1
|
Fukuda A, Sato K, Fujimori C, Yamashita T, Takeuchi A, Ohuchi H, Umatani C, Kanda S. Direct photoreception by pituitary endocrine cells regulates hormone release and pigmentation. Science 2025; 387:43-48. [PMID: 39745961 DOI: 10.1126/science.adj9687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/10/2024] [Accepted: 11/01/2024] [Indexed: 01/04/2025]
Abstract
The recent discovery of nonvisual photoreceptors in various organs has raised expectations for uncovering their roles and underlying mechanisms. In this work, we identified a previously unrecognized hormone-releasing mechanism in the pituitary of the Japanese rice fish (medaka) induced by light. Ca2+ imaging analysis revealed that melanotrophs, a type of pituitary endocrine cell that secretes melanocyte-stimulating hormone, robustly increase the concentration of intracellular Ca2+ during short-wavelength light exposure. Moreover, we identified Opn5m as the key molecule that drives this response. Knocking out opn5m attenuated melanogenesis by reducing tyrosinase expression in the skin. Our findings suggest a mechanism in which direct reception of short-wavelength light by pituitary melanotrophs triggers a pathway that might contribute to protection from ultraviolet radiation in medaka.
Collapse
Affiliation(s)
- Ayaka Fukuda
- Department of Marine Bioscience, Atmosphere and Ocean Research Institute, The University of Tokyo, Kashiwa, Japan
| | - Keita Sato
- Department of Cytology and Histology, Faculty of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Chika Fujimori
- Department of Marine Bioscience, Atmosphere and Ocean Research Institute, The University of Tokyo, Kashiwa, Japan
| | - Takahiro Yamashita
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto, Japan
| | - Atsuko Takeuchi
- Division of Analytical Laboratory, Kobe Pharmaceutical University, Kobe, Japan
| | - Hideyo Ohuchi
- Department of Cytology and Histology, Faculty of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Chie Umatani
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Shinji Kanda
- Department of Marine Bioscience, Atmosphere and Ocean Research Institute, The University of Tokyo, Kashiwa, Japan
| |
Collapse
|
2
|
Alves AC, Bissig C, Mayer A. FRAP Assay to Trace Lipid Mixing of the Inner and Outer Leaflet of Yeast Vacuoles: Assessing the Fusion State in Live Cells. Methods Mol Biol 2025; 2887:197-206. [PMID: 39806156 DOI: 10.1007/978-1-0716-4314-3_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Fluorescence recovery after photobleaching (FRAP) can be employed to investigate membrane lipid mixing of vacuoles in live budding yeast cells and distinguish the fused, hemi-fused or non-fused states of these organelles under physiological conditions. Here, we describe a protocol for labeling the outer and inner leaflets of vacuoles in live cells that allow to detect hemifusion intermediates and, thus, identify components necessary for fusion pore opening.
Collapse
Affiliation(s)
- Ana Catarina Alves
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Christin Bissig
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Andreas Mayer
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland.
| |
Collapse
|
3
|
Cheng KJ, Shi J, Pogorelov TV, Capponi S. Investigating the Bromoform Membrane Interactions Using Atomistic Simulations and Machine Learning: Implications for Climate Change Mitigation. J Phys Chem B 2024; 128:12493-12506. [PMID: 39641917 DOI: 10.1021/acs.jpcb.4c04930] [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: 12/07/2024]
Abstract
Methane emissions from livestock contribute to global warming. Seaweeds used as food additive offer a promising emission mitigation strategy because seaweeds are enriched in bromoform─a methanogenesis inhibitor. Therefore, understanding bromoform storage and production in seaweeds and particularly in a cell-like environment is crucial. As a first step toward this aim, we present an atomistic description of bromoform dynamics, diffusion, and aggregation in the presence of lipid membranes. Using all-atom molecular dynamics simulations with customized CHARMM-formatted bromoform force field files, we investigate the interactions of bromoform and lipid bilayer across various concentrations. Bromoform penetrates membranes and at high concentrations forms aggregates outside the membrane without affecting membrane thickness or lipid tail order. Aggregates outside the membrane influence the membrane curvature. Within the membrane, bromoform preferentially localizes in the membrane hydrophobic core and diffuses the slowest along the membrane normal. Employing general local-atomic descriptors and unsupervised machine learning, we demonstrate the similarity of bromoform local structures between the liquid and aggregated forms.
