dynamics, and the interaction between tumors and the surrounding microenvironment.This conference served as a pivotal opportunity to strengthen international collaborations and spark innovative translational approaches. Its goal was to accelerate the shift from preclinical research to clinical application, paving the way for increasingly personalized and effective cancer therapies.
Collapse
Affiliation(s)
- Giovanni Blandino
- Translational Research Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy.
| | - Ronit Satchi-Fainaro
- Deapartment of Physiology and Pharmacology, Tel Aviv University, Tel Aviv, Israel
| | - Ingeborg Tinhofer
- Department of Radiooncology and Radiotherapy, Charité University Medicin, Berlin, Germany
| | - Giovanni Tonon
- Center of Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, USA
| | - Yong-Jun Kwon
- Luxembourg Institute of Health, Strassen, Luxembourg
| | - Ana Pestana
- Department of Radiooncology and Radiotherapy, Charité University Medicin, Berlin, Germany
| | - Carlotta Frascolla
- Translational Research Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Luca Pompili
- Translational Research Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Aurora Puce
- Cellular Network and Molecular Therapeutic Target Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Sara Iachettini
- Translational Research Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Annalisa Tocci
- Tumor of Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Sofia Karkampouna
- Department for BioMedical Research, University of Bern, Swiss, Switzerland
| | | | - Piera Tocci
- Unit of Preclinical Models and New Therapeutic Agents, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Nicla Porciello
- Tumor of Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Klizia Maccaroni
- Cellular Network and Molecular Therapeutic Target Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Daniela Rutigliano
- Translational Research Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Xiling Shen
- GI Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
- Terasaki Institute of Biomedical Innovation, Los Angeles, CA, USA
| | - Gennaro Ciliberto
- Scientific Direction, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
14
|
Sho T, Li Y, Jiao H, Yu L. Migratory autolysosome disposal mitigates lysosome damage. J Cell Biol 2024; 223:e202403195. [PMID: 39347717 PMCID: PMC11457477 DOI: 10.1083/jcb.202403195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 06/26/2024] [Accepted: 08/14/2024] [Indexed: 10/01/2024] Open
Abstract
Lysosomes, essential for intracellular degradation and recycling, employ damage-control strategies such as lysophagy and membrane repair mechanisms to maintain functionality and cellular homeostasis. Our study unveils migratory autolysosome disposal (MAD), a response to lysosomal damage where cells expel LAMP1-LC3 positive structures via autolysosome exocytosis, requiring autophagy machinery, SNARE proteins, and cell migration. This mechanism, crucial for mitigating lysosomal damage, underscores the role of cell migration in lysosome damage control and facilitates the release of small extracellular vesicles, highlighting the intricate relationship between cell migration, organelle quality control, and extracellular vesicle release.
Collapse
Affiliation(s)
- Takami Sho
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Ying Li
- Cryo-EM Facility of Tsinghua University, Branch of National Protein Science Facility, Tsinghua University, Beijing, China
| | - Haifeng Jiao
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Li Yu
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
15
|
Rajendran D, Oon CE. Navigating therapeutic prospects by modulating autophagy in colorectal cancer. Life Sci 2024; 358:123121. [PMID: 39389340 DOI: 10.1016/j.lfs.2024.123121] [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: 06/13/2024] [Revised: 09/25/2024] [Accepted: 10/05/2024] [Indexed: 10/12/2024]
Abstract
Colorectal cancer (CRC) remains a leading cause of death globally despite the improvements in cancer treatment. Autophagy is an evolutionarily conserved lysosomal-dependent degradation pathway that is critical in maintaining cellular homeostasis. However, in cancer, autophagy may have conflicting functions in preventing early tumour formation versus the maintenance of advanced-stage tumours. Defective autophagy has a broad and dynamic effect not just on cancer cells, but also on the tumour microenvironment which influences tumour progression and response to treatment. To add to the layer of complexity, somatic mutations in CRC including tumour protein p53 (TP53), v-raf murine sarcoma viral oncogene homolog B1 (BRAF), Kirsten rat sarcoma viral oncogene homolog (KRAS), and phosphatase and tensin homolog (PTEN) can render chemoresistance by promoting a pro-survival advantage through autophagy. Recent studies have also reported autophagy-related cell deaths that are distinct from classical autophagy by employing parts of the autophagic machinery, which impacts strategies for autophagy regulation in cancer therapy. This review discusses the molecular processes of autophagy in the evolution of CRC and its role in the tumour microenvironment, as well as prospective therapeutic methods based on autophagy suppression or promotion. It also highlights clinical trials using autophagy modulators for treating CRC, underscoring the importance of autophagy regulation in CRC therapy.
Collapse
Affiliation(s)
- Deepa Rajendran
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Gelugor, 11800, Penang, Malaysia.
| | - Chern Ein Oon
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Gelugor, 11800, Penang, Malaysia.
| |
Collapse
|
16
|
Nam HY, Park SH, Lee GH, Kim EY, Lee S, Chang HW, Chang EJ, Choi KC, Kim SW. TIGAR coordinates senescence-associated secretory phenotype via lysosome repositioning and α-tubulin deacetylation. Exp Mol Med 2024; 56:2726-2738. [PMID: 39633033 DOI: 10.1038/s12276-024-01362-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/06/2024] [Accepted: 09/25/2024] [Indexed: 12/07/2024] Open
Abstract
TP53-induced glycolysis and apoptosis regulator (TIGAR) regulates redox homeostasis and provides the intermediates necessary for cell growth by reducing the glycolytic rate. During cellular senescence, cells undergo metabolic rewiring towards the glycolytic pathway, along with the development of the senescence-associated secretory phenotype (SASP), also known as the secretome. We observed that TIGAR expression increased during replicative senescence following the in vitro expansion of human mesenchymal stromal cells (MSCs) and that TIGAR knockout (KO) decreased SASP factors and triggered premature senescence with decelerated progression. Additionally, TIGAR KO impaired flexible lysosomal movement to the perinuclear region and decreased the autophagic flux of MSCs. Research on the mechanism of lysosomal movement revealed that, while native senescent MSCs presented low levels of Ac-α-tubulin (lysine 40) and increased sirtuin 2 (SIRT2) activity compared with those in growing cells, TIGAR KO-MSCs maintained Ac-α-tubulin levels and exhibited decreased SIRT2 activity despite being in a senescent state. The overexpression of SIRT2 reduced Ac-α-tubulin as a protein target of SIRT2 and induced the positioning of lysosomes at the perinuclear region, restoring the cytokine secretion of TIGAR KO-MSCs. Furthermore, TIGAR expression was positively correlated with SIRT2 activity, indicating that TIGAR affects SIRT2 activity partly by modulating the NAD+ level. Thus, our study demonstrated that TIGAR provides a foundation that translates the regulation of energy metabolism into lysosome positioning, affecting the secretome for senescence development. Considering the functional value of the cell-secretome in aging-related diseases, these findings suggest the feasibility of TIGAR for the regulation of secretory phenotypes.
Collapse
Affiliation(s)
- Hae Yun Nam
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea.
| | - Seung-Ho Park
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - Geun-Hee Lee
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - Eun-Young Kim
- Department Hematology and Medical Oncology, Whinship Cancer Institute of Emory University, Atlanta, GA, 30322, USA
| | - SangEun Lee
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - Hyo Won Chang
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - Eun-Ju Chang
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - Kyung-Chul Choi
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea.
| | - Seong Who Kim
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea.
| |
Collapse
|
17
|
Skach K, Boserle J, Nuta GC, Břehová P, Bialik S, Carvalho S, Kozer N, Barr H, Chaloupecká E, Kimchi A, Nencka R. Structure-activity relationship study of small-molecule inhibitor of Atg12-Atg3 protein-protein interaction. Bioorg Med Chem Lett 2024; 112:129939. [PMID: 39218407 DOI: 10.1016/j.bmcl.2024.129939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Autophagy is a catabolic process that was described to play a critical role in advanced stages of cancer, wherein it maintains tumor cell homeostasis and growth by supplying nutrients. Autophagy is also described to support alternative cellular trafficking pathways, providing a non-canonical autophagy-dependent inflammatory cytokine secretion mechanism. Therefore, autophagy inhibitors have high potential in the treatment of cancer and acute inflammation. In our study, we identified compound 1 as an inhibitor of the ATG12-ATG3 protein-protein interaction. We focused on the systematic modification of the original hit 1, a casein kinase 2 (CK2) inhibitor, to find potent disruptors of ATG12-ATG3 protein-protein interaction. A systematic modification of the hit structure led us to a wide plethora of compounds that maintain its ATG12-ATG3 inhibitory activity, which could act as a viable starting point to design new compounds with diverse therapeutic applications.
Collapse
Affiliation(s)
- Krystof Skach
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 16610 Prague, Czech Republic; Department of Chemistry of Natural Compounds, University of Chemistry and Technology, Prague, 166 28 Prague, Czech Republic
| | - Jiri Boserle
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 16610 Prague, Czech Republic
| | - Gal Chaim Nuta
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Petra Břehová
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 16610 Prague, Czech Republic
| | - Shani Bialik
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Silvia Carvalho
- The Nancy and Stephen Grand Israel National Center for Personalized Medicine (G-INCPM), Weizmann Institute of Science, Rehovot 76100, Israel
| | - Noga Kozer
- The Nancy and Stephen Grand Israel National Center for Personalized Medicine (G-INCPM), Weizmann Institute of Science, Rehovot 76100, Israel
| | - Haim Barr
- The Nancy and Stephen Grand Israel National Center for Personalized Medicine (G-INCPM), Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ema Chaloupecká
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 16610 Prague, Czech Republic
| | - Adi Kimchi
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Radim Nencka
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 16610 Prague, Czech Republic.
| |
Collapse
|
18
|
Wang C, Luo H. Crosstalk Between Innate Immunity and Autophagy in Viral Myocarditis Leading to Dilated Cardiomyopathy. Rev Med Virol 2024; 34:e2586. [PMID: 39349889 DOI: 10.1002/rmv.2586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/02/2024] [Accepted: 09/12/2024] [Indexed: 11/08/2024]
Abstract
Viral myocarditis, characterised by inflammation of the heart muscle, presents a significant challenge to global public health, particularly affecting younger individuals and often progressing to dilated cardiomyopathy (DCM), a leading cause of heart failure. Despite ongoing research efforts, viable treatments for this condition remain elusive. Recent studies have shed light on the complex interplay between the innate immune response and autophagy mechanisms, revealing their pivotal roles in the pathogenesis of viral myocarditis and subsequent DCM development. This review aims to delve into the recent advancements in understanding the molecular mechanisms and pathways that intersect innate immunity and autophagy in the context of viral myocarditis. Furthermore, it explores the potential therapeutic implications of these findings, offering insights into promising avenues for the management and treatment of this debilitating condition.
