1
|
Li L, He A, Zhao H, Tian C, Liu S, Stuart MAC, Wang J, Liu W. Rational design and structure-activity relationship of random copolymers for enhanced siRNA delivery. J Colloid Interface Sci 2025; 690:137273. [PMID: 40088818 DOI: 10.1016/j.jcis.2025.137273] [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: 12/25/2024] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 03/17/2025]
Abstract
HYPOTHESIS Cationic polymers and their derivatives have garnered significant interest as advanced vectors for siRNA delivery. Recently, we developed a robust diblock copolymer featuring an innovative binding block and a stealth block that work synergistically to facilitate efficient delivery of biotherapeutics. However, the fundamental mechanisms underlying its superior delivery capacity remain to be fully elucidated. EXPERIMENTS Since the binding block dominantly regulate the delivery performance, we synthesized a series of adapted copolymers, P(AAPBAm-co-DMAPMAn), by solely incorporating the key involved units, namely 3-acrylamidophenylboronic acid (AAPBA) and N-(3-dimethylaminopropyl)methacrylamide (DMAPMA). We thoroughly varied the block combinations, sequences and lengths, and investigated their effects on siRNA delivery. FINDINGS AAPBA and DMAPMA can bound to siRNA through reversible ester bonds and electrostatic interactions, respectively. The former enhanced siRNA release due to its responsive properties, while the cationic DMAPMA promoted endosomal escape of the complexes through its inherent interaction with membrane. Notably, only the rational combination of 20 units of each monomer, defined as copolymer P(AAPBA20-co-DMAPMA20), integrated the multiple yet balanced functions that sequentially promoted siRNA loading, endocytosis, endosome escape, and cytoplasmic release, ultimately leading to superior gene silencing. The clarified structure-activity relationships and revealed principles are valuable for the rational design of novel polymeric vectors to improve siRNA delivery and therapeutic applications.
Collapse
Affiliation(s)
- Lingshu Li
- State-Key Laboratory of Chemical Engineering, and Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China
| | - Axiang He
- Department of National Orthopaedic Medical Center, Shanghai Jiaotong University School of Medicine Affiliated Sixth People's Hospital, No. 222, West Huanhu Third Road, Pudong New Area, Shanghai 201306, People's Republic of China
| | - Hongyang Zhao
- State-Key Laboratory of Chemical Engineering, and Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China
| | - Chang Tian
- State-Key Laboratory of Chemical Engineering, and Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China
| | - Sishuo Liu
- State-Key Laboratory of Chemical Engineering, and Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China
| | - Martien A Cohen Stuart
- State-Key Laboratory of Chemical Engineering, and Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China
| | - Junyou Wang
- State-Key Laboratory of Chemical Engineering, and Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China.
| | - Wanjun Liu
- Department of National Orthopaedic Medical Center, Shanghai Jiaotong University School of Medicine Affiliated Sixth People's Hospital, No. 222, West Huanhu Third Road, Pudong New Area, Shanghai 201306, People's Republic of China.
| |
Collapse
|
2
|
Okafor M, Schmitt D, Ory S, Gasman S, Hureau C, Faller P, Vitale N. The Different Cellular Entry Routes for Drug Delivery Using Cell Penetrating Peptides. Biol Cell 2025; 117:e70012. [PMID: 40490965 DOI: 10.1111/boc.70012] [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: 01/31/2025] [Revised: 04/18/2025] [Accepted: 04/23/2025] [Indexed: 06/11/2025]
Abstract
The cell plasma membrane acts as a semi-permeable barrier essential for cellular protection and function, posing a challenge for therapeutic molecule delivery. Conventional techniques for crossing this barrier, including biophysical and biochemical methods, often exhibit limitations such as cytotoxicity and the risk of genomic integration when viral vectors are involved. In contrast, cell-penetrating peptides (CPPs) offer a promising non-invasive means to deliver a broad range of molecular cargoes, including proteins, nucleic acids and small molecules, into cells. CPPs, typically 5 to 30 amino acids long and rich in basic or non-polar residues, interact favourably with different cell membranes. These peptides have evolved since the discovery of the HIV-1 TAT peptide in the 1980s, expanding into various CPP families with diverse therapeutic applications. CPPs can form covalent or non-covalent complexes with their cargo, influencing their stability and efficacy. Based on their sequence properties and interactions, CPPs can be amphipathic or non-amphipathic, with distinct mechanisms of membrane penetration, such as direct penetration and endocytosis. While their uptake mechanisms are complex and not fully elucidated, ongoing optimization aims to enhance CPP specificity and efficacy. CPPs have demonstrated potential in drug delivery, gene therapy, cancer treatment and vaccine development, addressing key safety and efficiency concerns associated with viral vectors. This review explores the classification, mechanisms of action and therapeutic potential. It focuses on the intracellular vesicular trafficking of CPPs, highlighting their role as transformative tools in advancing cellular therapies and medical treatments.
Collapse
Affiliation(s)
- Michael Okafor
- Institut des Neurosciences Cellulaires et Intégratives-CNRS UPR3212, Université de Strasbourg, Strasbourg, France
- Institut de Chimie-UMR7177, Université de Strasbourg/CNRS, Strasbourg, France
| | - David Schmitt
- Institut des Neurosciences Cellulaires et Intégratives-CNRS UPR3212, Université de Strasbourg, Strasbourg, France
- Laboratoire de Chimie de Coordination-CNRS UPR8241, Université de Toulouse, Toulouse, France
| | - Stéphane Ory
- Institut des Neurosciences Cellulaires et Intégratives-CNRS UPR3212, Université de Strasbourg, Strasbourg, France
| | - Stéphane Gasman
- Institut des Neurosciences Cellulaires et Intégratives-CNRS UPR3212, Université de Strasbourg, Strasbourg, France
| | - Christelle Hureau
- Laboratoire de Chimie de Coordination-CNRS UPR8241, Université de Toulouse, Toulouse, France
| | - Peter Faller
- Institut de Chimie-UMR7177, Université de Strasbourg/CNRS, Strasbourg, France
| | - Nicolas Vitale
- Institut des Neurosciences Cellulaires et Intégratives-CNRS UPR3212, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
3
|
Debnath M, Malhotra M, Kulkarni A. Protein corona formation on supramolecular polymer nanoparticles causes differential endosomal sorting resulting in an attenuated NLRP3 inflammasome activation. Biomater Sci 2025; 13:3030-3047. [PMID: 40244934 DOI: 10.1039/d5bm00244c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
Upon introduction into biological environments, nanoparticles undergo the spontaneous formation of a dynamic protein corona, which continually evolves and significantly modifies their physicochemical properties and interactions with biological systems. This evolving protein corona can critically impact the nanoparticles' endocytic pathways and targeting efficiency, potentially altering their functional characteristics and obscuring their intended therapeutic effects. Despite considerable focus on the characterization of corona proteins and their impact on nanoparticle uptake, the intracellular processes and their effects on immunogenicity are not yet thoroughly understood. Supramolecular polymer nanoparticles (SNPs) with a highly hydrophobic core are recognized for triggering NLRP3 inflammasome activation, a key component of the innate immune system. Here, it is reported that the protein corona formation on SNPs exerts an inhibitory effect on the activation pathway of NLRP3 inflammasome. The protein corona impairs the intrinsic capacity of SNPs to induce lysosomal membrane rupture, thereby diminishing the cellular stress signals necessary for the formation of the NLRP3 inflammasome complex. Furthermore, the cells transport SNPs with an attached protein corona to recycling endosomes, where they are sorted and prepared for exocytosis. Conversely, nascent SNPs are primarily confined to late endosomes and lysosomes, leading to lysosomal rupture and inflammasome activation. This differential routing reflects the significant impact of the protein corona on the cellular handling and subsequent biological activity of nanoparticles. In summary, this study elucidates the fundamental role of the protein corona in shaping the intracellular disposition of nanoparticles, with implications for modulating their interactions with the immune system.
Collapse
Affiliation(s)
- Maharshi Debnath
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA.
| | - Mehak Malhotra
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA.
| | - Ashish Kulkarni
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA.
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA, USA
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, USA
| |
Collapse
|
4
|
Zaiki Y, Yap PG, Gan CY, Rani MFA, Traini D, Wong TW. "Actual" peptide properties required for nanoparticle development in precision cancer therapeutic delivery. J Control Release 2025:113866. [PMID: 40412661 DOI: 10.1016/j.jconrel.2025.113866] [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: 02/19/2025] [Revised: 04/27/2025] [Accepted: 05/18/2025] [Indexed: 05/27/2025]
Abstract
Functionalizing nanoparticles with peptides (3-30 amino acids) reduces premature clearance and increases colloidal stability and targeting capacity of cancer therapeutics. Glutamate/lysine-rich zwitterionic and hydrophilic/neutral peptides minimize reticuloendothelial digestion of nanomedicine through reducing particle hydrophobicity and depressing plasma anti-PEG immunoglobulin that disrupts the PEG-based particle stealth. Anionic peptides negate protein corona formation and subsequent particle aggregation in vivo enabling efficient nanoparticles biodistribution and drug targeting by facilitating their endothelial/extracellular matrix pore diffusion. Cationic and hydrophobic peptides display a strong affinity for anionic cancer cell membrane and mediate membrane porosification or receptor binding leading to particle uptake and endocytosis. The peptide ionic and hydrophobicity/hydrophilicity attributes collectively facilitate endosomal escape, and nuclear and mitochondria targeting of nanoparticles. Peptides are required to present with different physicochemical attributes from administration site, through blood and extracellular matrix, to cancer site of action. Charge/hydrophilicity-hydrophobicity switching and projection of receptor-specific domain of peptides are attainable through pH-pKa interplay and labile bond hydrolysis of "unwanted" domain to give rise to new functional domains in response to pH, thermal and enzymatic stimuli. Co-introducing all functional attributes on a single peptide is challenging. Use of peptide blends risks leaching during nanoparticles production. Peptides-nanoparticles conjugation risks peptide conformational alterations and loss of acidic/basic termini affecting their roles in nanoparticle stabilization, targeting, membrane permeabilization and subcellular delivery.
Collapse
Affiliation(s)
- Yazid Zaiki
- Non-Destructive Biomedical and Pharmaceutical Research Centre, Smart Manufacturing Research Institute, Universiti Teknologi MARA Selangor, Puncak Alam 42300, Selangor, Malaysia; Particle Design Research Group, Faculty of Pharmacy, Universiti Teknologi MARA Selangor, Puncak Alam 42300, Selangor, Malaysia
| | - Pei Gee Yap
- Analytical Biochemistry Research Centre (ABrC), Universiti Sains Malaysia, University Innovation Incubator Building, SAINS@USM campus, Lebuh Bukit Jambul, Bayan Lepas, 11900, Penang, Malaysia
| | - Chee Yuen Gan
- Analytical Biochemistry Research Centre (ABrC), Universiti Sains Malaysia, University Innovation Incubator Building, SAINS@USM campus, Lebuh Bukit Jambul, Bayan Lepas, 11900, Penang, Malaysia
| | | | - Daniela Traini
- Woolcock Institute of Medical Research, 431 Glebe Point Road, Glebe, Sydney 2037, Australia; Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Campus Macquarie Park, Sydney 2019, Australia
| | - Tin Wui Wong
- Non-Destructive Biomedical and Pharmaceutical Research Centre, Smart Manufacturing Research Institute, Universiti Teknologi MARA Selangor, Puncak Alam 42300, Selangor, Malaysia; Particle Design Research Group, Faculty of Pharmacy, Universiti Teknologi MARA Selangor, Puncak Alam 42300, Selangor, Malaysia; Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand.
| |
Collapse
|
5
|
Heiser BJ, Veyssi A, Ghosh D. Recent strategies for enhanced delivery of mRNA to the lungs. Nanomedicine (Lond) 2025; 20:1043-1069. [PMID: 40190037 PMCID: PMC12051540 DOI: 10.1080/17435889.2025.2485669] [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: 12/20/2024] [Accepted: 03/25/2025] [Indexed: 05/02/2025] Open
Abstract
mRNA-based therapies have emerged as a transformative tool in modern medicine, gaining significant attention following their successful use in COVID-19 vaccines. Delivery to the lungs offers several compelling advantages for mRNA delivery. The lungs are one of the most vascularized organs in the body, which provides an extensive surface area that can facilitate efficient drug transport. Local delivery to the lungs bypasses gastrointestinal degradation, potentially enhancing therapeutic efficacy. In addition, the extensive capillary network of the lungs provides an ideal target for systemic delivery. However, developing effective mRNA therapies for the lungs presents significant challenges. The complex anatomy of the lungs and the body's immune response to foreign particles create barriers to delivery. This review discusses key approaches for overcoming these challenges and improving mRNA delivery to the lungs. It examines both local and systemic delivery strategies aimed at improving lung delivery while mitigating off-target effects. Although substantial progress has been made in lung-targeted mRNA therapies, challenges remain in optimizing cellular uptake and achieving therapeutic efficacy within pulmonary tissues. The continued refinement of delivery strategies that enhance lung-specific targeting while minimizing degradation is critical for the clinical success of mRNA-based pulmonary therapies.
Collapse
Affiliation(s)
- Brittany J. Heiser
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Arian Veyssi
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Debadyuti Ghosh
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
6
|
Chen X, Wang YJ, Mu TW. Proteostasis regulation of GABA A receptors in neuronal function and disease. Biomed Pharmacother 2025; 186:117992. [PMID: 40112516 PMCID: PMC12068001 DOI: 10.1016/j.biopha.2025.117992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025] Open
Abstract
The γ-aminobutyric acid type A receptors (GABAARs) are ligand-gated anion channels that mediate fast inhibitory neurotransmission in the mammalian central nervous system. GABAARs form heteropentameric assemblies comprising two α1, two β2, and one γ2 subunits as the most common subtype in mammalian brains. Proteostasis regulation of GABAARs involves subunit folding within the endoplasmic reticulum, assembling into heteropentamers, receptor trafficking to the cell surface, and degradation of terminally misfolded subunits. As GABAARs are surface proteins, their trafficking to the plasma membrane is critical for proper receptor function. Thus, variants in the genes encoding GABAARs that disrupt proteostasis result in various neurodevelopmental disorders, ranging from intellectual disability to idiopathic generalized epilepsy. This review summarizes recent progress about how the proteostasis network regulates protein folding, assembly, degradation, trafficking, and synaptic clustering of GABAARs. Additionally, emerging pharmacological approaches that restore proteostasis of pathogenic GABAAR variants are presented, providing a promising strategy to treat related neurological diseases.