Collapse
Affiliation(s)
- Kevin J Cheng
- Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801 United States
- IBM Accelerated Discovery and Cellular Engineering, IBM Almaden Research Center, San Jose, California 95120 United States
- NSF Center for Cellular Construction, University of California in San Francisco, San Francisco, California 94158 United States
| | - Jie Shi
- IBM Accelerated Discovery and Cellular Engineering, IBM Almaden Research Center, San Jose, California 95120 United States
- NSF Center for Cellular Construction, University of California in San Francisco, San Francisco, California 94158 United States
| | - Taras V Pogorelov
- Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801 United States
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- School of Chemical Sciences, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- National Center for Supercomputing Applications, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Sara Capponi
- IBM Accelerated Discovery and Cellular Engineering, IBM Almaden Research Center, San Jose, California 95120 United States
- NSF Center for Cellular Construction, University of California in San Francisco, San Francisco, California 94158 United States
| |
Collapse
|
4
|
Pavlov RV, Akimov SA, Dashinimaev EB, Bashkirov PV. Boosting Lipofection Efficiency Through Enhanced Membrane Fusion Mechanisms. Int J Mol Sci 2024; 25:13540. [PMID: 39769303 PMCID: PMC11677079 DOI: 10.3390/ijms252413540] [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: 11/22/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Gene transfection is a fundamental technique in the fields of biological research and therapeutic innovation. Due to their biocompatibility and membrane-mimetic properties, lipid vectors serve as essential tools in transfection. The successful delivery of genetic material into the cytoplasm is contingent upon the fusion of the vector and cellular membranes, which enables hydrophilic polynucleic acids to traverse the hydrophobic barriers of two intervening membranes. This review examines the critical role of membrane fusion in lipofection efficiency, with a particular focus on the molecular mechanisms that govern lipoplex-membrane interactions. This analysis will examine the key challenges inherent to the fusion process, from achieving initial membrane proximity to facilitating final content release through membrane remodeling. In contrast to viral vectors, which utilize specialized fusion proteins, lipid vectors necessitate a strategic formulation and environmental optimization to enhance their fusogenicity. This review discusses recent advances in vector design and fusion-promoting strategies, emphasizing their potential to improve gene delivery yield. It highlights the importance of understanding lipoplex-membrane fusion mechanisms for developing next-generation delivery systems and emphasizes the need for continued fundamental research to advance lipid-mediated transfection technology.
Collapse
Affiliation(s)
- Rais V. Pavlov
- Research Institute for Systems Biology and Medicine, 18 Nauchniy Proezd, Moscow 117246, Russia
| | - Sergey A. Akimov
- Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Sciences, 31/4 Leninskiy Prospekt, Moscow 119071, Russia;
| | - Erdem B. Dashinimaev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow 117997, Russia;
| | - Pavel V. Bashkirov
- Research Institute for Systems Biology and Medicine, 18 Nauchniy Proezd, Moscow 117246, Russia
| |
Collapse
|
5
|
Zheng L, Wang S. Recent advances in solid-state nuclear magnetic resonance studies on membrane fusion proteins. FEBS J 2024. [PMID: 39552293 DOI: 10.1111/febs.17313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/13/2024] [Accepted: 10/17/2024] [Indexed: 11/19/2024]
Abstract
Membrane fusion is an essential biological process that merges two separate lipid bilayers into a whole one. Membrane fusion proteins facilitate this process by bringing lipid bilayers in close proximity to reduce the repulsive energy between membranes. Along with their interactions with membranes, the structures and dynamics of membrane fusion proteins are key to elucidating the mechanisms of membrane fusion. Solid-state NMR (SSNMR) spectroscopy has unique advantages in determining the structures and dynamics of membrane fusion proteins in their membrane-bound states. It has been extensively applied to reveal conformational changes in intermediate states of viral membrane fusion proteins and to characterize the critical lipid-membrane interactions that drive the fusion process. In this review, we summarize recent advancements in SSNMR techniques for studying membrane fusion proteins and their applications in elucidating the mechanisms of membrane fusion.