Collapse
Affiliation(s)
- Chen Wang
- Centre for Heart Lung Innovation, St. Paul's Hospital-University of British Columbia, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Honglin Luo
- Centre for Heart Lung Innovation, St. Paul's Hospital-University of British Columbia, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| |
Collapse
|
19
|
Nie J, Ma S, Wu L, Li Y, Cao J, Li M, Mei P, Cooper PR, Li A, Pei D. SEC31a-ATG9a Interaction Mediates the Recruitment of COPII Vesicles for Autophagosome Formation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405127. [PMID: 39361436 PMCID: PMC11600210 DOI: 10.1002/advs.202405127] [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: 05/11/2024] [Revised: 07/28/2024] [Indexed: 10/05/2024]
Abstract
Autophagy plays an important role in determining stem-cell differentiation. During the osteogenic differentiation of mesenchymal stem cells (MSCs), autophagosome formation is upregulated but the reason is unknown. A long-standing quest in the autophagy field is to find the membrane origin of autophagosomes. In this study, cytoplasmic coat protein complex II (COPII) vesicles, endoplasmic reticulum-derived vesicles responsible for the transport of storage proteins to the Golgi, are demonstrated to be a critical source of osteoblastic autophagosomal membrane. A significant correlation between the number of COPII vesicle and the autophagy level is identified in the rat bone tissues. Disruption of COPII vesicles restrained osteogenesis and decreased the number and size of autophagosomes. SEC31a (an outer coat protein of COPII vesicle) is found to be vital to regulate COPII vesicle-dependent autophagosome formation via interacting with ATG9a of autophagosomal seed vesicles. The interference of Sec31a inhibited autophagosome formation and osteogenesis in vitro and in vivo. These results identified a novel mechanism of autophagosome formation in osteogenic differentiation of stem cells and identified SEC31a as a critical protein that mediates the interplay between COPII and ATG9a vesicles. These findings broaden the understanding of the regulatory mechanism in the osteogenic differentiation of MSCs.
Collapse
Affiliation(s)
- Jiaming Nie
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine ResearchCollege of StomatologyXi'an Jiaotong UniversityXi'anShaanxi710004China
| | - Shaoyang Ma
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine ResearchCollege of StomatologyXi'an Jiaotong UniversityXi'anShaanxi710004China
| | - Linyue Wu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine ResearchCollege of StomatologyXi'an Jiaotong UniversityXi'anShaanxi710004China
| | - Ye Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine ResearchCollege of StomatologyXi'an Jiaotong UniversityXi'anShaanxi710004China
| | - Jiao Cao
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine ResearchCollege of StomatologyXi'an Jiaotong UniversityXi'anShaanxi710004China
| | - Meng Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine ResearchCollege of StomatologyXi'an Jiaotong UniversityXi'anShaanxi710004China
| | - Peter Mei
- Department of Oral SciencesFaculty of DentistryUniversity of OtagoDunedin9016New Zealand
| | - Paul R. Cooper
- Department of Oral SciencesFaculty of DentistryUniversity of OtagoDunedin9016New Zealand
| | - Ang Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine ResearchCollege of StomatologyXi'an Jiaotong UniversityXi'anShaanxi710004China
| | - Dandan Pei
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine ResearchCollege of StomatologyXi'an Jiaotong UniversityXi'anShaanxi710004China
| |
Collapse
|
20
|
Zhang Q, Jin W, Wang H, Tang C, Zhao X, Wang Y, Sun L, Piao C. Inhibition of endoplasmic reticulum stress and excessive autophagy by Jiedu Tongluo Tiaogan Formula via a CaMKKβ/AMPK pathway contributes to protect pancreatic β-cells. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118440. [PMID: 38885916 DOI: 10.1016/j.jep.2024.118440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/26/2024] [Accepted: 06/05/2024] [Indexed: 06/20/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Jiedu Tongluo Tiaogan Formula (JTTF), a traditional Chinese herbal decoction, exhibits the potential to treat type 2 diabetes mellitus (T2DM) by inhibiting endoplasmic reticulum stress (ERS) and excessive autophagy, which are the risk factors for the abnormal development and progression of β cells. AIM OF THE STUDY We aimed to assess the effect of JTTF on pancreatic glucotoxicity by inhibiting ERS and excessive autophagy, for which db/db mice and INS-1 insulinoma cells were used. MATERIALS AND METHODS The chemical composition of the JTTF was analyzed by UPLC-Q/TOF-MS. Diabetic (db/db) mice were treated with distilled water or JTTF (2.4 and 7.2 g/kg/day) for 8 weeks. Furthermore, INS-1 cells induced by high glucose (HG) levels were treated with or without JTTF (50, 100, and 200 μg/mL) for 48 h to elucidate the protective mechanism of JTTF on glucose toxicity. The experimental methods included an oral glucose tolerance test, hematoxylin-eosin staining, immunohistochemistry, western blotting, RT-qPCR, and acridine orange staining. RESULT 28 chemical components of JTTF were identified. Additionally, treatment with JTTF significantly decreased the severity of glycemic symptoms in the db/db mice. Moreover, the treatment partially restored glucose homeostasis in the db/db mice and protected the pancreatic β-cell function. JTTF protected INS-1 cells from HG injury by upregulating GSIS and PDX1, MafA mRNA expression. Further, treatment with JTTF downregulated GRP78 and ATF6 expression, whereas it inhibited Beclin-1 and LC3 activation. The treatment protected the cells from HG-induced ERS and excessive autophagy by downregulating the CaMKKβ/AMPK pathway. CONCLUSIONS The present study findings show that JTTF may protects β-cells by inhibiting the CaMKKβ/AMPK pathway, which deepens our understanding of the effectiveness of JTTF as a treatment strategy against T2DM.
Collapse
Affiliation(s)
- Qi Zhang
- Shenzhen Hospital (Fu Tian) of Guangzhou University of Chinese Medicine, Shenzhen, 518000, Guangdong, China
| | - Wenqi Jin
- College of traditional Chinese medicine, Changchun University of Chinese Medicine, Changchun, 130012, Jilin, China
| | - Han Wang
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Cheng Tang
- College of traditional Chinese medicine, Changchun University of Chinese Medicine, Changchun, 130012, Jilin, China
| | - Xiaohua Zhao
- Shenzhen Hospital (Fu Tian) of Guangzhou University of Chinese Medicine, Shenzhen, 518000, Guangdong, China
| | - Yu Wang
- College of traditional Chinese medicine, Changchun University of Chinese Medicine, Changchun, 130012, Jilin, China
| | - Liwei Sun
- College of traditional Chinese medicine, Changchun University of Chinese Medicine, Changchun, 130012, Jilin, China.
| | - Chunli Piao
- Shenzhen Hospital (Fu Tian) of Guangzhou University of Chinese Medicine, Shenzhen, 518000, Guangdong, China.
| |
Collapse
|
21
|
Néel E, Chiritoiu-Butnaru M, Fargues W, Denus M, Colladant M, Filaquier A, Stewart SE, Lehmann S, Zurzolo C, Rubinsztein DC, Marin P, Parmentier ML, Villeneuve J. The endolysosomal system in conventional and unconventional protein secretion. J Cell Biol 2024; 223:e202404152. [PMID: 39133205 PMCID: PMC11318669 DOI: 10.1083/jcb.202404152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/12/2024] [Accepted: 07/26/2024] [Indexed: 08/13/2024] Open
Abstract
Most secreted proteins are transported through the "conventional" endoplasmic reticulum-Golgi apparatus exocytic route for their delivery to the cell surface and release into the extracellular space. Nonetheless, formative discoveries have underscored the existence of alternative or "unconventional" secretory routes, which play a crucial role in exporting a diverse array of cytosolic proteins outside the cell in response to intrinsic demands, external cues, and environmental changes. In this context, lysosomes emerge as dynamic organelles positioned at the crossroads of multiple intracellular trafficking pathways, endowed with the capacity to fuse with the plasma membrane and recognized for their key role in both conventional and unconventional protein secretion. The recent recognition of lysosomal transport and exocytosis in the unconventional secretion of cargo proteins provides new and promising insights into our understanding of numerous physiological processes.
Collapse
Affiliation(s)
- Eloïse Néel
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM , Montpellier, France
| | | | - William Fargues
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM , Montpellier, France
| | - Morgane Denus
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM , Montpellier, France
| | - Maëlle Colladant
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM , Montpellier, France
| | - Aurore Filaquier
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM , Montpellier, France
| | - Sarah E Stewart
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Sylvain Lehmann
- Laboratoire de Biochimie-Protéomique Clinique-Plateforme de Protéomique Clinique, Université de Montpellier, Institute for Regenerative Medicine and Biotherapy Centre Hospitalier Universitaire de Montpellier, Institute for Neurosciences of Montpellier INSERM , Montpellier, France
| | - Chiara Zurzolo
- Unité de Trafic Membranaire et Pathogenèse, Institut Pasteur, UMR3691 CNRS , Paris, France
| | - David C Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute , Cambridge, UK
| | - Philippe Marin
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM , Montpellier, France
| | - Marie-Laure Parmentier
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM , Montpellier, France
| | - Julien Villeneuve
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM , Montpellier, France
| |
Collapse
|
22
|
Trojani MC, Santucci-Darmanin S, Breuil V, Carle GF, Pierrefite-Carle V. Lysosomal exocytosis: From cell protection to protumoral functions. Cancer Lett 2024; 597:217024. [PMID: 38871244 DOI: 10.1016/j.canlet.2024.217024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/27/2024] [Accepted: 06/03/2024] [Indexed: 06/15/2024]
Abstract
Lysosomes are single membrane bounded group of acidic organelles that can be involved in a process called lysosomal exocytosis which leads to the extracellular release of their content. Lysosomal exocytosis is required for plasma membrane repair or remodeling events such as bone resorption, antigen presentation or mitosis, and for protection against toxic agents such as heavy metals. Recently, it has been showed that to fulfill this protective role, lysosomal exocytosis needs some autophagic proteins, in an autophagy-independent manner. In addition to these crucial physiological roles, lysosomal exocytosis plays a major protumoral role in various cancers. This effect is exerted through tumor microenvironment modifications, including extracellular matrix remodeling, acidosis, oncogenic and profibrogenic signals. This review provides a comprehensive overview of the different elements released in the microenvironment during lysosomal exocytosis, i.e. proteases, exosomes, and protons, and their effects in the context of tumor development and treatment.