Collapse
Affiliation(s)
- Xi Chen
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| | - Ya-Juan Wang
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| | - Ting-Wei Mu
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| |
Collapse
|
7
|
de Freitas Oliveira-Tore C, de Moraes AG, Plácido HMBS, Signorini NMDL, Fontana PD, da Piedade Batista Godoy T, Boldt ABW, de Messias I. Non-canonical extracellular complement pathways and the complosome paradigm in cancer: a scoping review. Front Immunol 2025; 16:1519465. [PMID: 40370471 PMCID: PMC12075386 DOI: 10.3389/fimmu.2025.1519465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 03/13/2025] [Indexed: 05/16/2025] Open
Abstract
The Complement System (CS) comprises three catalytic pathways that can be activated by specific immune triggers. However, within the tumor microenvironment (TME), CS intracellular components, recently named as complosome, play roles that extend beyond the activation and regulation of its pathways. The interaction between TME elements and tumor cells alters the local immune response, leading to inflammation, cell proliferation, and tumor invasion. Our focus is on understanding the significance of complosome and non-canonical pathways in cancer. In this scoping review, we analyzed 45 articles that discussed the various roles of CS components in carcinogenesis. Many CS components, including C1q, C3a-C3aR, C5a-C5aR, factor H, and properdin, some of them at the intracellular level, may play a dual role in tumor progression, demonstrating either anti-tumor or pro-tumor activity independent of complement pathway activation. The specific function of each component can influence both the type and stage of tumor cells. There is a notable lack of studies on the role of the lectin pathway in tumor development, and this knowledge gap must be addressed to fully understand the role of complosome in cancer. Nevertheless, the activation of CS and the roles of its components in complosome pathways are crucial steps in tumor development.
Collapse
Affiliation(s)
- Camila de Freitas Oliveira-Tore
- Laboratory Molecular Immunopathology, Postgraduate Program in Internal Medicine and Health Sciences, Federal University of Paraná (UFPR), Curitiba, PR, Brazil
| | - Amarilis Giaretta de Moraes
- Laboratory Human Molecular Genetics, Postgraduate Program in Genetics, Department of Genetics, Federal University of Paraná (UFPR), Curitiba, PR, Brazil
| | - Helena Musetti B. S. Plácido
- Laboratory Molecular Immunopathology, Postgraduate Program in Internal Medicine and Health Sciences, Federal University of Paraná (UFPR), Curitiba, PR, Brazil
| | - Nathalia M. D. L. Signorini
- Laboratory Molecular Immunopathology, Postgraduate Program in Internal Medicine and Health Sciences, Federal University of Paraná (UFPR), Curitiba, PR, Brazil
| | - Pamela Dias Fontana
- Laboratory Molecular Immunopathology, Postgraduate Program in Internal Medicine and Health Sciences, Federal University of Paraná (UFPR), Curitiba, PR, Brazil
| | - Tatiane da Piedade Batista Godoy
- Laboratory Molecular Immunopathology, Postgraduate Program in Internal Medicine and Health Sciences, Federal University of Paraná (UFPR), Curitiba, PR, Brazil
| | - Angelica Beate Winter Boldt
- Laboratory Human Molecular Genetics, Postgraduate Program in Genetics, Department of Genetics, Federal University of Paraná (UFPR), Curitiba, PR, Brazil
| | - Iara de Messias
- Laboratory Molecular Immunopathology, Postgraduate Program in Internal Medicine and Health Sciences, Federal University of Paraná (UFPR), Curitiba, PR, Brazil
| |
Collapse
|
8
|
Jian L, Wang B, Gao Y, Xue Y, Guan Y, Qu Y, Liu K, Yan L, Li S, Luo H. Rab8a and Vps35 influence intracellular transport of vitamin E via α-Tocopherol transport protein in hepatocytes. Int J Biol Macromol 2025; 311:143021. [PMID: 40258550 DOI: 10.1016/j.ijbiomac.2025.143021] [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: 10/01/2024] [Revised: 04/08/2025] [Accepted: 04/08/2025] [Indexed: 04/23/2025]
Abstract
Of all the forms of vitamin E, α-tocopherol is distinguished as the primary ligand for the α-tocopherol transport protein (α-TTP), a pivotal factor in its secretion into the bloodstream and subsequent systemic distribution. Nevertheless, the intricate molecular mechanisms governing the transport of α-tocopherol via α-TTP have yet to be fully elucidated. In this research, Co-Immunoprecipitation (Co-IP)/LC-MS and His-pull-down assays were utilized to identify proteins interacting with α-TTP. Immunofluorescence staining and Co-IP/Western blotting further confirmed these interactions. Meanwhile, RNA-seq was utilized to discover α-tocopherol-related genes. Genes knockdown was conducted to examine the influence of related genes on vitamin E transport. The concentrations of intracellular and extracellular vitamin E were quantified using LC-MS and specific assay kits. Immunofluorescence staining showed colocalization of Rab8a, SNX3, SNX5, SNX17, and SNX27 with α-TTP, whereas Co-IP/Western blot analysis indicated a specific interaction among Vps35, Rab8a and α-TTP. Notably, the knockdown of Rab8a, SNX5, SNX17, and SNX27 individually influenced the vitamin E content both intracellularly and extracellularly, whereas knockdown of SNX3 did not show such effects in hepatocytes. This research highlights the crucial roles of Rab8a and Vps35 in α-tocopherol's intracellular transport probably by direct interaction with α-TTP, and their association with Retromer-SNX27, Commander-SNX17, and ESCPE1 complexes.
Collapse
Affiliation(s)
- Luyang Jian
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Bing Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yuefeng Gao
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ying Xue
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yongjuan Guan
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yanghua Qu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Kun Liu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Leyan Yan
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shuanghong Li
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Hailing Luo
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China.
| |
Collapse
|
9
|
Du G, He J, Zhan Y, Chen L, Hu Y, Qian J, Huang H, Meng F, Shan L, Chen Z, Hu D, Zhu C, Yue M, Qi Y, Tan W. Changes and application prospects of biomolecular materials in small extracellular vesicles (sEVs) after flavivirus infection. Eur J Med Res 2025; 30:275. [PMID: 40229861 PMCID: PMC11998145 DOI: 10.1186/s40001-025-02539-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: 09/27/2024] [Accepted: 03/31/2025] [Indexed: 04/16/2025] Open
Abstract
Small extracellular vesicles (sEVs), also known as exosomes, are membranous vesicles filled with various proteins and nucleic acids, serving as a communication vector between cells. Recent research has highlighted their role in viral diseases. This review synthesizes current understanding of viral sEVs and includes recent findings on sEVs infected with flaviviruses. It discusses the implications of viral sEVs research for advancing arbovirus sEVs research and anticipates the potential applications of sEVs in flavivirus infections.
Collapse
Affiliation(s)
- Gengting Du
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, People's Republic of China
- Nanjing Bioengineering (Gene) Technology Center for Medicines, Nanjing, People's Republic of China
| | - Junhua He
- Nanjing Jinling Hospital, Nanjing, Jiangsu, People's Republic of China
| | - Yan Zhan
- Nanjing Jinling Hospital, Nanjing, Jiangsu, People's Republic of China
| | - Leru Chen
- Nanjing Bioengineering (Gene) Technology Center for Medicines, Nanjing, People's Republic of China
| | - Yue Hu
- Nanjing Jinling Hospital, Nanjing, Jiangsu, People's Republic of China
| | - Jiaojiao Qian
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Huan Huang
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Fanjin Meng
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Laiyou Shan
- Nanjing Jinling Hospital, Nanjing, Jiangsu, People's Republic of China
| | - Zhiyu Chen
- Nanjing Jinling Hospital, Nanjing, Jiangsu, People's Republic of China
| | | | - Changqiang Zhu
- Nanjing Bioengineering (Gene) Technology Center for Medicines, Nanjing, People's Republic of China
| | - Ming Yue
- Department of Infectious Diseases, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yong Qi
- Nanjing Bioengineering (Gene) Technology Center for Medicines, Nanjing, People's Republic of China
| | - Weilong Tan
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, People's Republic of China.
- Nanjing Bioengineering (Gene) Technology Center for Medicines, Nanjing, People's Republic of China.
- Nanjing Jinling Hospital, Nanjing, Jiangsu, People's Republic of China.
| |
Collapse
|
10
|
Xu G, Zhang Q, Cheng R, Qu J, Li W. Survival strategies of cancer cells: the role of macropinocytosis in nutrient acquisition, metabolic reprogramming, and therapeutic targeting. Autophagy 2025; 21:693-718. [PMID: 39817564 PMCID: PMC11925119 DOI: 10.1080/15548627.2025.2452149] [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: 10/09/2024] [Revised: 12/27/2024] [Accepted: 01/07/2025] [Indexed: 01/18/2025] Open
Abstract
Macropinocytosis is a nonselective form of endocytosis that allows cancer cells to largely take up the extracellular fluid and its contents, including nutrients, growth factors, etc. We first elaborate meticulously on the process of macropinocytosis. Only by thoroughly understanding this entire process can we devise targeted strategies against it. We then focus on the central role of the MTOR (mechanistic target of rapamycin kinase) complex 1 (MTORC1) in regulating macropinocytosis, highlighting its significance as a key signaling hub where various pathways converge to control nutrient uptake and metabolic processes. The article covers a comprehensive analysis of the literature on the molecular mechanisms governing macropinocytosis, including the initiation, maturation, and recycling of macropinosomes, with an emphasis on how these processes are hijacked by cancer cells to sustain their growth. Key discussions include the potential therapeutic strategies targeting macropinocytosis, such as enhancing drug delivery via this pathway, inhibiting macropinocytosis to starve cancer cells, blocking the degradation and recycling of macropinosomes, and inducing methuosis - a form of cell death triggered by excessive macropinocytosis. Targeting macropinocytosis represents a novel and innovative approach that could significantly advance the treatment of cancers that rely on this pathway for survival. Through continuous research and innovation, we look forward to developing more effective and safer anti-cancer therapies that will bring new hope to patients.Abbreviation: AMPK: AMP-activated protein kinase; ASOs: antisense oligonucleotides; CAD: carbamoyl-phosphate synthetase 2, aspartate transcarbamylase, and dihydroorotase; DC: dendritic cell; EGF: epidermal growth factor; EGFR: epidermal growth factor receptor; ERBB2: erb-b2 receptor tyrosine kinase 2; ESCRT: endosomal sorting complex required for transport; GAP: GTPase-activating protein; GEF: guanine nucleotide exchange factor; GRB2: growth factor receptor bound protein 2; LPP: lipopolyplex; MTOR: mechanistic target of rapamycin kinase; MTORC1: mechanistic target of rapamycin kinase complex 1; MTORC2: mechanistic target of rapamycin kinase complex 2; NSCLC: non-small cell lung cancer; PADC: pancreatic ductal adenocarcinoma; PDPK1: 3-phosphoinositide dependent protein kinase 1; PI3K: phosphoinositide 3-kinase; PIK3C3: phosphatidylinositol 3-kinase catalytic subunit type 3; PtdIns(3,4,5)P3: phosphatidylinositol-(3,4,5)-trisphosphate; PtdIns(4,5)P2: phosphatidylinositol-(4,5)-bisphosphate; PTT: photothermal therapies; RAC1: Rac family small GTPase 1; RPS6: ribosomal protein S6; RPS6KB1: ribosomal protein S6 kinase B1; RTKs: receptor tyrosine kinases; SREBF: sterol regulatory element binding transcription factor; TFEB: transcription factor EB; TNBC: triple-negative breast cancer; TSC2: TSC complex subunit 2; ULK1: unc-51 like autophagy activating kinase 1; UPS: ubiquitin-proteasome system.
Collapse
Affiliation(s)
- Guoshuai Xu
- Department of General Surgery, Aerospace Center Hospital, Beijing, China
| | - Qinghong Zhang
- Emergency Department, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Renjia Cheng
- Department of Intensive Care Medicine, The General Hospital of the Northern Theater Command of the People’s Liberation Army of China, Shenyang, Liaoning, China
| | - Jun Qu
- Department of General Surgery, Aerospace Center Hospital, Beijing, China
| | - Wenqiang Li
- Department of General Surgery, Aerospace Center Hospital, Beijing, China
| |
Collapse
|
11
|
Xu P, Yue J. Protocol for monitoring the endosomal trafficking of membrane proteins in mammalian cells. STAR Protoc 2025; 6:103686. [PMID: 40057947 PMCID: PMC11928761 DOI: 10.1016/j.xpro.2025.103686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/12/2024] [Accepted: 02/19/2025] [Indexed: 03/25/2025] Open
Abstract
Here, we present a protocol to study epidermal growth factor (EGF) receptor (EGFR) or transferrin trafficking in mammalian cells. We describe steps for using fluorescent ligands or antibodies, confocal imaging, and quantitative analysis to track their movement. We detail procedures for cell culture preparation, labeling membrane proteins, optimizing imaging, and isolating cell lysates for the biochemical analysis of EGFR degradation after EGF treatment. This protocol is adaptable to various cell types and for assessing genetic or pharmacological impacts on endosomal trafficking. For complete details on the use and execution of this protocol, please refer to Ye et al.,1 Ye et al.,2 and Wang et al.3.