Collapse
Affiliation(s)
- Lifen Zheng
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Shenlin Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| |
Collapse
|
6
|
Xu C, Ma C, Zhang W, Wei Y, Yang K, Yuan B. Membrane Fusion Mediated by Cationic Helical Peptide L-MMBen through Phosphatidylglycerol Recruitment. J Phys Chem Lett 2024; 15:11027-11034. [PMID: 39466831 DOI: 10.1021/acs.jpclett.4c02558] [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: 10/30/2024]
Abstract
Membrane fusion is the basis for many biological processes, which holds promise in biomedical applications including the creation of engineered hybrid cells and cell membrane functionalization. Extensive research efforts, including investigations into DNA zippers and carbon nanotubes, have been dedicated to the development of membrane fusion strategies inspired by natural SNARE proteins; nevertheless, achieving a delicate balance between membrane selectivity and high fusion efficiency through precise molecular engineering remains unclear. In our recent study, we successfully designed L-MMBen, a cationic helical antimicrobial peptide that exhibits remarkable antimicrobial efficacy while demonstrating moderate cytotoxicity. In this work, we demonstrate the effective and selective induction of fusion between phosphatidylglycerol (PG)-containing membranes by L-MMBen. By combining biophysical assays at the single-vesicle level with computer simulations at the molecular level, we discovered that L-MMBen can stably adsorb onto the surface of PG-containing membranes, leading to the formation of stalk structures between vesicles and ultimately resulting in membrane fusion. Furthermore, the occurrence of fusion is attributed to the unique ability of L-MMBen to recruit PG lipids and bridge adjacent vesicles. In contrast, its nonhelical counterpart DL-MMBen was found to lack this capability despite possessing an identical positive charge. These findings present an alternative molecule for achieving selective membrane fusion and provide insights for designing helical peptides with diverse applications.
Collapse
Affiliation(s)
- Cheng Xu
- Songshan Lake Materials Laboratory, Dongguan 523808, Guangdong, China
| | - Chiyun Ma
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, Jiangsu, China
| | - Wanting Zhang
- Songshan Lake Materials Laboratory, Dongguan 523808, Guangdong, China
- National Laboratory of Solid State Microstructures and Department of Physics, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210093, Jiangsu, China
| | - Yushuang Wei
- Songshan Lake Materials Laboratory, Dongguan 523808, Guangdong, China
| | - Kai Yang
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, Jiangsu, China
| | - Bing Yuan
- Songshan Lake Materials Laboratory, Dongguan 523808, Guangdong, China
| |
Collapse
|
7
|
Wang Y, Niu B, Tian Y, Lan H, Zhou Z, Li Y, Zhao S, Zhang Y, Yang C, Kong L, Zhang Z. Mitoxantrone Combined with Engineered TRAIL-Nanovesicles for Enhanced Cancer Immunotherapy Via Converting Apoptosis into Pyroptosis. Adv Healthc Mater 2024; 13:e2401723. [PMID: 39049538 DOI: 10.1002/adhm.202401723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/15/2024] [Indexed: 07/27/2024]
Abstract
Pyroptosis, a highly inflammatory form of programmed cell death, has emerged as a promising target for cancer immunotherapy. However, in the context of pyroptosis execution, while both caspase-3 and GSDME are essential, it is noteworthy that GSDME is frequently under-expressed in cold tumors. To overcome this limitation, engineered cellular nanovesicles (NVs) presenting TRAIL on their membranes (NVTRAIL) are developed to trigger the upregulation of cleaved caspase-3. When strategically combined with the chemotherapeutic agent mitoxantrone (MTO), known for its ability to enhance GSDME expression, MTO@NVTRAIL can convert cancer cells from apoptosis into pyroptosis, inhibit the tumor growth and metastasis successfully in primary tumor. The microparticles released by pyroptotic tumor cells also exhibited certain cytotoxicity against other tumor cells. In addition, tumor cells exposed to the combination treatment of MTO@NVTRAIL in vitro have also demonstrated potential utility as a novel form of vaccine for cancer immunotherapy. Flow analysis of the tumor microenvironment and draining lymph nodes reveals an increased proportion of matured dendritic cells and activation of T cells. In summary, the research provided a reference and alternative approach to induce cancer pyroptosis for clinical antitumor therapy based on engineered cellular nanovesicles and chemotherapy.