Collapse
Affiliation(s)
- Marie-Charlotte Trojani
- UMR E-4320 TIRO-MATOs CEA/DRF/Institut Joliot, Université Côte d'Azur, Faculté de Médecine Nice, France; Service de Rhumatologie, CHU de Nice, Nice, France
| | - Sabine Santucci-Darmanin
- UMR E-4320 TIRO-MATOs CEA/DRF/Institut Joliot, Université Côte d'Azur, Faculté de Médecine Nice, France; CNRS, Paris, France
| | - Véronique Breuil
- UMR E-4320 TIRO-MATOs CEA/DRF/Institut Joliot, Université Côte d'Azur, Faculté de Médecine Nice, France; Service de Rhumatologie, CHU de Nice, Nice, France
| | - Georges F Carle
- UMR E-4320 TIRO-MATOs CEA/DRF/Institut Joliot, Université Côte d'Azur, Faculté de Médecine Nice, France; CNRS, Paris, France
| | - Valérie Pierrefite-Carle
- UMR E-4320 TIRO-MATOs CEA/DRF/Institut Joliot, Université Côte d'Azur, Faculté de Médecine Nice, France; INSERM, Paris, France.
| |
Collapse
|
23
|
Yuan J, Guo L, Ma J, Zhang H, Xiao M, Li N, Gong H, Yan M. HMGB1 as an extracellular pro-inflammatory cytokine: Implications for drug-induced organic damage. Cell Biol Toxicol 2024; 40:55. [PMID: 39008169 PMCID: PMC11249443 DOI: 10.1007/s10565-024-09893-2] [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: 02/14/2024] [Accepted: 06/18/2024] [Indexed: 07/16/2024]
Abstract
Drug-induced organic damage encompasses various intricate mechanisms, wherein HMGB1, a non-histone chromosome-binding protein, assumes a significant role as a pivotal hub gene. The regulatory functions of HMGB1 within the nucleus and extracellular milieu are interlinked. HMGB1 exerts a crucial regulatory influence on key biological processes including cell survival, inflammatory regulation, and immune response. HMGB1 can be released extracellularly from the cell during these processes, where it functions as a pro-inflammation cytokine. HMGB1 interacts with multiple cell membrane receptors, primarily Toll-like receptors (TLRs) and receptor for advanced glycation end products (RAGE), to stimulate immune cells and trigger inflammatory response. The excessive or uncontrolled HMGB1 release leads to heightened inflammatory responses and cellular demise, instigating inflammatory damage or exacerbating inflammation and cellular demise in different diseases. Therefore, a thorough review on the significance of HMGB1 in drug-induced organic damage is highly important for the advancement of pharmaceuticals, ensuring their effectiveness and safety in treating inflammation as well as immune-related diseases. In this review, we initially outline the characteristics and functions of HMGB1, emphasizing their relevance in disease pathology. Then, we comprehensively summarize the prospect of HMGB1 as a promising therapeutic target for treating drug-induced toxicity. Lastly, we discuss major challenges and propose potential avenues for advancing the development of HMGB1-based therapeutics.
Collapse
Affiliation(s)
- JianYe Yuan
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Hunan, China
- Xiangya School of Medicine, Central South University, Changsha, China
- Department of Pathology, The Eight Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Lin Guo
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Hunan, China
| | - JiaTing Ma
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Hunan, China
| | - HeJian Zhang
- Xiangya School of Medicine, Central South University, Changsha, China
| | - MingXuan Xiao
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Hunan, China
| | - Ning Li
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Hui Gong
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Hunan, China
| | - Miao Yan
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China.
- Institute of Clinical Pharmacy, Central South University, Changsha, China.
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Hunan, China.
| |
Collapse
|
24
|
Nakamura Y, Sawai T, Kakiuchi K, Arawaka S. Neuronal activity promotes secretory autophagy for the extracellular release of α-synuclein. J Biol Chem 2024; 300:107419. [PMID: 38815862 PMCID: PMC11253543 DOI: 10.1016/j.jbc.2024.107419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 05/15/2024] [Accepted: 05/19/2024] [Indexed: 06/01/2024] Open
Abstract
Extracellular secretion is an essential mechanism for α-synuclein (α-syn) proteostasis. Although it has been reported that neuronal activity affects α-syn secretion, the underlying mechanisms remain unclear. Here, we investigated the autophagic processes that regulate the physiological release of α-syn in mouse primary cortical neurons and SH-SY5Y cells. Stimulating neuronal activity with glutamate or depolarization with high KCl enhanced α-syn secretion. This glutamate-induced α-syn secretion was blocked by a mixture of NMDA receptor antagonist AP5 and AMPA receptor antagonist NBQX, as well as by cytosolic Ca2+ chelator BAPTA-AM. Additionally, mTOR inhibitor rapamycin increased α-syn and p62/SQSTM1 (p62) secretion, and this effect of rapamycin was reduced in primary cortical neurons deficient in the autophagy regulator beclin 1 (derived from BECN1+/- mice). Glutamate-induced α-syn and p62 secretion was suppressed by the knockdown of ATG5, which is required for autophagosome formation. Glutamate increased LC3-II generation and decreased intracellular p62 levels, and the increase in LC3-II levels was blocked by BAPTA-AM. Moreover, glutamate promoted co-localization of α-syn with LC3-positive puncta, but not with LAMP1-positive structures in the neuronal somas. Glutamate-induced α-syn and p62 secretion were also reduced by the knockdown of RAB8A, which is required for autophagosome fusion with the plasma membrane. Collectively, these findings suggest that stimulating neuronal activity mediates autophagic α-syn secretion in a cytosolic Ca2+-dependent manner, and autophagosomes may participate in autophagic secretion by functioning as α-syn carriers.
Collapse
Affiliation(s)
- Yoshitsugu Nakamura
- Division of Neurology, Department of Internal Medicine IV, Osaka Medical and Pharmaceutical University Faculty of Medicine, Takatsuki, Osaka, Japan
| | - Taiki Sawai
- Division of Neurology, Department of Internal Medicine IV, Osaka Medical and Pharmaceutical University Faculty of Medicine, Takatsuki, Osaka, Japan
| | - Kensuke Kakiuchi
- Division of Neurology, Department of Internal Medicine IV, Osaka Medical and Pharmaceutical University Faculty of Medicine, Takatsuki, Osaka, Japan
| | - Shigeki Arawaka
- Division of Neurology, Department of Internal Medicine IV, Osaka Medical and Pharmaceutical University Faculty of Medicine, Takatsuki, Osaka, Japan.
| |
Collapse
|
25
|
Lin PW, Chu ML, Liu YW, Chen YC, Shih YH, Lan SH, Wu SY, Kuo IY, Chang HY, Liu HS, Lee YR. Revealing potential Rab proteins participate in regulation of secretory autophagy machinery. Kaohsiung J Med Sci 2024; 40:642-649. [PMID: 38804615 DOI: 10.1002/kjm2.12848] [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: 03/11/2024] [Revised: 04/11/2024] [Accepted: 05/02/2024] [Indexed: 05/29/2024] Open
Abstract
Autophagy can be classified as degradative and secretory based on distinct functions. The small GTPase proteins Rab8a and Rab37 are responsible for secretory autophagy-mediated exocytosis of IL-1β, insulin, and TIMP1 (tissue inhibitor of 54 metalloproteinase 1). Other Rab family members participating in secretory autophagy are poorly understood. Herein, we identified 26 overlapped Rab proteins in purified autophagosomes of mouse pancreatic β-cell "Min-6" and human lung cancer cell "CL1-5-Q89L" with high secretory autophagy tendency by LC-MS/MS proteomics analysis. Six Rab proteins (Rab8a, Rab11b, Rab27a, Rab35, Rab37, and Rab7a) were detected in autophagosomes of four cell lines, associating them with autophagy-related vesicle trafficking. We used CL1-5-Q89L cell line model to evaluate the levels of Rab proteins colocalization with autophagy LC3 proteins and presence in purified autophagosomes. We found five Rab proteins (Rab8a, Rab11b, Rab27a, Rab35, and Rab37) are highly expressed in the autophagosome compared to the normal control by immunoblotting under active secretion conditions. However, only Rab8a, Rab35, and Rab37 showing high colocalization with LC3 protein by cofocal microscopy. Despite the discrepancy between the image and immunoblotting analysis, our data sustains the speculation that Rab8a, Rab11b, Rab27a, Rab35, and Rab37 are possibly associated with the secretory autophagy machinery. In contrast, Rab7a shows low colocalization with LC3 puncta and low level in the autophagosome, suggesting it regulates different vesicle trafficking machineries. Our findings open a new direction toward exploring the role of Rab proteins in secretory autophagy-related cargo exocytosis and identifying the cargoes and effectors regulated by specific Rab proteins.
Collapse
Affiliation(s)
- Pei-Wen Lin
- Master of Science Program in Tropical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Man-Ling Chu
- Master of Science Program in Tropical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Wen Liu
- Master of Science Program in Tropical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yu-Cing Chen
- Master of Science Program in Tropical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yao-Hsiang Shih
- Department of Anatomy, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Sheng-Hui Lan
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shang-Ying Wu
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - I-Ying Kuo
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hong-Yi Chang
- Department of Anatomy, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hsiao-Sheng Liu
- Master of Science Program in Tropical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
- Teaching and Research Center, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung, Taiwan
| | - Ying-Ray Lee
- Master of Science Program in Tropical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Microbiology and Immunology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Faculty of Post-Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
26
|
Yang X, Li J, Shan C, Song X, Yang J, Xu H, Ou D. Baicalin reduced injury of and autophagy-related gene expression in RAW264.7 cells infected with H6N6 avian influenza virus. Heliyon 2024; 10:e32645. [PMID: 38988579 PMCID: PMC11233939 DOI: 10.1016/j.heliyon.2024.e32645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 05/14/2024] [Accepted: 06/06/2024] [Indexed: 07/12/2024] Open
Abstract
In the present study, we investigated whether baicalin could reduce the damage caused to RAW264.7 cells following infection with H6N6 avian influenza virus. In addition, we studied the expression of autophagy-related genes. The morphological changes in cells were observed by hematoxylin and eosin (H&E) staining, and the inflammatory factors in the cell supernatant were detected by enzyme-linked immunosorbent assay (ELISA). Transmission electron microscopy (TEM) was used to detect the levels of RAW264.7 autophagosomes, and western blotting and immunofluorescence were used to detect the protein expression of autophagy marker LC3. Quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) was used to detect the mRNA transcription levels of autophagy key factors. The results showed that different doses of baicalin significantly reduced the H6N6 virus-induced damage of RAW264.7 cells. The contents of interleukin (IL)-1β, IL-2, IL-6, and tumor necrosis factor (TNF)-α in the cell supernatant significantly decreased. In addition, the protein expression of LC3 and Beclin-1, ATG12, ATG5 the mRNA levels were significantly decreased. This study showed that baicalin can reduce cell damage and affect the H6N6-induced autophagy level of RAW264.7 cells.
Collapse
Affiliation(s)
- Xin Yang
- College of Animal Science, Guizhou University, Guiyang, 550025, Guizhou Province, China
- Tongren Center for Prevention and Control of Animal Disease, Tongren, 554300, Guizhou Province, China
| | - Junxian Li
- College of Animal Science, Guizhou University, Guiyang, 550025, Guizhou Province, China
| | - Chunlan Shan
- College of Animal Science, Guizhou University, Guiyang, 550025, Guizhou Province, China
| | - Xuqin Song
- College of Animal Science, Guizhou University, Guiyang, 550025, Guizhou Province, China
| | - Jian Yang
- College of Animal Science, Guizhou University, Guiyang, 550025, Guizhou Province, China
| | - Hao Xu
- College of Animal Science, Guizhou University, Guiyang, 550025, Guizhou Province, China
| | - Deyuan Ou
- College of Animal Science, Guizhou University, Guiyang, 550025, Guizhou Province, China
| |
Collapse
|
27
|
Li Q, Peng G, Liu H, Wang L, Lu R, Li L. Molecular mechanisms of secretory autophagy and its potential role in diseases. Life Sci 2024; 347:122653. [PMID: 38663839 DOI: 10.1016/j.lfs.2024.122653] [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: 03/28/2024] [Accepted: 04/17/2024] [Indexed: 04/29/2024]
Abstract
Autophagy is a cellular degradation system that recycles or degrades damaged organelles, viral particles, and aggregated proteins through the lysosomal pathway. Autophagy plays an indispensable role in cellular homeostasis and communication processes. An interesting aspect is that autophagy also mediates the secretion of cellular contents, a process known as secretory autophagy. Secretory autophagy differs from macroautophagy, which sequesters recruited proteins, organelles, or viral particles into autophagosomes and degrades these sequesters in lysosomes, while the secretory autophagy pathway participates in the extracellular export of cellular contents sequestered by autophagosomes through autophagy and endosomal modulators. Recent evidence reveals that secretory autophagy is pivotal in the occurrence and progression of diseases. In this review, we summarize the molecular mechanisms of secretory autophagy. Furthermore, we review the impact of secretory autophagy on diseases, including cancer, viral infectious diseases, neurodegenerative diseases, and cardiovascular diseases. Considering the pleiotropic actions of secretory autophagy on diseases, studying the mechanism of secretory autophagy may help to understand the relevant pathophysiological processes.