Collapse
Affiliation(s)
- Peng Xu
- Division of Natural and Applied Sciences, Synear Molecular Biology Lab, Global Health Research Center, Jiangsu Provincial University Key (Construction) Laboratory for Smart Diagnosis and Treatment of Lung Cancer, Duke Kunshan University, Kunshan 215316, China.
| | - Jianbo Yue
- Division of Natural and Applied Sciences, Synear Molecular Biology Lab, Global Health Research Center, Jiangsu Provincial University Key (Construction) Laboratory for Smart Diagnosis and Treatment of Lung Cancer, Duke Kunshan University, Kunshan 215316, China.
| |
Collapse
|
12
|
Milne SM, Edeen PT, Fay DS. TAT-1, a phosphatidylserine flippase, affects molting and regulates membrane trafficking in the epidermis of Caenorhabditis elegans. Genetics 2025; 229:iyae216. [PMID: 39722491 PMCID: PMC12086690 DOI: 10.1093/genetics/iyae216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024] Open
Abstract
Membrane trafficking is a conserved process required for the import, export, movement, and distribution of proteins and other macromolecules within cells. The Caenorhabditis elegans NIMA-related kinases NEKL-2 (human NEK8/9) and NEKL-3 (human NEK6/7) are conserved regulators of membrane trafficking and are required for the completion of molting. Using a genetic approach, we identified reduction-of-function mutations in tat-1 that suppress nekl-associated molting defects. tat-1 encodes the C. elegans ortholog of mammalian ATP8A1/2, a phosphatidylserine flippase that promotes the asymmetric distribution of phosphatidylserine on the cytosolic leaflet of lipid membrane bilayers. CHAT-1 (human CDC50), a conserved chaperone, was required for the correct localization of TAT-1, and chat-1 inhibition strongly suppressed nekl defects. Using a phosphatidylserine sensor, we found that TAT-1 was required for the normal localization of phosphatidylserine at apical endosomes and that loss of TAT-1 led to aberrant endosomal morphologies. Consistent with these data, TAT-1 localized to early endosomes and to recycling endosomes marked with RME-1, the C. elegans ortholog of the human EPS15 homology domain-containing protein, EHD1. TAT-1, phosphatidylserine biosynthesis, and the phosphatidylserine-binding protein RFIP-2 (human RAB11-FIP2) were all required for the normal localization of RME-1 to apical endosomes. Consistent with these proteins functioning together, inhibition of RFIP-2 or RME-1 led to the partial suppression of nekl molting defects, as did inhibition of phosphatidylserine biosynthesis. We propose that TAT-1 flippase activity, in conjunction with RFIP-2, promotes the recruitment of RME-1 to apical recycling endosomes and that inhibition of TAT-1-RFIP-2-RME-1 can compensate for a reduction in NEKL activities.
Collapse
Affiliation(s)
- Shae M Milne
- Department of Molecular Biology, College of Agriculture, Life Sciences and Natural Resources, University of Wyoming, Laramie, WY 82071, United States
| | - Philip T Edeen
- Department of Molecular Biology, College of Agriculture, Life Sciences and Natural Resources, University of Wyoming, Laramie, WY 82071, United States
| | - David S Fay
- Department of Molecular Biology, College of Agriculture, Life Sciences and Natural Resources, University of Wyoming, Laramie, WY 82071, United States
| |
Collapse
|
13
|
Long Y, Li Y, Xue J, Geng W, Ma M, Wang X, Wang L. Mechanisms by which SNX-BAR subfamily controls the fate of SNXs' cargo. Front Physiol 2025; 16:1559313. [PMID: 40144551 PMCID: PMC11936996 DOI: 10.3389/fphys.2025.1559313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 02/20/2025] [Indexed: 03/28/2025] Open
Abstract
The SNX-BAR subfamily is a component of the sorting nexins (SNXs) superfamily. Distinct from other SNXs, which feature a PX domain for phosphoinositide binding, the SNX-BAR subfamily includes a BAR domain that induces membrane curvature. Members of the SNX-BAR subfamily work together to recognize and select specific cargo, regulate receptor signaling, and manage cargo sorting both with and without the involvement of sorting complexes. They play a crucial role in maintaining cellular homeostasis by directing intracellular cargo to appropriate locations through endo-lysosomal, autophagolysosomal, and ubiquitin-proteasome pathways. This subfamily thus links various protein homeostasis pathways. This review examines the established and hypothesized functions of the SNX-BAR subfamily, its role in intracellular protein sorting and stability, and explores the potential involvement of subfamily dysfunction in the pathophysiology of cardiovascular and neurodegenerative diseases.
Collapse
Affiliation(s)
- Yaolin Long
- Basic Medical Research Center, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yang Li
- Basic Medical Research Center, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jin Xue
- Basic Medical Research Center, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Wanqing Geng
- Department of Ophthalmology, Shanxi Medical University Second Affiliated Hospital, Taiyuan, Shanxi, China
| | - Mingxia Ma
- Basic Medical Research Center, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaohui Wang
- Basic Medical Research Center, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Li Wang
- Basic Medical Research Center, Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
14
|
Peng W, Chen S, Ma J, Wei W, Lin N, Xing J, Guo W, Li H, Zhang L, Chan K, Yen A, Zhu G, Yue J. Endosomal trafficking participates in lipid droplet catabolism to maintain lipid homeostasis. Nat Commun 2025; 16:1917. [PMID: 39994216 PMCID: PMC11850777 DOI: 10.1038/s41467-025-57038-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 02/07/2025] [Indexed: 02/26/2025] Open
Abstract
The interplay between lipid droplets (LDs) and endosomes remains unknown. Here, we screen and synthesize AP1-coumarin, an LD-specific probe, by conjugating a fluorescent dye coumarin to a triazine compound AP1. AP1-coumarin labels all stages of LDs in live cells and markedly induces the accumulation of enlarged RAB5-RAB7 double-positive intermediate endosomes. The AP1-coumarin-labeled LDs contact these intermediate endosomes, with some LDs even being engulfed in them. When LD biogenesis is inhibited, the ability of AP1-coumarin to label LDs is markedly reduced, and the accumulation of enlarged intermediate endosomes is abolished. Moreover, blocking the biogenesis of LDs decreases the number of late endosomes while increasing the number of early endosomes and inhibits the endosomal trafficking of low-density lipoprotein (LDL) and transferrin. Correspondingly, interference with RAB5 or RAB7, either through knockdown or using dominant-negative mutants, inhibits LD catabolism, whereas the expression of a RAB7 constitutively active mutant accelerates LD catabolism. Additionally, CCZ1 knockdown not only induces the accumulation of intermediate endosomes but also inhibits LD catabolism. These results collectively suggest that LDs and endosomes interact and influence each other's functions, and endosomal trafficking participates in the catabolic process of LDs to maintain lipid homeostasis.
Collapse
Affiliation(s)
- Wang Peng
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, USA
- Division of Natural and Applied Sciences, Synear Molecular Biology Lab, Jiangsu Provincial University Key (Construction) Laboratory for Smart Diagnosis and Treatment of Lung Cancer, Duke Kunshan University, Kunshan, China
| | - Shu Chen
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
- Department of Chemistry, City University of Hong Kong, Hong Kong, China
| | - Jingyu Ma
- Division of Natural and Applied Sciences, Synear Molecular Biology Lab, Jiangsu Provincial University Key (Construction) Laboratory for Smart Diagnosis and Treatment of Lung Cancer, Duke Kunshan University, Kunshan, China
| | - Wenjie Wei
- Core Research Facilities, Southern University of Science and Technology, Shenzhen, China
| | - Naixin Lin
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Jinchao Xing
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Wenjing Guo
- Analysis and Testing Center, Guangzhou Institute of Biomedicine and Health (GIBH) Chinese Academy of Sciences, Guangzhou, China
| | - Heying Li
- Analysis and Testing Center, Guangzhou Institute of Biomedicine and Health (GIBH) Chinese Academy of Sciences, Guangzhou, China
| | - Liang Zhang
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Kuiming Chan
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Andrew Yen
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, USA
| | - Guangyu Zhu
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China.
- Department of Chemistry, City University of Hong Kong, Hong Kong, China.
| | - Jianbo Yue
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China.
- Division of Natural and Applied Sciences, Synear Molecular Biology Lab, Jiangsu Provincial University Key (Construction) Laboratory for Smart Diagnosis and Treatment of Lung Cancer, Duke Kunshan University, Kunshan, China.
- College of Life Sciences, Wuhan University, Wuhan, China.
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China.
| |
Collapse
|
15
|
Rahma MN, Suhandi C, Mohammed AFA, El-Rayyes A, Elamin KM, Sulastri E, Wathoni N. The Role and Advancement of Liposomes for Oral Diseases Therapy. Int J Nanomedicine 2025; 20:1865-1880. [PMID: 39975418 PMCID: PMC11837752 DOI: 10.2147/ijn.s492353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/25/2024] [Indexed: 02/21/2025] Open
Abstract
As many as 48.0% of the global population suffers from disabilities caused by oral conditions. These conditions encompass dental caries, periodontal diseases, oral cancers, and other pathologies affecting the hard and soft tissues of the oral and maxillofacial regions. Topical drug treatments in the oral cavity are often ineffective due to the short contact time, which prevents the drug from reaching optimal concentrations necessary for therapeutic effect. Conventional liposomes have several limitations, including low stability, challenges in long-term storage, and rapid clearance by the reticuloendothelial system (RES). These factors significantly reduce their effectiveness in maintaining sustained drug delivery and achieving desired therapeutic outcomes. To overcome these challenges, advanced drug delivery systems have been developed. Among these systems, liposomes have been extensively explored as nanocarriers in targeted drug delivery systems, particularly in mucosal drug delivery, due to their biocompatibility and degradability, making them promising agents for the treatment of oral diseases. To address these issues, extensive research has been conducted to modify the surface of liposomes, optimizing their efficacy, and understanding their mechanisms of action. This review article discusses the role and recent advancements of liposomes in the treatment of oral diseases, highlighting their potential to revolutionize oral health care through improved drug delivery and therapeutic outcomes.
Collapse
Affiliation(s)
- Maya Nurul Rahma
- Department of Pharmaceutics and Pharmaceutical Technology, Universitas Padjadjaran, Sumedang, West Java, 45363, Indonesia
| | - Cecep Suhandi
- Department of Pharmaceutics and Pharmaceutical Technology, Universitas Padjadjaran, Sumedang, West Java, 45363, Indonesia
| | - Ahmed F A Mohammed
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| | - Ali El-Rayyes
- Center for Scientific Research and Entrepreneurship, Northern Border University, Arar, 73213, Saudi Arabia
| | - Khaled M Elamin
- Graduated School of Pharmaceutical Science, Kumamoto University, Kumamoto, 862-0973, Japan
| | - Evi Sulastri
- Department of Pharmaceutics and Pharmaceutical Technology, Universitas Padjadjaran, Sumedang, West Java, 45363, Indonesia
- Department of Pharmacy, Faculty of Mathematics and Natural Sciences, Tadulako University, Palu, Central Sulawesi, 94118, Indonesia
| | - Nasrul Wathoni
- Department of Pharmaceutics and Pharmaceutical Technology, Universitas Padjadjaran, Sumedang, West Java, 45363, Indonesia
| |
Collapse
|
16
|
Gonzalez-Lozano MA, Schmid EW, Whelan EM, Jiang Y, Paulo JA, Walter JC, Harper JW. EndoMAP.v1, a Structural Protein Complex Landscape of Human Endosomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.07.636106. [PMID: 39975243 PMCID: PMC11839024 DOI: 10.1101/2025.02.07.636106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Early/sorting endosomes are dynamic organelles that play key roles in proteome control by triaging plasma membrane proteins for either recycling or degradation in the lysosome1,2,3. These events are coordinated by numerous transiently-associated regulatory complexes and integral membrane components that contribute to organelle identity during endosome maturation4. While a subset of the several hundred protein components and cargoes known to associate with endosomes have been studied at the biochemical and/or structural level, interaction partners and higher order molecular assemblies for many endosomal components remain unknown. Here, we combine cross-linking and native gel mass spectrometry5-8 of purified early endosomes with AlphaFold9,10 and computational analysis to create a systematic human endosomal structural interactome. We present dozens of structural models for endosomal protein pairs and higher order assemblies supported by experimental cross-links from their native subcellular context, suggesting structural mechanisms for previously reported regulatory processes. Using induced neurons, we validate two candidate complexes whose interactions are supported by crosslinks and structural predictions: TMEM230 as a subunit of ATP8/11 lipid flippases11 and TMEM9/9B as subunits of CLCN3/4/5 chloride-proton antiporters12. This resource and its accompanying structural network viewer provide an experimental framework for understanding organellar structural interactomes and large-scale validation of structural predictions.
Collapse
Affiliation(s)
- Miguel A Gonzalez-Lozano
- Department of Cell Biology, Harvard Medical School, Boston MA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Ernst W Schmid
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston MA, USA
| | - Enya Miguel Whelan
- Department of Cell Biology, Harvard Medical School, Boston MA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Yizhi Jiang
- Department of Cell Biology, Harvard Medical School, Boston MA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Initiative in Trafficking and Neurogeneration, Department of Cell Biology, Harvard Medical School, Boston MA, USA
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston MA, USA
| | - Johannes C Walter
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - J Wade Harper
- Department of Cell Biology, Harvard Medical School, Boston MA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| |
Collapse
|
17
|
Lučin P, Mahmutefendić Lučin H. The Cell Biologist Potential of Cytomegalovirus to Solve Biogenesis and Maintenance of the Membrane Recycling System. Biomedicines 2025; 13:326. [PMID: 40002739 PMCID: PMC11853475 DOI: 10.3390/biomedicines13020326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 12/27/2024] [Accepted: 01/16/2025] [Indexed: 02/27/2025] Open
Abstract
Cytomegalovirus (CMV) is an important pathogen that extensively remodels the nucleus and cytosol of an infected cell to establish a productive infection [...].
