1
|
Han Y, Sun Y, Peng S, Tang T, Zhang B, Yu R, Sun X, Guo S, Ma L, Li P, Yang P. PI3K/AKT pathway: A potential therapeutic target in cerebral ischemia-reperfusion injury. Eur J Pharmacol 2025; 998:177505. [PMID: 40118329 DOI: 10.1016/j.ejphar.2025.177505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 02/21/2025] [Accepted: 03/10/2025] [Indexed: 03/23/2025]
Abstract
Cerebral ischemia is a prevalent cerebrovascular disorder, with the restoration of blocked blood vessels serving as the current standard clinical treatment. However, reperfusion can exacerbate neuronal damage and neurological dysfunction, resulting in cerebral ischemia-reperfusion (I/R) injury. Presently, clinical treatment strategies for cerebral I/R injury are limited, creating an urgent need to identify new effective therapeutic targets. The PI3K/AKT signaling pathway, a pro-survival pathway associated with cerebral I/R injury, has garnered significant attention. We conducted a comprehensive review of the literature on the PI3K/AKT pathway in the context of cerebral I/R. Our findings indicate that activation of the PI3K/AKT signaling pathway following cerebral I/R can alleviate oxidative stress, reduce endoplasmic reticulum stress (ERS), inhibit inflammatory responses, decrease neuronal apoptosis, autophagy, and pyroptosis, mitigate blood-brain barrier (BBB) damage, and promote neurological function recovery. Consequently, this pathway ultimately reduces neuronal death, alleviates brain tissue damage, decreases the volume of cerebral infarction, and improves behavioral impairments. These results suggest that the PI3K/AKT signaling pathway is a promising therapeutic target for further research and drug development, holding significant potential for the treatment of cerebral I/R injury.
Collapse
Affiliation(s)
- Yiming Han
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Yu Sun
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Shiyu Peng
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Tingting Tang
- First Clinical College, Xinxiang Medical University, Xinxiang, China
| | - Beibei Zhang
- First Clinical College, Xinxiang Medical University, Xinxiang, China
| | - Ruonan Yu
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Xiaoyan Sun
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Shanshan Guo
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China; Staff Hospital of Henan Fifth Construction Group Co., Ltd, Zhengzhou, Henan, China
| | - Lijuan Ma
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.
| | - Peng Li
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.
| | - Pengfei Yang
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.
| |
Collapse
|
2
|
He X, Liu J, Zhang Y, Xie B. Transcriptomic profiling of autophagy and apoptosis pathways in liver cancer cells treated with a new tyrosine kinase inhibitor PD161570. Mol Med Rep 2025; 32:175. [PMID: 40242947 PMCID: PMC12046374 DOI: 10.3892/mmr.2025.13540] [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: 06/10/2024] [Accepted: 02/10/2025] [Indexed: 04/18/2025] Open
Abstract
Liver cancer is the third most lethal and prevalent cancer in the Asia‑Pacific regions. Despite the use of tyrosine kinase inhibitors as first‑ and second‑line therapies, the overall survival rate for advanced liver cancer remains dismal and has not improved over the past decade. The present study, through high‑throughput screening, identified and demonstrated that PD161570, a new tyrosine kinase inhibitor, inhibited cell growth and proliferation in liver cancer cells. Mechanistically, PD161570 induced autophagy and enhanced autophagic flux in an autophagy‑related gene (ATG5)‑dependent and mammalian target of rapamycin kinase‑independent manner. Furthermore, when combined with chloroquine treatment, PD161570 not only suppressed cell proliferation but also increased cell apoptosis due to autophagy inhibition. RNA sequencing analysis revealed 1,121 differentially expressed genes in liver cancer cells following PD161570 treatment under autophagy inhibition via ATG5 knockdown. Notably, key molecules involved in autophagy (such as Damage Regulated Autophagy Modulator 1) and apoptosis regulators (including HRK, CTSS, BIRC3, BBC3, DDIT3 and GADD45B), were identified. Functional enrichment analyses, including Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG), demonstrated enrichment in apoptotic and cell death signaling pathways, highlighting the critical role of the mitogen‑activated protein kinases signaling pathway. In conclusion, PD161570 elicited an ATG5‑dependent autophagic process in liver cancer cells, while simultaneously enhancing apoptosis under conditions of autophagy inhibition.
Collapse
Affiliation(s)
- Xingxing He
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jianping Liu
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yulian Zhang
- Jiangxi Provincial Key Laboratory of Digestive Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Jiangxi Clinical Research Center for Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Bushan Xie
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Jiangxi Provincial Key Laboratory of Digestive Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Jiangxi Clinical Research Center for Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
3
|
Guo Z, He L, Wang W, Tian S, Lin R. FUT2-dependent fucosylation of LAMP1 promotes the apoptosis of colorectal cancer cells by regulating the autophagy-lysosomal pathway. Cancer Lett 2025; 619:217643. [PMID: 40112906 DOI: 10.1016/j.canlet.2025.217643] [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: 12/23/2024] [Revised: 03/06/2025] [Accepted: 03/15/2025] [Indexed: 03/22/2025]
Abstract
Fucosyltransferase 2 (FUT2) is an enzyme that adds fucose to proteins or lipids via α-1,2-fucosylation in the intestinal mucosa. While FUT2 deficiency is linked to increased susceptibility to inflammatory bowel disease (IBD), its role in colorectal cancer (CRC) is unclear, and the molecular mechanisms involved remain largely unknown. We established an azoxymethane (AOM) and dextran sulfate sodium (DSS) model to induce CRC. FUT2 expression was assessed in human CRC tissues, AOM/DSS-induced mouse models, and CRC cell lines using qRT-PCR, western blotting, and UEA-I staining. FUT2 knockout (FUT2△IEC) mice were treated with AOM/DSS, and FUT2-overexpressing CRC cells were created to evaluate the effects of FUT2 on apoptosis in both in vitro and in vivo settings through Western blot analyses and functional assays. N-glycoproteomics, UEA-I chromatography, and co-immunoprecipitation were utilized to identify regulatory mechanisms and target fucosylated proteins. FUT2 expression and α-1,2-fucosylation were significantly decreased in CRC. FUT2 deficiency worsened AOM/DSS-induced CRC and reduced tumor apoptosis, while FUT2 overexpression induced apoptosis and inhibited proliferation in CRC cells and xenografts. Mechanistically, FUT2 appears to suppress autophagy by impairing lysosomal function and directly targeting and fucosylating LAMP1, contributing to lysosomal dysfunction. Our study reveals a fucosylation-dependent antitumor mechanism of FUT2 in CRC, suggesting potential therapeutic strategies for CRC treatment.
Collapse
Affiliation(s)
- Zijun Guo
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lingnan He
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong New Area, Shanghai, China
| | - Weijun Wang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuxin Tian
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rong Lin
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
4
|
Zarro PR, De Felice S, Sabbieti MG, Agas D. The Inflamed Bone Marrow Scenery Amongst the Symplegades of Ageing and Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025. [PMID: 40279001 DOI: 10.1007/5584_2025_860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
Upon inflammation, the bone marrow (BM) landscape undergoes significant architectural and functional modifications. Stimulation of the hematopoietic niche triggers a series of lightning events, which begin with stem/progenitor blood elements mobilization and culminates with the activation of immune responses. Ageing partially mirrors this process, albeit with a propensity towards chronic inflammation and immune dysfunction. Age-related chronic inflammation disrupts bone homeostasis and accompanies impaired tissue regeneration. Thus, focusing on the bone marrow's dynamics during inflammatory bone diseases could lay the way for the development of novel therapeutic platforms aimed at niche reprogramming. Herein, we summarize inflammatory and age-induced processes in multiple BM compartments, with particular reference to hematopoietic, stromal stem/progenitor cells, and mature immunocytes. Finally, we focus on autophagy and its potential to clinically re-modulate the pathological "flogistic" bias, possibly by restoring functional phenotypes within the bone marrow niche elements.
Collapse
Affiliation(s)
- Pier Raffaele Zarro
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, MC, Italy
| | - Simona De Felice
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, MC, Italy
| | | | - Dimitrios Agas
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, MC, Italy.
| |
Collapse
|
5
|
Ngana GS, Di Bernardo MA, Surette MG, MacNeil LT. Actinomyces viscosus promotes neuroprotection in C. elegans models of Parkinson's disease. Mech Ageing Dev 2025; 225:112061. [PMID: 40258426 DOI: 10.1016/j.mad.2025.112061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 04/04/2025] [Accepted: 04/11/2025] [Indexed: 04/23/2025]
Abstract
Parkinson's Disease is characterized by selective degeneration of dopaminergic neurons, primarily in the substantia nigra pars compacta, as well as accumulation of alpha-synuclein enriched protein aggregates within neurons. The pathogenesis of PD is still not completely understood, and no treatments exist that alter disease progression. Obvious genetic causes are detected in only a small number of PD patients (5-10 %), suggesting that environmental factors play a significant role the development of PD. Correlative studies suggest that the microbiota could be an important environmental modifier of neurodegeneration. We identified a microbiotal isolate, Actinomyces viscosus, that reduced neurodegeneration in C. elegans expressing a pathological mutant form (G2019S) of leucine-rich repeat kinase 2 (LRRK2) in dopaminergic neurons. A. viscosus also suppressed autophagic dysfunction in these animals and reduced alpha-synuclein aggregation in a synucleinopathy model. Global gene expression analysis revealed increased expression of aspartic cathepsins in response to A. viscosus. Consistent with the involvement of these proteins in neuroprotection, we found that reducing aspartic cathepsin function increased neurodegeneration in the LRRK2 transgenic model. Our findings contribute to the current understanding of how the gut microbiota may influence PD, elucidating one potential mechanism of microbiota-mediated neuroprotection.
Collapse
Affiliation(s)
- G Sophie Ngana
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W., Hamilton, ON, Canada
| | - Mercedes A Di Bernardo
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W., Hamilton, ON, Canada
| | - Michael G Surette
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W., Hamilton, ON, Canada; Farncombe Family Digestive Health Research Institute, McMaster University, 1280 Main St W, Hamilton, ON, Canada; Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main St W, Hamilton ON, Canada; Department of Medicine, McMaster University, 1280 Main St W, Hamilton, ON, Canada
| | - Lesley T MacNeil
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W., Hamilton, ON, Canada; Farncombe Family Digestive Health Research Institute, McMaster University, 1280 Main St W, Hamilton, ON, Canada; Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main St W, Hamilton ON, Canada.
| |
Collapse
|
6
|
Canova PN, Katzenell S, Cerón S, Charron AJ, Pesola JM, Oh HS, Coen DM, Knipe DM, Leib DA. Regulation of the innate immune response in human neurons by ICP34.5 maintains herpes simplex virus 1 latency. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.04.647253. [PMID: 40291710 PMCID: PMC12026746 DOI: 10.1101/2025.04.04.647253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Herpes simplex virus 1 (HSV-1) establishes latent infections in sensory neurons, from which HSV sporadically reactivates due to external stress and other stimuli. Latency and reactivation are studied through in vivo models in a variety of hosts, as well as in vitro models using primary neurons, and neurons derived from pluripotent stem cells (iPSCs). These systems behave disparately, but the reasons remain unknown. The interferon (IFN)-based neuronal innate immune response is critical in controlling HSV-1 replication and HSV-1 counters these responses in part through infectedcell protein 34.5 (ICP34.5). ICP34.5 also promotes neurovirulence by preventing host translational shutoff and interfering with host cell autophagy. Here we demonstrate in a human iPSC neuronal model that sustaining host translation is the key activity of ICP34.5 for enhancement of reactivation. Specifically, our data shows that ICP34.5 was key for maintenance of HSV-1 latency. While interaction of ICP34.5 with the autophagy regulator Beclin 1 was important for maintaining latency, this was not due to modulation of bulk autophagy. Our work from primary mouse neurons suggested that the major effect of ICP34.5 on latency maintenance occurs in an IRF3/7-dependent manner. Notably, the role of ICP34.5 in regulating latency and reactivation differs between neurons derived from human iPSCs (iNeurons) and primary mouse trigeminal (TG) neurons. This highlights the importance of selecting an appropriate neuronal model and validating experimental outcomes in multiple models.
Collapse
|
7
|
Islam S, Sarkar O, Mukherjee S, Chattopadhyay A. Long-Term Impact of Cr(VI) Exposure in Swiss Albino Mice: ROS-Driven Modulation of Autophagy and Cellular Fate. Biol Trace Elem Res 2025:10.1007/s12011-025-04599-w. [PMID: 40180680 DOI: 10.1007/s12011-025-04599-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
Hexavalent chromium [Cr(VI)], due to its high solubility and permeability, is significantly more toxic than trivalent chromium [Cr(III)] as it generates reactive oxygen species (ROS) during cellular reduction. Industrial discharges have led to increasing Cr(VI) contamination in surface and groundwater, posing serious environmental and public health concerns. In our previous study, we demonstrated that exposure to 5 ppm Cr(VI) for 4 and 8 months adversely affected body weight, water consumption, and liver function in Swiss albino mice. Histological analyses revealed tissue alterations, disrupted DNA repair gene expression in liver tissue, and a marked increase in apoptotic gene expression after 8 months of exposure. Building on these findings, we employed the same Cr(VI) concentration (5 ppm via drinking water) over 4 and 8 months in the present study. Our results showed a significant increase in ROS generation in the liver, brain, and kidney tissues at both time intervals. Additionally, the presence of autophagolysosomes was markedly elevated after chronic Cr(VI) exposure in each tissue. We also observed altered expression patterns of key autophagy-related genes (Atg5, Beclin1, and Lc3) and mTor in these tissues. Immunohistochemical analysis further confirmed a significant increase in LC3B expression after 4 months of exposure. Our findings suggest that heightened intracellular oxidative stress triggers a protective autophagy response, mediated via mTOR signaling, to maintain cellular integrity. However, prolonged toxic insult and ROS accumulation may eventually shift pro-survival autophagy toward apoptotic cell death in the liver and brain tissues.
Collapse
Affiliation(s)
- Shehnaz Islam
- Department of Zoology, Visva-Bharati, Santiniketan, 731235, West Bengal, India
| | - Olivia Sarkar
- Department of Zoology, Visva-Bharati, Santiniketan, 731235, West Bengal, India
| | - Sunanda Mukherjee
- Department of Zoology, Visva-Bharati, Santiniketan, 731235, West Bengal, India
| | | |
Collapse
|
8
|
Chen Z, Wang J, Lu B, Meng W, Zhu Y, Jiang Q, Gao D, Ma Z, Zeng H, Chen J, Liu S, Wang Z, Jia K. Reduction of microRNA-221 in BVDV infection enhances viral replication by targeting the ATG7-mediated autophagy pathway. Ir Vet J 2025; 78:10. [PMID: 40176193 PMCID: PMC11963565 DOI: 10.1186/s13620-025-00286-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 01/06/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND Bovine viral diarrhoea (BVD), a condition triggered by bovine viral diarrhoea virus (BVDV), is recognized globally as a prevalent pathogen among ruminants and markedly affects the economics of animal husbandry. MicroRNAs, a class of small noncoding RNAs, play pivotal roles in regulating a myriad of biological processes.The ATG7-LC3 pathway, a canonical autophagy mechanism, is integral in defending against pathogenic invasion and maintaining cellular homeostasis. RESULTS In this study, we observed significant downregulation of bta-miR-221 in cells infected with BVDV. We further established that overexpression of bta-miR-221 markedly attenuated BVDV replication in Madin‒Darby bovine kidney (MDBK) cells. Through bioinformatics prediction analysis, we identified ATG7, an autophagy-related gene, as a direct downstream target of bta-miR-221. However, the intricate relationships among bta-miR-221, the ATG7-LC3 pathway, and BVDV infection remained unclear. Our study revealed that ATG7 expression was significantly elevated in BVDV-infected cells, whereas bta-miR-221 mimics repressed both endogenous and exogenous ATG7 expression. Following BVDV infection, we noted a decrease in LC3I expression, its conversion to LC3II, a significant increase in ATG7 expression, and a notable decrease in SQSTM1/p62 expression. By employing laser confocal microscopy and immunoprecipitation assays, we elucidated the regulation of the ATG7-LC3 pathway by bta-miR-221 in MDBK cells. Our findings recealed that BVDV infection enhanced the ATG7-LC3 interaction, inducing autophagy through the suppression of bta-miR-221 in MDBK cells. Consequently, bta-miR-221 emerged as a potent inhibitor of BVDV, impacting its proliferation and replication within the host. CONCLUSIONS This research sheds light on novel aspects of virus-host interactions and lays a foundation for the development of antiviral therapeutics.