Collapse
Affiliation(s)
- Qin Li
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang 421001, Hunan, China
| | - Guolong Peng
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang 421001, Hunan, China
| | - Huimei Liu
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang 421001, Hunan, China
| | - Liwen Wang
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang 421001, Hunan, China
| | - Ruirui Lu
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang 421001, Hunan, China.
| | - Lanfang Li
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang 421001, Hunan, China.
| |
Collapse
|
28
|
Cucinotta L, Mannino D, Filippone A, Romano A, Esposito E, Paterniti I. The role of autophagy in Parkinson's disease: a gender difference overview. Front Pharmacol 2024; 15:1408152. [PMID: 38933683 PMCID: PMC11199695 DOI: 10.3389/fphar.2024.1408152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
Recent studies have demonstrated dysregulation of the autophagy pathway in patients with Parkinson's disease (PD) and in animal models of PD, highlighting its emerging role in disease. In particular, several studies indicate that autophagy, which is an essential degradative process for the damaged protein homeostasis and the management of cell balance, can manifest significant variations according to gender. While some evidence suggests increased autophagic activation in men with PD, women may have distinct regulatory patterns. In this review, we examined the existing literature on gender differences in PD-associated autophagic processes, focusing on the autophagy related proteins (ATGs) and leucine rich repeat kinase 2 (LRRK2) genes. Also, this review would suggest that an in-depth understanding of these gender differences in autophagic processes could open new perspectives for personalized therapeutic strategies, promoting more effective and targeted management of PD.
Collapse
Affiliation(s)
- Laura Cucinotta
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Deborah Mannino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Alessia Filippone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Adele Romano
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, Rome, Italy
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Irene Paterniti
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| |
Collapse
|
29
|
Yuan SHC, Wu CC, Wang YC, Chan XY, Chu HW, Yang Y, Liu HP. AGR2-mediated unconventional secretion of 14-3-3ε and α-actinin-4, responsive to ER stress and autophagy, drives chemotaxis in canine mammary tumor cells. Cell Mol Biol Lett 2024; 29:84. [PMID: 38822246 PMCID: PMC11140979 DOI: 10.1186/s11658-024-00601-w] [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: 12/25/2023] [Accepted: 05/21/2024] [Indexed: 06/02/2024] Open
Abstract
BACKGROUND Canine mammary tumors (CMTs) in intact female dogs provide a natural model for investigating metastatic human cancers. Our prior research identified elevated expression of Anterior Gradient 2 (AGR2), a protein disulfide isomerase (PDI) primarily found in the endoplasmic reticulum (ER), in CMT tissues, highly associated with CMT progression. We further demonstrated that increased AGR2 expression actively influences the extracellular microenvironment, promoting chemotaxis in CMT cells. Unraveling the underlying mechanisms is crucial for assessing the potential of therapeutically targeting AGR2 as a strategy to inhibit a pro-metastatic microenvironment and impede tumor metastasis. METHODS To identify the AGR2-modulated secretome, we employed proteomics analysis of the conditioned media (CM) from two CMT cell lines ectopically expressing AGR2, compared with corresponding vector-expressing controls. AGR2-regulated release of 14-3-3ε (gene: YWHAE) and α-actinin 4 (gene: ACTN4) was validated through ectopic expression, knockdown, and knockout of the AGR2 gene in CMT cells. Extracellular vesicles derived from CMT cells were isolated using either differential ultracentrifugation or size exclusion chromatography. The roles of 14-3-3ε and α-actinin 4 in the chemotaxis driven by the AGR2-modulated CM were investigated through gene knockdown, antibody-mediated interference, and recombinant protein supplement. Furthermore, the clinical relevance of the release of 14-3-3ε and α-actinin 4 was assessed using CMT tissue-immersed saline and sera from CMT-afflicted dogs. RESULTS Proteomics analysis of the AGR2-modulated secretome revealed increased abundance in 14-3-3ε and α-actinin 4. Ectopic expression of AGR2 significantly increased the release of 14-3-3ε and α-actinin 4 in the CM. Conversely, knockdown or knockout of AGR2 expression remarkably reduced their release. Silencing 14-3-3ε or α-actinin 4 expression diminished the chemotaxis driven by AGR2-modulated CM. Furthermore, AGR2 controls the release of 14-3-3ε and α-actinin 4 primarily via non-vesicular routes, responding to the endoplasmic reticulum (ER) stress and autophagy activation. Knockout of AGR2 resulted in increased α-actinin 4 accumulation and impaired 14-3-3ε translocation in autophagosomes. Depletion of extracellular 14-3-3ε or α-actinin 4 reduced the chemotaxis driven by AGR2-modulated CM, whereas supplement with recombinant 14-3-3ε in the CM enhanced the CM-driven chemotaxis. Notably, elevated levels of 14-3-3ε or α-actinin 4 were observed in CMT tissue-immersed saline compared with paired non-tumor samples and in the sera of CMT dogs compared with healthy dogs. CONCLUSION This study elucidates AGR2's pivotal role in orchestrating unconventional secretion of 14-3-3ε and α-actinin 4 from CMT cells, thereby contributing to paracrine-mediated chemotaxis. The insight into the intricate interplay between AGR2-involved ER stress, autophagy, and unconventional secretion provides a foundation for refining strategies aimed at impeding metastasis in both canine mammary tumors and potentially human cancers.
Collapse
Affiliation(s)
- Stephen Hsien-Chi Yuan
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Chih-Ching Wu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Otolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan
- Department of Medical Biotechnology and Laboratory Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Chih Wang
- Graduate Institute of Veterinary Pathology, College of Veterinary Medicine, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Xiu-Ya Chan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hao-Wei Chu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Youngsen Yang
- Department of Oncology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Hao-Ping Liu
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung Hsing University, Taichung, 40227, Taiwan.
- Biotechnology Center, National Chung Hsing University, Taichung, 40227, Taiwan.
| |
Collapse
|
30
|
Zhang Y, Ding N, Li Y, Ouyang M, Fu P, Peng Y, Tan Y. Transcription factor FOXM1 specifies chromatin DNA to extracellular vesicles. Autophagy 2024; 20:1054-1071. [PMID: 37974331 PMCID: PMC11135825 DOI: 10.1080/15548627.2023.2284523] [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: 08/13/2023] [Revised: 10/31/2023] [Accepted: 11/10/2023] [Indexed: 11/19/2023] Open
Abstract
Extracellular vesicle DNAs (evDNAs) hold significant diagnostic value for various diseases and facilitate transcellular transfer of genetic material. Our study identifies transcription factor FOXM1 as a mediator for directing chromatin genes or DNA fragments (termed FOXM1-chDNAs) to extracellular vesicles (EVs). FOXM1 binds to MAP1LC3/LC3 in the nucleus, and FOXM1-chDNAs, such as the DUX4 gene and telomere DNA, are designated by FOXM1 binding and translocated to the cytoplasm before being released to EVs through the secretory autophagy during lysosome inhibition (SALI) process involving LC3. Disrupting FOXM1 expression or the SALI process impairs FOXM1-chDNAs incorporation into EVs. FOXM1-chDNAs can be transmitted to recipient cells via EVs and expressed in recipient cells when they carry functional genes. This finding provides an example of how chromatin DNA fragments are specified to EVs by transcription factor FOXM1, revealing its contribution to the formation of evDNAs from nuclear chromatin. It provides a basis for further exploration of the roles of evDNAs in biological processes, such as horizontal gene transfer.Abbreviation: ATG5: autophagy related 5; CCFs: cytoplasmic chromatin fragments; ChIP: chromatin immunoprecipitation; cytoDNA: cytoplasmic DNA; CQ: chloroquine; FOXM1-DBD: FOXM1 DNA binding domain; DUX4:double homeobox 4; EVs: extracellular vesicles; evDNAs: extracellular vesicle DNAs; FOXM1: forkhead box M1; FOXM1-chDNAs: chromatin DNA fragments directed by FOXM1 to EVs; HGT: horizontal gene transfer; LC3-II: lipid modified LC3; LMNB1: lamin B1; LIR: LC3-interacting region; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MVBs: multivesicular bodies; M1-binding DNA: a linear DNA containing 72× FOXM1 binding sites; SALI: secretory autophagy during lysosome inhibition; siRNA: small interfering RNA; TetO-DUX4: TetO array-containing DUX4 DNA; TetO: tet operator; TetR: tet repressor.
Collapse
Affiliation(s)
- Yunsheng Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, Changsha, Hunan, PR China
- The Second Affiliated Hospital, University of South China, Hengyang, Hunan, PR China
| | - Nana Ding
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, Changsha, Hunan, PR China
| | - Yizhen Li
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, Changsha, Hunan, PR China
| | - Min Ouyang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, Changsha, Hunan, PR China
| | - Ping Fu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, Changsha, Hunan, PR China
| | - Yousong Peng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, Changsha, Hunan, PR China
| | - Yongjun Tan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, Changsha, Hunan, PR China
| |
Collapse
|
31
|
Chang YC, Gao Y, Lee JY, Peng YJ, Langen J, Chang KT. Identification of secretory autophagy as a mechanism modulating activity-induced synaptic remodeling. Proc Natl Acad Sci U S A 2024; 121:e2315958121. [PMID: 38588427 PMCID: PMC11032469 DOI: 10.1073/pnas.2315958121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 02/23/2024] [Indexed: 04/10/2024] Open
Abstract
The ability of neurons to rapidly remodel their synaptic structure and strength in response to neuronal activity is highly conserved across species and crucial for complex brain functions. However, mechanisms required to elicit and coordinate the acute, activity-dependent structural changes across synapses are not well understood, as neurodevelopment and structural plasticity are tightly linked. Here, using an RNAi screen in Drosophila against genes affecting nervous system functions in humans, we uncouple cellular processes important for synaptic plasticity and synapse development. We find mutations associated with neurodegenerative and mental health disorders are 2-times more likely to affect activity-induced synaptic remodeling than synapse development. We report that while both synapse development and activity-induced synaptic remodeling at the fly NMJ require macroautophagy (hereafter referred to as autophagy), bifurcation in the autophagy pathway differentially impacts development and synaptic plasticity. We demonstrate that neuronal activity enhances autophagy activation but diminishes degradative autophagy, thereby driving the pathway towards autophagy-based secretion. Presynaptic knockdown of Snap29, Sec22, or Rab8, proteins implicated in the secretory autophagy pathway, is sufficient to abolish activity-induced synaptic remodeling. This study uncovers secretory autophagy as a transsynaptic signaling mechanism modulating synaptic plasticity.