Collapse
Affiliation(s)
- Pero Lučin
- Department of Physiology, Immunology and Pathophysiology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia;
- Department of Nursing, University Center Varaždin, University North, Jurja Križanića 31b, 42000 Varaždin, Croatia
| | - Hana Mahmutefendić Lučin
- Department of Physiology, Immunology and Pathophysiology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia;
- Department of Nursing, University Center Varaždin, University North, Jurja Križanića 31b, 42000 Varaždin, Croatia
| |
Collapse
|
18
|
Baroi R, Ahmad Reshi H, Maddika S. Mitochondrial phosphatase PPTC7 promotes EGFR recycling by facilitating VPS4A endosomal localization. J Cell Sci 2025; 138:jcs263676. [PMID: 39776116 DOI: 10.1242/jcs.263676] [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: 06/06/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025] Open
Abstract
PPTC7 is a mitochondrial phosphatase that is essential for mitochondrial biogenesis, metabolism, protein content maintenance and transport. Although the mitochondrial roles of PPTC7 are well characterized, its roles outside the mitochondria are unclear. Here we identified a non-mitochondrial role for PPTC7 in regulating epidermal growth factor receptor (EGFR) trafficking. PPTC7 interacts with and dephosphorylates VPS4A, a crucial ESCRT and multivesicular body-associated protein. We found that PPTC7-mediated dephosphorylation of VPS4A at serine 335 is required for VPS4A stability and its early endosomal localization. Either loss of PPTC7 or presence of constitutively phosphorylated VPS4A led to defective recycling of EGFR, thus leading to EGFR re-routing to lysosomes for degradation. Further, we demonstrate that PPTC7-VPS4A-dependent EGFR recycling promotes the AKT signaling pathway, thus enhancing cell proliferation and migration. Overall, our studies unveil an important mechanism where the PPTC7-VPS4A complex orchestrates an endosomal switch to promote EGFR recycling.
Collapse
Affiliation(s)
- Rahul Baroi
- Laboratory of Cell Death & Cell Survival, Centre for DNA Fingerprinting and Diagnostics (CDFD), Uppal, Hyderabad 500039, India
- Graduate studies, Regional Centre for Biotechnology, Faridabad 121001, India
| | - Hilal Ahmad Reshi
- Laboratory of Cell Death & Cell Survival, Centre for DNA Fingerprinting and Diagnostics (CDFD), Uppal, Hyderabad 500039, India
- Graduate studies, Regional Centre for Biotechnology, Faridabad 121001, India
| | - SubbaReddy Maddika
- Laboratory of Cell Death & Cell Survival, Centre for DNA Fingerprinting and Diagnostics (CDFD), Uppal, Hyderabad 500039, India
| |
Collapse
|
19
|
D'Souza-Schorey C, Stahl PD. Resolving the two-body problem: A postulated role for the V0 sector of the V0V1-ATPase in exosome biogenesis and multivesicular body fate. Mol Biol Cell 2025; 36:pe1. [PMID: 39705591 PMCID: PMC11742106 DOI: 10.1091/mbc.e24-09-0412] [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: 09/18/2024] [Revised: 11/04/2024] [Accepted: 11/22/2024] [Indexed: 12/22/2024] Open
Abstract
Because the discovery of the multivesicular body (MVB) as the origin of secreted vesicles or exosomes, the question arose and still looms-what distinguishes an MVB destined for fusion with the plasma membrane (EXO-MVB) facilitating exosome release from an MVB involved in transport of content to the lysosome (LYSO-MVB). Do they have independent origins? Hence, the two-body problem. We hypothesize that a key to this conundrum is the membrane spanning V0 sector of the proton pump, V0V1-ATPase. The V0V1-ATPase participates in the acidification of intracellular compartments, although V0 can function separately from V1 and different V0 isoforms are endowed with membrane binding capabilities that allow the V0V1-ATPase to selectively localize to different endocytic compartments including early and late endosomes and lysosomes. We propose that V0, in collaboration with cholesterol and phosphoinositides, plays a central role in the early endosome as a nucleation center to direct the de novo assembly of an EXO-MVB scaffold. The EXO-MVB scaffold may play multiple roles-operating as an assembly platform, participating in membrane fission as well as providing downstream navigational queues necessary for exosome secretion. Thus, V0 may represent an influential nexus, a starting point, in exosome biogenesis.
Collapse
Affiliation(s)
| | - Philip D. Stahl
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
20
|
Da Graça J, Delevoye C, Morel E. Morphodynamical adaptation of the endolysosomal system to stress. FEBS J 2025; 292:248-260. [PMID: 38706230 PMCID: PMC11734881 DOI: 10.1111/febs.17154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/28/2024] [Accepted: 04/25/2024] [Indexed: 05/07/2024]
Abstract
In eukaryotes, the spatiotemporal control of endolysosomal organelles is central to the maintenance of homeostasis. By providing an interface between the cytoplasm and external environment, the endolysosomal system is placed at the forefront of the response to a wide range of stresses faced by cells. Endosomes are equipped with a dedicated set of membrane-associated proteins that ensure endosomal functions as well as crosstalk with the secretory or the autophagy pathways. Morphodynamical processes operate through local spatialization of subdomains, enabling specific remodeling and membrane contact capabilities. Consequently, the plasticity of endolysosomal organelles can be considered a robust and flexible tool exploited by cells to cope with homeostatic deviations. In this review, we provide insights into how the cellular responses to various stresses (osmotic, UV, nutrient deprivation, or pathogen infections) rely on the adaptation of the endolysosomal system morphodynamics.
Collapse
Affiliation(s)
- Juliane Da Graça
- Université Paris Cité, INSERM UMR‐S1151, CNRS UMR‐S8253, Institut Necker Enfants MaladesFrance
| | - Cédric Delevoye
- Université Paris Cité, INSERM UMR‐S1151, CNRS UMR‐S8253, Institut Necker Enfants MaladesFrance
- Institut Curie, PSL Research University, CNRS, UMR144, Structure and Membrane CompartmentsParisFrance
| | - Etienne Morel
- Université Paris Cité, INSERM UMR‐S1151, CNRS UMR‐S8253, Institut Necker Enfants MaladesFrance
| |
Collapse
|
21
|
Jain K, Prelic S, Hansson BS, Wicher D. Expression of Drosophila melanogaster V-ATPases in Olfactory Sensillum Support Cells. INSECTS 2024; 15:1016. [PMID: 39769617 PMCID: PMC11676623 DOI: 10.3390/insects15121016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025]
Abstract
V-ATPases are ubiquitous and evolutionarily conserved rotatory proton pumps, which are crucial for maintaining various biological functions. Previous investigations have shown that a V-ATPase is present in the support cells of moth trichoid sensilla and influences their olfactory sensory neuron performance. Generally, V-ATPases are thought to regulate the pH value within the sensillum lymph, and aid K+ homeostasis within the sensillum. This, in turn, could influence various mechanisms involved within the support cells, like maintaining the receptor membrane potential (receptor current), nutrient and ion transport, odorant solubility, and various signaling mechanisms. In this study, we identify V-ATPase expression and localization in the Drosophila melanogaster antenna using bioinformatics and immunohistochemistry. Elucidating an olfactory V-ATPase function will improve our current understanding of how support cells contribute to Drosophila's sense of smell.
Collapse
Affiliation(s)
| | | | | | - Dieter Wicher
- Department Evolutionary Neuroethology, Max Planck Institute for Chemical Ecology, 07745 Jena, Germany; (K.J.); (S.P.); (B.S.H.)
| |
Collapse
|
22
|
Gillett DA, Tigro H, Wang Y, Suo Z. FMR1 Disorders: Basics of Biology and Therapeutics in Development. Cells 2024; 13:2100. [PMID: 39768191 PMCID: PMC11674747 DOI: 10.3390/cells13242100] [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: 10/24/2024] [Revised: 12/04/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Fragile X Syndrome (FXS) presents with a constellation of phenotypes, including trouble regulating emotion and aggressive behaviors, disordered sleep, intellectual impairments, and atypical physical development. Genetic study of the X chromosome revealed that substantial repeat expansion of the 5' end of the gene fragile X messenger ribonucleoprotein 1 (FMR1) promoted DNA methylation and, consequently, silenced expression of FMR1. Further analysis proved that shorter repeat expansions in FMR1 also manifested in disease at later stages in life. Treatment and therapy options do exist, but they only manage symptoms. Up to now, no cure for FMR1 disorders exists. In this review, we aim to provide an overview of FMR1 biology and the latest research focused on developing therapeutic interventions that can potentially prevent and/or reverse FXS.
Collapse
Affiliation(s)
| | | | | | - Zucai Suo
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
23
|
Kobayashi J, Higashisaka K, Muranaka M, Xie Y, Okuno W, Haga Y, Tsutsumi Y. Localization of silica nanoparticles to lysosome causes lysosomal dysfunction in JEG-3 cells. Biochem Biophys Res Commun 2024; 736:150488. [PMID: 39111054 DOI: 10.1016/j.bbrc.2024.150488] [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: 07/23/2024] [Accepted: 07/31/2024] [Indexed: 11/10/2024]
Abstract
Nanoparticles have useful functions due to the characteristics conferred on them by an increase in their specific surface area, and they have already been put into practical use in products in various industrial fields. Although exposure to nanoparticles in daily life is unavoidable for pregnant women, studies that evaluate the toxicity of nanoparticles in pregnant women are lacking. To redress this, we have focused on the placenta and have previously revealed that nanoparticles can show placental toxicity. However, there is still little knowledge regarding the behavior of nanoparticles within placental cells, which would enable us to understand their mode of action. Here, we tried to clarify the intracellular localization of silica nanoparticles in placental cells and how this affects placental toxicity. We analyzed the uptake of silica nanoparticles with a diameter of 10 nm (nSP10) into JEG-3 cells, a human choriocarcinoma cell line. Flow cytometry analysis showed that nSP10 labelled with red fluorescence were taken up into JEG-3 cells, and that pre-treatment with the endocytosis inhibitor cytochalasin D inhibited their uptake, suggesting that nSP10 are taken up into JEG-3 cells by the endocytic pathway. Moreover, confocal microscopy revealed that nSP10 are prominently localized in lysosomes. Staining with LysoTracker showed that nSP10 treatment increased the acidic compartment of JEG-3 cells, suggesting lysosome accumulation and swelling. These results indicate that nSP10 taken into placental cells are transferred to lysosomes and may cause lysosomal dysfunction.
Collapse
Affiliation(s)
- Jundai Kobayashi
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Kazuma Higashisaka
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan; School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan; Institute for Advanced Co-Creation Studies, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Mizuki Muranaka
- School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Yankun Xie
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Wakako Okuno
- School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Yuya Haga
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan; School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Yasuo Tsutsumi
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan; School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan; Global Center for Medical Engineering and Informatics, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan; Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
24
|
Guajardo-Contreras GI, Abdalla AL, Chen A, Niu M, Beauchamp E, Berthiaume LG, Cochrane AW, Mouland AJ. HIV-1 N-myristoylation-dependent hijacking of late endosomes/lysosomes to drive Gag assembly in macrophages. J Cell Sci 2024; 137:jcs263588. [PMID: 39439384 DOI: 10.1242/jcs.263588] [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: 10/03/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024] Open
Abstract
Macrophages represent an important viral reservoir in HIV-1-infected individuals. Different from T cells, HIV-1 assembly in macrophages occurs at intracellular compartments termed virus-containing compartments (VCCs). Our previous research in HeLa cells - in which assembly resembles that found in infected T cells - suggested that late endosomes/lysosomes (LELs) play a role in HIV-1 trafficking towards its assembly sites. However, the role of LELs during assembly at VCCs is not fully understood. Herein, we used the HIV-1-inducible cell line THP-1 GagZip as a model to study HIV-1 Gag intracellular trafficking and assembly in macrophages. We demonstrated LEL involvement at VCCs using various microscopy techniques and biochemical approaches. Live-cell imaging revealed that HIV-1 repositions LELs towards the plasma membrane and modulates their motility. We showed that Arl8b-mediated LEL repositioning is not responsible for Gag trafficking to VCCs. Additionally, the inhibition of myristoylation by PCLX-001 decreased the presence of Gag on endosomes and inhibited VCC formation in both the THP-1 cell line and primary macrophages. In conclusion, we present evidence supporting the idea that HIV-1 manipulates the LEL trajectory to guide Gag to VCCs in an N-myristoylation-dependent manner.
Collapse
Affiliation(s)
- Gabriel I Guajardo-Contreras
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada
- Lady Davis Institute at the Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Ana L Abdalla
- Lady Davis Institute at the Jewish General Hospital, Montreal, QC H3T 1E2, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada
| | - Alex Chen
- The Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Meijuan Niu
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada
| | | | - Luc G Berthiaume
- Pacylex Pharmaceuticals Inc., Edmonton, AB T5J 4P6, Canada
- Department of Cell Biology, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Alan W Cochrane
- The Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Andrew J Mouland
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada
| |
Collapse
|
25
|
Larose A, Miller CCJ, Mórotz GM. The lemur tail kinase family in neuronal function and disfunction in neurodegenerative diseases. Cell Mol Life Sci 2024; 81:447. [PMID: 39520508 PMCID: PMC11550312 DOI: 10.1007/s00018-024-05480-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 09/12/2024] [Accepted: 10/12/2024] [Indexed: 11/16/2024]
Abstract
The complex neuronal architecture and the long distance of synapses from the cell body require precisely orchestrated axonal and dendritic transport processes to support key neuronal functions including synaptic signalling, learning and memory formation. Protein phosphorylation is a major regulator of both intracellular transport and synaptic functions. Some kinases and phosphatases such as cyclin dependent kinase-5 (cdk5)/p35, glycogen synthase kinase-3β (GSK3β) and protein phosphatase-1 (PP1) are strongly involved in these processes. A primary pathological hallmark of neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis/frontotemporal dementia, is synaptic degeneration together with disrupted intracellular transport. One attractive possibility is that alterations to key kinases and phosphatases may underlie both synaptic and axonal transport damages. The brain enriched lemur tail kinases (LMTKs, formerly known as lemur tyrosine kinases) are involved in intracellular transport and synaptic functions, and are also centrally placed in cdk5/p35, GSK3β and PP1 signalling pathways. Loss of LMTKs is documented in major neurodegenerative diseases and thus can contribute to pathological defects in these disorders. However, whilst function of their signalling partners became clearer in modulating both synaptic signalling and axonal transport progress has only recently been made around LMTKs. In this review, we describe this progress with a special focus on intracellular transport, synaptic functions and neurodegenerative diseases.