Collapse
Affiliation(s)
- Zihan Chen
- College of Veterinary Medicine, South China Agricultural University, Guangdong, 510642, China
| | - Jingyu Wang
- College of Veterinary Medicine, South China Agricultural University, Guangdong, 510642, China
| | - Baochun Lu
- College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Wenxin Meng
- College of Veterinary Medicine, South China Agricultural University, Guangdong, 510642, China
| | - Yufan Zhu
- College of Veterinary Medicine, South China Agricultural University, Guangdong, 510642, China
| | - Qifeng Jiang
- College of Veterinary Medicine, South China Agricultural University, Guangdong, 510642, China
| | - Duo Gao
- College of Veterinary Medicine, South China Agricultural University, Guangdong, 510642, China
| | - Zihang Ma
- College of Veterinary Medicine, South China Agricultural University, Guangdong, 510642, China
| | - Huijuan Zeng
- College of Veterinary Medicine, South China Agricultural University, Guangdong, 510642, China
| | - Jinping Chen
- College of Veterinary Medicine, South China Agricultural University, Guangdong, 510642, China
| | - Shizhe Liu
- College of Veterinary Medicine, South China Agricultural University, Guangdong, 510642, China
| | - Zhen Wang
- College of Veterinary Medicine, South China Agricultural University, Guangdong, 510642, China
| | - Kun Jia
- College of Veterinary Medicine, South China Agricultural University, Guangdong, 510642, China.
| |
Collapse
|
9
|
Robinson KS, Sennhenn P, Yuan DS, Liu H, Taddei D, Qian Y, Luo W. TMBIM6/BI-1 is an intracellular environmental regulator that induces paraptosis in cancer via ROS and Calcium-activated ERAD II pathways. Oncogene 2025; 44:494-512. [PMID: 39609612 PMCID: PMC11832424 DOI: 10.1038/s41388-024-03222-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: 06/15/2023] [Revised: 10/28/2024] [Accepted: 11/05/2024] [Indexed: 11/30/2024]
Abstract
Transmembrane B cell lymphoma 2-associated X protein inhibitor motif-containing (TMBIM) 6, also known as Bax Inhibitor-1 (BI-1), has been heavily researched for its cytoprotective functions. TMBIM6 functional diversity includes modulating cell survival, stress, metabolism, cytoskeletal dynamics, organelle function, regulating cytosolic acidification, calcium, and reactive oxygen species (ROS). Clinical research shows TMBIM6 plays a key role in many of the world's top diseases/injuries (i.e., Alzheimer's, Parkinson's, diabetes, obesity, brain injury, liver disease, heart disease, aging, etc.), including cancer, where TMBIM6 expression impacts patient survival, chemoresistance, cancer progression, and metastasis. We show TMBIM6 is activated by, and undergoes, different conformational changes that dictate its function following a significant change in the cell's IntraCellular Environment (ICE). TMBIM6 agonism, following ICE change, can help the cell overcome multiple stresses including toxin exposure, viral infection, wound healing, and excitotoxicity. However, in cancer cells TMBIM6 agonism results in rapid paraptotic induction irrespective of the cancer type, sub-type, genotype or phenotype. Furthermore, the level of TMBIM6 expression in cancer did not dictate the level of paraptotic induction; however, it did dictate the rate at which paraptosis occurred. TMBIM6 agonism did not induce paraptosis in cancer via canonical routes involving p38 MAPK, JNK, ERK, UPR, autophagy, proteasomes, or Caspase-9. Instead, TMBIM6 agonism in cancer upregulates cytosolic Ca2+ and ROS, activates lysosome biogenesis, and induces paraptosis via ERAD II mechanisms. In xenograft models, we show TMBIM6 agonism induces rapid cancer cell death with no toxicity, even at high doses of TMBIM6 agonist (>450 mg/kg). In summary, this study shows TMBIM6's functional diversity is only activated by severe ICE change in diseased/injured cells, highlighting its transformative potential as a therapeutic target across various diseases and injuries, including cancer.
Collapse
Affiliation(s)
| | | | | | - Hai Liu
- Viva Biotech, Shanghai, China
| | | | | | - Wei Luo
- MicroQuin, Cambridge, MA, USA
| |
Collapse
|
10
|
Wang Y, Ge Y, Hua S, Shen C, Cai B, Zhao H. Aloe-Emodin Improves Mitophagy in Alzheimer's Disease via Activating the AMPK/PGC-1α/SIRT3 Signaling Pathway. CNS Neurosci Ther 2025; 31:e70346. [PMID: 40125832 PMCID: PMC11931456 DOI: 10.1111/cns.70346] [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: 12/19/2024] [Revised: 02/22/2025] [Accepted: 03/11/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Impaired mitophagy results in the accumulation of defective mitochondria that are unable to be cleared effectively in Alzheimer's disease (AD). Aloe-emodin (AE), a key component of the traditional Chinese medicine Rhubarb, exhibits neuroprotective effects against Alzheimer's disease, though the underlying mechanism remains unclear. Studying aloe-emodin's role in enhancing mitophagy is vital for improving cognitive function and reducing neuronal damage in Alzheimer's disease. METHODS The APP/PS1 double transgenic mice were adopted as models for AD to assess the effects of aloe-emodin upon cognitive function and its neuroprotective impact on hippocampal neurons. Additionally, we investigated the regulatory mechanisms of proteins within the aforementioned pathway, and the morphological characteristics of mitophagy-related proteins. An AD hippocampal neuron model was developed using Aβ25-35 to evaluate the mitochondrial function, the protein expression of such a pathway and the mitophagy. This approach aims to elucidate the effects and underlying mechanisms of aloe-emodin in relation to AD. RESULTS AE activates mitophagy in neurons, improves cognitive dysfunction, reduces hippocampal damage, and alleviates AD symptoms in model mice. AE activates the expression of AMPK, PGC-1α and SIRT3. Increased expression of SIRT3 in mitochondria promotes mitophagy and regulates the function of mitochondrial proteins. When mitochondrial autophagy is enhanced, the expression of Beclin1, LC3, P62, Parkin, and PINK1-related proteins changes. Further in vitro experiments showed that AE can enhance mitochondrial function in Alzheimer's disease cell models. The mitochondrial membrane potential, GSH, ROS and Ca2+ levels gradually recover, alleviating the pathological manifestations of AD. Knocking down SIRT3 leads to increased mitochondrial damage and a reduction in mitophagy in HT22 cells. CONCLUSION Experimental results show that AE can activate mitophagy through AMPK/PGC-1α/SIRT3 pathway, alleviate cognitive dysfunction in AD, and reduce damage to hippocampal neurons.
Collapse
Affiliation(s)
- Yulu Wang
- College of Integrated Chinese and Western MedicineAnhui University of Chinese MedicineHefeiChina
| | - Yunzhi Ge
- College of Integrated Chinese and Western MedicineAnhui University of Chinese MedicineHefeiChina
| | - Siyu Hua
- College of Integrated Chinese and Western MedicineAnhui University of Chinese MedicineHefeiChina
| | - Chenrui Shen
- College of Integrated Chinese and Western MedicineAnhui University of Chinese MedicineHefeiChina
| | - Biao Cai
- College of Integrated Chinese and Western MedicineAnhui University of Chinese MedicineHefeiChina
- Institute of Integrated Chinese and Western MedicineAnhui Academy of Chinese MedicineHefeiChina
- Key Laboratory of Xin'an MedicineAnhui University of Chinese Medicine, Ministry of EducationHefeiChina
- Anhui Province Key Laboratory of Chinese Medicinal FormulaHefeiChina
| | - Han Zhao
- College of Integrated Chinese and Western MedicineAnhui University of Chinese MedicineHefeiChina
- Institute of Integrated Chinese and Western MedicineAnhui Academy of Chinese MedicineHefeiChina
- Key Laboratory of Xin'an MedicineAnhui University of Chinese Medicine, Ministry of EducationHefeiChina
- Anhui Province Key Laboratory of Chinese Medicinal FormulaHefeiChina
| |
Collapse
|
11
|
Wu Y, Xu R, Zhuang X. Multifaceted Roles of the ATG8 Protein Family in Plant Autophagy: From Autophagosome Biogenesis to Cargo Recognition. J Mol Biol 2025:168981. [PMID: 39909236 DOI: 10.1016/j.jmb.2025.168981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/25/2025] [Accepted: 01/30/2025] [Indexed: 02/07/2025]
Abstract
In plant cells, autophagy is an essential quality control process by forming a double-membrane structure named the autophagosome, which envelopes and transports the cargoes to the vacuole for degradation/recycling. Autophagy-related (ATG) 8, a key regulator in autophagy, exerts multifunctional roles during autophagy. ATG8 anchors on the phagophore membrane through the ATG8 conjugation system and participates in different steps during autophagosome formation. Accumulating evidence has demonstrated that ATG8 cooperates with other ATG or non-ATG proteins in autophagosome biogenesis. Meanwhile, ATG8 plays an important role in cargo recognition, which is mainly attributed by the specific interactions between ATG8 and the selective autophagy receptors (SARs) or cargos for selective autophagy. Emerging roles of ATG8 in non-canonical autophagy have been recently reported in plants for different stress adaptations. Here, we review the diverse functions of ATG8 in plants, focusing on autophagosome biogenesis and cargo recognition in canonical and non-canonical autophagy.
Collapse
Affiliation(s)
- Yixin Wu
- AoE Centre for Organelle Biogenesis and Function, Centre for Cell & Developmental Biology and State Key Laboratory of Agrobiotechnology, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Rui Xu
- AoE Centre for Organelle Biogenesis and Function, Centre for Cell & Developmental Biology and State Key Laboratory of Agrobiotechnology, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xiaohong Zhuang
- AoE Centre for Organelle Biogenesis and Function, Centre for Cell & Developmental Biology and State Key Laboratory of Agrobiotechnology, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
12
|
Tian L, Su F, Zhu X, Zou X. The Effect of Maternal Obesity on Placental Autophagy in Lean Breed Sows. Vet Sci 2025; 12:97. [PMID: 40005857 PMCID: PMC11861729 DOI: 10.3390/vetsci12020097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/17/2025] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
This study aimed to evaluate the influence of back-fat thickness (BF), at mating of sows, on autophagy in placenta and the potential mechanism. The sows were divided into two groups according to their BF at mating: BFI (15-20 mm, n = 14) and BFII (21-27 mm, n = 14) as the maternal obesity group. The placental samples used for investigating autophagic function and fatty acid profiles were obtained by vaginal delivery. Our results demonstrated that autophagy defects were observed in placenta from BFII sows along with altered circulating and placental fatty acid profiles. Indicative of impaired autophagy, reduced autophagic vesicles as well as LC3-positive puncta were linked to decreased mRNA or protein expression of autophagy-related genes, including ATG5, ATG7, Beclin1, ATG12, LC3, LAMP1 and LAMP2 in the placenta of BFII sows (p < 0.05). Meanwhile, we found reduced conversion of LC3-I to LC3-II and up-regulated protein content of p62 in the placenta from BFII group (p < 0.05). Furthermore, excessive back-fat was also associated with increased activation of AKT/mTOR signaling and decreased mRNA content of transcription factors regulating the autophagic pathway, including PPARα and PGC1α, but increased mRNA expression of NcoR1 in placenta. Together, these findings indicate that maternal obesity incites autophagy injury in pig term placenta, which may contribute to augmented placental lipid accumulation and therefore impaired placental function.
Collapse
Affiliation(s)
- Liang Tian
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | | | | | | |
Collapse
|
13
|
Jia N, Ganesan D, Guan H, Jeong YY, Han S, Rajapaksha G, Nissenbaum M, Kusnecov AW, Cai Q. Mitochondrial bioenergetics stimulates autophagy for pathological MAPT/Tau clearance in tauopathy neurons. Autophagy 2025; 21:54-79. [PMID: 39171695 DOI: 10.1080/15548627.2024.2392408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 08/04/2024] [Accepted: 08/09/2024] [Indexed: 08/23/2024] Open
Abstract
Hyperphosphorylation and aggregation of MAPT (microtubule-associated protein tau) is a pathogenic hallmark of tauopathies and a defining feature of Alzheimer disease (AD). Pathological MAPT/tau is targeted by macroautophagy/autophagy for clearance after being sequestered within autophagosomes, but autophagy dysfunction is indicated in tauopathy. While mitochondrial bioenergetic deficits have been shown to precede MAPT/tau pathology in tauopathy brains, it is unclear whether energy metabolism deficiency is involved in the pathogenesis of autophagy defects. Here, we reveal that stimulation of anaplerotic metabolism restores defective oxidative phosphorylation (OXPHOS) in tauopathy neurons which, strikingly, leads to pronounced MAPT/tau clearance by boosting autophagy functionality through enhancements of mitochondrial biosynthesis and supply of phosphatidylethanolamine for autophagosome biogenesis. Furthermore, early anaplerotic stimulation of OXPHOS elevates autophagy activity and attenuates MAPT/tau pathology, thereby counteracting memory impairment in tauopathy mice. Taken together, our study sheds light on a pivotal role of mitochondrial bioenergetic deficiency in tauopathy-related autophagy defects and suggests a new therapeutic strategy to prevent the buildup of pathological MAPT/tau in AD and other tauopathy diseases.Abbreviation: AA: antimycin A; AD, Alzheimer disease; ATP, adenosine triphosphate; AV, autophagosome/autophagic vacuole; AZ, active zone; Baf-A1: bafilomycin A1; CHX, cycloheximide; COX, cytochrome c oxidase; DIV, days in vitro; DRG, dorsal root ganglion; ETN, ethanolamine; FRET, Förster/fluorescence resonance energy transfer; FTD, frontotemporal dementia; Gln, glutamine; HA: hydroxylamine; HsMAPT/Tau, human MAPT; IMM, inner mitochondrial membrane; LAMP1, lysosomal-associated membrane protein 1; LIs, lysosomal inhibitors; MDAV, mitochondria-derived autophagic vacuole; MmMAPT/Tau, murine MAPT; NFT, neurofibrillary tangle; OCR, oxygen consumption rate; Omy: oligomycin; OXPHOS, oxidative phosphorylation; PPARGC1A/PGC-1alpha: peroxisome proliferative activated receptor, gamma, coactivator 1 alpha; PE, phosphatidylethanolamine; phospho-MAPT/tau, hyperphosphorylated MAPT; PS, phosphatidylserine; PISD, phosphatidylserine decarboxylase;SQSTM1/p62, sequestosome 1; STX1, syntaxin 1; SYP, synaptophysin; Tg, transgenic; TCA, tricarboxylic acid; TEM, transmission electron microscopy.