Collapse
Affiliation(s)
- Yi Wang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Boning Niu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yinmei Tian
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hongbing Lan
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhanhao Zhou
- Liyuan Hospital, Huazhong University of Science and Technology, Wuhan, 430077, China
| | - Yang Li
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Siyu Zhao
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Conglian Yang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li Kong
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Engineering Research Centre for Novel Drug Delivery System, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhiping Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Engineering Research Centre for Novel Drug Delivery System, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
8
|
Esparza D, Lima C, Abuelreich S, Ghaeli I, Hwang J, Oh E, Lenz A, Gu A, Jiang N, Kandeel F, Thurmond DC, Jovanovic-Talisman T. Pancreatic β-cells package double C2-like domain beta protein into extracellular vesicles via tandem C2 domains. Front Endocrinol (Lausanne) 2024; 15:1451279. [PMID: 39497805 PMCID: PMC11532064 DOI: 10.3389/fendo.2024.1451279] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/10/2024] [Indexed: 11/07/2024] Open
Abstract
Introduction Double C2-like domain beta (DOC2B) is a vesicle priming protein critical for glucose-stimulated insulin secretion in β-cells. Individuals with type 1 diabetes (T1D) have lower levels of DOC2B in their residual functional β-cell mass and platelets, a phenotype also observed in a mouse model of T1D. Thus, DOC2B levels could provide important information on β-cell dys(function). Objective Our objective was to evaluate the DOC2B secretome of β-cells. In addition to soluble extracellular protein, we assessed DOC2B localized within membrane-delimited nanoparticles - extracellular vesicles (EVs). Moreover, in rat clonal β-cells, we probed domains required for DOC2B sorting into EVs. Method Using Single Extracellular VEsicle Nanoscopy, we quantified EVs derived from clonal β-cells (human EndoC-βH1, rat INS-1 832/13, and mouse MIN6); two other cell types known to regulate glucose homeostasis and functionally utilize DOC2B (skeletal muscle rat myotube L6-GLUT4myc and human neuronal-like SH-SY5Y cells); and human islets sourced from individuals with no diabetes (ND). EVs derived from ND human plasma, ND human islets, and cell lines were isolated with either size exclusion chromatography or differential centrifugation. Isolated EVs were comprehensively characterized using dotblots, transmission electron microscopy, nanoparticle tracking analysis, and immunoblotting. Results DOC2B was present within EVs derived from ND human plasma, ND human islets, and INS-1 832/13 β-cells. Compared to neuronal-like SH-SY5Y cells and L6-GLUT4myc myotubes, clonal β-cells (EndoC-βH1, INS-1 832/13, and MIN6) produced significantly more EVs. DOC2B levels in EVs (over whole cell lysates) were higher in INS-1 832/13 β-cells compared to L6-GLUT4myc myotubes; SH-SY5Y neuronal-like cells did not release appreciable DOC2B. Mechanistically, we show that DOC2B was localized to the EV lumen; the tandem C2 domains were sufficient to confer sorting to INS-1 832/13 β-cell EVs. Discussion Clonal β-cells and ND human islets produce abundant EVs. In cell culture, appreciable DOC2B can be packaged into EVs, and a small fraction is excreted as a soluble protein. While DOC2B-laden EVs and soluble protein are present in ND plasma, further studies will be necessary to determine if DOC2B originating from β-cells significantly contributes to the plasma secretome.