Collapse
Affiliation(s)
- Angelique Larose
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4, Budapest, H-1089, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
| | - Christopher C J Miller
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 125 Coldharbour Lane Camberwell, London, SE5 9RX, UK.
| | - Gábor M Mórotz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4, Budapest, H-1089, Hungary.
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
26
|
Ishemgulova A, Mukhamedova L, Trebichalská Z, Rájecká V, Payne P, Šmerdová L, Moravcová J, Hrebík D, Buchta D, Škubník K, Füzik T, Vaňáčová Š, Nováček J, Plevka P. Endosome rupture enables enteroviruses from the family Picornaviridae to infect cells. Commun Biol 2024; 7:1465. [PMID: 39511383 PMCID: PMC11543853 DOI: 10.1038/s42003-024-07147-9] [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: 04/02/2024] [Accepted: 10/24/2024] [Indexed: 11/15/2024] Open
Abstract
Membrane penetration by non-enveloped viruses is diverse and generally not well understood. Enteroviruses, one of the largest groups of non-enveloped viruses, cause diseases ranging from the common cold to life-threatening encephalitis. Enteroviruses enter cells by receptor-mediated endocytosis. However, how enterovirus particles or RNA genomes cross the endosome membrane into the cytoplasm remains unknown. Here we used cryo-electron tomography of infected cells to show that endosomes containing enteroviruses deform, rupture, and release the virus particles into the cytoplasm. Blocking endosome acidification with bafilomycin A1 reduced the number of particles that released their genomes, but did not prevent them from reaching the cytoplasm. Inhibiting post-endocytic membrane remodeling with wiskostatin promoted abortive enterovirus genome release in endosomes. The rupture of endosomes also occurs in control cells and after the endocytosis of very low-density lipoprotein. In summary, our results show that cellular membrane remodeling disrupts enterovirus-containing endosomes and thus releases the virus particles into the cytoplasm to initiate infection. Since the studied enteroviruses employ different receptors for cell entry but are delivered into the cytoplasm by cell-mediated endosome disruption, it is likely that most if not all enteroviruses, and probably numerous other viruses from the family Picornaviridae, can utilize endosome rupture to infect cells.
Collapse
Affiliation(s)
- Aygul Ishemgulova
- Central European Institute of Technology, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic.
| | - Liya Mukhamedova
- Central European Institute of Technology, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - Zuzana Trebichalská
- Central European Institute of Technology, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Veronika Rájecká
- Central European Institute of Technology, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - Pavel Payne
- Central European Institute of Technology, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - Lenka Šmerdová
- Central European Institute of Technology, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - Jana Moravcová
- Central European Institute of Technology, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - Dominik Hrebík
- Central European Institute of Technology, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - David Buchta
- Central European Institute of Technology, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - Karel Škubník
- Central European Institute of Technology, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - Tibor Füzik
- Central European Institute of Technology, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - Štěpánka Vaňáčová
- Central European Institute of Technology, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - Jiří Nováček
- Central European Institute of Technology, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - Pavel Plevka
- Central European Institute of Technology, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic.
| |
Collapse
|
27
|
Frisby D, Murakonda AB, Ashraf B, Dhawan K, Almeida-Souza L, Naslavsky N, Caplan S. Endosomal actin branching, fission, and receptor recycling require FCHSD2 recruitment by MICAL-L1. Mol Biol Cell 2024; 35:ar144. [PMID: 39382837 PMCID: PMC11617095 DOI: 10.1091/mbc.e24-07-0324] [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: 07/29/2024] [Revised: 09/25/2024] [Accepted: 10/04/2024] [Indexed: 10/10/2024] Open
Abstract
Endosome fission is required for the release of carrier vesicles and the recycling of receptors to the plasma membrane. Early events in endosome budding and fission rely on actin branching to constrict the endosomal membrane, ultimately leading to nucleotide hydrolysis and enzymatic fission. However, our current understanding of this process is limited, particularly regarding the coordination between the early and late steps of endosomal fission. Here we have identified a novel interaction between the endosomal scaffolding protein, MICAL-L1, and the human homologue of the Drosophila Nervous Wreck (Nwk) protein, FCH and double SH3 domains protein 2 (FCHSD2). We demonstrate that MICAL-L1 recruits FCHSD2 to the endosomal membrane, where it is required for ARP2/3-mediated generation of branched actin, endosome fission and receptor recycling to the plasma membrane. Because MICAL-L1 first recruits FCHSD2 to the endosomal membrane, and is subsequently responsible for recruitment of the ATPase and fission protein EHD1 to endosomes, our findings support a model in which MICAL-L1 orchestrates endosomal fission by connecting between the early actin-driven and subsequent nucleotide hydrolysis steps of the process.
Collapse
Affiliation(s)
- Devin Frisby
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Ajay B. Murakonda
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Bazella Ashraf
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Kanika Dhawan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla 92093, CA
| | - Leonardo Almeida-Souza
- Helsinki Institute of Life Science, University of Helsinki, Helsinki 00790, Finland
- Institute of Biotechnology, University of Helsinki, Helsinki 00790, Finland
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki 00790, Finland
| | - Naava Naslavsky
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Steve Caplan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198
| |
Collapse
|
28
|
Endo A, Komada M, Yoshida Y. Ubiquitin-mediated endosomal stress: A novel organelle stress of early endosomes that initiates cellular signaling pathways: USP8 serves as a gatekeeper of ubiquitin-mediated endosomal stress to counteract the activation of cellular signaling pathways. Bioessays 2024; 46:e2400127. [PMID: 39194376 DOI: 10.1002/bies.202400127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/08/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024]
Abstract
Cells utilize diverse organelles to maintain homeostasis and to respond to extracellular stimuli. Recently, multifaceted aspects of organelle stress caused by various factors have been emerging. The endosome is an essential organelle, functioning as the central hub for membrane trafficking in cooperation with the ubiquitin system. However, knowledge regarding endosomal stress, which refers to organelle stress of the endosome, is currently limited. We recently revealed ubiquitin-mediated endosomal stress of early endosomes (EEs) and its responsive signaling pathways. These findings shed light on the relevance of ubiquitin-mediated endosomal stress to physiological and pathological processes. Here, we present a hypothesis that ubiquitin-mediated endosomal stress may have significant roles in biological contexts and that ubiquitin-specific protease 8 is a key regulator of ubiquitin clearance from EEs.
Collapse
Affiliation(s)
- Akinori Endo
- Laboratory of Protein Metabolism, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Masayuki Komada
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Yukiko Yoshida
- Laboratory of Protein Metabolism, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| |
Collapse
|
29
|
Wang J, Liu P, Zhang R, Xing B, Chen G, Han L, Yu J. VASH2 enhances KIF3C-mediated EGFR-endosomal recycling to promote aggression and chemoresistance of lung squamous cell carcinoma by increasing tubulin detyrosination. Cell Death Dis 2024; 15:772. [PMID: 39443476 PMCID: PMC11499603 DOI: 10.1038/s41419-024-07155-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: 06/03/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024]
Abstract
Lung squamous cell carcinoma (LUSC) is associated with high mortality and has few therapeutic options. Chemotherapy remains the main treatment for LUSC patients, but multi-drug resistance has become the dominant challenge in the failure of chemotherapy in various cancers. Therefore, the effective therapeutic strategy for LUSC patients is an urgent unmet need. Here, we found vasohibin-2 (VASH2) was a prognostic biomarker for LUSC patients, and VASH2 promoted the malignant biological behaviors of LUSC cells and chemoresistance by increasing the detyrosination of α-tubulin. The high level of detyrosinated-tubulin was negatively associated with patient prognosis. Blocking the tubulin carboxypeptidase (TCP) activity of VASH2 inhibited the xenograft tumor growth and improved the treatment efficacy of paclitaxel in vivo. Results revealed that VASH2-induced increase in tubulin detyrosination boosted the binding of kinesin family member 3C (KIF3C) to microtubules and enhanced KIF3C-dependent endosomal recycling of EGFR, leading to the prolonged activation of PI3K/Akt/mTOR signaling. This study demonstrated that VASH2 was not only a prognostic biomarker but also a promising therapeutic target in LUSC, which offers a novel insight that combination of chemotherapy and EpoY, a TCP inhibitor, may be a promising treatment strategy for LUSC patients.
Collapse
Affiliation(s)
- Jing Wang
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, 300202, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, 300202, Tianjin, China
| | - Pengpeng Liu
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, 300202, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, 300202, Tianjin, China
| | - Rui Zhang
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, 300202, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, 300202, Tianjin, China
| | - Biyuan Xing
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, 300202, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, 300202, Tianjin, China
| | - Guidong Chen
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, 300202, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, 300202, Tianjin, China
| | - Lei Han
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, 300202, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, 300202, Tianjin, China
| | - Jinpu Yu
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, 300202, Tianjin, China.
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, 300202, Tianjin, China.
| |
Collapse
|
30
|
Hebchen DM, Schröder K. Redox Signaling in Endosomes Using the Example of EGF Receptors: A Graphical Review. Antioxidants (Basel) 2024; 13:1215. [PMID: 39456468 PMCID: PMC11504029 DOI: 10.3390/antiox13101215] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Early endosomes represent first-line sorting compartments or even organelles for internalized molecules. They enable the transport of molecules or ligands to other compartments of the cell, such as lysosomes, for degradation or recycle them back to the membrane by various mechanisms. Moreover, early endosomes function as signaling and scaffolding platforms to initiate or prolong distinct signaling pathways. Accordingly, early endosomes have to be recognized as either part of a degradation or recycling pathway. The physical proximity of many ligand-binding receptors with other membrane-bound proteins or complexes such as NADPH oxidases may result in an interaction of second messengers, like reactive oxygen species (ROS) and early endosomes, that promote the correct recognition of individual early endosomes. In fact, redoxosomes comprise an endosomal subsection of signaling endosomes. One example of such potential interaction is epidermal growth factor receptor (EGFR) signaling. Here we summarize recent findings on EGFR signaling as a well-studied example for receptor trafficking and trans-activation and illustrate the interplay between cellular and endosomal ROS.
Collapse
Affiliation(s)
| | - Katrin Schröder
- Institute of Physiology, Medical Faculty, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany;
| |
Collapse
|
31
|
Zhang MQ, Li JR, Yang L, Peng ZG, Wu S, Zhang JP. ATG10S promotes IFNL1 expression and autophagic degradation of multiple viral proteins mediated by IFNL1. Autophagy 2024; 20:2238-2254. [PMID: 38842055 PMCID: PMC11423677 DOI: 10.1080/15548627.2024.2361580] [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: 10/07/2023] [Revised: 05/21/2024] [Accepted: 05/24/2024] [Indexed: 06/07/2024] Open
Abstract
ATG10S is a newly discovered subtype of the autophagy protein ATG10. It promotes complete macroautophagy/autophagy, degrades multiple viral proteins, and increases the expression of type III interferons. Here, we aimed to investigate the mechanism of ATG10S cooperation with IFNL1 to degrade viral proteins from different viruses. Using western blot, immunoprecipitation (IP), tandem sensor RFP-GFP-LC3B and in situ proximity ligation assays, we showed that exogenous recombinant ATG10S protein (rHsATG10S) could enter into cells through clathrin, and ATG10S combined with ATG7 with IFNL1 assistance to facilitate ATG12-ATG5 conjugation, thereby contributing to the autophagosome formation in multiple cell lines containing different virions or viral proteins. The results of DNA IP and luciferase assays also showed that ATG10S was able to directly bind to a core motif (CAAGGG) within a binding site of transcription factor ZNF460 on the IFNL1 promoter, by which IFNL1 transcription was activated. These results clarified that ATG10S promoted autophagosome formation with the assistance of IFNL1 to ensure autophagy flux and autophagic degradation of multiple viral proteins and that ATG10S could also act as a novel transcription factor to promote IFNL1 gene expression. Importantly, this study further explored the antiviral mechanism of ATG10S interaction with type III interferon and provided a theoretical basis for the development of ATG10S into a new broad-spectrum antiviral protein drug.Abbreviation: ATG: autophagy related; ATG10S: the shorter isoform of autophagy-related 10; CC50: half cytotoxicity concentration; CCV: clathrin-coated transport vesicle; CLTC: clathrin heavy chain; CM: core motif; co-IP: co-immunoprecipitation; CPZ: chlorpromazine; ER: endoplasmic reticulum; HCV: hepatitis C virus; HBV: hepatitis B virus; HsCoV-OC43: Human coronavirus OC43; IFN: interferon; PLA: proximity ligation assay; rHsATG10S: recombinant human ATG10S protein; RLU: relative light unit; SQSTM1: sequestosome 1; ZNF: zinc finger protein.