Collapse
Affiliation(s)
- Nuo Jia
- Department of Cell Biology, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Dhasarathan Ganesan
- Department of Cell Biology, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Hongyuan Guan
- Department of Cell Biology, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Yu Young Jeong
- Department of Cell Biology, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Sinsuk Han
- Department of Cell Biology, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Gavesh Rajapaksha
- Department of Cell Biology, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Marialaina Nissenbaum
- Department of Psychology, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Alexander W Kusnecov
- Department of Psychology, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Qian Cai
- Department of Cell Biology, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| |
Collapse
|
14
|
Zhang JX, Lan MF, Shang JZ, Lai XL, Li LS, Duan TT, Xu RH, Chen KL, Duan X. DMT1 Maintains Iron Homeostasis to Regulate Mitochondrial Function in Porcine Oocytes. J Cell Physiol 2025; 240:e31494. [PMID: 39639679 DOI: 10.1002/jcp.31494] [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: 06/30/2024] [Revised: 10/29/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024]
Abstract
Iron plays critical roles in many cellular functions, including energy production, metabolism, and cell proliferation. However, the role of iron in maintaining oocyte quality remains unclear. In this study, DMT1 was identified as a key iron transporter during porcine oocyte maturation. The results demonstrated that iron deficiency in porcine oocyte led to aberrant meiotic progression, accompanied by increased gene expression of DMT1. Inhibition of DMT1 resulted in the failure of cumulus cell expansion and oocyte maturation, along by the abnormal actin and microtubule assembly. Furthermore, loss of DMT1 function caused disruption in mitochondrial function and dynamics, resulting in oxidative stress and Ca2+ dyshomeostasis. Additionally, the absence of DMT1 function activated PINK1/Parkin-dependent mitophagy in porcine oocyte. These findings suggested that DMT1 played a crucial role in safeguarding oocyte quality by protecting against iron-deficiency-induced mitochondrial dysfunction and autophagy. This study provided compelling evidence that DMT1 and iron homeostasis were crucial for maintaining the capacity of porcine oocyte maturation. Moreover, the results hinted at the potential of DMT1 as a novel therapeutic target for treating iron deficiency-related female reproductive disorders.
Collapse
Affiliation(s)
- Jin-Xin Zhang
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, Zhejiang, China
| | - Meng-Fan Lan
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, Zhejiang, China
| | - Jian-Zhou Shang
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, Zhejiang, China
| | - Xin-Le Lai
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, Zhejiang, China
| | - Li-Shu Li
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, Zhejiang, China
| | - Tong-Tong Duan
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, Zhejiang, China
| | - Ru-Hai Xu
- Key Laboratory of Animal Genetics and Breeding of Zhejiang Province, Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Kun-Lin Chen
- Key Laboratory of Crop and Animal Integrated Farming, Ministry of Agriculture, Jiangsu Academy of Agricultural Sciences, Nanjing, Jiangsu, China
| | - Xing Duan
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, Zhejiang, China
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| |
Collapse
|
15
|
Lim RM, Lu A, Chuang BM, Anaraki C, Chu B, Halbrook CJ, Edinger AL. CARMIL1-AA selectively inhibits macropinocytosis while sparing autophagy. Mol Biol Cell 2025; 36:ar4. [PMID: 39602282 PMCID: PMC11742120 DOI: 10.1091/mbc.e24-09-0434] [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: 10/01/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024] Open
Abstract
Macropinocytosis is reported to fuel tumor growth and drug resistance by allowing cancer cells to scavenge extracellular macromolecules. However, accurately defining the role of macropinocytosis in cancer depends on our ability to selectively block this process. 5-(N-ethyl-N-isopropyl)-amiloride (EIPA) is widely used to inhibit macropinocytosis but affects multiple Na+/H+ exchangers (NHE) that regulate cytoplasmic and organellar pH. Consistent with this, we report that EIPA slows proliferation to a greater extent than can be accounted for by macropinocytosis inhibition and triggers conjugation of ATG8 to single membranes (CASM). Knocking down only NHE1 would not avoid macropinocytosis-independent effects on pH. Moreover, contrary to published reports, NHE1 loss did not block macropinocytosis in multiple cell lines. Knocking down CARMIL1 with CRISPR-Cas9 editing limited macropinocytosis, but only by 50%. In contrast, expressing the CARMIL1-AA mutant inhibits macropinocytosis induced by a wide range of macropinocytic stimuli to a similar extent as EIPA. CARMIL1-AA expression did not inhibit proliferation, highlighting the shortcomings of EIPA as a macropinocytosis inhibitor. Importantly, autophagy, another actin dependent, nutrient-producing process, was not affected by CARMIL1-AA expression. In sum, constitutive or inducible CARMIL1-AA expression reduced macropinocytosis without affecting proliferation, RAC activation, or autophagy, other processes that drive tumor initiation and progression.
Collapse
Affiliation(s)
- Rebecca M. Lim
- Department of Developmental and Cell Biology, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA 92617
| | - Alexa Lu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, CA 92617
| | - Brennan M. Chuang
- Department of Developmental and Cell Biology, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA 92617
| | - Cecily Anaraki
- Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Irvine, CA 92617
| | - Brandon Chu
- Department of Developmental and Cell Biology, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA 92617
| | - Christopher J. Halbrook
- Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Irvine, CA 92617
- Chao Family Comprehensive Cancer Center, University of California, Irvine, Orange, CA 92868
| | - Aimee L. Edinger
- Department of Developmental and Cell Biology, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA 92617
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, CA 92617
- Chao Family Comprehensive Cancer Center, University of California, Irvine, Orange, CA 92868
| |
Collapse
|
16
|
Georgoulis I, Giantsis IA, Michaelidis B, Feidantsis K. Heat Hardening Ameliorates Apoptotic and Inflammatory Effects Through Increased Autophagy in Mussels. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2024; 26:1271-1286. [PMID: 39240443 DOI: 10.1007/s10126-024-10371-3] [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: 05/02/2024] [Accepted: 08/30/2024] [Indexed: 09/07/2024]
Abstract
The severity, frequency, and duration of extreme events, in the context of global warming, have placed many marine ecosystems at high risk. Therefore, the application of methods that can mediate the impacts of global warming on marine organisms seems to be an emerging necessity in the near term. In this context, enhancing the thermal resilience of marine organisms may be crucial for their sustainability. It has been shown that the repeated time-limited exposure of an organism to an environmental stimulus modifies its response mode, thus enhancing resilience and allowing adaptation of the physiological and developmental phenotype to environmental stress. In the present study, we investigated the "stress memory" effect caused by heat hardening on Mytilus galloprovincialis cellular pathways to identify the underlying biochemical mechanisms that enhance mussel thermal tolerance. Heat hardening resulted in increased ETS activity and ATP production and increased autophagic performance at all elevated temperatures (24 °C, 26 °C, and 28 °C). Furthermore, at these increased temperatures, apoptosis and inflammation remain at significantly lower levels in pregnant individuals than in nonhardened individuals. Autophagy, as a negative regulator of apoptosis, may lead to decreased damage to surrounding cells, which in turn alleviates inflammatory effects. In conclusion, the exposure of mussels to heat hardening seems to provide a physiological response that enhances heat tolerance and increases cell survival through increased energy production and reduced cell death and inflammatory responses. The latter can be utilized for the management and conservation of aquatic species of economic value or endangered status.
Collapse
Affiliation(s)
- Ioannis Georgoulis
- Laboratory of Animal Physiology, Department of Zoology, School of Biology, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece.
| | - Ioannis A Giantsis
- Department of Animal Science, Faculty of Agriculture, Forestry and Natural Environment, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Basile Michaelidis
- Laboratory of Animal Physiology, Department of Zoology, School of Biology, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Konstantinos Feidantsis
- Department of Fisheries & Aquaculture, School of Agricultural Sciences, University of Patras, 26504, Mesolonghi, Greece.
| |
Collapse
|
17
|
Pimentel JM, Zhou JY, Wu GS. Autophagy and cancer therapy. Cancer Lett 2024; 605:217285. [PMID: 39395780 DOI: 10.1016/j.canlet.2024.217285] [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: 08/02/2024] [Revised: 09/25/2024] [Accepted: 10/03/2024] [Indexed: 10/14/2024]
Abstract
Autophagy is an intracellular degradation process that sequesters cytoplasmic components in double-membrane vesicles known as autophagosomes, which are degraded upon fusion with lysosomes. This pathway maintains the integrity of proteins and organelles while providing energy and nutrients to cells, particularly under nutrient deprivation. Deregulation of autophagy can cause genomic instability, low protein quality, and DNA damage, all of which can contribute to cancer. Autophagy can also be overactivated in cancer cells to aid in cancer cell survival and drug resistance. Emerging evidence indicates that autophagy has functions beyond cargo degradation, including roles in tumor immunity and cancer stem cell survival. Additionally, autophagy can also influence the tumor microenvironment. This feature warrants further investigation of the role of autophagy in cancer, in which autophagy manipulation can improve cancer therapies, including cancer immunotherapy. This review discusses recent findings on the regulation of autophagy and its role in cancer therapy and drug resistance.
Collapse
Affiliation(s)
- Julio M Pimentel
- Department of Pharmacology, University of California San Diego, La Jolla, CA, 92093, USA; Institutional Research Academic Career Development Award Program, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jun Ying Zhou
- Molecular Therapeutics Program, Karmanos Cancer Institute, Detroit, MI, 48201, USA; Department of Oncology, Wayne State University, Detroit, MI, 48201, USA
| | - Gen Sheng Wu
- Molecular Therapeutics Program, Karmanos Cancer Institute, Detroit, MI, 48201, USA; Department of Oncology, Wayne State University, Detroit, MI, 48201, USA; Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
18
|
Wang HD, Lv CL, Feng L, Guo JX, Zhao SY, Jiang P. The role of autophagy in brain health and disease: Insights into exosome and autophagy interactions. Heliyon 2024; 10:e38959. [PMID: 39524893 PMCID: PMC11546156 DOI: 10.1016/j.heliyon.2024.e38959] [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: 09/21/2023] [Revised: 09/27/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024] Open
Abstract
Effective management of cellular components is essential for maintaining brain health, and studies have identified several crucial biological processes in the brain. Among these, autophagy and the role of exosomes in cellular communication are critical for brain health and disease. The interaction between autophagy and exosomes in the nervous system, as well as their contributions to brain damage, have garnered significant attention. This review summarizes that exosomes and their cargoes have been implicated in the autophagy process in the pathophysiology of nervous system diseases. Furthermore, the onset and progression of neurological disorders may be affected by autophagy regulation of the secretion and release of exosomes. These findings may provide new insights into the potential mechanism by which autophagy mediates different exosome secretion and release, as well as the valuable biomedical applications of exosomes in the prevention and treatment of various brain diseases by targeting autophagy.
Collapse
Affiliation(s)
- Hai-Dong Wang
- Department of Pharmacy, The Affiliated Lianyungang Hospital of Xuzhou Medical University/Nanjing Medical University Kangda College First Affiliated Hospital/The First People's Hospital of Lianyungang, Lianyungang, 222000, China
| | - Chao-Liang Lv
- Department of Spine Surgery, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
| | - Lei Feng
- Department of Neurosurgery, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
| | - Jin-Xiu Guo
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, 272000, China
| | - Shi-Yuan Zhao
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, 272000, China
| | - Pei Jiang
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, 272000, China
| |
Collapse
|
19
|
Afroz S, Preet R, Vishwakarma V, Evans AE, Magstadt AN, Dixon DA. Regulation of autophagy by Rab27B in colorectal cancer. Int J Biochem Cell Biol 2024:106693. [PMID: 39542128 DOI: 10.1016/j.biocel.2024.106693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 10/29/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
Autophagy is a cellular recycling process that is associated with tumor growth, anti-tumor immune responses, and therapy resistance in colorectal cancer (CRC). In this report, we identify the small GTPase Rab27B to control the autophagy process in CRC. Depletion of Rab27B showed an abnormal accumulation of autophagy vesicles and increased autophagy markers in CRC cells, indicating autophagy dysregulation. Image analysis indicated that autophagy flux is blocked at the autophagosome/lysosome fusion step when Rab27B is lost. While Rab27B deficient cells are proficient at growth under 2D in vitro conditions, cell growth was significantly impacted in both in vitro 3D growth and in vivo tumorigenesis studies. Together, these results demonstrate a new role of Rab27B in the autophagy trafficking process in CRC and identify Rab27B as a potential therapeutic target for CRC.
Collapse
Affiliation(s)
- Sahida Afroz
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, USA
| | - Ranjan Preet
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, USA
| | - Vikalp Vishwakarma
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, USA
| | - Andrew E Evans
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, USA
| | - Alexa N Magstadt
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, USA
| | - Dan A Dixon
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, USA; University of Kansas Cancer Center, Kansas City, KS, USA.
| |
Collapse
|
20
|
Sun F, Ali NN, Londoño-Vásquez D, Simintiras CA, Qiao H, Ortega MS, Agca Y, Takahashi M, Rivera RM, Kelleher AM, Sutovsky P, Patterson AL, Balboula AZ. Increased DNA damage in full-grown oocytes is correlated with diminished autophagy activation. Nat Commun 2024; 15:9463. [PMID: 39487138 PMCID: PMC11530536 DOI: 10.1038/s41467-024-53559-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: 08/17/2023] [Accepted: 10/14/2024] [Indexed: 11/04/2024] Open
Abstract
Unlike mild DNA damage exposure, DNA damage repair (DDR) is reported to be ineffective in full-grown mammalian oocytes exposed to moderate or severe DNA damage. The underlying mechanisms of this weakened DDR are unknown. Here, we show that moderate DNA damage in full-grown oocytes leads to aneuploidy. Our data reveal that DNA-damaged oocytes have an altered, closed, chromatin state, and suggest that the failure to repair damaged DNA could be due to the inability of DDR proteins to access damaged loci. Our data also demonstrate that, unlike somatic cells, mouse and porcine oocytes fail to activate autophagy in response to DNA double-strand break-inducing treatment, which we suggest may be the cause of the altered chromatin conformation and inefficient DDR. Importantly, autophagy activity is further reduced in maternally aged oocytes (which harbor severe DNA damage), and its induction is correlated with reduced DNA damage in maternally aged oocytes. Our findings provide evidence that reduced autophagy activation contributes to weakened DDR in oocytes, especially in those from aged females, offering new possibilities to improve assisted reproductive therapy in women with compromised oocyte quality.
Collapse
Affiliation(s)
- Fei Sun
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Nourhan Nashat Ali
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
- Department of Physiology, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | | | - Constantine A Simintiras
- School of Animal Sciences, Agricultural Center, Louisiana State University, Baton Rouge, LA, USA
| | - Huanyu Qiao
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - M Sofia Ortega
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Yuksel Agca
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
| | - Masashi Takahashi
- Research Faculty of Agriculture, Hokkaido University, Hokkaido, Japan
| | - Rocío M Rivera
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Andrew M Kelleher
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, MO, USA
| | - Peter Sutovsky
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, MO, USA
| | - Amanda L Patterson
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, MO, USA
| | - Ahmed Z Balboula
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
21
|
Ma K, Zhang C, Zhang H, An C, Li G, Cheng L, Li M, Ren M, Bai Y, Liu Z, Ji S, Liu X, Gao J, Zhang Z, Wu X, Chen X. High-Salt Diet Accelerates Neuron Loss and Anxiety in APP/PS1 Mice Through Serpina3n. Int J Mol Sci 2024; 25:11731. [PMID: 39519278 PMCID: PMC11546851 DOI: 10.3390/ijms252111731] [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: 09/20/2024] [Revised: 10/29/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
High salt (HS) consumption is an independent risk factor for neurodegenerative diseases such as dementia, stroke, and cerebral small vessel disease related to cognitive decline. Recently, Alzheimer's disease-like pathology changes have been reported as consequences of a HS diet in wild-type (wt) mice. However, it has not been revealed how HS diets accelerate the progress of Alzheimer's disease (AD) in APP/PS1 mice. Here, we fed APP/PS1 mice a HS diet or normal diet (ND) for six months; the effects of the HS/ND on wt mice were also observed. The results of our behavior test reveal that the HS diet exacerbates anxiety, β-amyloid overload, neuron loss, and synapse damage in the hippocampi of APP/PS1 mice; this was not observed in HS-treated wt mice. RNA sequencing shows that nearly all serpin family members were increased in the hippocampus of HS-treated APP/PS1 mice. Gene function analysis showed that a HS diet induces neurodegeneration, including axon dysfunction and neuro-ligand-based dysfunction, and regulates serine protein inhibitor activities. The mRNA and protein levels of Serpina3n were dramatically increased. Upregulated Serpina3n may be the key for β-amyloid aggregation and neuronal loss in the hippocampus of HS-treated APP/PS1 mice. Serpina3n inhibition attenuated the anxiety and increased the number of neurons in the hippocampal CA1(cornu ammonis) region of APP/PS1 mice. Our study provides novel insights into the mechanisms by which excessive HS diet deteriorates anxiety in AD mice. Therefore, decreasing daily dietary salt consumption constitutes a pivotal public health intervention for mitigating the progression of neuropathology, especially for old patients and those with neurodegenerative disease.