Collapse
Affiliation(s)
- Yen-Ching Chang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| | - Yuan Gao
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| | - Joo Yeun Lee
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| | - Yi-Jheng Peng
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| | - Jennifer Langen
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| | - Karen T. Chang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| |
Collapse
|
32
|
Pavlyuchenkova AN, Smirnov MS, Chernyak BV, Chelombitko MA. The Role Played by Autophagy in FcεRI-Dependent Activation of Mast Cells. Cells 2024; 13:690. [PMID: 38667305 PMCID: PMC11049365 DOI: 10.3390/cells13080690] [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: 02/29/2024] [Revised: 04/10/2024] [Accepted: 04/14/2024] [Indexed: 04/28/2024] Open
Abstract
The significant role of mast cells in the development of allergic and inflammatory diseases is well-established. Among the various mechanisms of mast cell activation, the interaction of antigens/allergens with IgE and the subsequent binding of this complex to the high-affinity IgE receptor FcεRI stand out as the most studied and fundamental pathways. This activation process leads to the rapid exocytosis of granules containing preformed mediators, followed by the production of newly synthesized mediators, including a diverse array of cytokines, chemokines, arachidonic acid metabolites, and more. While conventional approaches to allergy control primarily focus on allergen avoidance and the use of antihistamines (despite their associated side effects), there is increasing interest in exploring novel methods to modulate mast cell activity in modern medicine. Recent evidence suggests a role for autophagy in mast cell activation, offering potential avenues for utilizing low-molecular-weight autophagy regulators in the treatment of allergic diseases. More specifically, mitochondria, which play an important role in the regulation of autophagy as well as mast cell activation, emerge as promising targets for drug development. This review examines the existing literature regarding the involvement of the molecular machinery associated with autophagy in FcεRI-dependent mast cell activation.
Collapse
Affiliation(s)
- Anastasia N. Pavlyuchenkova
- Belozersky Institute of Physicochemical Biology, Moscow State University, Moscow 119992, Russia; (A.N.P.)
- Faculty of Bioengineering and Bioinformatics, Moscow State University, Moscow 119992, Russia
| | - Maxim S. Smirnov
- Belozersky Institute of Physicochemical Biology, Moscow State University, Moscow 119992, Russia; (A.N.P.)
- Faculty of Bioengineering and Bioinformatics, Moscow State University, Moscow 119992, Russia
| | - Boris V. Chernyak
- Belozersky Institute of Physicochemical Biology, Moscow State University, Moscow 119992, Russia; (A.N.P.)
| | - Maria A. Chelombitko
- Belozersky Institute of Physicochemical Biology, Moscow State University, Moscow 119992, Russia; (A.N.P.)
| |
Collapse
|
33
|
Hartmann J, Bajaj T, Otten J, Klengel C, Ebert T, Gellner AK, Junglas E, Hafner K, Anderzhanova EA, Tang F, Missig G, Rexrode L, Trussell DT, Li KX, Pöhlmann ML, Mackert S, Geiger TM, Heinz DE, Lardenoije R, Dedic N, McCullough KM, Próchnicki T, Rhomberg T, Martinelli S, Payton A, Robinson AC, Stein V, Latz E, Carlezon WA, Hausch F, Schmidt MV, Murgatroyd C, Berretta S, Klengel T, Pantazopoulos H, Ressler KJ, Gassen NC. SKA2 regulated hyperactive secretory autophagy drives neuroinflammation-induced neurodegeneration. Nat Commun 2024; 15:2635. [PMID: 38528004 PMCID: PMC10963788 DOI: 10.1038/s41467-024-46953-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 03/15/2024] [Indexed: 03/27/2024] Open
Abstract
High levels of proinflammatory cytokines induce neurotoxicity and catalyze inflammation-driven neurodegeneration, but the specific release mechanisms from microglia remain elusive. Here we show that secretory autophagy (SA), a non-lytic modality of autophagy for secretion of vesicular cargo, regulates neuroinflammation-mediated neurodegeneration via SKA2 and FKBP5 signaling. SKA2 inhibits SA-dependent IL-1β release by counteracting FKBP5 function. Hippocampal Ska2 knockdown in male mice hyperactivates SA resulting in neuroinflammation, subsequent neurodegeneration and complete hippocampal atrophy within six weeks. The hyperactivation of SA increases IL-1β release, contributing to an inflammatory feed-forward vicious cycle including NLRP3-inflammasome activation and Gasdermin D-mediated neurotoxicity, which ultimately drives neurodegeneration. Results from protein expression and co-immunoprecipitation analyses of male and female postmortem human brains demonstrate that SA is hyperactivated in Alzheimer's disease. Overall, our findings suggest that SKA2-regulated, hyperactive SA facilitates neuroinflammation and is linked to Alzheimer's disease, providing mechanistic insight into the biology of neuroinflammation.
Collapse
Affiliation(s)
- Jakob Hartmann
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA.
| | - Thomas Bajaj
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University of Bonn, 53127, Bonn, Germany
| | - Joy Otten
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, 37075, Göttingen, Germany
| | - Claudia Klengel
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Tim Ebert
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University of Bonn, 53127, Bonn, Germany
| | - Anne-Kathrin Gellner
- Department of Psychiatry and Psychotherapy, University of Bonn, 53127, Bonn, Germany
| | - Ellen Junglas
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University of Bonn, 53127, Bonn, Germany
| | - Kathrin Hafner
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Elmira A Anderzhanova
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University of Bonn, 53127, Bonn, Germany
- Research Group Neuronal Plasticity, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Fiona Tang
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Galen Missig
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Lindsay Rexrode
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Daniel T Trussell
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Katelyn X Li
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Max L Pöhlmann
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Sarah Mackert
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University of Bonn, 53127, Bonn, Germany
| | - Thomas M Geiger
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, 64287, Darmstadt, Germany
| | - Daniel E Heinz
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University of Bonn, 53127, Bonn, Germany
- Research Group Neuronal Plasticity, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Roy Lardenoije
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, 37075, Göttingen, Germany
| | - Nina Dedic
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Kenneth M McCullough
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Tomasz Próchnicki
- Institute of Innate Immunity, University Hospital Bonn, 53127, Bonn, Germany
| | - Thomas Rhomberg
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Silvia Martinelli
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Antony Payton
- Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, M13 9PL, UK
| | - Andrew C Robinson
- Division of Neuroscience, Faculty of Biology, Medicine and Health, School of Biological Sciences, The University of Manchester, Salford Royal Hospital, Salford, M6 8HD, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre (MAHSC), Manchester, UK
| | - Valentin Stein
- Institute of Physiology II, University of Bonn, 53127, Bonn, Germany
| | - Eicke Latz
- Institute of Innate Immunity, University Hospital Bonn, 53127, Bonn, Germany
- Deutsches Rheuma Forschungszentrum Berlin (DRFZ), 10117, Berlin, Germany
| | - William A Carlezon
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Felix Hausch
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, 64287, Darmstadt, Germany
| | - Mathias V Schmidt
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Chris Murgatroyd
- Department of Life Sciences, Manchester Metropolitan University, Manchester, M15 6BH, UK
| | - Sabina Berretta
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Torsten Klengel
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, 37075, Göttingen, Germany
| | - Harry Pantazopoulos
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Kerry J Ressler
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA.
| | - Nils C Gassen
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University of Bonn, 53127, Bonn, Germany.
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804, Munich, Germany.
| |
Collapse
|
34
|
Wang L, Xu Y, Fukushige T, Saidi L, Wang X, Yu C, Lee JG, Krause M, Huang L, Ye Y. Mono-UFMylation promotes misfolding-associated secretion of α-synuclein. SCIENCE ADVANCES 2024; 10:eadk2542. [PMID: 38489364 PMCID: PMC10942102 DOI: 10.1126/sciadv.adk2542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 02/12/2024] [Indexed: 03/17/2024]
Abstract
Stressed cells secret misfolded proteins lacking signaling sequence via an unconventional protein secretion (UcPS) pathway, but how misfolded proteins are targeted selectively in UcPS is unclear. Here, we report that misfolded UcPS clients are subject to modification by a ubiquitin-like protein named ubiquitin-fold modifier 1 (UFM1). Using α-synuclein (α-Syn) as a UcPS model, we show that mutating the UFMylation sites in α-Syn or genetic inhibition of the UFMylation system mitigates α-Syn secretion, whereas overexpression of UFBP1, a component of the endoplasmic reticulum-associated UFMylation ligase complex, augments α-Syn secretion in mammalian cells and in model organisms. UFM1 itself is cosecreted with α-Syn, and the serum UFM1 level correlates with that of α-Syn. Because UFM1 can be directly recognized by ubiquitin specific peptidase 19 (USP19), a previously established UcPS stimulator known to associate with several chaperoning activities, UFMylation might facilitate substrate engagement by USP19, allowing stringent and regulated selection of misfolded proteins for secretion and proteotoxic stress alleviation.
Collapse
Affiliation(s)
- Lihui Wang
- Laboratory of Molecular Biology, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yue Xu
- Laboratory of Molecular Biology, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tetsunari Fukushige
- Laboratory of Molecular Biology, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Layla Saidi
- Laboratory of Molecular Biology, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Xiaorong Wang
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA 92697, USA
| | - Clinton Yu
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA 92697, USA
| | - Jin-Gu Lee
- Laboratory of Molecular Biology, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael Krause
- Laboratory of Molecular Biology, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lan Huang
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA 92697, USA
| | - Yihong Ye
- Laboratory of Molecular Biology, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
35
|
Li X, Gao L, Li X, Xia J, Pan Y, Bai C. Autophagy, Pyroptosis and Ferroptosis are Rising Stars in the Pathogenesis of Diabetic Nephropathy. Diabetes Metab Syndr Obes 2024; 17:1289-1299. [PMID: 38505538 PMCID: PMC10949337 DOI: 10.2147/dmso.s450695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 03/01/2024] [Indexed: 03/21/2024] Open
Abstract
Diabetic nephropathy (DN) is one of the most common microvascular complications in diabetes and can potentially develop into end-stage renal disease. Its pathogenesis is complex and not fully understood. Podocytes, glomerular endothelial cells (GECs), glomerular mesangial cells (GMCs) and renal tubular epithelial cells (TECs) play important roles in the normal function of glomerulus and renal tubules, and their injury is involved in the progression of DN. Although our understanding of the mechanisms leading to DN has substantially improved, we still need to find more effective therapeutic targets. Autophagy, pyroptosis and ferroptosis are programmed cell death processes that are associated with inflammation and are closely related to a variety of diseases. Recently, a growing number of studies have reported that autophagy, pyroptosis and ferroptosis regulate the function of podocytes, GECs, GMCs and TECs. This review highlights the contributions of autophagy, pyroptosis, and ferroptosis to DN injury in these cells, offering potential therapeutic targets for DN treatment.