Collapse
Affiliation(s)
- Miao-Qing Zhang
- Key Laboratory of Biotechnology of Antibiotics, the National Health Commission (NHC), Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jian-Rui Li
- Key Laboratory of Biotechnology of Antibiotics, the National Health Commission (NHC), Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lu Yang
- Key Laboratory of Biotechnology of Antibiotics, the National Health Commission (NHC), Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zong-Gen Peng
- Key Laboratory of Biotechnology of Antibiotics, the National Health Commission (NHC), Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuo Wu
- Key Laboratory of Biotechnology of Antibiotics, the National Health Commission (NHC), Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing-Pu Zhang
- Key Laboratory of Biotechnology of Antibiotics, the National Health Commission (NHC), Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
32
|
Milne SM, Edeen PT, Fay DS. TAT-1, a phosphatidylserine flippase, affects molting and regulates membrane trafficking in the epidermis of C. elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.15.613099. [PMID: 39314363 PMCID: PMC11419146 DOI: 10.1101/2024.09.15.613099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Membrane trafficking is a conserved process required for the movement and distribution of proteins and other macromolecules within cells. The Caenorhabditis elegans NIMA-related kinases NEKL-2 (human NEK8/9) and NEKL-3 (human NEK6/7) are conserved regulators of membrane trafficking and are required for the completion of molting. We used a genetic approach to identify reduction-of-function mutations in tat-1 that suppress nekl -associated molting defects. tat-1 encodes the C. elegans ortholog of mammalian ATP8A1/2, a phosphatidylserine (PS) flippase that promotes the asymmetric distribution of PS to the cytosolic leaflet of lipid membrane bilayers. CHAT-1 (human CDC50), a conserved chaperone, was required for the correct localization of TAT-1, and chat-1 inhibition strongly suppressed nekl defects. Using a PS sensor, we found that TAT-1 was required for the normal localization of PS at apical endosomes and that loss of TAT-1 led to aberrant endosomal morphologies. Consistent with this, TAT-1 localized to early endosomes and to recycling endosomes marked with RME-1, the C. elegans ortholog of the human EPS15 homology (EH) domain-containing protein, EHD1. TAT-1, PS biosynthesis, and the PS-binding protein RFIP-2 (human RAB11-FIP2) were all required for the normal localization of RME-1 to apical endosomes. Consistent with these proteins functioning together, inhibition of RFIP-2 or RME-1 led to the partial suppression of nekl molting defects, as did the inhibition of PS biosynthesis. Using the auxin-inducible degron system, we found that depletion of NEKL-2 or NEKL-3 led to defects in RME-1 localization and that a reduction in TAT-1 function partially restored RME-1 localization in NEKL-3-depleted cells. ARTICLE SUMMARY Endocytosis is an essential process required for the movement of proteins and lipids within cells. NEKL-2 and NEKL-3, two evolutionarily conserved proteins in the nematode Caenorhabditis elegans , are important regulators of endocytosis. In the current study, the authors describe a new functional link between the NEKLs and several proteins with known roles in endocytosis including TAT-1, a conserved enzyme that moves lipids between the bilayers of cellular membranes. As previous work implicated NEKLs in developmental defects and cancer, the present study can provide new insights into how the misregulation of endocytosis affects human health and disease.
Collapse
|
33
|
Gu M, Carvalho EJ, Read KA, Nardo DP, Riley JL. Rab5 Overcomes CAR T Cell Dysfunction Induced by Tumor-Mediated CAR Capture. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.26.605334. [PMID: 39211164 PMCID: PMC11361039 DOI: 10.1101/2024.07.26.605334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Continuous interaction between chimeric antigen receptor (CAR) T cell (CART) and tumors often result in CART dysfunction and tumor escape. We observed that tumors can take up CAR molecules, leaving CARTs without surface-expressed CARs and thus unable to kill tumors after prolonged exposure. Overexpression of Rab5 resulted in augmented clathrin-independent endocytosis, preventing loss of surface-expressed CARs, and enhanced CART activity. Interestingly, we observed membrane protrusions on the CART cell surface which disappeared after multiple tumor challenges. Rab5 maintained these protrusions after repeated tumor engagements and their presence correlated with effective tumor clearance, suggesting a link between endocytosis, membrane protrusions, and cytolytic activity. In vivo , Rab5-expressing CARTs demonstrated improved activity and were able to clear an otherwise refractory mesothelin-expressing solid cancer in humanized mice by maintaining CAR surface expression within the tumor. Thus, pairing Rab5 with CAR expression could improve the clinical efficacy of CART therapy. Highlights "CAR-jacking" occurs when surface CAR is internalized by target tumor cells.Rab5 overexpression prevents "CAR-jacking" and enhances CART function.Rab5 promotes CAR endocytic recycling and maintains membrane protrusions.Rab5-expressing CARTs exhibit enhanced therapeutic efficacy against solid tumors.
Collapse
|
34
|
Kourkoulou A, Martzoukou O, Fischer R, Amillis S. A type II phosphatidylinositol-4-kinase coordinates sorting of cargo polarizing by endocytic recycling. Commun Biol 2024; 7:855. [PMID: 38997419 PMCID: PMC11245547 DOI: 10.1038/s42003-024-06553-3] [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: 10/30/2023] [Accepted: 07/04/2024] [Indexed: 07/14/2024] Open
Abstract
Depending on their phosphorylation status, derivatives of phosphatidylinositol play important roles in vesicle identity, recognition and intracellular trafficking processes. In eukaryotic cells, phosphatidylinositol-4 phosphate pools generated by specific kinases are key determinants of the conventional secretion pathways. Earlier work in yeast has classified phosphatidylinositol-4 kinases in two types, Stt4p and Pik1p belonging to type III and Lsb6p to type II, with distinct cellular localizations and functions. Eurotiomycetes appear to lack Pik1p homologues. In Aspergillus nidulans, unlike homologues in other fungi, AnLsb6 is associated to late Golgi membranes and when heterologously overexpressed, it compensates for the thermosensitive phenotype in a Saccharomyces cerevisiae pik1 mutant, whereas its depletion leads to disorganization of Golgi-associated PHOSBP-labelled membranes, that tend to aggregate dependent on functional Rab5 GTPases. Evidence provided herein, indicates that the single type II phosphatidylinositol-4 kinase AnLsb6 is the main contributor for decorating secretory vesicles with relevant phosphatidylinositol-phosphate species, which navigate essential cargoes following the route of apical polarization via endocytic recycling.
Collapse
Affiliation(s)
- Anezia Kourkoulou
- National and Kapodistrian University of Athens, Department of Biology, Athens, Hellas, Greece
| | - Olga Martzoukou
- National and Kapodistrian University of Athens, Department of Biology, Athens, Hellas, Greece
| | - Reinhard Fischer
- Karlsruhe Institute of Technology - South Campus, Institute for Applied Biosciences, Department of Microbiology, Karlsruhe, Germany
| | - Sotiris Amillis
- National and Kapodistrian University of Athens, Department of Biology, Athens, Hellas, Greece.
- Karlsruhe Institute of Technology - South Campus, Institute for Applied Biosciences, Department of Microbiology, Karlsruhe, Germany.
| |
Collapse
|
35
|
Ansari I, Mandal A, Kansal K, Walling P, Khan S, Aijaz S. The C-terminal proline-rich repeats of Enteropathogenic E. coli effector EspF are sufficient for the depletion of tight junction membrane proteins and interactions with early and recycling endosomes. Gut Pathog 2024; 16:36. [PMID: 38972985 PMCID: PMC11229284 DOI: 10.1186/s13099-024-00626-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 06/28/2024] [Indexed: 07/09/2024] Open
Abstract
BACKGROUND Enteropathogenic E. coli (EPEC) causes acute infantile diarrhea accounting for significant morbidity and mortality in developing countries. EPEC uses a type three secretion system to translocate more than twenty effectors into the host intestinal cells. At least four of these effectors, namely EspF, Map, EspG1/G2 and NleA, are reported to disrupt the intestinal tight junction barrier. We have reported earlier that the expression of EspF and Map in MDCK cells causes the depletion of the TJ membrane proteins and compromises the integrity of the intestinal barrier. In the present study, we have examined the role of the proline-rich repeats (PRRs) within the C-terminus of EspF in the depletion of the tight junction membrane proteins and identified key endocytosis markers that interact with EspF via these repeats. RESULTS We generated mutant EspF proteins which lacked one or more proline-rich repeats (PRRs) from the N-terminus of EspF and examined the effect of their expression on the cellular localization of tight junction membrane proteins. In lysates derived from cells expressing the mutant EspF proteins, we found that the C-terminal PRRs of EspF are sufficient to cause the depletion of TJ membrane proteins. Pull-down assays revealed that the PRRs mediate interactions with the TJ adaptor proteins ZO-1 and ZO-2 as well as with the proteins involved in endocytosis such as caveolin-1, Rab5A and Rab11. CONCLUSIONS Our study demonstrates the direct role of the proline-rich repeats of EspF in the depletion of the TJ membrane proteins and a possible involvement of the PRRs in the endocytosis of host proteins. New therapeutic strategies can target these PRR domains to prevent intestinal barrier dysfunction in EPEC infections.
Collapse
Affiliation(s)
- Imran Ansari
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Anupam Mandal
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Kritika Kansal
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Pangertoshi Walling
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Sumbul Khan
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Saima Aijaz
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
36
|
Palmulli R, Couty M, Piontek MC, Ponnaiah M, Dingli F, Verweij FJ, Charrin S, Tantucci M, Sasidharan S, Rubinstein E, Kontush A, Loew D, Lhomme M, Roos WH, Raposo G, van Niel G. CD63 sorts cholesterol into endosomes for storage and distribution via exosomes. Nat Cell Biol 2024; 26:1093-1109. [PMID: 38886558 DOI: 10.1038/s41556-024-01432-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 05/01/2024] [Indexed: 06/20/2024]
Abstract
Extracellular vesicles such as exosomes are now recognized as key players in intercellular communication. Their role is influenced by the specific repertoires of proteins and lipids, which are enriched when they are generated as intraluminal vesicles (ILVs) in multivesicular endosomes. Here we report that a key component of small extracellular vesicles, the tetraspanin CD63, sorts cholesterol to ILVs, generating a pool that can be mobilized by the NPC1/2 complex, and exported via exosomes to recipient cells. In the absence of CD63, cholesterol is retrieved from the endosomes by actin-dependent vesicular transport, placing CD63 and cholesterol at the centre of a balance between inward and outward budding of endomembranes. These results establish CD63 as a lipid-sorting mechanism within endosomes, and show that ILVs and exosomes are alternative providers of cholesterol.
Collapse
Affiliation(s)
- Roberta Palmulli
- Institut Curie, PSL Research University, CNRS, UMR144, 26 rue d'Ulm, 75248, Paris Cedex 05, France
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université de Paris, Paris, France
| | - Mickaël Couty
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université de Paris, Paris, France
- CRCI2NA, Nantes Université, Inserm UMR1307, CNRS UMR6075, Université d'Angers, Nantes, France
| | - Melissa C Piontek
- Moleculaire Biofysica, Zernike Instituut, Rijksuniversiteit Groningen, Groningen, The Netherlands
| | - Maharajah Ponnaiah
- Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN, ICAN OMICS and ICAN I/O), F-75013, Paris, France
| | - Florent Dingli
- CurieCoreTech Spectrométrie de Masse Protéomique, Institut Curie, PSL Research University, Paris, France
| | - Frederik J Verweij
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université de Paris, Paris, France
| | - Stéphanie Charrin
- Centre d'Immunologie et des Maladies Infectieuses (CIMI), Sorbonne Université, Inserm, Paris, France
| | - Matteo Tantucci
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université de Paris, Paris, France
| | - Sajitha Sasidharan
- Moleculaire Biofysica, Zernike Instituut, Rijksuniversiteit Groningen, Groningen, The Netherlands
| | - Eric Rubinstein
- Centre d'Immunologie et des Maladies Infectieuses (CIMI), Sorbonne Université, Inserm, Paris, France
| | - Anatol Kontush
- ICAN, National Institute for Health and Medical Research, Paris, France
| | - Damarys Loew
- CurieCoreTech Spectrométrie de Masse Protéomique, Institut Curie, PSL Research University, Paris, France
| | - Marie Lhomme
- Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN, ICAN OMICS and ICAN I/O), F-75013, Paris, France
| | - Wouter H Roos
- Moleculaire Biofysica, Zernike Instituut, Rijksuniversiteit Groningen, Groningen, The Netherlands
| | - Graça Raposo
- Institut Curie, PSL Research University, CNRS, UMR144, 26 rue d'Ulm, 75248, Paris Cedex 05, France
- Institut Curie, PSL Research University, CNRS, UMR144, Cell and Tissue Imaging Facility (PICT-IBiSA), 26, rue d'Ulm, 75248, Paris Cedex 05, France
| | - Guillaume van Niel
- Institut Curie, PSL Research University, CNRS, UMR144, 26 rue d'Ulm, 75248, Paris Cedex 05, France.
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université de Paris, Paris, France.
- CRCI2NA, Nantes Université, Inserm UMR1307, CNRS UMR6075, Université d'Angers, Nantes, France.
- GHU-Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, Paris, France.
| |
Collapse
|
37
|
Frisby D, Murakonda AB, Ashraf B, Dhawan K, Almeida-Souza L, Naslavsky N, Caplan S. Endosomal actin branching, fission and receptor recycling require FCHSD2 recruitment by MICAL-L1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.601011. [PMID: 38979241 PMCID: PMC11230409 DOI: 10.1101/2024.06.27.601011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Endosome fission is required for the release of carrier vesicles and the recycling of receptors to the plasma membrane. Early events in endosome budding and fission rely on actin branching to constrict the endosomal membrane, ultimately leading to nucleotide hydrolysis and enzymatic fission. However, our current understanding of this process is limited, particularly regarding the coordination between the early and late steps of endosomal fission. Here we have identified a novel interaction between the endosomal scaffolding protein, MICAL-L1, and the human homolog of the Drosophila Nervous Wreck (Nwk) protein, FCH and double SH3 domains protein 2 (FCHSD2). We demonstrate that MICAL-L1 recruits FCHSD2 to the endosomal membrane, where it is required for ARP2/3-mediated generation of branched actin, endosome fission and receptor recycling to the plasma membrane. Since MICAL-L1 first recruits FCHSD2 to the endosomal membrane, and is subsequently responsible for recruitment of the ATPase and fission protein EHD1 to endosomes, our findings support a model in which MICAL-L1 orchestrates endosomal fission by connecting between the early actin-driven and subsequent nucleotide hydrolysis steps of the process.