Collapse
Affiliation(s)
- Kaige Ma
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (K.M.); (C.Z.); (H.Z.); (C.A.); (G.L.); (L.C.); (M.L.); (M.R.); (Y.B.); (Z.L.); (S.J.); (X.L.); (J.G.); (Z.Z.)
- Key Laboratory of Ministry of Education for Environment and Genes Related to Diseases, Xi’an Jiaotong University, Xi’an 710061, China
| | - Chenglin Zhang
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (K.M.); (C.Z.); (H.Z.); (C.A.); (G.L.); (L.C.); (M.L.); (M.R.); (Y.B.); (Z.L.); (S.J.); (X.L.); (J.G.); (Z.Z.)
| | - Hanyue Zhang
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (K.M.); (C.Z.); (H.Z.); (C.A.); (G.L.); (L.C.); (M.L.); (M.R.); (Y.B.); (Z.L.); (S.J.); (X.L.); (J.G.); (Z.Z.)
- Key Laboratory of Ministry of Education for Environment and Genes Related to Diseases, Xi’an Jiaotong University, Xi’an 710061, China
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China
| | - Chanyuan An
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (K.M.); (C.Z.); (H.Z.); (C.A.); (G.L.); (L.C.); (M.L.); (M.R.); (Y.B.); (Z.L.); (S.J.); (X.L.); (J.G.); (Z.Z.)
| | - Ge Li
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (K.M.); (C.Z.); (H.Z.); (C.A.); (G.L.); (L.C.); (M.L.); (M.R.); (Y.B.); (Z.L.); (S.J.); (X.L.); (J.G.); (Z.Z.)
| | - Lixue Cheng
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (K.M.); (C.Z.); (H.Z.); (C.A.); (G.L.); (L.C.); (M.L.); (M.R.); (Y.B.); (Z.L.); (S.J.); (X.L.); (J.G.); (Z.Z.)
- Key Laboratory of Ministry of Education for Environment and Genes Related to Diseases, Xi’an Jiaotong University, Xi’an 710061, China
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China
| | - Mai Li
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (K.M.); (C.Z.); (H.Z.); (C.A.); (G.L.); (L.C.); (M.L.); (M.R.); (Y.B.); (Z.L.); (S.J.); (X.L.); (J.G.); (Z.Z.)
| | - Minghe Ren
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (K.M.); (C.Z.); (H.Z.); (C.A.); (G.L.); (L.C.); (M.L.); (M.R.); (Y.B.); (Z.L.); (S.J.); (X.L.); (J.G.); (Z.Z.)
| | - Yudan Bai
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (K.M.); (C.Z.); (H.Z.); (C.A.); (G.L.); (L.C.); (M.L.); (M.R.); (Y.B.); (Z.L.); (S.J.); (X.L.); (J.G.); (Z.Z.)
- Key Laboratory of Ministry of Education for Environment and Genes Related to Diseases, Xi’an Jiaotong University, Xi’an 710061, China
| | - Zichang Liu
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (K.M.); (C.Z.); (H.Z.); (C.A.); (G.L.); (L.C.); (M.L.); (M.R.); (Y.B.); (Z.L.); (S.J.); (X.L.); (J.G.); (Z.Z.)
| | - Shengfeng Ji
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (K.M.); (C.Z.); (H.Z.); (C.A.); (G.L.); (L.C.); (M.L.); (M.R.); (Y.B.); (Z.L.); (S.J.); (X.L.); (J.G.); (Z.Z.)
| | - Xiyue Liu
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (K.M.); (C.Z.); (H.Z.); (C.A.); (G.L.); (L.C.); (M.L.); (M.R.); (Y.B.); (Z.L.); (S.J.); (X.L.); (J.G.); (Z.Z.)
| | - Jinman Gao
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (K.M.); (C.Z.); (H.Z.); (C.A.); (G.L.); (L.C.); (M.L.); (M.R.); (Y.B.); (Z.L.); (S.J.); (X.L.); (J.G.); (Z.Z.)
| | - Zhichao Zhang
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (K.M.); (C.Z.); (H.Z.); (C.A.); (G.L.); (L.C.); (M.L.); (M.R.); (Y.B.); (Z.L.); (S.J.); (X.L.); (J.G.); (Z.Z.)
| | - Xiaolin Wu
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China
- Institute of Neuroscience, Translational Medicine Institute, Xi’an Jiaotong University, Xi’an 710061, China
| | - Xinlin Chen
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (K.M.); (C.Z.); (H.Z.); (C.A.); (G.L.); (L.C.); (M.L.); (M.R.); (Y.B.); (Z.L.); (S.J.); (X.L.); (J.G.); (Z.Z.)
- Key Laboratory of Ministry of Education for Environment and Genes Related to Diseases, Xi’an Jiaotong University, Xi’an 710061, China
| |
Collapse
|
22
|
Chen Y, Barylko B, Eichorst J, Mueller J, Albanesi J. Identification of the GABARAP binding determinant in PI4K2A. Biosci Rep 2024; 44:BSR20240200. [PMID: 39344512 PMCID: PMC11499380 DOI: 10.1042/bsr20240200] [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: 02/22/2024] [Revised: 09/17/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024] Open
Abstract
GABARAP is a member of the ATG8 family of ubiquitin-like autophagy related proteins. It was initially discovered as a facilitator of GABA-A receptor translocation to the plasma membrane and has since been shown to promote the intracellular transport of a variety of other proteins under non-autophagic conditions. We and others have shown that GABARAP interacts with the Type II phosphatidylinositol 4-kinase, PI4K2A, and that this interaction is important for autophagosome-lysosome fusion. Here, we identify a 7-amino acid segment within the PI4K2A catalytic domain that contains the GABARAP interaction motif (GIM). This segment resides in an exposed loop that is not conserved in the other mammalian Type II PI 4-kinase, PI4K2B, explaining the specificity of GABARAP binding to the PI4K2A isoform. Mutation of the PI4K2A GIM inhibits GABARAP binding and PI4K2A-mediated recruitment of cytosolic GABARAP to subcellular organelles. We further show that GABARAP binds to mono-phosphorylated phosphoinositides, PI3P, PI4P, and PI5P, raising the possibility that these lipids contribute to the binding energies that drive GABARAP-protein interactions on membranes.
Collapse
Affiliation(s)
- Yan Chen
- School of Physics and Astronomy, University of Minnesota, Minneapolis, MN 55455, U.S.A
| | - Barbara Barylko
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, U.S.A
| | - John P. Eichorst
- School of Physics and Astronomy, University of Minnesota, Minneapolis, MN 55455, U.S.A
| | - Joachim D. Mueller
- School of Physics and Astronomy, University of Minnesota, Minneapolis, MN 55455, U.S.A
| | - Joseph P. Albanesi
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, U.S.A
| |
Collapse
|
23
|
Li M, Feng J, Zhao K, Huang T, Zhang B, Yang Y, Sun A, Lin Q, Shao G. LSD1 Demethylates and Destabilizes Autophagy Protein LC3B in Ovarian Cancer. Biomolecules 2024; 14:1377. [PMID: 39595554 PMCID: PMC11591952 DOI: 10.3390/biom14111377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 11/28/2024] Open
Abstract
Autophagy is a complex cellular process that can either promote or inhibit cancer progression and development, depending on the context and molecular regulation involved. This study investigates how LSD1 regulates autophagy in ovarian cancer by interacting with the autophagy protein LC3B. Utilizing the bioinformatic analysis of TCGA, CPTAC, and GEO datasets, as well as immunohistochemistry in ovarian cancer patients, we explored the expression association between LSD1 and LC3B. Molecular mechanisms were further analyzed using Western blotting, immunoprecipitation, and GST pull-down assays. Our findings reveal that LSD1 binds to LC3B via its SWIRM domain, and high levels of LSD1 are closely associated with aggressive ovarian cancer and poor patient outcomes. Mechanistically, LSD1 demethylates LC3B, leading to decreased LC3B stability. The observed inverse correlation between LSD1 expression and LC3B protein levels in clinical samples underscores the need for further investigation to elucidate how reduced LC3B protein levels induced by LSD1 demethylation may contribute to ovarian cancer.
Collapse
Affiliation(s)
- Mingyang Li
- Institute of Urinary System Diseases, The Affiliated People’s Hospital, Jiangsu University, 8 Dianli Road, Zhenjiang 212002, China;
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Jie Feng
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Kangrong Zhao
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Ting Huang
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Bowen Zhang
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Yifan Yang
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Aiqin Sun
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Qiong Lin
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Genbao Shao
- Institute of Urinary System Diseases, The Affiliated People’s Hospital, Jiangsu University, 8 Dianli Road, Zhenjiang 212002, China;
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
24
|
Guo Y, Xu N, Yan H, Li J, Huang L, Zhu L, Du W, Liu Z, Zhao M. Splice Variant of Retinal G-Protein-Coupled Receptor Deletion-Mediated Dysregulation of Autophagy Increases the Susceptibility to Age-Related Macular Degeneration-Like Defects. Ophthalmic Res 2024; 67:611-624. [PMID: 39406195 DOI: 10.1159/000541991] [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: 03/29/2024] [Accepted: 09/30/2024] [Indexed: 11/14/2024]
Abstract
INTRODUCTION The splice variant of retinal G-protein-coupled receptor deletion (RGR-d) is a persistent component of drusen and may be involved in the pathogenesis of dry age-related macular degeneration (AMD). Increasing evidence has demonstrated the critical role of autophagy in AMD. In this study, we investigated whether RGR-d disrupts autophagy in early dry AMD in vivo and in vitro. METHODS Fundus imaging and fluoroscopy were performed on RGR-d mice created by multiplex gene editing. The retina microstructure was evaluated by performing hematoxylin and eosin (H&E) staining as well as transmission electron microscopy (TEM). Retinal function was assessed by full-field electroretinography (ERG). After lentivirus transfection and stimulation, the permeability, phagocytosis, and tight junctions of ARPE-19 cells were evaluated. Western blotting of ATG5, Beclin-1, LC3II/I, and P62 was performed to detect the changes in autophagy pathways. RESULTS Atrophy and patchy penetrating hyperfluorescent foci, consistent with early AMD-like defects, were observed in the fundus of 12-month-old RGR-d mice. H&E staining of retinal tissues indicated thinning of each layer of the retinal structure. H&E staining of retinal tissues indicated thinning of each layer of the retinal structure. TEM analysis showed some diffuse granular deposits. And the morphology of choroidal microvascular endothelial cells was degraded and distorted. The morphology of the photoreceptor outer segments showed structural damage, and Bruch's membrane was thickened. ERG indicated that the photoreceptor of RGR-d mice were dysfunctional. Changes in autophagy-related protein expression were observed in the retinal pigment epithelium and retinal neurepithelium, and autophagy regulation was decreased. Palmitic acid (PA) stimulation caused permeability, phagocytosis, and tight junction dysfunction in cells overexpressing RGR-d. Beclin-1 and LC3II/I expression levels were significantly decreased and that of P62 was elevated in RGR-d cells after PA stimulation. CONCLUSION RGR-d disrupts the autophagy pathway, causing the development of an early AMD-like pathophysiology.
Collapse
Affiliation(s)
- Yue Guo
- Department of Ophthalmology, Peking University People's Hospital, Beijing, China,
- Eye Diseases and Optometry Institute, Beijing, China,
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, China,
- College of Optometry, Peking University Health Science Center, Beijing, China,
| | - Ningda Xu
- Department of Ophthalmology, Peking University People's Hospital, Beijing, China
- Eye Diseases and Optometry Institute, Beijing, China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, China
- College of Optometry, Peking University Health Science Center, Beijing, China
| | - Huichao Yan
- Department of Ophthalmology, Peking University People's Hospital, Beijing, China
- Eye Diseases and Optometry Institute, Beijing, China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, China
- College of Optometry, Peking University Health Science Center, Beijing, China
| | - Jiarui Li
- Department of Ophthalmology, Peking University People's Hospital, Beijing, China
- Eye Diseases and Optometry Institute, Beijing, China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, China
- College of Optometry, Peking University Health Science Center, Beijing, China
| | - Lvzhen Huang
- Department of Ophthalmology, Peking University People's Hospital, Beijing, China
- Eye Diseases and Optometry Institute, Beijing, China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, China
- College of Optometry, Peking University Health Science Center, Beijing, China
| | - Li Zhu
- Department of Ophthalmology, Peking University People's Hospital, Beijing, China
- Eye Diseases and Optometry Institute, Beijing, China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, China
- College of Optometry, Peking University Health Science Center, Beijing, China
| | - Wei Du
- Department of Ophthalmology, Peking University People's Hospital, Beijing, China
- Eye Diseases and Optometry Institute, Beijing, China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, China
- College of Optometry, Peking University Health Science Center, Beijing, China
| | - Zhiming Liu
- Department of Ophthalmology, Peking University People's Hospital, Beijing, China
- Eye Diseases and Optometry Institute, Beijing, China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, China
- College of Optometry, Peking University Health Science Center, Beijing, China
| | - Mingwei Zhao
- Department of Ophthalmology, Peking University People's Hospital, Beijing, China
- Eye Diseases and Optometry Institute, Beijing, China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, China
- College of Optometry, Peking University Health Science Center, Beijing, China
| |
Collapse
|
25
|
Wang L, Chen H, Deng L, Hu M, Wang Z, Zhang K, Lian C, Wang X, Zhang J. Roburic acid inhibits lung cancer metastasis and triggers autophagy as verified by network pharmacology, molecular docking techniques and experiments. Front Oncol 2024; 14:1449143. [PMID: 39450260 PMCID: PMC11499198 DOI: 10.3389/fonc.2024.1449143] [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: 06/22/2024] [Accepted: 09/23/2024] [Indexed: 10/26/2024] Open
Abstract
Background Roburic acid (ROB) is a newly discovered tetracyclic triterpene acid extracted from oak galls, which has anti-inflammatory effects, but the mechanism of its anticancer effect is not clear. Our study focuses on exploring the potential mechanism of action of ROB in the treatment of lung cancer using a combination of network pharmacological prediction, molecular docking technique and experimental validation. Methods A network pharmacology approach was used to screen the protein targets of ROB and lung cancer, and PPI network analysis and enrichment analysis were performed on the intersecting genes. The tissue and organ distribution of the targets was also evaluated based on the BioGPS database. To ensure the reliability of the network pharmacology prediction results, we proceeded to use molecular docking technique to determine the relationship between drugs and targets. Finally, in vitro experiments with cell lines were performed to further reveal the potential mechanism of ROB for the treatment of lung cancer. Results A total of 83 potential targets of ROB in lung cancer were collected and further screened by using Cytoscape software, and 7 targets of PTGS2, CYP19A1, PTGS1, AR, CYP17A1, PTGES and SRD5A1 were obtained as hub genes and 7 hub targets had good binding energy with ROB. GO and KEGG analysis showed that ROB treatment of lung cancer mainly involves Arachidonic acid metabolism, Notch signaling pathway, cancer pathway and PPAR signaling pathway. The results of in vitro experiments indicated that ROB may inhibit the proliferation and metastasis of lung cancer cells and activate the PPARγ signaling pathway, as well as induce cellular autophagy. Conclusions The results of this study comprehensively elucidated the potential targets and molecular mechanisms of ROB for the treatment of lung cancer, providing new ideas for further lung cancer therapy.