Collapse
Affiliation(s)
- Xiudan Li
- Department of Endocrinology, Affiliated Hospital of Chifeng University, Chifeng, 024000, China
- Inner Mongolia Key Laboratory of Human Genetic Disease Research, Chifeng University, Chifeng, 024000, China
| | - Lifeng Gao
- Inner Mongolia Key Laboratory of Human Genetic Disease Research, Chifeng University, Chifeng, 024000, China
| | - Xuyang Li
- Inner Mongolia Key Laboratory of Human Genetic Disease Research, Chifeng University, Chifeng, 024000, China
| | - Jingdong Xia
- Department of Endocrinology, Affiliated Hospital of Chifeng University, Chifeng, 024000, China
| | - Yurong Pan
- Department of Endocrinology, Affiliated Hospital of Chifeng University, Chifeng, 024000, China
| | - Chunying Bai
- Inner Mongolia Key Laboratory of Human Genetic Disease Research, Chifeng University, Chifeng, 024000, China
| |
Collapse
|
36
|
Zubkova E, Kalinin A, Bolotskaya A, Beloglazova I, Menshikov M. Autophagy-Dependent Secretion: Crosstalk between Autophagy and Exosome Biogenesis. Curr Issues Mol Biol 2024; 46:2209-2235. [PMID: 38534758 DOI: 10.3390/cimb46030142] [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: 11/30/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 03/28/2024] Open
Abstract
The cellular secretome is pivotal in mediating intercellular communication and coordinating responses to stressors. Exosomes, initially recognized for their role in waste disposal, have now emerged as key intercellular messengers with significant therapeutic and diagnostic potential. Similarly, autophagy has transcended its traditional role as a waste removal mechanism, emerging as a regulator of intracellular communication pathways and a contributor to a unique autophagy-dependent secretome. Secretory authophagy, initiated by various stress stimuli, prompts the selective release of proteins implicated in inflammation, including leaderless proteins that bypass the conventional endoplasmic reticulum-Golgi secretory pathway. This reflects the significant impact of stress-induced autophagy on cellular secretion profiles, including the modulation of exosome release. The convergence of exosome biogenesis and autophagy is exemplified by the formation of amphisomes, vesicles that integrate autophagic and endosomal pathways, indicating their synergistic interplay. Regulatory proteins common to both pathways, particularly mTORC1, emerge as potential therapeutic targets to alter cellular secretion profiles involved in various diseases. This review explores the dynamic interplay between autophagy and exosome formation, highlighting the potential to influence the secretome composition. While the modulation of exosome secretion and cytokine preconditioning is well-established in regenerative medicine, the strategic manipulation of autophagy is still underexplored, presenting a promising but uncharted therapeutic landscape.
Collapse
Affiliation(s)
- Ekaterina Zubkova
- National Medical Research Centre of Cardiology Named after Academician E.I. Chazov, 121552 Moscow, Russia
| | - Alexander Kalinin
- National Medical Research Centre of Cardiology Named after Academician E.I. Chazov, 121552 Moscow, Russia
- Faculty of Fundamental Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Anastasya Bolotskaya
- National Medical Research Centre of Cardiology Named after Academician E.I. Chazov, 121552 Moscow, Russia
- Institute of Clinical Medicine, Sechenov University, 119435 Moscow, Russia
| | - Irina Beloglazova
- National Medical Research Centre of Cardiology Named after Academician E.I. Chazov, 121552 Moscow, Russia
| | - Mikhail Menshikov
- National Medical Research Centre of Cardiology Named after Academician E.I. Chazov, 121552 Moscow, Russia
| |
Collapse
|
37
|
Pan Y, Liu J, Wang D, Zhou P, Chen T, Tang Y, Ji C. DADS Inhibits the Proliferation of MDCC-MSB-1 Cells by Inducing Autophagy via the MEK/ERK Signalling Pathway. Cell Biochem Biophys 2024; 82:271-278. [PMID: 38214811 DOI: 10.1007/s12013-023-01214-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 12/26/2023] [Indexed: 01/13/2024]
Abstract
Diallyl disulfide (DADS) is effective at suppressing tumour cell growth and proliferation. This study verified the morphology and growth activity of MDCC-MSB-1 cells by using an MTT assay to detect the effect of DADS on the proliferation of MDCC-MSB-1 cells and a CCK8 assay to detect the effect of DADS on the viability and proliferation of MDCC-MSB-1 cells. We found that the viability and proliferation of MDCC-MSB-1 cells decreased with increasing DADS concentrations. MDC staining and Western blotting were used to analyse autophagy, the associated protein LC3 and the MEK/ERK pathway proteins MEK and ERK and to investigate changes in cellular autophagy based on cell morphology and molecular biology. With increasing concentrations of DADS, MDCC-MSB-1 cell autophagy increased in a gradient manner. Additionally, the conversion of the autophagy marker protein LC3-I increased with increasing drug concentrations, and the relative expression of LC3-II steadily increased, as did the expression of key protein components of the MEK/ERK signalling pathway, including P-MEK1/2 and P-ERK1/2. These results suggest that DADS induces autophagy through the MEK/ERK pathway, thereby inhibiting the proliferation of MDCC-MSB-1 cells.
Collapse
Affiliation(s)
- Yanying Pan
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
- Hunan Engineering Technology Research Center of Veterinary Drugs, Hunan Agricultural University, Changsha, 410128, China
| | - Jianying Liu
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
- Hunan Engineering Technology Research Center of Veterinary Drugs, Hunan Agricultural University, Changsha, 410128, China
| | - Dongliang Wang
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
- Hunan Engineering Technology Research Center of Veterinary Drugs, Hunan Agricultural University, Changsha, 410128, China
| | - Pengfei Zhou
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
- Hunan Engineering Technology Research Center of Veterinary Drugs, Hunan Agricultural University, Changsha, 410128, China
| | - Tao Chen
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
- Hunan Engineering Technology Research Center of Veterinary Drugs, Hunan Agricultural University, Changsha, 410128, China
| | - Yulong Tang
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan, Changsha, 410125, China
| | - Chunxiao Ji
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China.
- Hunan Engineering Technology Research Center of Veterinary Drugs, Hunan Agricultural University, Changsha, 410128, China.
| |
Collapse
|
38
|
Gao M, Shen H, Li Q, Gu X, Jia T, Wang Y. Perfluorooctane sulfonate (PFOS) induces apoptosis and autophagy by inhibition of PI3K/AKT/mTOR pathway in human granulosa cell line KGN. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 344:123333. [PMID: 38211877 DOI: 10.1016/j.envpol.2024.123333] [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: 10/17/2023] [Revised: 12/22/2023] [Accepted: 01/07/2024] [Indexed: 01/13/2024]
Abstract
Perfluorooctane sulfonate (PFOS) is recognized as an environmental endocrine disruptor with widespread use in industrial manufacturing and daily life, contributing to various public health concerns. However, the precise impacts of PFOS on the ovary and its regulatory mechanisms remain unclear. This study aims to delineate the ovarian toxicity of PFOS and scrutinize its effects on apoptosis and autophagy through modulation of the PI3K/AKT/mTOR pathway in the human granulosa cell line (KGN). Cell viability, assessed via the Cell Counting Kit-8 (CCK8), revealed a dose-dependent reduction in cell viability upon PFOS exposure. Flow cytometry analysis demonstrated an elevated proportion of apoptotic cells following PFOS treatment. Western blot analyses unveiled increased expression of Bax, Cyt c, cleaved caspase-9, and LC3-II/I, coupled with decreased expression of Bcl-2 and p62. Transmission electron microscopy (TEM) observations illustrated a heightened number of autophagosomes induced by PFOS. Molecular docking investigations, in conjunction with Western blot experiments, substantiated PFOS's significant inhibition of the PI3K/AKT/mTOR signaling pathway. These findings collectively underscore that PFOS induces apoptosis and autophagy in KGN cells through modulation of the PI3K/AKT/mTOR pathway, providing experimental evidence for PFOS-induced ovarian toxicity and elucidating the underlying regulatory mechanisms in KGN cells.
Collapse
Affiliation(s)
- Min Gao
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Haofei Shen
- The First Clinical Medical College, Lanzhou University, Lanzhou, China; The First Hospital of Lanzhou University, Lanzhou, China
| | - Qiuyuan Li
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Xuzhao Gu
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Tianyu Jia
- The First Clinical Medical College, Lanzhou University, Lanzhou, China; The First Hospital of Lanzhou University, Lanzhou, China
| | - Yiqing Wang
- The First School of Clinical Medicine & Research Unit of Peptide Science, Chinese Academy of Medical Science, 2019RU066, Lanzhou University, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Reproductive Medicine Transformation Application, Key Laboratory for Reproductive Medicine and Embryo of Gansu Province & Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou, China.
| |
Collapse
|
39
|
Hu M, Fan JX, He ZY, Zeng J. The regulatory role of autophagy between TAMs and tumor cells. Cell Biochem Funct 2024; 42:e3984. [PMID: 38494666 DOI: 10.1002/cbf.3984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 03/19/2024]
Abstract
Cancer has become a global public health problem and its harmful effects have received widespread attention. Conventional treatments such as surgical resection, radiotherapy and other techniques are applicable to clinical practice, but new drugs are constantly being developed and other therapeutic approaches, such as immunotherapy are being applied. In addition to studying the effects on individual tumor cells, it is important to explore the role of tumor microenvironment on tumor cell development since tumor cells do not exist alone but in the tumor microenvironment. In the tumor microenvironment, tumor cells are interconnected with other stromal cells and influence each other, among which tumor-associated macrophages (TAMs) are the most numerous immune cells. At the same time, it was found that cancer cells have different levels of autophagy from normal cells. In cancer therapy, the occurrence of autophagy plays an important role in promoting tumor cell death or inhibiting tumor cell death, and is closely related to the environment. Therefore, elucidating the regulatory role of autophagy between TAMs and tumor cells may be an important breakthrough, providing new perspectives for further research on antitumor immune mechanisms and improving the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Min Hu
- College of Life Sciences, Chongqing Normal University, Chongqing, 401331, China
| | - Jiao-Xiu Fan
- College of Life Sciences, Chongqing Normal University, Chongqing, 401331, China
| | - Zi-Yue He
- College of Life Sciences, Chongqing Normal University, Chongqing, 401331, China
| | - Jun Zeng
- College of Life Sciences, Chongqing Normal University, Chongqing, 401331, China
- Animal Biology Key Laboratory of Chongqing Education Commission of China
| |
Collapse
|
40
|
Pan M, Hu T, Lyu J, Yin Y, Sun J, Wang Q, Xu L, Hu H, Wang C. CSNK1A1/CK1α suppresses autoimmunity by restraining the CGAS-STING1 signaling. Autophagy 2024; 20:311-328. [PMID: 37723657 PMCID: PMC10813568 DOI: 10.1080/15548627.2023.2256135] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 08/22/2023] [Accepted: 08/31/2023] [Indexed: 09/20/2023] Open
Abstract
STING1 (stimulator of interferon response cGAMP interactor 1) is the quintessential protein in the CGAS-STING1 signaling pathway, crucial for the induction of type I IFN (interferon) production and eliciting innate immunity. Nevertheless, the overactivation or sustained activation of STING1 has been closely associated with the onset of autoimmune disorders. Notably, the majority of these disorders manifest as an upregulated expression of type I interferons and IFN-stimulated genes (ISGs). Hence, strict regulation of STING1 activity is paramount to preserve immune homeostasis. Here, we reported that CSNK1A1/CK1α, a serine/threonine protein kinase, was essential to prevent the overactivation of STING1-mediated type I IFN signaling through autophagic degradation of STING1. Mechanistically, CSNK1A1 interacted with STING1 upon the CGAS-STING1 pathway activation and promoted STING1 autophagic degradation by enhancing the phosphorylation of SQSTM1/p62 at serine 351 (serine 349 in human), which was critical for SQSTM1-mediated STING1 autophagic degradation. Consistently, SSTC3, a selective CSNK1A1 agonist, significantly attenuated the response of the CGAS-STING1 signaling by promoting STING1 autophagic degradation. Importantly, pharmacological activation of CSNK1A1 using SSTC3 markedly repressed the systemic autoinflammatory responses in the trex1-/- mouse autoimmune disease model and effectively suppressed the production of IFNs and ISGs in the PBMCs of SLE patients. Taken together, our study reveals a novel regulatory role of CSNK1A1 in the autophagic degradation of STING1 to maintain immune homeostasis. Manipulating CSNK1A1 through SSTC3 might be a potential therapeutic strategy for alleviating STING1-mediated aberrant type I IFNs in autoimmune diseases.Abbreviations: BMDMs: bone marrow-derived macrophages; cGAMP: cyclic GMP-AMP; CGAS: cyclic GMP-AMP synthase; HTDNA: herring testes DNA; IFIT1: interferon induced protein with tetratricopeptide repeats 1; IFNA4: interferon alpha 4; IFNB: interferon beta; IRF3: interferon regulatory factor 3; ISD: interferon stimulatory DNA; ISGs: IFN-stimulated genes; MEFs: mouse embryonic fibroblasts; PBMCs: peripheral blood mononuclear cells; RSAD2: radical S-adenosyl methionine domain containing 2; SLE: systemic lupus erythematosus; STING1: stimulator of interferon response cGAMP interactor 1; TBK1: TANK binding kinase 1.