Collapse
Affiliation(s)
- Diana Esparza
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Carinna Lima
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Sarah Abuelreich
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Ima Ghaeli
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Jinhee Hwang
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Eunjin Oh
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Ayelet Lenz
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Angel Gu
- Department of Translational Research and Cellular Therapeutics, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Nan Jiang
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Fouad Kandeel
- Department of Translational Research and Cellular Therapeutics, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Debbie C. Thurmond
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Tijana Jovanovic-Talisman
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute at City of Hope, Duarte, CA, United States
| |
Collapse
|
9
|
Mandal P, Roy S, Karmakar M, Bhatta SR, Ghosh CC, Thakur A, Parui PP. Determination of divalent metal ion-regulated proton concentration and polarity at the interface of anionic phospholipid membranes. SOFT MATTER 2024; 20:7646-7656. [PMID: 39291663 DOI: 10.1039/d4sm00876f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
We studied the influence of trace quantities of divalent metal ions (M2+: Ca2+, Mg2+, and Zn2+) on proton concentration (-log[H+], designated as pH') and polarity at the interface of anionic PG-phospholipid membranes comprising saturated and unsaturated acrylic chains. A spiro-rhodamine-6G-gallic acid (RGG) pH-probe was synthesized to monitor the interfacial pH' of large unilamellar vesicles (LUVs) at a physiologically appropriate bulk pH (6.0-7.5). 1H-NMR spectroscopy and fluorescence microscopy showed that RGG interacted with the LUV interface. The pH-dependent equilibrium between the spiro-closed and spiro-open forms of RGG at the interface from the bulk phase was compared using fluorescence spectra to obtain interfacial pH'. Interfacial dielectric constant (κ) was estimated using a porphyrin-based polarity-probe (GPP) that exhibits a κ-induced equilibrium between monomeric and oligomeric forms. M2+ interaction decreased LUV interfacial κ from ∼67 to 61, regardless of lipid/M2+ types. Fluorescence spectral and microscopic analysis revealed that low Ca2+ and Mg2+ amounts (M2+/lipid = 1 : 20 for unsaturated DOPG and POPG and ∼1 : 10 for saturated DMPG lipids), but not Zn2+, decreased LUV interfacial acidity from pH' ∼3.8 to 4.4 at bulk pH 7.0. Although membrane surface charges are normally responsible for pH' deviation from the bulk to the interface, they cannot explain M2+-mediated interfacial pH' increase since there is little change in surface charges up to a low M2+/lipid ratio of <1/10. M2+-induced tight lipid headgroup packing and the resulting increased surface rigidity inhibit interfacial H+/H2O penetration, reducing interfacial acidity and polarity. Our findings revealed that in certain cases, essential M2+ ion-induced bio-membrane reactivity can be attributed to the influence of interfacial pH'/polarity.
Collapse
Affiliation(s)
- Pratima Mandal
- Department of Chemistry, Jadavpur University, Kolkata 700032, India.
| | - Snigdha Roy
- Department of Chemistry, Jadavpur University, Kolkata 700032, India.
| | - Manisha Karmakar
- Department of Chemistry, Jadavpur University, Kolkata 700032, India.
| | | | | | - Arunabha Thakur
- Department of Chemistry, Jadavpur University, Kolkata 700032, India.