Collapse
Affiliation(s)
- Luyao Wang
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
- Department of Genetics, School of Life Sciences, Bengbu Medical University, Bengbu, China
| | - Huili Chen
- Research Center of Clinical Laboratory Science, Bengbu Medical University, Bengbu, China
| | - Lili Deng
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Mengling Hu
- Department of Genetics, School of Life Sciences, Bengbu Medical University, Bengbu, China
| | - Ziqiang Wang
- Research Center of Clinical Laboratory Science, Bengbu Medical University, Bengbu, China
| | - Kai Zhang
- Research Center of Clinical Medicine, Bengbu Medical University, Bengbu, China
| | - Chaoqun Lian
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
- Research Center of Clinical Laboratory Science, Bengbu Medical University, Bengbu, China
| | - Xiaojing Wang
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
- Joint Research Center for Regional Diseases of Institute of Healthcare Management (IHM), The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Jing Zhang
- Department of Genetics, School of Life Sciences, Bengbu Medical University, Bengbu, China
| |
Collapse
|
26
|
Lin TH, Chen WL, Hsu SF, Chen IC, Lin CH, Chang KH, Wu YR, Chen YR, Yao CF, Lin W, Lee-Chen GJ, Chen CM. Small Molecules Inducing Autophagic Degradation of Expanded Polyglutamine Protein through Interaction with Both Mutant ATXN3 and LC3. Int J Mol Sci 2024; 25:10707. [PMID: 39409036 PMCID: PMC11477298 DOI: 10.3390/ijms251910707] [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: 09/10/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024] Open
Abstract
Polyglutamine (polyQ)-mediated spinocerebellar ataxia (SCA), including SCA1, 2, 3, 6, 7, and 17, are caused by mutant genes with expanded CAG repeats, leading to the intracellular accumulation of aggregated proteins, the production of reactive oxygen species, and cell death. Among SCA, SCA3 is caused by a mutation in the ATXN3 (ataxin-3) gene. In a circumstance of polyQ aggregation, the autophagic pathway is induced to degrade the aggregated proteins, thereby suppressing downstream deleterious effects and promoting neuronal survival. In this study, we tested the effects of synthetic indole (NC009-1, -2, -3, -6) and coumarin (LM-022, -031) derivatives as chemical chaperones to assist mutant ATXN3-Q75 folding, as well as autophagy inducers to clear aggregated protein. Among the tested compounds, NC009-1, -2, and -6 and LM-031 interfered with Escherichia coli-derived ATXN3-Q75 aggregation in thioflavin T binding and filter trap assays. In SH-SY5Y cells expressing GFP-fused ATXN3-Q75, these compounds displayed aggregation-inhibitory and neurite growth-promoting potentials compared to untreated cells. Furthermore, these compounds activated autophagy by increasing the phosphatidylethanolamine-conjugated LC3 (microtubule associated protein 1 light chain 3)-II:cytosolic LC3-I ratio in these cells. A biochemical co-immunoprecipitation assay by using a mixture of HEK 293T cell lysates containing recombinant ATXN3-Q75-Venus-C-terminus (VC) or Venus-N-terminus (VN)-LC3 protein indicated that NC009-1 and -2 and LM-031 served as an autophagosome-tethering compound (ATTEC) to interact with ATXN3-Q75 and LC3, and the interaction was further confirmed by bimolecular fluorescence complementation analysis in cells co-expressing both ATXN3-Q75-VC and VN-LC3 proteins. The study results suggest the potential of NC009-1 and -2 and LM-031 as an ATTEC in treating SCA3 and, probably, other polyQ diseases.
Collapse
Affiliation(s)
- Te-Hsien Lin
- Department of Neurology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan 33302, Taiwan; (T.-H.L.); (W.-L.C.); (C.-H.L.); (K.-H.C.); (Y.-R.W.)
| | - Wan-Ling Chen
- Department of Neurology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan 33302, Taiwan; (T.-H.L.); (W.-L.C.); (C.-H.L.); (K.-H.C.); (Y.-R.W.)
| | - Shao-Fan Hsu
- Department of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan; (S.-F.H.); (I.-C.C.)
| | - I-Cheng Chen
- Department of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan; (S.-F.H.); (I.-C.C.)
| | - Chih-Hsin Lin
- Department of Neurology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan 33302, Taiwan; (T.-H.L.); (W.-L.C.); (C.-H.L.); (K.-H.C.); (Y.-R.W.)
| | - Kuo-Hsuan Chang
- Department of Neurology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan 33302, Taiwan; (T.-H.L.); (W.-L.C.); (C.-H.L.); (K.-H.C.); (Y.-R.W.)
| | - Yih-Ru Wu
- Department of Neurology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan 33302, Taiwan; (T.-H.L.); (W.-L.C.); (C.-H.L.); (K.-H.C.); (Y.-R.W.)
| | - Yi-Ru Chen
- Department of Chemistry, National Taiwan Normal University, Taipei 11677, Taiwan; (Y.-R.C.); (C.-F.Y.); (W.L.)
| | - Ching-Fa Yao
- Department of Chemistry, National Taiwan Normal University, Taipei 11677, Taiwan; (Y.-R.C.); (C.-F.Y.); (W.L.)
| | - Wenwei Lin
- Department of Chemistry, National Taiwan Normal University, Taipei 11677, Taiwan; (Y.-R.C.); (C.-F.Y.); (W.L.)
| | - Guey-Jen Lee-Chen
- Department of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan; (S.-F.H.); (I.-C.C.)
| | - Chiung-Mei Chen
- Department of Neurology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan 33302, Taiwan; (T.-H.L.); (W.-L.C.); (C.-H.L.); (K.-H.C.); (Y.-R.W.)
| |
Collapse
|
27
|
Kudo Y, Nakamura K, Tsuzuki H, Hirota K, Kawai M, Takaya D, Fukuzawa K, Honma T, Yoshino Y, Nakamura M, Shiota M, Fujimoto N, Ikari A, Endo S. Docosahexaenoic acid enhances the treatment efficacy for castration-resistant prostate cancer by inhibiting autophagy through Atg4B inhibition. Arch Biochem Biophys 2024; 760:110135. [PMID: 39181384 DOI: 10.1016/j.abb.2024.110135] [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: 07/26/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Autophagy induction in cancer is involved in cancer progression and the acquisition of resistance to anticancer agents. Therefore, autophagy is considered a potential therapeutic target in cancer therapy. In this study, we found that long-chain fatty acids (LCFAs) have inhibitory effects on Atg4B, which is essential for autophagosome formation, through screening based on the pharmacophore of 21f, a recently developed Atg4B inhibitor. Among these fatty acids, docosahexaenoic acid (DHA), a polyunsaturated fatty acid, exhibited the most potent Atg4B inhibitory activity. DHA inhibited autophagy induced by androgen receptor signaling inhibitors (ARSI) in LNCaP and 22Rv1 prostate cancer cells and significantly increased apoptotic cell death. Furthermore, we investigated the effect of DHA on resistance to ARSI by establishing darolutamide-resistant prostate cancer 22Rv1 (22Rv1/Dar) cells, which had developed resistance to darolutamide, a novel ARSI. At baseline, 22Rv1/Dar cells showed a higher autophagy level than parental 22Rv1 cells. DHA significantly suppressed Dar-induced autophagy and sensitized 22Rv1/Dar cells by inducing apoptotic cell death through mitochondrial dysfunction. These results suggest that DHA supplementation may improve prostate cancer therapy with ARSI.
Collapse
Affiliation(s)
- Yudai Kudo
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Kana Nakamura
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Honoka Tsuzuki
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Kotaro Hirota
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Mina Kawai
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Daisuke Takaya
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan
| | - Kaori Fukuzawa
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan
| | - Teruki Honma
- Center for Biosystems Dynamics Research, RIKEN, Kanagawa, 230-0045, Japan
| | - Yuta Yoshino
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Mitsuhiro Nakamura
- Laboratories of Drug Informatics, Department of Pharmacy Practice and Science, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Masaki Shiota
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Naohiro Fujimoto
- Department of Urology, University of Occupational and Environmental Health, Fukuoka, 807-8555, Japan
| | - Akira Ikari
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Satoshi Endo
- The United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu, 501-1194, Japan; Center for One Medicine Innovative Translational Research (COMIT), Gifu University, Gifu, 501-1193, Japan.
| |
Collapse
|
28
|
Dhavarasa P, Sack T, Cerrato CP, Cheng AP, Zhang YY, Chen K, Kelley SO. Mitochondrial Probe for Glutathione Depletion Reveals NME3 Essentiality for Mitochondrial Redox Response. ACS Chem Biol 2024; 19:2012-2022. [PMID: 39133631 DOI: 10.1021/acschembio.4c00287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
Maintenance of the mitochondrial thiol redox state is essential for cell survival. However, we lack a comprehensive understanding of the redox response to mitochondrial glutathione depletion. We developed a mitochondria-penetrating peptide, mtCDNB, to specifically deplete mitochondrial glutathione. A genome-wide CRISPR/Cas9 screen in tandem with mtCDNB treatment was employed to uncover regulators of the redox response to mitochondrial glutathione depletion. We identified nucleoside diphosphate kinase 3 (NME3) as a regulator of mitochondrial dynamics. We show that NME3 is recruited to the mitochondrial outer membrane when under redox stress. In the absence of NME3, there is impaired mitophagy, which leads to the accumulation of dysfunctional mitochondria. NME3 knockouts depleted of mitochondrial glutathione have increased mitochondrial ROS production, accumulate mtDNA lesions, and present a senescence-associated secretory phenotype. Our findings suggest a novel role for NME3 in selecting mitochondria for degradation through mitophagy under conditions of mitochondrial redox stress.
Collapse
Affiliation(s)
- Piriththiv Dhavarasa
- Department of Biochemistry, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Tanja Sack
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada
| | - Carmine P Cerrato
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada
| | - Ashley P Cheng
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada
| | - Yi Y Zhang
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada
| | - Kangfu Chen
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60607, United States
| | - Shana O Kelley
- Department of Biochemistry, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada
- Department of Chemistry, Northwestern University, Evanston, Illinois 60607, United States
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60607, United States
- Chan Zuckerberg Biohub Chicago, Chicago, Illinois 60642, United States
| |
Collapse
|
29
|
Gökerküçük EB, Tramier M, Bertolin G. Protocol for quantifying LC3B FRET biosensor activity in living cells using a broad-to-sensitive data analysis pipeline. STAR Protoc 2024; 5:103181. [PMID: 39178110 PMCID: PMC11387700 DOI: 10.1016/j.xpro.2024.103181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 04/10/2024] [Accepted: 06/18/2024] [Indexed: 08/25/2024] Open
Abstract
Here, we present a protocol to comprehensively quantify autophagy initiation using the readout of the microtubule associated protein 1 light chain 3 beta (LC3B) Förster's resonance energy transfer (FRET) biosensor. We describe steps for cell seeding, transfection, FRET/FLIM (fluorescence lifetime imaging microscopy) imaging, and image analysis. This protocol can be useful in any physiology- or disease-related paradigm where the LC3B biosensor can be expressed to determine whether autophagy has been initiated or is stalled. The analysis pipeline presented here can be applied to any other genetically encoded FRET sensor imaged using FRET/FLIM. For complete details on the use and execution of this protocol, please refer to Gökerküçük et al.1.
Collapse
Affiliation(s)
- Elif Begüm Gökerküçük
- Univ Rennes, CNRS, IGDR (Institute of Genetics and Development of Rennes), UMR 6290, F-35000 Rennes, France
| | - Marc Tramier
- Univ Rennes, CNRS, IGDR (Institute of Genetics and Development of Rennes), UMR 6290, F-35000 Rennes, France.
| | - Giulia Bertolin
- Univ Rennes, CNRS, IGDR (Institute of Genetics and Development of Rennes), UMR 6290, F-35000 Rennes, France.
| |
Collapse
|
30
|
McMann E, Gorski SM. Last but not least: emerging roles of the autophagy-related protein ATG4D. Autophagy 2024; 20:1916-1927. [PMID: 38920354 PMCID: PMC11346562 DOI: 10.1080/15548627.2024.2369436] [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: 11/27/2023] [Revised: 06/05/2024] [Accepted: 06/13/2024] [Indexed: 06/27/2024] Open
Abstract
The evolutionarily conserved ATG4 cysteine proteases regulate macroautophagy/autophagy through the priming and deconjugation of the Atg8-family proteins. In mammals there are four ATG4 family members (ATG4A, ATG4B, ATG4C, ATG4D) but ATG4D has been relatively understudied. Heightened interest in ATG4D has been stimulated by recent links to human disease. Notably, genetic variations in human ATG4D were implicated in a heritable neurodevelopmental disorder. Genetic analyses in dogs, along with loss-of-function zebrafish and mouse models, further support a neuroprotective role for ATG4D. Here we discuss the evidence connecting ATG4D to neurological diseases and other pathologies and summarize its roles in both autophagy-dependent and autophagy-independent cellular processes.Abbrevation: ATG: autophagy related; BafA1: bafilomycin A1; BCL2: BCL2 apoptosis regulator; BH3: BCL2 homology region 3; CASP3: caspase 3; EV: extracellular vesicle; GABA: gamma aminobutyric acid; GABARAP: GABA type A receptor-associated protein; GABARAPL1: GABA type A receptor associated protein like 1; GABARAPL2: GABA type A receptor associated protein like 2; GFP: green fluorescent protein; LIR: LC3-interacting region; MAP1LC3: microtubule associated protein 1 light chain 3; MEF: mouse embryonic fibroblast; MYC: MYC proto-oncogene, bHLH transcription factor; PE: phosphatidylethanolamine; PS: phosphatidylserine; QKO: quadruple knockout; SDS-PAGE: sodium dodecyl sulfate-polyacrylamide gel; SQSTM1: sequestosome 1.
Collapse
Affiliation(s)
- Emily McMann
- Canada’s Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, British Columbia, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Sharon M. Gorski
- Canada’s Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, British Columbia, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
- Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, British Columbia, Canada
| |
Collapse
|
31
|
Lee YM, Vucic D. The role of autophagy in RIP1 mediated cell death and intestinal inflammation. Adv Immunol 2024; 163:1-20. [PMID: 39271257 DOI: 10.1016/bs.ai.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
Autophagy, a highly conserved catabolic process that targets various types of cellular cargoes to lysosomal degradation, is one of the most important biological mechanisms critical for cellular homeostasis. Components of these cellular cargoes can range from individual proteins to invading pathogens, and degrading these materials is important for maintaining organismal health and survival. The process of autophagy is carried out by complex molecular mechanisms, and a growing body of evidence indicates that these mechanisms intersect with those involved in the cell death pathways. In this review, we examine several emerging studies elucidating the role of autophagy in RIP1-mediated cell death signaling, with particular emphasis on impaired autophagy caused by ATG16L1 deficiency. We also discuss how autophagy in RIP1-mediated cell death affects intestinal homeostasis in preclinical models, and the implications of the intersection between RIP1 and autophagy for understanding the intestinal pathologies associated with inflammatory bowel disease (IBD). Finally, we highlight the potential benefits of therapeutic targeting of RIP1 and autophagy proteins, while also proposing areas of research that will likely elucidate new links between autophagy and cell death signaling.