Collapse
|
38
|
Moghassemi S, Dadashzadeh A, Sousa MJ, Vlieghe H, Yang J, León-Félix CM, Amorim CA. Extracellular vesicles in nanomedicine and regenerative medicine: A review over the last decade. Bioact Mater 2024; 36:126-156. [PMID: 38450204 PMCID: PMC10915394 DOI: 10.1016/j.bioactmat.2024.02.021] [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: 12/01/2023] [Revised: 02/15/2024] [Accepted: 02/19/2024] [Indexed: 03/08/2024] Open
Abstract
Small extracellular vesicles (sEVs) are known to be secreted by a vast majority of cells. These sEVs, specifically exosomes, induce specific cell-to-cell interactions and can activate signaling pathways in recipient cells through fusion or interaction. These nanovesicles possess several desirable properties, making them ideal for regenerative medicine and nanomedicine applications. These properties include exceptional stability, biocompatibility, wide biodistribution, and minimal immunogenicity. However, the practical utilization of sEVs, particularly in clinical settings and at a large scale, is hindered by the expensive procedures required for their isolation, limited circulation lifetime, and suboptimal targeting capacity. Despite these challenges, sEVs have demonstrated a remarkable ability to accommodate various cargoes and have found extensive applications in the biomedical sciences. To overcome the limitations of sEVs and broaden their potential applications, researchers should strive to deepen their understanding of current isolation, loading, and characterization techniques. Additionally, acquiring fundamental knowledge about sEVs origins and employing state-of-the-art methodologies in nanomedicine and regenerative medicine can expand the sEVs research scope. This review provides a comprehensive overview of state-of-the-art exosome-based strategies in diverse nanomedicine domains, encompassing cancer therapy, immunotherapy, and biomarker applications. Furthermore, we emphasize the immense potential of exosomes in regenerative medicine.
Collapse
Affiliation(s)
- Saeid Moghassemi
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Arezoo Dadashzadeh
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Maria João Sousa
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Hanne Vlieghe
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Jie Yang
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Cecibel María León-Félix
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Christiani A. Amorim
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
39
|
Schott MB, Rozeveld CN, Bhatt S, Crossman B, Krueger EW, Weller SG, Rasineni K, Casey CA, McNiven MA. Ethanol disrupts hepatocellular lipophagy by altering Rab5-centric LD-lysosome trafficking. Hepatol Commun 2024; 8:e0446. [PMID: 38780316 PMCID: PMC11124685 DOI: 10.1097/hc9.0000000000000446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 02/23/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Previous reports suggest that lipid droplets (LDs) in the hepatocyte can be catabolized by a direct engulfment from nearby endolysosomes (microlipophagy). Further, it is likely that this process is compromised by chronic ethanol (EtOH) exposure leading to hepatic steatosis. This study investigates the hepatocellular machinery supporting microlipophagy and EtOH-induced alterations in this process with a focus on the small, endosome-associated, GTPase Rab5. METHODS AND RESULTS Here we report that this small Ras-related GTPase is a resident component of LDs, and its activity is important for hepatocellular LD-lysosome proximity and physical interactions. We find that Rab5 siRNA knockdown causes an accumulation of LDs in hepatocytes by inhibiting lysosome dependent LD catabolism. Importantly, Rab5 appears to support this process by mediating the recruitment of early endosomal and or multivesicular body compartments to the LD surface before lysosome fusion. Interestingly, while wild-type or a constituently active GTPase form (Q79L) of Rab5 supports LD-lysosome transport, this process is markedly reduced in cells expressing a GTPase dead (S34N) Rab5 protein or in hepatocytes exposed to chronic EtOH. CONCLUSIONS These findings support the novel premise of an early endosomal/multivesicular body intermediate compartment on the LD surface that provides a "docking" site for lysosomal trafficking, not unlike the process that occurs during the hepatocellular degradation of endocytosed ligands that is also known to be compromised by EtOH exposure.
Collapse
Affiliation(s)
- Micah B. Schott
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Cody N. Rozeveld
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Saumya Bhatt
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Bridget Crossman
- Department of Biochemistry and Molecular Biology, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Eugene W. Krueger
- Department of Biochemistry and Molecular Biology, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Shaun G. Weller
- Department of Biochemistry and Molecular Biology, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Karuna Rasineni
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Veterans’ Affairs, VA-Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA
| | - Carol A. Casey
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Veterans’ Affairs, VA-Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA
| | - Mark A. McNiven
- Department of Biochemistry and Molecular Biology, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
40
|
Ma G, Crowley AR, Heyndrickx L, Rogiers I, Parthoens E, Van Santbergen J, Ober RJ, Bobkov V, de Haard H, Ulrichts P, Hofman E, Louagie E, Balbino B, Ward ES. Differential effects of FcRn antagonists on the subcellular trafficking of FcRn and albumin. JCI Insight 2024; 9:e176166. [PMID: 38713534 PMCID: PMC11141909 DOI: 10.1172/jci.insight.176166] [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: 10/04/2023] [Accepted: 04/10/2024] [Indexed: 05/09/2024] Open
Abstract
The homeostasis of IgG is maintained by the neonatal Fc receptor, FcRn. Consequently, antagonism of FcRn to reduce endogenous IgG levels is an emerging strategy for treating antibody-mediated autoimmune disorders using either FcRn-specific antibodies or an engineered Fc fragment. For certain FcRn-specific antibodies, this approach has resulted in reductions in the levels of serum albumin, the other major ligand transported by FcRn. Cellular and molecular analyses of a panel of FcRn antagonists have been carried out to elucidate the mechanisms leading to their differential effects on albumin homeostasis. These analyses have identified 2 processes underlying decreases in albumin levels during FcRn blockade: increased degradation of FcRn and competition between antagonist and albumin for FcRn binding. These findings have potential implications for the design of drugs to modulate FcRn function.
Collapse
Affiliation(s)
- Guanglong Ma
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Andrew R. Crowley
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | | | | | - Eef Parthoens
- VIB BioImaging Core, Center for Inflammation Research, Ghent, Belgium
| | | | - Raimund J. Ober
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | | | | | | | | | | | | | - E. Sally Ward
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
41
|
Migliano SM, Schultz SW, Wenzel EM, Takáts S, Liu D, Mørk S, Tan KW, Rusten TE, Raiborg C, Stenmark H. Removal of hypersignaling endosomes by simaphagy. Autophagy 2024; 20:769-791. [PMID: 37840274 PMCID: PMC11062362 DOI: 10.1080/15548627.2023.2267958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 09/14/2023] [Accepted: 10/01/2023] [Indexed: 10/17/2023] Open
Abstract
Activated transmembrane receptors continue to signal following endocytosis and are only silenced upon ESCRT-mediated internalization of the receptors into intralumenal vesicles (ILVs) of the endosomes. Accordingly, endosomes with dysfunctional receptor internalization into ILVs can cause sustained receptor signaling which has been implicated in cancer progression. Here, we describe a surveillance mechanism that allows cells to detect and clear physically intact endosomes with aberrant receptor accumulation and elevated signaling. Proximity biotinylation and proteomics analyses of ESCRT-0 defective endosomes revealed a strong enrichment of the ubiquitin-binding macroautophagy/autophagy receptors SQSTM1 and NBR1, a phenotype that was confirmed in cell culture and fly tissue. Live cell microscopy demonstrated that loss of the ESCRT-0 subunit HGS/HRS or the ESCRT-I subunit VPS37 led to high levels of ubiquitinated and phosphorylated receptors on endosomes. This was accompanied by dynamic recruitment of NBR1 and SQSTM1 as well as proteins involved in autophagy initiation and autophagosome biogenesis. Light microscopy and electron tomography revealed that endosomes with intact limiting membrane, but aberrant receptor downregulation were engulfed by phagophores. Inhibition of autophagy caused increased intra- and intercellular signaling and directed cell migration. We conclude that dysfunctional endosomes are surveyed and cleared by an autophagic process, simaphagy, which serves as a failsafe mechanism in signal termination.Abbreviations: AKT: AKT serine/threonine kinase; APEX2: apurinic/apyrimidinic endodoexyribonuclease 2; ctrl: control; EEA1: early endosome antigen 1; EGF: epidermal growth factor; EGFR: epidermal growth factor receptor; ESCRT: endosomal sorting complex required for transport; GFP: green fluorescent protein; HGS/HRS: hepatocyte growth factor-regulated tyrosine kinase substrate; IF: immunofluorescence; ILV: intralumenal vesicle; KO: knockout; LIR: LC3-interacting region; LLOMe: L-leucyl-L-leucine methyl ester (hydrochloride); MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MAPK1/ERK2: mitogen-activated protein kinase 1; MAPK3/ERK1: mitogen-activated protein kinase 3; NBR1: NBR1 autophagy cargo receptor; PAG10: Protein A-conjugated 10-nm gold; RB1CC1/FIP200: RB1 inducible coiled-coil 1; siRNA: small interfering RNA; SQSTM1: sequestosome 1; TUB: Tubulin; UBA: ubiquitin-associated; ULK1: unc-51 like autophagy activating kinase 1; VCL: Vinculin; VPS37: VPS37 subunit of ESCRT-I; WB: western blot; WT: wild-type.
Collapse
Affiliation(s)
- Simona M. Migliano
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Sebastian W. Schultz
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Eva M. Wenzel
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Szabolcs Takáts
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, Budapest, Hungary
| | - Dan Liu
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Silje Mørk
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Kia Wee Tan
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Medical Cell Biology, University of Uppsala, Uppsala, Sweden
| | - Tor Erik Rusten
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Camilla Raiborg
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Harald Stenmark
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
42
|
Abstract
Lipid nanoparticles (LNPs) have recently emerged as a powerful and versatile clinically approved platform for nucleic acid delivery, specifically for mRNA vaccines. A major bottleneck in the field is the release of mRNA-LNPs from the endosomal pathways into the cytosol of cells where they can execute their encoded functions. The data regarding the mechanism of these endosomal escape processes are limited and contradicting. Despite extensive research, there is no consensus regarding the compartment of escape, the cause of the inefficient escape and are currently lacking a robust method to detect the escape. Here, we review the currently known mechanisms of endosomal escape and the available methods to study this process. We critically discuss the limitations and challenges of these methods and the possibilities to overcome these challenges. We propose that the development of currently lacking robust, quantitative high-throughput techniques to study endosomal escape is timely and essential. A better understanding of this process will enable better RNA-LNP designs with improved efficiency to unlock new therapeutic modalities.
Collapse
Affiliation(s)
- Sushmita Chatterjee
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Edo Kon
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Preeti Sharma
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Dan Peer
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
43
|
El Safadi D, Mokhtari A, Krejbich M, Lagrave A, Hirigoyen U, Lebeau G, Viranaicken W, Krejbich-Trotot P. Exosome-Mediated Antigen Delivery: Unveiling Novel Strategies in Viral Infection Control and Vaccine Design. Vaccines (Basel) 2024; 12:280. [PMID: 38543914 PMCID: PMC10974137 DOI: 10.3390/vaccines12030280] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 01/03/2025] Open
Abstract
Exosomes are small subtypes of extracellular vesicles (EVs) naturally released by different types of cells into their environment. Their physiological roles appear to be multiple, yet many aspects of their biological activities remain to be understood. These vesicles can transport and deliver a variety of cargoes and may serve as unconventional secretory vesicles. Thus, they play a crucial role as important vectors for intercellular communication and the maintenance of homeostasis. Exosome production and content can vary under several stresses or modifications in the cell microenvironment, influencing cellular responses and stimulating immunity. During infectious processes, exosomes are described as double-edged swords, displaying both beneficial and detrimental effects. Owing to their tractability, the analysis of EVs from multiple biofluids has become a booming tool for monitoring various pathologies, from infectious to cancerous origins. In this review, we present an overview of exosome features and discuss their particular and ambiguous functions in infectious contexts. We then focus on their properties as diagnostic or therapeutic tools. In this regard, we explore the capacity of exosomes to vectorize immunogenic viral antigens and their function in mounting adaptive immune responses. As exosomes provide interesting platforms for antigen presentation, we further review the available data on exosome engineering, which enables peptides of interest to be exposed at their surface. In the light of all these data, exosomes are emerging as promising avenues for vaccine strategies.
Collapse
Affiliation(s)
- Daed El Safadi
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD, Plateforme Technologique CYROI, 94791 Sainte Clotilde, La Réunion, France; (D.E.S.); (A.M.); (G.L.); (W.V.)
| | - Alexandre Mokhtari
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD, Plateforme Technologique CYROI, 94791 Sainte Clotilde, La Réunion, France; (D.E.S.); (A.M.); (G.L.); (W.V.)
| | - Morgane Krejbich
- Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes-Angers, CRCI2NA, INSERM U1307, CNRS UMR 6075, Université de Nantes, Université d’Angers, 8 Quai Moncousu, P.O. Box 70721, Cedex 1, 44007 Nantes, France; (M.K.); (U.H.)
| | - Alisé Lagrave
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD, Plateforme Technologique CYROI, 94791 Sainte Clotilde, La Réunion, France; (D.E.S.); (A.M.); (G.L.); (W.V.)
- National Reference Center for Arboviruses, Institut Pasteur de la Guyane, Cayenne 97300, French Guiana
| | - Ugo Hirigoyen
- Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes-Angers, CRCI2NA, INSERM U1307, CNRS UMR 6075, Université de Nantes, Université d’Angers, 8 Quai Moncousu, P.O. Box 70721, Cedex 1, 44007 Nantes, France; (M.K.); (U.H.)
| | - Grégorie Lebeau
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD, Plateforme Technologique CYROI, 94791 Sainte Clotilde, La Réunion, France; (D.E.S.); (A.M.); (G.L.); (W.V.)
- Unité Mixte Diabète Athérothrombose Réunion Océan Indien (DéTROI), Université de La Réunion, INSERM U1188, Campus Santé de Terre Sainte, 97410 Saint-Pierre, La Réunion, France
| | - Wildriss Viranaicken
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD, Plateforme Technologique CYROI, 94791 Sainte Clotilde, La Réunion, France; (D.E.S.); (A.M.); (G.L.); (W.V.)