| | | |
Collapse
|
10
|
Pandia S, Chakraborty H. Strategic Design of Tryptophan-Aspartic Acid-Containing Peptide Inhibitors Using Coronin 1 as a Template: Inhibition of Fusion by Enhancing Acyl Chain Order. J Phys Chem B 2024; 128:9163-9171. [PMID: 39268813 DOI: 10.1021/acs.jpcb.4c03853] [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: 09/15/2024]
Abstract
Enveloped viruses enter the host cell by fusing at the cell membrane or entering the cell via endocytosis and fusing at the endosome. Conventional inhibitors target the viral fusion protein to inactivate it for inducing fusion. These target-specific vis-à-vis virus-specific inhibitors fail to display their inhibitory efficacy against emerging and remerging viral infections. This necessitates the need to develop broad-spectrum entry inhibitors that are effective irrespective of the virus. Using a broad range of targeting techniques, the fusion inhibitors can modify the physical characteristics of the viral membrane, making it less prone to fusion. We have previously shown that two tryptophan-aspartic acid (WD)-containing hydrophobic peptides, TG-23 and GG-21, from coronin 1, a phagosomal protein, inhibit membrane fusion by modulating membrane organization and dynamics. In the present work, we designed two WD-containing hydrophilic peptides, QG-22 and AG-22, using coronin 1 as a template and evaluated their fusion inhibitory efficacies in the absence and presence of membrane cholesterol. Our results demonstrate that QG-22 and AG-22 inhibit membrane fusion irrespective of the concentration of membrane cholesterol. Our measurements of depth-dependent membrane organization and dynamics reveal that they impede fusion by enhancing the acyl chain order. Overall, our results validate the hypothesis of designing fusion inhibitors by modulating the membrane's physical properties. In addition, it demonstrates that chain hydrophobicity might not be a critical determinant for the development of peptide-based fusion inhibitors.
Collapse
Affiliation(s)
- Swaratmika Pandia
- School of Chemistry, Sambalpur University, Jyoti Vihar, Burla, Odisha 768 019, India
| | - Hirak Chakraborty
- School of Chemistry, Sambalpur University, Jyoti Vihar, Burla, Odisha 768 019, India
| |
Collapse
|
11
|
Tagoe DNA, Ribeiro E Silva A, Drozda AA, Coppens I, Coleman BI, Gubbels MJ. Toxoplasma FER1 is a versatile and dynamic mediator of differential microneme trafficking and microneme exocytosis. Sci Rep 2024; 14:21819. [PMID: 39294204 PMCID: PMC11410953 DOI: 10.1038/s41598-024-72628-0] [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: 06/11/2024] [Accepted: 09/09/2024] [Indexed: 09/20/2024] Open
Abstract
Toxoplasma gondii is a polarized cell concentrating several secretory organelles at the apical pole. The secretory micronemes come in two sub-populations differentiated by dependence on Rab5A/C in their biogenesis. Calcium-dependent exocytosis of micronemes occurs at the very apical tip and is critical for parasite egress from its host cell, adhesion and invasion of the next cell. Ferlins represent a protein family with roles in exocytosis containing multiple Ca2+-sensing C2 domains. We determined that T. gondii's ferlin 1 (FER1) localized dynamically to the parasite's secretory pathway. FER1 function was dissected by dominant negative overexpression strategies. We demonstrated that FER1 traffics microneme organelles along the following trajectories: (1) Along the cortex to the apical end; (2) To the apical tip for fusion with the plasma membrane; (3) Differential microneme sub-population traffic, and that FER1 could putatively be responsible for microneme protein trafficking. (4) From the trans-Golgi-endosomal network to the subpellicular cortex; (5) Retrograde transport allowing microneme recycling from mother to daughter. Finally, FER1 overexpression triggers a microneme exocytosis burst, supporting the notion that the radially organized micronemes at the apical tip comprise a readily-releasable microneme pool. In summary, FER1 is pivotal for dynamic microneme trafficking, acts differently on the two microneme subpopulations, and acts on the plasma membrane fusion step during microneme exocytosis.