Collapse
Affiliation(s)
| | - Domagoj Vucic
- Immunology Discovery, Genentech, South San Francisco, CA, United States.
| |
Collapse
|
32
|
Knupp J, Chen YJ, Wang E, Arvan P, Tsai B. Sigma-1 receptor recruits LC3 mRNA to ER-associated omegasomes to promote localized LC3 translation enabling functional autophagy. Cell Rep 2024; 43:114619. [PMID: 39128005 PMCID: PMC11376464 DOI: 10.1016/j.celrep.2024.114619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/14/2024] [Accepted: 07/25/2024] [Indexed: 08/13/2024] Open
Abstract
Autophagosome formation initiated on the endoplasmic reticulum (ER)-associated omegasome requires LC3. Translational regulation of LC3 biosynthesis is unexplored. Here we demonstrate that LC3 mRNA is recruited to omegasomes by directly binding to the ER transmembrane Sigma-1 receptor (S1R). Cell-based and in vitro reconstitution experiments show that S1R interacts with the 3' UTR of LC3 mRNA and ribosomes to promote LC3 translation. Strikingly, the 3' UTR of LC3 is also required for LC3 protein lipidation, thereby linking the mRNA-3' UTR to LC3 function. An autophagy-defective S1R mutant responsible for amyotrophic lateral sclerosis cannot bind LC3 mRNA or induce LC3 translation. We propose a model wherein S1R de-represses LC3 mRNA via its 3' UTR at the ER, enabling LC3 biosynthesis and lipidation. Because several other LC3-related proteins use the same mechanism, our data reveal a conserved pathway for localized translation essential for autophagosome biogenesis with insights illuminating the molecular basis of a neurodegenerative disease.
Collapse
Affiliation(s)
- Jeffrey Knupp
- Department of Cell & Developmental Biology, University of Michigan Medical School, 109 Zina Pitcher Place, BSRB 3043, Ann Arbor, MI 48109, USA; Cellular and Molecular Biology Program, University of Michigan Medical School, 1135 Catherine Street, Ann Arbor, MI 48109 USA
| | - Yu-Jie Chen
- Department of Cell & Developmental Biology, University of Michigan Medical School, 109 Zina Pitcher Place, BSRB 3043, Ann Arbor, MI 48109, USA
| | - Emily Wang
- Department of Cell & Developmental Biology, University of Michigan Medical School, 109 Zina Pitcher Place, BSRB 3043, Ann Arbor, MI 48109, USA
| | - Peter Arvan
- Cellular and Molecular Biology Program, University of Michigan Medical School, 1135 Catherine Street, Ann Arbor, MI 48109 USA; Division of Metabolism Endocrinology & Diabetes, University of Michigan Medical School, 1000 Wall Street, Ann Arbor, MI 48105, USA.
| | - Billy Tsai
- Department of Cell & Developmental Biology, University of Michigan Medical School, 109 Zina Pitcher Place, BSRB 3043, Ann Arbor, MI 48109, USA; Cellular and Molecular Biology Program, University of Michigan Medical School, 1135 Catherine Street, Ann Arbor, MI 48109 USA.
| |
Collapse
|
33
|
Barz S, Hofmann K, Reggiori F, Kraft C. Beyond the C-terminal Glycine of ATG8 Proteins - The Story of Some Neglected Amino Acids. J Mol Biol 2024; 436:168588. [PMID: 38663545 DOI: 10.1016/j.jmb.2024.168588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/12/2024] [Accepted: 04/17/2024] [Indexed: 05/07/2024]
Abstract
ATG8 proteins form a family of small ubiquitin-like modifiers, well-known for their importance in both macroautophagy and autophagy-independent processes. A unique feature of this protein family is their conjugation to membrane lipids through the covalent attachment of a glycine residue at the C-terminus of ATG8 proteins. Notably, most ATG8 proteins are expressed with additional amino acids at their C-terminus, shielding the key glycine residue. Consequently, lipidation requires the activation of the ATG8 precursors through proteolytic cleavage, known as priming. ATG4 proteases catalyze this priming process, and under physiological conditions, unprimed forms of ATG8 are not detected. This raises the question about the purpose of the C-terminal extension of ATG8 proteins. While the roles of lipidated and free, primed ATG8 proteins have been extensively studied, the potential function of their precursor form or the priming process itself remains largely unexplored. Here, we summarize information from existing literature and our own experiments to contribute to the understanding of these neglected amino acids.
Collapse
Affiliation(s)
- Saskia Barz
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Kay Hofmann
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany
| | - Fulvio Reggiori
- Department of Biomedicine, Aarhus University, Ole Worms Allé 4, 8000 Aarhus C, Denmark
| | - Claudine Kraft
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany.
| |
Collapse
|
34
|
Pino-Belmar C, Aguilar R, Valenzuela-Nieto GE, Cavieres VA, Cerda-Troncoso C, Navarrete VC, Salazar P, Burgos PV, Otth C, Bustamante HA. An Intrinsic Host Defense against HSV-1 Relies on the Activation of Xenophagy with the Active Clearance of Autophagic Receptors. Cells 2024; 13:1256. [PMID: 39120287 PMCID: PMC11311385 DOI: 10.3390/cells13151256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 06/10/2024] [Indexed: 08/10/2024] Open
Abstract
Autophagy engulfs cellular components in double-membrane-bound autophagosomes for clearance and recycling after fusion with lysosomes. Thus, autophagy is a key process for maintaining proteostasis and a powerful cell-intrinsic host defense mechanism, protecting cells against pathogens by targeting them through a specific form of selective autophagy known as xenophagy. In this context, ubiquitination acts as a signal of recognition of the cargoes for autophagic receptors, which direct them towards autophagosomes for subsequent breakdown. Nevertheless, autophagy can carry out a dual role since numerous viruses including members of the Orthoherpesviridae family can either inhibit or exploit autophagy for its own benefit and to replicate within host cells. There is growing evidence that Herpes simplex virus type 1 (HSV-1), a highly prevalent human pathogen that infects epidermal keratinocytes and sensitive neurons, is capable of negatively modulating autophagy. Since the effects of HSV-1 infection on autophagic receptors have been poorly explored, this study aims to understand the consequences of HSV-1 productive infection on the levels of the major autophagic receptors involved in xenophagy, key proteins in the recruitment of intracellular pathogens into autophagosomes. We found that productive HSV-1 infection in human neuroglioma cells and keratinocytes causes a reduction in the total levels of Ub conjugates and decreases protein levels of autophagic receptors, including SQSTM1/p62, OPTN1, NBR1, and NDP52, a phenotype that is also accompanied by reduced levels of LC3-I and LC3-II, which interact directly with autophagic receptors. Mechanistically, we show these phenotypes are the result of xenophagy activation in the early stages of productive HSV-1 infection to limit virus replication, thereby reducing progeny HSV-1 yield. Additionally, we found that the removal of the tegument HSV-1 protein US11, a recognized viral factor that counteracts autophagy in host cells, enhances the clearance of autophagic receptors, with a significant reduction in the progeny HSV-1 yield. Moreover, the removal of US11 increases the ubiquitination of SQSTM1/p62, indicating that US11 slows down the autophagy turnover of autophagy receptors. Overall, our findings suggest that xenophagy is a potent host defense against HSV-1 replication and reveals the role of the autophagic receptors in the delivery of HSV-1 to clearance via xenophagy.
Collapse
Affiliation(s)
- Camila Pino-Belmar
- Instituto de Microbiología Clínica, Facultad de Medicina, Universidad Austral de Chile, Valdivia 5110566, Chile; (C.P.-B.); (R.A.); (V.C.N.); (P.S.)
| | - Rayén Aguilar
- Instituto de Microbiología Clínica, Facultad de Medicina, Universidad Austral de Chile, Valdivia 5110566, Chile; (C.P.-B.); (R.A.); (V.C.N.); (P.S.)
| | - Guillermo E. Valenzuela-Nieto
- Instituto de Medicina, Facultad de Medicina, Universidad Austral de Chile, Valdivia 5110566, Chile;
- Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Universidad Austral de Chile, Valdivia 5110566, Chile
| | - Viviana A. Cavieres
- Organelle Phagy Lab, Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Lota 2465, Santiago 7510157, Chile; (V.A.C.); (C.C.-T.); (P.V.B.)
- Departamento de Ciencias Biológicas y Químicas, Facultad de Medicina y Ciencia, Universidad San Sebastián, Lota 2465, Santiago 7510157, Chile
| | - Cristóbal Cerda-Troncoso
- Organelle Phagy Lab, Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Lota 2465, Santiago 7510157, Chile; (V.A.C.); (C.C.-T.); (P.V.B.)
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago 7750000, Chile
| | - Valentina C. Navarrete
- Instituto de Microbiología Clínica, Facultad de Medicina, Universidad Austral de Chile, Valdivia 5110566, Chile; (C.P.-B.); (R.A.); (V.C.N.); (P.S.)
| | - Paula Salazar
- Instituto de Microbiología Clínica, Facultad de Medicina, Universidad Austral de Chile, Valdivia 5110566, Chile; (C.P.-B.); (R.A.); (V.C.N.); (P.S.)
| | - Patricia V. Burgos
- Organelle Phagy Lab, Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Lota 2465, Santiago 7510157, Chile; (V.A.C.); (C.C.-T.); (P.V.B.)
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago 7750000, Chile
| | - Carola Otth
- Instituto de Microbiología Clínica, Facultad de Medicina, Universidad Austral de Chile, Valdivia 5110566, Chile; (C.P.-B.); (R.A.); (V.C.N.); (P.S.)
- Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Universidad Austral de Chile, Valdivia 5110566, Chile
| | - Hianara A. Bustamante
- Instituto de Microbiología Clínica, Facultad de Medicina, Universidad Austral de Chile, Valdivia 5110566, Chile; (C.P.-B.); (R.A.); (V.C.N.); (P.S.)
| |
Collapse
|
35
|
Ashok S, Ramachandra Rao S. Updates on protein-prenylation and associated inherited retinopathies. FRONTIERS IN OPHTHALMOLOGY 2024; 4:1410874. [PMID: 39026984 PMCID: PMC11254824 DOI: 10.3389/fopht.2024.1410874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 06/13/2024] [Indexed: 07/20/2024]
Abstract
Membrane-anchored proteins play critical roles in cell signaling, cellular architecture, and membrane biology. Hydrophilic proteins are post-translationally modified by a diverse range of lipid molecules such as phospholipids, glycosylphosphatidylinositol, and isoprenes, which allows their partition and anchorage to the cell membrane. In this review article, we discuss the biochemical basis of isoprenoid synthesis, the mechanisms of isoprene conjugation to proteins, and the functions of prenylated proteins in the neural retina. Recent discovery of novel prenyltransferases, prenylated protein chaperones, non-canonical prenylation-target motifs, and reversible prenylation is expected to increase the number of inherited systemic and blinding diseases with aberrant protein prenylation. Recent important investigations have also demonstrated the role of several unexpected regulators (such as protein charge, sequence/protein-chaperone interaction, light exposure history) in the photoreceptor trafficking of prenylated proteins. Technical advances in the investigation of the prenylated proteome and its application in vision research are discussed. Clinical updates and technical insights into known and putative prenylation-associated retinopathies are provided herein. Characterization of non-canonical prenylation mechanisms in the retina and retina-specific prenylated proteome is fundamental to the understanding of the pathogenesis of protein prenylation-associated inherited blinding disorders.
Collapse
Affiliation(s)
- Sudhat Ashok
- Department of Ophthalmology, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, NY, United States
| | - Sriganesh Ramachandra Rao
- Department of Ophthalmology, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, NY, United States
- Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, NY, United States
- Research Service, VA Western New York Healthcare System, Buffalo, NY, United States
| |
Collapse
|
36
|
Jiang T, Ma C, Chen H. Unraveling the ultrastructure and dynamics of autophagic vesicles: Insights from advanced imaging techniques. FASEB Bioadv 2024; 6:189-199. [PMID: 38974114 PMCID: PMC11226998 DOI: 10.1096/fba.2024-00035] [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: 03/01/2024] [Revised: 04/18/2024] [Accepted: 04/22/2024] [Indexed: 07/09/2024] Open
Abstract
Autophagy, an intracellular self-degradation process, is governed by a complex interplay of signaling pathways and interactions between proteins and organelles. Its fundamental purpose is to efficiently clear and recycle cellular components that are damaged or redundant. Central to this process are autophagic vesicles, specialized structures that encapsulate targeted cellular elements, playing a pivotal role in autophagy. Despite growing interest in the molecular components of autophagic machinery and their regulatory mechanisms, capturing the detailed ultrastructural dynamics of autophagosome formation continues to present significant challenges. However, recent advancements in microscopy, particularly in electron microscopy, have begun to illuminate the dynamic regulatory processes underpinning autophagy. This review endeavors to provide an exhaustive overview of contemporary research on the ultrastructure of autophagic processes. By synthesizing observations from diverse technological methodologies, this review seeks to deepen our understanding of the genesis of autophagic vesicles, their membrane origins, and the dynamic alterations that transpire during the autophagy process. The aim is to bridge gaps in current knowledge and foster a more comprehensive comprehension of this crucial cellular mechanism.
Collapse
Affiliation(s)
- Ting Jiang
- Institute of Reproductive MedicineMedical School of Nantong UniversityNantongPR China
| | - Chaoye Ma
- Institute of Reproductive MedicineMedical School of Nantong UniversityNantongPR China
| | - Hao Chen
- Institute of Reproductive MedicineMedical School of Nantong UniversityNantongPR China
- Guangzhou Women and Children’s Medical Center, GMU‐GIBH Joint School of Life ScienceGuangzhou Medical UniversityGuangzhouChina
| |
Collapse
|
37
|
Ferretti GDS, Quaas CE, Bertolini I, Zuccotti A, Saatci O, Kashatus JA, Sharmin S, Lu DY, Poli ANR, Quesnelle AF, Rodriguez-Blanco J, de Cubas AA, Hobbs GA, Liu Q, O'Bryan JP, Salvino JM, Kashatus DF, Sahin O, Barnoud T. HSP70-mediated mitochondrial dynamics and autophagy represent a novel vulnerability in pancreatic cancer. Cell Death Differ 2024; 31:881-896. [PMID: 38802657 PMCID: PMC11239841 DOI: 10.1038/s41418-024-01310-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 04/29/2024] [Accepted: 05/01/2024] [Indexed: 05/29/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), the most prevalent type of pancreatic cancer, is one of the deadliest forms of cancer with limited therapy options. Overexpression of the heat shock protein 70 (HSP70) is a hallmark of cancer that is strongly associated with aggressive disease and worse clinical outcomes. However, the underlying mechanisms by which HSP70 allows tumor cells to thrive under conditions of continuous stress have not been fully described. Here, we report that PDAC has the highest expression of HSP70 relative to normal tissue across all cancers analyzed. Furthermore, HSP70 expression is associated with tumor grade and is further enhanced in metastatic PDAC. We show that genetic or therapeutic ablation of HSP70 alters mitochondrial subcellular localization, impairs mitochondrial dynamics, and promotes mitochondrial swelling to induce apoptosis. Mechanistically, we find that targeting HSP70 suppresses the PTEN-induced kinase 1 (PINK1) mediated phosphorylation of dynamin-related protein 1 (DRP1). Treatment with the HSP70 inhibitor AP-4-139B was efficacious as a single agent in primary and metastatic mouse models of PDAC. In addition, we demonstrate that HSP70 inhibition promotes the AMP-activated protein kinase (AMPK) mediated phosphorylation of Beclin-1, a key regulator of autophagic flux. Accordingly, we find that the autophagy inhibitor hydroxychloroquine (HCQ) enhances the ability of AP-4-139B to mediate anti-tumor activity in vivo. Collectively, our results suggest that HSP70 is a multi-functional driver of tumorigenesis that orchestrates mitochondrial dynamics and autophagy. Moreover, these findings support the rationale for concurrent inhibition of HSP70 and autophagy as a novel therapeutic approach for HSP70-driven PDAC.