Collapse
Affiliation(s)
- Mingyu Pan
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
- Department of Biomedical Science, City University of Hong Kong, Hong Kong, Hong Kong, China
| | - Tongyu Hu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Jiao Lyu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yue Yin
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Jing Sun
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Quanyi Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Lingxiao Xu
- Department of Rheumatology, The affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, Jiangsu, China
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Haiyang Hu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Chen Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
41
|
Chen R, Zou J, Zhong X, Li J, Kang R, Tang D. HMGB1 in the interplay between autophagy and apoptosis in cancer. Cancer Lett 2024; 581:216494. [PMID: 38007142 DOI: 10.1016/j.canlet.2023.216494] [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: 07/21/2023] [Revised: 10/25/2023] [Accepted: 11/08/2023] [Indexed: 11/27/2023]
Abstract
Lysosome-mediated autophagy and caspase-dependent apoptosis are dynamic processes that maintain cellular homeostasis, ensuring cell health and functionality. The intricate interplay and reciprocal regulation between autophagy and apoptosis are implicated in various human diseases, including cancer. High-mobility group box 1 (HMGB1), a nonhistone chromosomal protein, plays a pivotal role in coordinating autophagy and apoptosis levels during tumor initiation, progression, and therapy. The regulation of autophagy machinery and the apoptosis pathway by HMGB1 is influenced by various factors, including the protein's subcellular localization, oxidative state, and interactions with binding partners. In this narrative review, we provide a comprehensive overview of the structure and function of HMGB1, with a specific focus on the interplay between autophagic degradation and apoptotic death in tumorigenesis and cancer therapy. Gaining a comprehensive understanding of the significance of HMGB1 as a biomarker and its potential as a therapeutic target in tumor diseases is crucial for advancing our knowledge of cell survival and cell death.
Collapse
Affiliation(s)
- Ruochan Chen
- Department of Infectious Diseases, Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Ju Zou
- Department of Infectious Diseases, Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Xiao Zhong
- Department of Infectious Diseases, Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Jie Li
- Department of Infectious Diseases, Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
42
|
Kim WD, DiGiacinto AF, Huber RJ. Assaying Lysosomal Enzyme Activity in Dictyostelium discoideum. Methods Mol Biol 2024; 2814:55-79. [PMID: 38954197 DOI: 10.1007/978-1-0716-3894-1_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Lysosomes are membrane-enclosed organelles that digest intracellular material. They contain more than 50 different enzymes that can degrade a variety of macromolecules including nucleic acids, proteins, polysaccharides, and lipids. In addition to functioning within lysosomes, lysosomal enzymes are also secreted. Alterations in the levels and activities of lysosomal enzymes dysregulates lysosomes, which can lead to the intralysosomal accumulation of biological material and the development of lysosomal storage diseases (LSDs) in humans. Dictyostelium discoideum has a long history of being used to study the trafficking and functions of lysosomal enzymes. More recently, it has been used as a model system to study several LSDs. In this chapter, we outline the methods for assessing the activity of several lysosomal enzymes in D. discoideum (α-galactosidase, β-galactosidase, α-glucosidase, β-glucosidase, β-N-acetylglucosaminidase, α-mannosidase, cathepsin B, cathepsin D, cathepsin F, palmitoyl protein thioesterase 1, and tripeptidyl peptidase 1).
Collapse
Affiliation(s)
- William D Kim
- Environmental and Life Sciences Graduate Program, Trent University, Peterborough, ON, Canada
| | | | - Robert J Huber
- Environmental and Life Sciences Graduate Program, Trent University, Peterborough, ON, Canada.
- Department of Biology, Trent University, Peterborough, ON, Canada.
| |
Collapse
|
43
|
Zhang Y, Ma S, Nie J, Liu Z, Chen F, Li A, Pei D. Journey of Mineral Precursors in Bone Mineralization: Evolution and Inspiration for Biomimetic Design. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2207951. [PMID: 37621037 DOI: 10.1002/smll.202207951] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 06/27/2023] [Indexed: 08/26/2023]
Abstract
Bone mineralization is a ubiquitous process among vertebrates that involves a dynamic physical/chemical interplay between the organic and inorganic components of bone tissues. It is now well documented that carbonated apatite, an inorganic component of bone, is proceeded through transient amorphous mineral precursors that transforms into the crystalline mineral phase. Here, the evolution on mineral precursors from their sources to the terminus in the bone mineralization process is reviewed. How organisms tightly control each step of mineralization to drive the formation, stabilization, and phase transformation of amorphous mineral precursors in the right place, at the right time, and rate are highlighted. The paradigm shifts in biomineralization and biomaterial design strategies are intertwined, which promotes breakthroughs in biomineralization-inspired material. The design principles and implementation methods of mineral precursor-based biomaterials in bone graft materials such as implant coatings, bone cements, hydrogels, and nanoparticles are detailed in the present manuscript. The biologically controlled mineralization mechanisms will hold promise for overcoming the barriers to the application of biomineralization-inspired biomaterials.
Collapse
Affiliation(s)
- Yuchen Zhang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Shaoyang Ma
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Jiaming Nie
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zhongbo Liu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Faming Chen
- School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Ang Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Dandan Pei
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China
| |
Collapse
|
44
|
Giansanti M, Theinert T, Boeing SK, Haas D, Schlegel PG, Vacca P, Nazio F, Caruana I. Exploiting autophagy balance in T and NK cells as a new strategy to implement adoptive cell therapies. Mol Cancer 2023; 22:201. [PMID: 38071322 PMCID: PMC10709869 DOI: 10.1186/s12943-023-01893-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 10/30/2023] [Indexed: 12/18/2023] Open
Abstract
Autophagy is an essential cellular homeostasis pathway initiated by multiple stimuli ranging from nutrient deprivation to viral infection, playing a key role in human health and disease. At present, a growing number of evidence suggests a role of autophagy as a primitive innate immune form of defense for eukaryotic cells, interacting with components of innate immune signaling pathways and regulating thymic selection, antigen presentation, cytokine production and T/NK cell homeostasis. In cancer, autophagy is intimately involved in the immunological control of tumor progression and response to therapy. However, very little is known about the role and impact of autophagy in T and NK cells, the main players in the active fight against infections and tumors. Important questions are emerging: what role does autophagy play on T/NK cells? Could its modulation lead to any advantages? Could specific targeting of autophagy on tumor cells (blocking) and T/NK cells (activation) be a new intervention strategy? In this review, we debate preclinical studies that have identified autophagy as a key regulator of immune responses by modulating the functions of different immune cells and discuss the redundancy or diversity among the subpopulations of both T and NK cells in physiologic context and in cancer.
Collapse
Affiliation(s)
- Manuela Giansanti
- Immunology Research Area, Innate Lymphoid Cells Unit, Bambino Gesù Children's Hospital (IRCCS), Rome, Italy
| | - Tobias Theinert
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Sarah Katharina Boeing
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Dorothee Haas
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Paul-Gerhardt Schlegel
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Paola Vacca
- Immunology Research Area, Innate Lymphoid Cells Unit, Bambino Gesù Children's Hospital (IRCCS), Rome, Italy
| | - Francesca Nazio
- Immunology Research Area, Innate Lymphoid Cells Unit, Bambino Gesù Children's Hospital (IRCCS), Rome, Italy.
- Department of Biology, University of Rome Tor Vergata, 00133, Rome, Italy.
| | - Ignazio Caruana
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Würzburg, 97080, Würzburg, Germany.
| |
Collapse
|
45
|
Cui X, Yao A, Jia L. Starvation insult induces the translocation of high mobility group box 1 to cytosolic compartments in glioma. Oncol Rep 2023; 50:216. [PMID: 37888772 PMCID: PMC10636726 DOI: 10.3892/or.2023.8653] [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: 05/03/2023] [Accepted: 10/04/2023] [Indexed: 10/28/2023] Open
Abstract
High mobility group box 1 (HMGB1) is a highly conserved and ubiquitous nuclear protein in eukaryotic cells. In response to stress, it transfers from the nucleus to the cytoplasm and finally, to the extracellular matrix, participating in inflammation and carcinogenesis. Increased HMGB1 protein levels are frequently associated with the reduced survival of patients with glioma. HMGB1 plays contextual roles depending on its subcellular localization. However, the mechanisms underlying its subcellular localization and secretion remain unclear. In the present study, the subcellular localization and secretion of HMGB1 in starved glioma cells were investigated using immunofluorescence microscopy, enzyme‑linked immunosorbent assay, subcellular fractionation, western blotting and immunoelectron microscopy. The results demonstrated that starvation induced HMGB1 translocation from the nucleus to the cytoplasm and finally, to the extracellular milieu in glioma cells. HMGB1 was localized in the mitochondria, endoplasmic reticulum (ER), peroxisomes, autophagosomes, lysosomes, endosomes and the cytoskeleton. Immunoelectron microscopy confirmed that HMGB1 was present within or around cytosolic compartments. Subcellular fractionation further demonstrated that HMGB1 transferred to membrane‑bound compartments. In addition, HMGB1 was localized to specific contact areas between the ER and mitochondria, known as mitochondria‑associated membranes. On the whole, the results of the present study suggest that starvation induces HMGB1 secretion, which can be inhibited through the suppression of autophagy. Starvation insult induces HMGB1 translocation to the cytosolic compartments of glioma cells, and autophagy may be involved in the extracellular secretion of HMGB1 in starved glioma cells.