Collapse
Affiliation(s)
- Daniel N A Tagoe
- Department of Biology, Boston College, Chestnut Hill, MA, 02467, USA
- CANbridge Pharmaceuticals Inc., Burlington, MA, USA
| | | | - Allison A Drozda
- Department of Biology, Boston College, Chestnut Hill, MA, 02467, USA
- KromaTiD, Longmont, CO, USA
| | - Isabelle Coppens
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Bradley I Coleman
- Department of Biology, Boston College, Chestnut Hill, MA, 02467, USA
- Flagship Pioneering, Cambridge, MA, USA
| | - Marc-Jan Gubbels
- Department of Biology, Boston College, Chestnut Hill, MA, 02467, USA.
| |
Collapse
|
12
|
Peñalva DA, Monnappa AK, Natale P, López-Montero I. Mfn2-dependent fusion pathway of PE-enriched micron-sized vesicles. Proc Natl Acad Sci U S A 2024; 121:e2313609121. [PMID: 39012824 PMCID: PMC11287154 DOI: 10.1073/pnas.2313609121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 06/17/2024] [Indexed: 07/18/2024] Open
Abstract
Mitofusins (Mfn1 and Mfn2) are the mitochondrial outer-membrane fusion proteins in mammals and belong to the dynamin superfamily of multidomain GTPases. Recent structural studies of truncated variants lacking alpha helical transmembrane domains suggested that Mfns dimerize to promote the approximation and the fusion of the mitochondrial outer membranes upon the hydrolysis of guanine 5'-triphosphate disodium salt (GTP). However, next to the presence of GTP, the fusion activity seems to require multiple regulatory factors that control the dynamics and kinetics of mitochondrial fusion through the formation of Mfn1-Mfn2 heterodimers. Here, we purified and reconstituted the full-length murine Mfn2 protein into giant unilamellar vesicles (GUVs) with different lipid compositions. The incubation with GTP resulted in the fusion of Mfn2-GUVs. High-speed video-microscopy showed that the Mfn2-dependent membrane fusion pathway progressed through a zipper mechanism where the formation and growth of an adhesion patch eventually led to the formation of a membrane opening at the rim of the septum. The presence of physiological concentration (up to 30 mol%) of dioleoyl-phosphatidylethanolamine (DOPE) was shown to be a requisite to observe GTP-induced Mfn2-dependent fusion. Our observations show that Mfn2 alone can promote the fusion of micron-sized DOPE-enriched vesicles without the requirement of regulatory cofactors, such as membrane curvature, or the assistance of other proteins.
Collapse
Affiliation(s)
- Daniel A. Peñalva
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional del Sur, Bahía BlancaB8000, Argentina
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, Bahía BlancaB8000, Argentina
| | - Ajay K. Monnappa
- Instituto de Investigación Biomédica Hospital Doce de Octubre (imas12), Madrid28041, Spain
| | - Paolo Natale
- Instituto de Investigación Biomédica Hospital Doce de Octubre (imas12), Madrid28041, Spain
- Departamento Química Física, Universidad Complutense de Madrid, Madrid28041, Spain
- Instituto Pluridisciplinar, Universidad Complutense de Madrid, Madrid28041, Spain
| | - Iván López-Montero
- Instituto de Investigación Biomédica Hospital Doce de Octubre (imas12), Madrid28041, Spain
- Departamento Química Física, Universidad Complutense de Madrid, Madrid28041, Spain
- Instituto Pluridisciplinar, Universidad Complutense de Madrid, Madrid28041, Spain
| |
Collapse
|
13
|
Kim BK, Kang DH, Woo J, Yoon W, Ryu H, Han K, Chung S, Kim TS. Control of artificial membrane fusion in physiological ionic solutions beyond the limits of electroformation. Nat Commun 2024; 15:4524. [PMID: 38806492 PMCID: PMC11133453 DOI: 10.1038/s41467-024-48875-0] [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: 09/26/2023] [Accepted: 05/15/2024] [Indexed: 05/30/2024] Open
Abstract
Membrane fusion, merging two lipid bilayers, is crucial for fabricating artificial membrane structures. Over the past 40 years, in contrast to precise and controllable membrane fusion in-vivo through specific molecules such as SNAREs, controlling the fusion in-vitro while fabricating artificial membrane structures in physiological ionic solutions without fusion proteins has been a challenge, becoming a significant obstacle to practical applications. We present an approach consisting of an electric field and a few kPa hydraulic pressure as an additional variable to physically control the fusion, enabling tuning of the shape and size of the 3D freestanding lipid bilayers in physiological ionic solutions. Mechanical model analysis reveals that pressure-induced parallel/normal tensions enhance fusion among membranes in the microwell. In-vitro peptide-membrane assay, mimicking vesicular transport via pressure-assisted fusion, and stability of 38 days with in-chip pressure control via pore size-regulated hydrogel highlight the potential for diverse biological applications.