- Unité Mixte Diabète Athérothrombose Réunion Océan Indien (DéTROI), Université de La Réunion, INSERM U1188, Campus Santé de Terre Sainte, 97410 Saint-Pierre, La Réunion, France
| | - Pascale Krejbich-Trotot
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD, Plateforme Technologique CYROI, 94791 Sainte Clotilde, La Réunion, France; (D.E.S.); (A.M.); (G.L.); (W.V.)
| |
Collapse
|
44
|
Guo H, Zhou C, Zheng M, Zhang J, Wu H, He Q, Ding L, Yang B. Insights into the role of derailed endocytic trafficking pathway in cancer: From the perspective of cancer hallmarks. Pharmacol Res 2024; 201:107084. [PMID: 38295915 DOI: 10.1016/j.phrs.2024.107084] [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: 10/19/2023] [Revised: 01/12/2024] [Accepted: 01/25/2024] [Indexed: 02/06/2024]
Abstract
The endocytic trafficking pathway is a highly organized cellular program responsible for the regulation of membrane components and uptake of extracellular substances. Molecules internalized into the cell through endocytosis will be sorted for degradation or recycled back to membrane, which is determined by a series of sorting events. Many receptors, enzymes, and transporters on the membrane are strictly regulated by endocytic trafficking process, and thus the endocytic pathway has a profound effect on cellular homeostasis. However, the endocytic trafficking process is typically dysregulated in cancers, which leads to the aberrant retention of receptor tyrosine kinases and immunosuppressive molecules on cell membrane, the loss of adhesion protein, as well as excessive uptake of nutrients. Therefore, hijacking endocytic trafficking pathway is an important approach for tumor cells to obtain advantages of proliferation and invasion, and to evade immune attack. Here, we summarize how dysregulated endocytic trafficking process triggers tumorigenesis and progression from the perspective of several typical cancer hallmarks. The impact of endocytic trafficking pathway to cancer therapy efficacy is also discussed.
Collapse
Affiliation(s)
- Hongjie Guo
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chen Zhou
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Mingming Zheng
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jie Zhang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Honghai Wu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiaojun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; The Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou 310018, China; Cancer Center of Zhejiang University, Hangzhou 310058, China
| | - Ling Ding
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Nanhu Brain-computer Interface Institute, Hangzhou 311100, China.
| | - Bo Yang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; School of Medicine, Hangzhou City University, Hangzhou 310015, China; The Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou 310018, China.
| |
Collapse
|
45
|
Ouyang Y, Nauwynck HJ. Molecular basis for the different PCV2 susceptibility of T-lymphoblasts in Landrace and Piétrain pigs. Vet Res 2024; 55:22. [PMID: 38374131 PMCID: PMC10875804 DOI: 10.1186/s13567-024-01275-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 01/29/2024] [Indexed: 02/21/2024] Open
Abstract
Clinically, Landrace pigs are more susceptible to porcine circovirus-associated diseases (PCVADs) than Piétrain pigs. We previously found that porcine circovirus type 2 (PCV2) can infect T-lymphoblasts. The present study examined the replication kinetics of six PCV2 strains in the lymphoblasts of Landrace and Piétrain pigs. The results showed that T-lymphoblasts from Landrace pigs are much more susceptible to PCV2 infection than those from Piétrain pigs. In addition, PCV2 replication was strain-dependent. PCV2 binding to T-lymphoblasts was partially mediated by chondroitin sulfate (CS) and dermatan sulfate (DS). Phosphacan, an effective internalization mediator in monocytes that contains several CS chains, was also demonstrated to be involved in PCV2 internalization. Viral binding and internalization were not different between the two breeds, however, the subsequent step, the disassembly was. Although inhibition of serine proteases blocked PCV2 replication in both Landrace and Piétrain pigs, this only occurred at a neutral pH in Piétrain pigs, whereas this occurred also at a low pH in Landrace. This suggested that more proteases can cleave PCV2 in Landrace lymphoblasts than in Piétrain lymphoblasts, explaining the better replication. Through co-localization studies of viral particles with endo-lysosomal markers, and quantitative analysis of organelle sizes during viral internalization, it was observed that PCV2 may exhibit a higher propensity for viral escape from late endosomes in Landrace pigs (smaller) compared to Piétrain pigs. These results provide new understandings of the different PCV2 susceptibility in Landrace and Piétrain pigs.
Collapse
Affiliation(s)
- Yueling Ouyang
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium.
| | - Hans J Nauwynck
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| |
Collapse
|
46
|
Xie S, Naslavsky N, Caplan S. Emerging insights into CP110 removal during early steps of ciliogenesis. J Cell Sci 2024; 137:jcs261579. [PMID: 38415788 PMCID: PMC10941660 DOI: 10.1242/jcs.261579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024] Open
Abstract
The primary cilium is an antenna-like projection from the plasma membrane that serves as a sensor of the extracellular environment and a crucial signaling hub. Primary cilia are generated in most mammalian cells, and their physiological significance is highlighted by the large number of severe developmental disorders or ciliopathies that occur when primary ciliogenesis is impaired. Primary ciliogenesis is a tightly regulated process, and a central early regulatory step is the removal of a key mother centriole capping protein, CP110 (also known as CCP110). This uncapping allows vesicles docked on the distal appendages of the mother centriole to fuse to form a ciliary vesicle, which is bent into a ciliary sheath as the microtubule-based axoneme grows and extends from the mother centriole. When the mother centriole migrates toward the plasma membrane, the ciliary sheath fuses with the plasma membrane to form the primary cilium. In this Review, we outline key early steps of primary ciliogenesis, focusing on several novel mechanisms for removal of CP110. We also highlight examples of ciliopathies caused by genetic variants that encode key proteins involved in the early steps of ciliogenesis.
Collapse
Affiliation(s)
- Shuwei Xie
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Naava Naslavsky
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Steve Caplan
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
47
|
Zhu H, Wang D, Ye Z, Huang L, Wei W, Chan KM, Zhang R, Zhang L, Yue J. The temporal association of CapZ with early endosomes regulates endosomal trafficking and viral entry into host cells. BMC Biol 2024; 22:12. [PMID: 38273307 PMCID: PMC10809671 DOI: 10.1186/s12915-024-01819-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 01/09/2024] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND Many viruses enter host cells by hijacking endosomal trafficking. CapZ, a canonical actin capping protein, participates in endosomal trafficking, yet its precise role in endocytosis and virus infection remains elusive. RESULTS Here, we showed that CapZ was transiently associated with early endosomes (EEs) and was subsequently released from the matured EEs after the fusion of two EEs, which was facilitated by PI(3)P to PI(3,5)P2 conversion. Vacuolin-1 (a triazine compound) stabilized CapZ at EEs and thus blocked the transition of EEs to late endosomes (LEs). Likewise, artificially tethering CapZ to EEs via a rapamycin-induced protein-protein interaction system blocked the early-to-late endosome transition. Remarkably, CapZ knockout or artificially tethering CapZ to EEs via rapamycin significantly inhibited flaviviruses, e.g., Zika virus (ZIKV) and dengue virus (DENV), or beta-coronavirus, e.g., murine hepatitis virus (MHV), infection by preventing the escape of RNA genome from endocytic vesicles. CONCLUSIONS These results indicate that the temporal association of CapZ with EEs facilitates early-to-late endosome transition (physiologically) and the release of the viral genome from endocytic vesicles (pathologically).
Collapse
Affiliation(s)
- Huazhang Zhu
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
| | - Dawei Wang
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
| | - Zuodong Ye
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
| | - Lihong Huang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Wenjie Wei
- Research Core Facilities, Southern University of Science and Technology of China, Shenzhen, 518052, China
| | - Kui Ming Chan
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Rongxin Zhang
- Laboratory of Immunology and Inflammation, Institute of Basic Medical Sciences and Department of Biotechnology, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Liang Zhang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Jianbo Yue
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China.
- Divison of Natural and Applied Sciences, Synear Molecular Biology Lab, Duke Kunshan University, Kunshan, China.
| |
Collapse
|
48
|
Gandek TB, van der Koog L, Nagelkerke A. A Comparison of Cellular Uptake Mechanisms, Delivery Efficacy, and Intracellular Fate between Liposomes and Extracellular Vesicles. Adv Healthc Mater 2023; 12:e2300319. [PMID: 37384827 PMCID: PMC11469107 DOI: 10.1002/adhm.202300319] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 06/21/2023] [Accepted: 06/21/2023] [Indexed: 07/01/2023]
Abstract
A key aspect for successful drug delivery via lipid-based nanoparticles is their internalization in target cells. Two prominent examples of such drug delivery systems are artificial phospholipid-based carriers, such as liposomes, and their biological counterparts, the extracellular vesicles (EVs). Despite a wealth of literature, it remains unclear which mechanisms precisely orchestrate nanoparticle-mediated cargo delivery to recipient cells and the subsequent intracellular fate of therapeutic cargo. In this review, internalization mechanisms involved in the uptake of liposomes and EVs by recipient cells are evaluated, also exploring their intracellular fate after intracellular trafficking. Opportunities are highlighted to tweak these internalization mechanisms and intracellular fates to enhance the therapeutic efficacy of these drug delivery systems. Overall, literature to date shows that both liposomes and EVs are predominantly internalized through classical endocytosis mechanisms, sharing a common fate: accumulation inside lysosomes. Studies tackling the differences between liposomes and EVs, with respect to cellular uptake, intracellular delivery and therapy efficacy, remain scarce, despite its importance for the selection of an appropriate drug delivery system. In addition, further exploration of functionalization strategies of both liposomes and EVs represents an important avenue to pursue in order to control internalization and fate, thereby improving therapeutic efficacy.
Collapse
Affiliation(s)
- Timea B. Gandek
- Pharmaceutical AnalysisGroningen Research Institute of PharmacyUniversity of GroningenP.O. Box 196, XB20Groningen9700 ADThe Netherlands
| | - Luke van der Koog
- Molecular PharmacologyGroningen Research Institute of PharmacyUniversity of GroningenP.O. Box 196, XB10Groningen9700 ADThe Netherlands
| | - Anika Nagelkerke
- Pharmaceutical AnalysisGroningen Research Institute of PharmacyUniversity of GroningenP.O. Box 196, XB20Groningen9700 ADThe Netherlands
| |
Collapse
|
49
|
Soha SA, Santhireswaran A, Huq S, Casimir-Powell J, Jenkins N, Hodgson GK, Sugiyama M, Antonescu CN, Impellizzeri S, Botelho RJ. Improved imaging and preservation of lysosome dynamics using silver nanoparticle-enhanced fluorescence. Mol Biol Cell 2023; 34:ar96. [PMID: 37405751 PMCID: PMC10551705 DOI: 10.1091/mbc.e22-06-0200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 07/06/2023] Open
Abstract
The dynamics of living cells can be studied by live-cell fluorescence microscopy. However, this requires the use of excessive light energy to obtain good signal-to-noise ratio, which can then photobleach fluorochromes, and more worrisomely, lead to phototoxicity. Upon light excitation, noble metal nanoparticles such as silver nanoparticles (AgNPs) generate plasmons, which can then amplify excitation in direct proximity of the nanoparticle's surface and couple to the oscillating dipole of nearby radiating fluorophores, modifying their rate of emission and thus, enhancing their fluorescence. Here, we show that AgNPs fed to cells to accumulate within lysosomes enhanced the fluorescence of lysosome-targeted Alexa488-conjugated dextran, BODIPY-cholesterol, and DQ-BSA. Moreover, AgNP increased the fluorescence of GFP fused to the cytosolic tail of LAMP1, showing that metal enhanced fluorescence can occur across the lysosomal membrane. The inclusion of AgNPs in lysosomes did not disturb lysosomal properties such as lysosomal pH, degradative capacity, autophagy and autophagic flux, and membrane integrity, though AgNP seemed to increase basal lysosome tubulation. Importantly, by using AgNP, we could track lysosome motility with reduced laser power without damaging and altering lysosome dynamics. Overall, AgNP-enhanced fluorescence may be a useful tool to study the dynamics of the endo-lysosomal pathway while minimizing phototoxicity.
Collapse
Affiliation(s)
- Sumaiya A. Soha
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario, Canada, M5B 2K3
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada, M5B 2K3
| | - Araniy Santhireswaran
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada, M5B 2K3
| | - Saaimatul Huq
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada, M5B 2K3
| | - Jayde Casimir-Powell
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario, Canada, M5B 2K3
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada, M5B 2K3
| | - Nicala Jenkins
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario, Canada, M5B 2K3
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada, M5B 2K3
| | - Gregory K. Hodgson
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada, M5B 2K3
| | - Michael Sugiyama
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada, M5B 2K3
| | - Costin N. Antonescu
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario, Canada, M5B 2K3
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada, M5B 2K3
| | - Stefania Impellizzeri
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario, Canada, M5B 2K3
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada, M5B 2K3
| | - Roberto J. Botelho
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario, Canada, M5B 2K3
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada, M5B 2K3
| |
Collapse
|
50
|
Naslavsky N, Caplan S. Advances and challenges in understanding endosomal sorting and fission. FEBS J 2023; 290:4187-4195. [PMID: 36413090 PMCID: PMC10200825 DOI: 10.1111/febs.16687] [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: 09/20/2022] [Revised: 11/04/2022] [Accepted: 11/21/2022] [Indexed: 11/23/2022]
Abstract
Endosomes play crucial roles in the cell, serving as focal and 'triage' points for internalized lipids and receptors. As such, endosomes are a critical branching point that determines whether receptors are sorted for degradation or recycling. This Viewpoint aims to highlight recent advances in endosome research, including key endosomal functions such as sorting and fission. Moreover, the Viewpoint addresses key technical and conceptual challenges in studying endosomes.
Collapse
Affiliation(s)
- Naava Naslavsky
- Department of Biochemistry & Molecular Biology and Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Steve Caplan
- Department of Biochemistry & Molecular Biology and Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|