Collapse
Affiliation(s)
- Giulia D S Ferretti
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Colleen E Quaas
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Irene Bertolini
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA, USA
| | - Alessandro Zuccotti
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Ozge Saatci
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Jennifer A Kashatus
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, VA, USA
| | - Salma Sharmin
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, VA, USA
| | - David Y Lu
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA, USA
| | | | - Abigail F Quesnelle
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Jezabel Rodriguez-Blanco
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
- Darby Children's Research Institute, Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Aguirre A de Cubas
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - G Aaron Hobbs
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA
| | - Qin Liu
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA, USA
| | - John P O'Bryan
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA
- Ralph H. Johnson VA Medical Center, Charleston, SC, USA
| | - Joseph M Salvino
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA, USA
| | - David F Kashatus
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, VA, USA
| | - Ozgur Sahin
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Thibaut Barnoud
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA.
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
38
|
Ling Z, Pan J, Zhang Z, Chen G, Geng J, Lin Q, Zhang T, Cao S, Chen C, Lin J, Yuan H, Ding W, Xiao F, Xu X, Li F, Wang G, Zhang Y, Li J. Small-molecule Molephantin induces apoptosis and mitophagy flux blockage through ROS production in glioblastoma. Cancer Lett 2024; 592:216927. [PMID: 38697460 DOI: 10.1016/j.canlet.2024.216927] [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: 02/27/2024] [Revised: 04/15/2024] [Accepted: 04/27/2024] [Indexed: 05/05/2024]
Abstract
Glioblastoma (GBM), one of the most malignant brain tumors in the world, has limited treatment options and a dismal survival rate. Effective and safe disease-modifying drugs for glioblastoma are urgently needed. Here, we identified a small molecule, Molephantin (EM-5), effectively penetrated the blood-brain barrier (BBB) and demonstrated notable antitumor effects against GBM with good safety profiles both in vitro and in vivo. Mechanistically, EM-5 not only inhibits the proliferation and invasion of GBM cells but also induces cell apoptosis through the reactive oxygen species (ROS)-mediated PI3K/Akt/mTOR pathway. Furthermore, EM-5 causes mitochondrial dysfunction and blocks mitophagy flux by impeding the fusion of mitophagosomes with lysosomes. It is noteworthy that EM-5 does not interfere with the initiation of autophagosome formation or lysosomal function. Additionally, the mitophagy flux blockage caused by EM-5 was driven by the accumulation of intracellular ROS. In vivo, EM-5 exhibited significant efficacy in suppressing tumor growth in a xenograft model. Collectively, our findings not only identified EM-5 as a promising, effective, and safe lead compound for treating GBM but also uncovered its underlying mechanisms from the perspective of apoptosis and mitophagy.
Collapse
Affiliation(s)
- Zhipeng Ling
- Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy, and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China; Department of Pharmacology, School of Medicine, and Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, China
| | - Junping Pan
- Guangdong Second Provincial General Hospital, Integrated Chinese and Western Medicine Postdoctoral Research Station, School of Medicine, Jinan University, Guangzhou, China
| | - Zhongfei Zhang
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Guisi Chen
- Department of Pharmacology, School of Medicine, and Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, China
| | - Jiayuan Geng
- Department of Microbial and Biochemical Pharmacy, College of Pharmacy, Jinan University, Guangzhou, China
| | - Qiang Lin
- Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy, and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
| | - Tao Zhang
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shuqin Cao
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Cheng Chen
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Jinrong Lin
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Hongyao Yuan
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Weilong Ding
- Department of Pharmacology, School of Medicine, and Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, China
| | - Fei Xiao
- Department of Pharmacology, School of Medicine, and Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, China
| | - Xinke Xu
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Fangcheng Li
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Guocai Wang
- Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy, and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China.
| | - Yubo Zhang
- Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy, and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China; Department of Pharmacology, School of Medicine, and Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, China.
| | - Junliang Li
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou, China.
| |
Collapse
|
39
|
Amin A, Perera ND, Tomas D, Cuic B, Radwan M, Hatters DM, Turner BJ, Shabanpoor F. Systemic administration of a novel Beclin 1-derived peptide significantly upregulates autophagy in the spinal motor neurons of autophagy reporter mice. Int J Pharm 2024; 659:124198. [PMID: 38816263 DOI: 10.1016/j.ijpharm.2024.124198] [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: 02/27/2024] [Revised: 04/09/2024] [Accepted: 05/01/2024] [Indexed: 06/01/2024]
Abstract
Autophagy, an intracellular degradation system, plays a vital role in protecting cells by clearing damaged organelles, pathogens, and protein aggregates. Autophagy upregulation through pharmacological interventions has gained significant attention as a potential therapeutic avenue for proteinopathies. Here, we report the development of an autophagy-inducing peptide (BCN4) derived from the Beclin 1 protein, the master regulator of autophagy. To deliver the BCN4 into cells and the central nervous system (CNS), it was conjugated to our previously developed cell and blood-brain barrier-penetrating peptide (CPP). CPP-BCN4 significantly upregulated autophagy and reduced protein aggregates in motor neuron (MN)-like cells. Moreover, its systemic administration in a reporter mouse model of autophagy resulted in a significant increase in autophagy activity in the spinal MNs. Therefore, this novel autophagy-inducing peptide with a demonstrated ability to upregulate autophagy in the CNS has significant potential for the treatment of various neurodegenerative diseases with protein aggregates as a characteristic feature.
Collapse
Affiliation(s)
- Azin Amin
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, VIC, Australia
| | - Nirma D Perera
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, VIC, Australia
| | - Doris Tomas
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, VIC, Australia
| | - Brittany Cuic
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, VIC, Australia
| | - Mona Radwan
- Walter and Eliza Hall Institute of Medical Research, University of Melbourne, Parkville 3010, VIC, Australia
| | - Danny M Hatters
- Department of Biochemistry and Pharmacology and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville 3010, VIC, Australia
| | - Bradley J Turner
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, VIC, Australia
| | - Fazel Shabanpoor
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, VIC, Australia; School of Chemistry, University of Melbourne, VIC 3010, Australia.
| |
Collapse
|
40
|
Ng ESY, Hu J, Jiang Z, Radu RA. Impaired cathepsin D in retinal pigment epithelium cells mediates Stargardt disease pathogenesis. FASEB J 2024; 38:e23720. [PMID: 38837708 PMCID: PMC11296957 DOI: 10.1096/fj.202400210rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 05/12/2024] [Accepted: 05/21/2024] [Indexed: 06/07/2024]
Abstract
Recessive Stargardt disease (STGD1) is an inherited juvenile maculopathy caused by mutations in the ABCA4 gene, for which there is no suitable treatment. Loss of functional ABCA4 in the retinal pigment epithelium (RPE) alone, without contribution from photoreceptor cells, was shown to induce STGD1 pathology. Here, we identified cathepsin D (CatD), the primary RPE lysosomal protease, as a key molecular player contributing to endo-lysosomal dysfunction in STGD1 using a newly developed "disease-in-a-dish" RPE model from confirmed STGD1 patients. Induced pluripotent stem cell (iPSC)-derived RPE originating from three STGD1 patients exhibited elevated lysosomal pH, as previously reported in Abca4-/- mice. CatD protein maturation and activity were impaired in RPE from STGD1 patients and Abca4-/- mice. Consequently, STGD1 RPE cells have reduced photoreceptor outer segment degradation and abnormal accumulation of α-synuclein, the natural substrate of CatD. Furthermore, dysfunctional ABCA4 in STGD1 RPE cells results in intracellular accumulation of autofluorescent material and phosphatidylethanolamine (PE). The altered distribution of PE associated with the internal membranes of STGD1 RPE cells presumably compromises LC3-associated phagocytosis, contributing to delayed endo-lysosomal degradation activity. Drug-mediated re-acidification of lysosomes in the RPE of STGD1 restores CatD functional activity and reduces the accumulation of immature CatD protein loads. This preclinical study validates the contribution of CatD deficiencies to STGD1 pathology and provides evidence for an efficacious therapeutic approach targeting RPE cells. Our findings support a cell-autonomous RPE-driven pathology, informing future research aimed at targeting RPE cells to treat ABCA4-mediated retinopathies.
Collapse
Affiliation(s)
- Eunice Sze Yin Ng
- UCLA Stein Eye Institute, University of California, Los Angeles, CA 90095, USA
- Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
- Molecular, Cellular, and Integrative Physiology Interdepartmental Program, University of California, Los Angeles, CA 90095, USA
| | - Jane Hu
- UCLA Stein Eye Institute, University of California, Los Angeles, CA 90095, USA
- Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Zhichun Jiang
- UCLA Stein Eye Institute, University of California, Los Angeles, CA 90095, USA
- Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Roxana A. Radu
- UCLA Stein Eye Institute, University of California, Los Angeles, CA 90095, USA
- Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
41
|
Lee A, Davis JH. NCOA4 initiates ferritinophagy by binding GATE16 using two highly avid short linear interaction motifs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.09.597909. [PMID: 38895392 PMCID: PMC11185777 DOI: 10.1101/2024.06.09.597909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Cells carefully regulate cytosolic iron, which is a vital enzymatic cofactor, yet is toxic in excess. In mammalian cells, surplus iron is sequestered in ferritin cages that, in iron limiting conditions, are degraded through the selective autophagy pathway ferritinophagy to liberate free iron. Prior work identified the ferritinophagy receptor protein NCOA4, which links ferritin and LC3/GABARAP-family member GATE16, effectively tethering ferritin to the autophagic machinery. Here, we elucidate the molecular mechanism underlying this interaction, discovering two short linear motifs in NCOA4 that each bind GATE16 with weak affinity. These binding motifs are highly avid and, in concert, support high-affinity NCOA4•GATE16 complex formation. We further find the minimal NCOA4383-522 fragment bearing these motifs is sufficient for ferritinophagy and that both motifs are necessary for this activity. This work suggests a general mechanism wherein selective autophagy receptors can distinguish between the inactive soluble pools of LC3/GABARAPs and the active membrane-conjugated forms that drive autophagy. Finally, we find that iron decreases NCOA4383-522's affinity for GATE16, providing a plausible mechanism for iron-dependent regulation of ferritinophagy.
Collapse
Affiliation(s)
- April Lee
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Joseph H. Davis
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
- Program in Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| |
Collapse
|
42
|
Kurganovs NJ, Engedal N. To eat or not to eat: a critical review on the role of autophagy in prostate carcinogenesis and prostate cancer therapeutics. Front Pharmacol 2024; 15:1419806. [PMID: 38910881 PMCID: PMC11190189 DOI: 10.3389/fphar.2024.1419806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 05/20/2024] [Indexed: 06/25/2024] Open
Abstract
Around 1 in 7 men will be diagnosed with prostate cancer during their lifetime. Many strides have been made in the understanding and treatment of this malignancy over the years, however, despite this; treatment resistance and disease progression remain major clinical concerns. Recent evidence indicate that autophagy can affect cancer formation, progression, and therapeutic resistance. Autophagy is an evolutionarily conserved process that can remove unnecessary or dysfunctional components of the cell as a response to metabolic or environmental stress. Due to the emerging importance of autophagy in cancer, targeting autophagy should be considered as a potential option in disease management. In this review, along with exploring the advances made on understanding the role of autophagy in prostate carcinogenesis and therapeutics, we will critically consider the conflicting evidence observed in the literature and suggest how to obtain stronger experimental evidence, as the application of current findings in clinical practice is presently not viable.
Collapse
Affiliation(s)
- Natalie Jayne Kurganovs
- Autophagy in Cancer Lab, Institute for Cancer Research, Department of Tumor Biology, Oslo University Hospital, Oslo, Norway
| | - Nikolai Engedal
- Autophagy in Cancer Lab, Institute for Cancer Research, Department of Tumor Biology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
43
|
Chen J, Zhao H, Liu M, Chen L. A new perspective on the autophagic and non-autophagic functions of the GABARAP protein family: a potential therapeutic target for human diseases. Mol Cell Biochem 2024; 479:1415-1441. [PMID: 37440122 DOI: 10.1007/s11010-023-04800-5] [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: 03/16/2023] [Accepted: 06/24/2023] [Indexed: 07/14/2023]
Abstract
Mammalian autophagy-related protein Atg8, including the LC3 subfamily and GABARAP subfamily. Atg8 proteins play a vital role in autophagy initiation, autophagosome formation and transport, and autophagy-lysosome fusion. GABARAP subfamily proteins (GABARAPs) share a high degree of homology with LC3 family proteins, and their unique roles are often overlooked. GABARAPs are as indispensable as LC3 in autophagy. Deletion of GABARAPs fails autophagy flux induction and autophagy lysosomal fusion, which leads to the failure of autophagy. GABARAPs are also involved in the transport of selective autophagy receptors. They are engaged in various particular autophagy processes, including mitochondrial autophagy, endoplasmic reticulum autophagy, Golgi autophagy, centrosome autophagy, and dorphagy. Furthermore, GABARAPs are closely related to the transport and delivery of the inhibitory neurotransmitter γ-GABAA and the angiotensin II AT1 receptor (AT1R), tumor growth, metastasis, and prognosis. GABARAPs also have been confirmed to be involved in various diseases, such as cancer, cardiovascular disease, and neurodegenerative diseases. In order to better understand the role and therapeutic potential of GABARAPs, this article comprehensively reviews the autophagic and non-autophagic functions of GABARAPs, as well as the research progress of the role and mechanism of GABARAPs in cancer, cardiovascular diseases and neurodegenerative diseases. It emphasizes the significance of GABARAPs in the clinical prevention and treatment of diseases, and may provide new therapeutic ideas and targets for human diseases. GABARAP and GABARAPL1 in the serum of cancer patients are positively correlated with the prognosis of patients, which can be used as a clinical biomarker, predictor and potential therapeutic target.
Collapse
Affiliation(s)
- Jiawei Chen
- Central Laboratory of Yan'nan Hospital Affiliated to Kunming, Medical University, Key Laboratory of Cardiovascular Diseases of Yunnan Province, Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, No. 245, Renmin East Road, Kunming, 650000, Yunnan, China
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Hong Zhao
- Central Laboratory of Yan'nan Hospital Affiliated to Kunming, Medical University, Key Laboratory of Cardiovascular Diseases of Yunnan Province, Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, No. 245, Renmin East Road, Kunming, 650000, Yunnan, China
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- School of Nursing, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Meiqing Liu
- Central Laboratory of Yan'nan Hospital Affiliated to Kunming, Medical University, Key Laboratory of Cardiovascular Diseases of Yunnan Province, Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, No. 245, Renmin East Road, Kunming, 650000, Yunnan, China.
| | - Linxi Chen
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
44
|
Coant N, Rendja K, Bellini L, Flamment M, Lherminier J, Portha B, Codogno P, Le Stunff H. Role of Sphingosine Kinase 1 in Glucolipotoxicity-Induced Early Activation of Autophagy in INS-1 Pancreatic β Cells. Cells 2024; 13:636. [PMID: 38607078 PMCID: PMC11011436 DOI: 10.3390/cells13070636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 03/04/2024] [Accepted: 03/27/2024] [Indexed: 04/13/2024] Open
Abstract
Insulin-producing pancreatic β cells play a crucial role in the regulation of glucose homeostasis, and their failure is a key event for diabetes development. Prolonged exposure to palmitate in the presence of elevated glucose levels, termed gluco-lipotoxicity, is known to induce β cell apoptosis. Autophagy has been proposed to be regulated by gluco-lipotoxicity in order to favor β cell survival. However, the role of palmitate metabolism in gluco-lipotoxcity-induced autophagy is presently unknown. We therefore treated INS-1 cells for 6 and 24 h with palmitate in the presence of low and high glucose concentrations and then monitored autophagy. Gluco-lipotoxicity induces accumulation of LC3-II levels in INS-1 at 6 h which returns to basal levels at 24 h. Using the RFP-GFP-LC3 probe, gluco-lipotoxicity increased both autophagosomes and autolysosmes structures, reflecting early stimulation of an autophagy flux. Triacsin C, a potent inhibitor of the long fatty acid acetyl-coA synthase, completely prevents LC3-II formation and recruitment to autophagosomes, suggesting that autophagic response requires palmitate metabolism. In contrast, etomoxir and bromo-palmitate, inhibitors of fatty acid mitochondrial β-oxidation, are unable to prevent gluco-lipotoxicity-induced LC3-II accumulation and recruitment to autophagosomes. Moreover, bromo-palmitate and etomoxir potentiate palmitate autophagic response. Even if gluco-lipotoxicity raised ceramide levels in INS-1 cells, ceramide synthase 4 overexpression does not potentiate LC3-II accumulation. Gluco-lipotoxicity also still stimulates an autophagic flux in the presence of an ER stress repressor. Finally, selective inhibition of sphingosine kinase 1 (SphK1) activity precludes gluco-lipotoxicity to induce LC3-II accumulation. Moreover, SphK1 overexpression potentiates autophagic flux induced by gluco-lipotxicity. Altogether, our results indicate that early activation of autophagy by gluco-lipotoxicity is mediated by SphK1, which plays a protective role in β cells.