Collapse
Affiliation(s)
- Xiaohang Cui
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Anhui Yao
- Department of Neurosurgery, 988th Hospital of Joint Logistic Support Force of PLA, Zhengzhou, Henan 450053, P.R. China
| | - Liyun Jia
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| |
Collapse
|
46
|
Wang W, Li X, Cui C, Yin G, Ren W, Wang X. Autophagy of umbilical cord mesenchymal stem cells induced by rapamycin conduces to pro-angiogenic function of the conditioned medium. Biochem Biophys Rep 2023; 36:101583. [PMID: 38053620 PMCID: PMC10694647 DOI: 10.1016/j.bbrep.2023.101583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/26/2023] [Accepted: 11/13/2023] [Indexed: 12/07/2023] Open
Abstract
Angiogenesis is critical for wound healing and tissue repair. Umbilical cord mesenchymal stem cells (UCMSCs)-conditioned medium has certain actions to promote angiogenesis, and is expected for wound healing and tissue repair. However, recent studies showed that the pro-angiogenic efficacy of unprocessed MSCs-conditioned medium is low, and insufficient for tissue repair. Autophagy is a process for protein recycling and a contributor for cell exocrine, which may enhance pro-angiogenic efficacy of the conditioned medium by stimulating cytokine release from UCMSCs. Therefore, in this study we attempted to obtain enhanced autophagy in UCMSCs using different concentrations of rapamycin and compared pro-angiogenic functions of the conditioned media. The in vitro data showed that although 100 nM-10 μM rapamycin all could induce autophagy in UCMSCs, 100 nM was the best dose to optimize the angiogenic effect of the conditioned medium. The in vivo data also showed that pro-angiogenic effect of the optimized conditioned medium was more obvious than that of the control conditioned medium (0 nM group) in the injected matrigel plaques. Further, the expressions of VEGF, FGF-2, MMP-9, PDGF-α and PDGF-β were markedly increased in UCMSCs treated with 100 nM rapamycin. In conclusion, appropriately enhancing autophagy of UCMSC can improve pro-angiogenic efficacy of the conditioned medium, which may optimize therapeutic applications of UCMSCs-conditioned medium in wound healing and tissue repair.
Collapse
Affiliation(s)
- Wenya Wang
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Xiao Li
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Chaochu Cui
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Guotian Yin
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Wenjie Ren
- Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Xianwei Wang
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
47
|
Matthews I, Birnbaum A, Gromova A, Huang AW, Liu K, Liu EA, Coutinho K, McGraw M, Patterson DC, Banks MT, Nobles AC, Nguyen N, Merrihew GE, Wang L, Baeuerle E, Fernandez E, Musi N, MacCoss MJ, Miranda HC, La Spada AR, Cortes CJ. Skeletal muscle TFEB signaling promotes central nervous system function and reduces neuroinflammation during aging and neurodegenerative disease. Cell Rep 2023; 42:113436. [PMID: 37952157 PMCID: PMC10841857 DOI: 10.1016/j.celrep.2023.113436] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/12/2023] [Accepted: 10/28/2023] [Indexed: 11/14/2023] Open
Abstract
Skeletal muscle has recently arisen as a regulator of central nervous system (CNS) function and aging, secreting bioactive molecules known as myokines with metabolism-modifying functions in targeted tissues, including the CNS. Here, we report the generation of a transgenic mouse with enhanced skeletal muscle lysosomal and mitochondrial function via targeted overexpression of transcription factor E-B (TFEB). We discovered that the resulting geroprotective effects in skeletal muscle reduce neuroinflammation and the accumulation of tau-associated pathological hallmarks in a mouse model of tauopathy. Muscle-specific TFEB overexpression significantly ameliorates proteotoxicity, reduces neuroinflammation, and promotes transcriptional remodeling of the aged CNS, preserving cognition and memory in aged mice. Our results implicate the maintenance of skeletal muscle function throughout aging in direct regulation of CNS health and disease and suggest that skeletal muscle originating factors may act as therapeutic targets against age-associated neurodegenerative disorders.
Collapse
Affiliation(s)
- Ian Matthews
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90007, USA
| | - Allison Birnbaum
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90007, USA
| | - Anastasia Gromova
- Department of Pathology and Laboratory Medicine, UCI Institute for Neurotherapeutics, University of California, Irvine, Irvine, CA 92697, USA
| | - Amy W Huang
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90007, USA
| | - Kailin Liu
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90007, USA
| | - Eleanor A Liu
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90007, USA
| | - Kristen Coutinho
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Megan McGraw
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Dalton C Patterson
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Macy T Banks
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Amber C Nobles
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Nhat Nguyen
- Department of Pathology and Laboratory Medicine, UCI Institute for Neurotherapeutics, University of California, Irvine, Irvine, CA 92697, USA
| | - Gennifer E Merrihew
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Lu Wang
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98195, USA
| | - Eric Baeuerle
- Department of Pharmacology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX 78229, USA; Geriatric Research, Education and Clinical Center, South Texas Veterans Health Care Network, San Antonio, TX 78229, USA
| | - Elizabeth Fernandez
- Department of Pharmacology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX 78229, USA; Geriatric Research, Education and Clinical Center, South Texas Veterans Health Care Network, San Antonio, TX 78229, USA
| | - Nicolas Musi
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Michael J MacCoss
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Helen C Miranda
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; RNA Center, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Albert R La Spada
- Department of Pathology and Laboratory Medicine, UCI Institute for Neurotherapeutics, University of California, Irvine, Irvine, CA 92697, USA; Department of Neurology and Department of Biological Chemistry, UCI Institute for Neurotherapeutics, University of California, Irvine, Irvine, CA 92697, USA.
| | - Constanza J Cortes
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90007, USA.
| |
Collapse
|
48
|
Chen Y, He S, Zeng A, He S, Jin X, Li C, Mei W, Lu Q. Inhibitory Effect of β-Sitosterol on the Ang II-Induced Proliferation of A7r5 Aortic Smooth Muscle Cells. Anal Cell Pathol (Amst) 2023; 2023:2677020. [PMID: 38028434 PMCID: PMC10645495 DOI: 10.1155/2023/2677020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 08/01/2023] [Accepted: 09/27/2023] [Indexed: 12/01/2023] Open
Abstract
Objective To explore the effects of β-sitosterol on VSMC proliferation. Materials and Methods A7r5 cells were pretreated with 2 µM angiotensin II (Ang II) for 24 hr to establish an excessive VSMC proliferation model, followed by treatment with β-sitosterol for 24 hr. Cells were divided into five groups: control, Ang II, and Ang II + β-sitosterol (2, 4, 8 µM). CCK-8 assay, flow cytometry, and Ad-mCherry-GFP-LC3B assay analyzed cell proliferation, cell cycle, cell apoptosis, and autophagic flux. Additionally, the expression of proteins was detected by the western blotting. Results β-Sitosterol effectively inhibited Ang II-induced A7r5 cell proliferation (IC50 : 6.841 µM at 24 hr). It achieved this by arresting cell cycle progression, promoting apoptosis, inhibiting autophagy, and suppressing the contractile-synthetic phenotypic switch. Mechanistically, β-sitosterol downregulated PCNA, Cyclin D1, and Bcl-2, while upregulating pro-caspase 3, cleaved-caspase 3, and Bax to induce cell cycle arrest and apoptosis. Additionally, it suppressed the contractile-synthetic phenotypic transformation by downregulating OPN and upregulating α-SMA. The Ad-mCherry-GFP-LC3B Assay and western blotting revealed β-sitosterol's autophagy inhibitory effects by downregulating LC3, ULK1, and Beclin-1 while upregulating P62 expression. Discussion and Conclusion. This study found for the first time that β-sitosterol could inhibit the proliferation of A7r5 cells induced by Ang II. β-Sitosterol treatment may be recommended as a therapeutic strategy to prevent the cardiovascular diseases.
Collapse
Affiliation(s)
- Yuankun Chen
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, No. 280 East Outer Ring Road, Panyu District, Guangzhou, China
- Department of Infectious and Tropical Diseases, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Shumiao He
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, No. 280 East Outer Ring Road, Panyu District, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ao Zeng
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, No. 280 East Outer Ring Road, Panyu District, Guangzhou, China
| | - Siqing He
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, No. 280 East Outer Ring Road, Panyu District, Guangzhou, China
| | - Xiaobao Jin
- Guangdong Province Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, China
| | - Chunmei Li
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, No. 280 East Outer Ring Road, Panyu District, Guangzhou, China
- Guangdong Province Engineering and Technology Center for Molecular Probe and Bio-medicine Imaging, Guangzhou, China
| | - Wenjie Mei
- Guangdong Province Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, China
| | - Qun Lu
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, No. 280 East Outer Ring Road, Panyu District, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Province Engineering and Technology Center for Molecular Probe and Bio-medicine Imaging, Guangzhou, China
| |
Collapse
|
49
|
Tan JX, Finkel T. Lysosomes in senescence and aging. EMBO Rep 2023; 24:e57265. [PMID: 37811693 PMCID: PMC10626421 DOI: 10.15252/embr.202357265] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 08/08/2023] [Accepted: 09/21/2023] [Indexed: 10/10/2023] Open
Abstract
Dysfunction of lysosomes, the primary hydrolytic organelles in animal cells, is frequently associated with aging and age-related diseases. At the cellular level, lysosomal dysfunction is strongly linked to cellular senescence or the induction of cell death pathways. However, the precise mechanisms by which lysosomal dysfunction participates in these various cellular or organismal phenotypes have remained elusive. The ability of lysosomes to degrade diverse macromolecules including damaged proteins and organelles puts lysosomes at the center of multiple cellular stress responses. Lysosomal activity is tightly regulated by many coordinated cellular processes including pathways that function inside and outside of the organelle. Here, we collectively classify these coordinated pathways as the lysosomal processing and adaptation system (LYPAS). We review evidence that the LYPAS is upregulated by diverse cellular stresses, its adaptability regulates senescence and cell death decisions, and it can form the basis for therapeutic manipulation for a wide range of age-related diseases and potentially for aging itself.
Collapse
Affiliation(s)
- Jay Xiaojun Tan
- Aging InstituteUniversity of Pittsburgh School of Medicine/University of Pittsburgh Medical CenterPittsburghPAUSA
- Department of Cell BiologyUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Toren Finkel
- Aging InstituteUniversity of Pittsburgh School of Medicine/University of Pittsburgh Medical CenterPittsburghPAUSA
- Department of MedicineUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| |
Collapse
|
50
|
Chang YC, Gao Y, Lee JY, Langen J, Chang KT. Identification of secretory autophagy as a novel mechanism modulating activity-induced synaptic remodeling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.06.560931. [PMID: 38328055 PMCID: PMC10849665 DOI: 10.1101/2023.10.06.560931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
The ability of neurons to rapidly remodel their synaptic structure and strength in response to neuronal activity is highly conserved across species and crucial for complex brain functions. However, mechanisms required to elicit and coordinate the acute, activity-dependent structural changes across synapses are not well understood. Here, using an RNAi screen in Drosophila against genes affecting nervous system functions in humans, we uncouple cellular processes important for synaptic plasticity from synapse development. We find mutations associated with neurodegenerative and mental health disorders are 2-times more likely to affect activity-induced synaptic remodeling than synapse development. We further demonstrate that neuronal activity stimulates autophagy activation but diminishes degradative autophagy, thereby driving the pathway towards autophagy-based secretion. Presynaptic knockdown of Snap29, Sec22, or Rab8, proteins implicated in the secretory autophagy pathway, is sufficient to abolish activity-induced synaptic remodeling. This study uncovers secretory autophagy as a novel trans-synaptic signaling mechanism modulating structural plasticity.
Collapse
|