Collapse
Affiliation(s)
- Nicolas Coant
- Unité BFA, Université Paris Cité, CNRS UMR 8251, 75006 Paris, France; (N.C.); (B.P.)
- Department of Pathology and Stony Brook Cancer Center, Stony Brook University Renaissance School of Medicine, Stony Brook, NY 11794, USA
| | - Karima Rendja
- Unité BFA, Université Paris Cité, CNRS UMR 8251, 75006 Paris, France; (N.C.); (B.P.)
| | - Lara Bellini
- Unité BFA, Université Paris Cité, CNRS UMR 8251, 75006 Paris, France; (N.C.); (B.P.)
| | - Mélissa Flamment
- Inserm, UMR-S 872, Centre de Recherche des Cordeliers, 75006 Paris, France
| | - Jeannine Lherminier
- INRA, UMR1347 Agroécologie, ERL CNRS 6300, Plateforme DImaCell, Centre de Microscopie INRA/Université de Bourgogne, 21065 Dijon, France
| | - Bernard Portha
- Unité BFA, Université Paris Cité, CNRS UMR 8251, 75006 Paris, France; (N.C.); (B.P.)
| | - Patrice Codogno
- INSERM U1151-CNRS UMR 8253, Institut Necker Enfants-Malades, University Paris Descartes, 75006 Paris, France
| | - Hervé Le Stunff
- Unité BFA, Université Paris Cité, CNRS UMR 8251, 75006 Paris, France; (N.C.); (B.P.)
- CNRS UMR 9197, Institut des Neurosciences Paris-Saclay, Saclay, University Paris, 91400 Saclay, France
| |
Collapse
|
45
|
Yin Y, Ahmed N, Hassan MF, Guo K, Shakir Y, Zang J, Lyu J. Effect of Nano-selenium on Biological Mechanism of Goblet Cells of the Small Intestine Within Laying Hen. Biol Trace Elem Res 2024; 202:1699-1710. [PMID: 37454307 DOI: 10.1007/s12011-023-03770-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/07/2023] [Indexed: 07/18/2023]
Abstract
Dietary selenium intake within the normal physiological range is critical for various supporting biological functions. However, the effect of nano-selenium on biological mechanism of goblet cells associated with autophagy is largely unknown.The purpose of this study was to investigate the effect of nano-selenium on the mucosal immune-defense mechanism of goblet cells (GCs) in the small intestine of laying hens.The autophagy was determined by using specific markers. Nano-selenium-treated group of immunohistochemistry (IHC), immunofluorescence (IF), and western blotting (WB) results indicated the strong positive immune signaling of microtubule-associated light chain (LC3) within the mucosal surface of the small intestine. However, weak expression of LC3 was observed in the 3-methyladenine autophagy inhibitor (3-MA) group. IHC and IF staining results showed the opposite tendency for LC3 of sequestosome 1 (P62/SQSTM1). P62/SQSTM1 showed strong positive immune signaling within the mucosal surface of the small intestine of the 3-MAgroup, and weak immune signaling of P62/SQSTM1 in the nano-selenium-treated group. Moreover, pinpointing autophagy was involved in the mucosal production and enrichment of mucosal immunity of the GCs. The morphology and ultrastructure evidence showed that the mucus secretion of GCs was significantly increased after nano-selenium treatment confirmed by light and transmission electron microscopy. Besides that, immunostaining of IHC, IF and WB showed that autophagy enhanced the secretion of Mucin2 (Muc2) protein in nano-selenium-treated group. This work illustrates that the nano-selenium particle might enhance the mucosal immune-defense mechanism via the protective role of GCs for intestinal homeostasis through autophagy.
Collapse
Affiliation(s)
- Yongxiang Yin
- Department of Pathology, Wuxi Maternal and Child Health Care Hospital, Womens Hospital of Jiangnan University, Jiangsu, 214002, China
| | - Nisar Ahmed
- Faculty of Veterinary and Animal Sciences, Lasbela University of Agriculture, Water and Marine Sciences, Uthal, 90150, Pakistan
| | - Mohammad Farooque Hassan
- Shaheed Benazir Bhutto University of Veterinary and Animal Sciences, Sakrand, Sindh, 67210, Pakistan
| | - Kai Guo
- Department of Pathology, Suzhou Science and Technology Town Hospital, Suzhou, 215153, China
| | - Yasmeen Shakir
- Department of Biochemistry, Hazara University, Mansehra, 21300, Pakistan
| | - Jia Zang
- Department of Laboratory Medicine, Wuxi Maternal and Child Health Care Hospital, Womens Hospital of Jiangnan University, Jiangsu, 214002, China.
| | - Jue Lyu
- Department of Laboratory Medicine, Wuxi No.2 Peoples Hospital, Jiangnan University Medical Center, Jiangsu, 214002, China.
| |
Collapse
|
46
|
Zhang Q, Guo F, Liu H, Hong L. Enhancing wound healing and overcoming cisplatin resistance in ovarian cancer. Int Wound J 2024; 21:e14569. [PMID: 38158767 PMCID: PMC10961880 DOI: 10.1111/iwj.14569] [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: 11/28/2023] [Revised: 12/04/2023] [Accepted: 12/04/2023] [Indexed: 01/03/2024] Open
Abstract
Ovarian cancer (OC) poses significant oncological challenges, notably impaired wound healing in the context of cisplatin (DDP) resistance. This study investigates the role of miR-200b in OC, emphasizing its impact on wound healing processes through DNMT3A/TGF-β1 pathway. The primary aim was to explore how miR-200b regulates autophagy and its consequential effects on wound healing in OC, alongside its influence on cisplatin resistance. Utilizing DDP-sensitive (A2780) and resistant (A2780/DDP) OC cell lines, along with human fibroblast cultures, the study employed an array of in vitro techniques. These included cell transfection with miR-200b mimic or inhibitor, chromatin immunoprecipitation (ChIP), dual-luciferase reporter (DLR) assays, quantitative PCR, Western blotting, MTT and particularly, wound healing assays. The research highlighted the role of miR-200b in wound healing within OC. Inhibition of miR-200b in A2780 cells and its mimic in A2780/DDP cells affected cell viability, indicating the link with DDP resistance. Crucially, miR-200b mimic significantly delayed fibroblast-mediated wound closure in assays, underscoring its impact on wound healing. Bioinformatics analysis and subsequent DLR assays confirmed miR-200b's interaction with DNMT3A, affecting TGF-β1 expression, the key factor in wound repair. Further, ChIP, quantitative PCR and Western blot analyses validated the interaction and expression changes in DNMT3A and TGF-β1. The study demonstrated that miR-200b played a pivotal role in OC by modulating autophagy, which in turn significantly affected wound healing through the DNMT3A/TGF-β1 pathway.
Collapse
Affiliation(s)
- Qifan Zhang
- Department of Obstetrics and GynecologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Fengqin Guo
- Department of Obstetrics and GynecologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Hua Liu
- Department of Obstetrics and GynecologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Li Hong
- Department of Obstetrics and GynecologyRenmin Hospital of Wuhan UniversityWuhanChina
| |
Collapse
|
47
|
Ke PY. Regulation of Autophagosome-Lysosome Fusion by Human Viral Infections. Pathogens 2024; 13:266. [PMID: 38535609 PMCID: PMC10974352 DOI: 10.3390/pathogens13030266] [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: 02/20/2024] [Revised: 03/13/2024] [Accepted: 03/20/2024] [Indexed: 02/11/2025] Open
Abstract
Autophagy plays a fundamental role in maintaining cellular homeostasis by eliminating intracellular components via lysosomes. Successful degradation through autophagy relies on the fusion of autophagosomes to lysosomes, which leads to the formation of autolysosomes containing acidic proteases that degrade the sequestered materials. Viral infections can exploit autophagy in infected cells to balance virus-host cell interactions by degrading the invading virus or promoting viral growth. In recent years, cumulative studies have indicated that viral infections may interfere with the fusion of autophagosomes and lysosomes, thus benefiting viral replication and associated pathogenesis. In this review, I provide an overview of the current understanding of the molecular mechanism by which human viral infections deregulate autophagosome-lysosome fusion and summarize the physiological significance in the virus life cycle and host cell damage.
Collapse
Affiliation(s)
- Po-Yuan Ke
- Department of Biochemistry & Molecular Biology and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; ; Tel.: +886-3-211-8800 (ext. 5115); Fax: +886-3-211-8700
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| |
Collapse
|
48
|
Ke PY. Molecular Mechanism of Autophagosome-Lysosome Fusion in Mammalian Cells. Cells 2024; 13:500. [PMID: 38534345 PMCID: PMC10968809 DOI: 10.3390/cells13060500] [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: 02/09/2024] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 03/28/2024] Open
Abstract
In eukaryotes, targeting intracellular components for lysosomal degradation by autophagy represents a catabolic process that evolutionarily regulates cellular homeostasis. The successful completion of autophagy initiates the engulfment of cytoplasmic materials within double-membrane autophagosomes and subsequent delivery to autolysosomes for degradation by acidic proteases. The formation of autolysosomes relies on the precise fusion of autophagosomes with lysosomes. In recent decades, numerous studies have provided insights into the molecular regulation of autophagosome-lysosome fusion. In this review, an overview of the molecules that function in the fusion of autophagosomes with lysosomes is provided. Moreover, the molecular mechanism underlying how these functional molecules regulate autophagosome-lysosome fusion is summarized.
Collapse
Affiliation(s)
- Po-Yuan Ke
- Department of Biochemistry & Molecular Biology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; ; Tel.: +886-3-211-8800 (ext. 5115); Fax: +886-3-211-8700
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| |
Collapse
|
49
|
Fan K, Liao Q, Yuan P, Xu R, Liu Z. Defective autophagy contributes to bupivacaine-induced aggravation of painful diabetic neuropathy in db/db mice. Neuropharmacology 2024; 245:109814. [PMID: 38104768 DOI: 10.1016/j.neuropharm.2023.109814] [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: 08/04/2023] [Revised: 11/08/2023] [Accepted: 12/08/2023] [Indexed: 12/19/2023]
Abstract
Current evidence suggests that hyperactivated or impaired autophagy can lead to neuronal death. The effect of local anesthetics on painful diabetic neuropathy (PDN) and the role of autophagy in the above pathological process remain unclear, warranting further studies. So, PDN models were established by assessing the paw withdrawal threshold (PWT) and paw withdrawal latency (PWL) in leptin gene-mutation (db/db) mice. Wild type (WT) and PDN mice received intrathecal 0.75% bupivacaine or/with intraperitoneal drug treatment (rapamycin or bafilomycin A1). Subsequently, the PWT and PWL were measured to assess hyperalgesia at 6 h, 24 h, 30 h, and 48 h after intrathecal bupivacaine. Also, sensory nerve conduction velocity (SNCV) and motor nerve conduction velocity (MNCV) were measured before and 48 h after intrathecal bupivacaine treatment. The spinal cord tissue of L4-L6 segments and serum were harvested to evaluate the change of autophagy, oxidative stress, oxidative injury, and apoptosis. We found that bupivacaine induced the activation of autophagy but did not affect the pain threshold, SNCV and MNCV in WT mice at predefined time points. Conversely, bupivacaine lowered autophagosome generation and degradation, slowed SNCV and aggravated spinal dorsal horn neuron oxidative injury and hyperalgesia in PDN mice. The autophagy activator (rapamycin) could decrease spinal dorsal horn neuron oxidative injury, alleviate the alterations in SNCV and hyperalgesia in bupivacaine-treated PDN mice. Meanwhile, the autophagy inhibitor (bafilomycin A1) could exacerbate spinal dorsal horn neuron oxidative injury, the alterations in SNCV and hyperalgesia in bupivacaine-treated PDN mice. Our results showed that bupivacaine could induce defective autophagy, slowed SNCV and aggravate spinal dorsal horn neuron oxidative injury and hyperalgesia in PDN mice. Restoring autophagy may represent a potential therapeutic approach against nerve injury in PDN patients with local anesthesia and analgesia.
Collapse
Affiliation(s)
- Keke Fan
- Department of Anesthesiology, Shenzhen Children's Hospital, Yantian Road 7019, Shenzhen, 518000, Guangdong Province, China.
| | - Qinming Liao
- Department of Neurosurgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong Province, China.
| | - Pengfei Yuan
- Department of Anesthesiology, South China Hospital of Shenzhen University, Fuxin Road 1, ShenZhen, 518116, Guangdong Province, China.
| | - Rui Xu
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Industrial Avenue Central 253, Guangzhou, 510282, Guangdong Province, China.
| | - Zhongjie Liu
- Department of Anesthesiology, Shenzhen Children's Hospital, Yantian Road 7019, Shenzhen, 518000, Guangdong Province, China; Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Industrial Avenue Central 253, Guangzhou, 510282, Guangdong Province, China.
| |
Collapse
|
50
|
Chen M, Zhu J, Luo H, Mu W, Guo L. The journey towards physiology and pathology: Tracing the path of neuregulin 4. Genes Dis 2024; 11:687-700. [PMID: 37692526 PMCID: PMC10491916 DOI: 10.1016/j.gendis.2023.03.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 02/11/2023] [Accepted: 03/05/2023] [Indexed: 09/12/2023] Open
Abstract
Neuregulin 4 (Nrg4), an epidermal growth factor (EGF) family member, can bind to and activate the ErbB4 receptor tyrosine kinase. Nrg4 has five different isoforms by alternative splicing and performs a wide variety of functions. Nrg4 is involved in a spectrum of physiological processes including neurobiogenesis, lipid metabolism, glucose metabolism, thermogenesis, and angiogenesis. In pathological processes, Nrg4 inhibits inflammatory factor levels and suppresses apoptosis in inflammatory diseases. In addition, Nrg4 could ameliorate obesity, insulin resistance, and cardiovascular diseases. Furthermore, Nrg4 improves non-alcoholic fatty liver disease (NAFLD) by promoting autophagy, improving lipid metabolism, and inhibiting cell death of hepatocytes. Besides, Nrg4 is closely related to the development of cancer, hyperthyroidism, and some other diseases. Therefore, elucidation of the functional role and mechanisms of Nrg4 will provide a clearer view of the therapeutic potential and possible risks of Nrg4.
Collapse
Affiliation(s)
- Min Chen
- School of Exercise and Health and Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China
| | - Jieying Zhu
- School of Exercise and Health and Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China
| | - Hongyang Luo
- School of Exercise and Health and Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China
| | - Wangjing Mu
- School of Exercise and Health and Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China
| | - Liang Guo
- School of Exercise and Health and Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China
| |
Collapse
|