Collapse
Affiliation(s)
- Bong Kyu Kim
- Center for Brain Technology, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
- Department of Mechanical Engineering, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Dong-Hyun Kang
- Center for Brain Technology, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
- Bionics Research Center, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Junhyuk Woo
- Center for Brain Technology, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Wooseung Yoon
- Center for Brain Technology, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Hyunil Ryu
- Center for Brain Technology, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Kyungreem Han
- Center for Brain Technology, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Seok Chung
- Department of Mechanical Engineering, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Tae Song Kim
- Center for Brain Technology, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea.
| |
Collapse
|
14
|
Pandia S, Mahapatra A, Chakraborty H. A Coronin 1-Derived Peptide Inhibits Membrane Fusion by Modulating Membrane Organization and Dynamics. J Phys Chem B 2024; 128:4986-4995. [PMID: 38739415 DOI: 10.1021/acs.jpcb.4c00295] [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: 05/14/2024]
Abstract
Membrane fusion is considered the first step in the entry of enveloped viruses into the host cell. Several targeted strategies have been implemented to block viral entry by limiting the fusion protein to form a six-helix bundle, which is a prerequisite for fusion. Nonetheless, the development of broad-spectrum fusion inhibitors is essential to combat emerging and re-emerging viral infections. TG-23, a coronin 1, a tryptophan-aspartate-rich phagosomal protein-derived peptide, demonstrated inhibition of fusion between small unilamellar vesicles (SUVs) by modulating the membrane's physical properties. However, its inhibitory efficacy reduces with an increasing concentration of membrane cholesterol. The present work aims to develop a fusion inhibitor whose efficacy would be unaltered in the presence of membrane cholesterol. A stretch of the tryptophan-aspartic acid-containing peptide with a similar secondary structure and hydrophobicity profile of TG-23 from coronin 1 was synthesized, and its ability to inhibit SUV-SUV fusion with varying concentrations of membrane cholesterol was evaluated. Our results demonstrate that the GG-21 peptide inhibits fusion irrespective of the cholesterol content of the membrane. We have further evaluated the peptide-induced change in the membrane organization and dynamics utilizing arrays of steady-state and time-resolved fluorescence measurements and correlated these results with their effect on fusion. Interestingly, GG-21 displays inhibitory efficacy in a wide variety of lipid compositions despite having a secondary structure and physical properties similar to those of TG-23. Overall, our results advocate that the secondary structure and physical properties of the peptide may not be sufficient to predict its inhibitory efficacy.
Collapse
Affiliation(s)
- Swaratmika Pandia
- School of Chemistry, Sambalpur University, Jyoti Vihar, Burla 768 019, Odisha, India
| | - Amita Mahapatra
- School of Chemical Sciences, National Institute of Science Education and Research (NISER), Jatni, Khurda, Bhubaneswar 752050, Odisha, India
- Homi Bhabha National Institute (HBNI), Mumbai 400094, India
| | - Hirak Chakraborty
- School of Chemistry, Sambalpur University, Jyoti Vihar, Burla 768 019, Odisha, India
| |
Collapse
|
15
|
Wang D, Guo Y, Heng BC, Zhang X, Wei Y, He Y, Xu M, Xia B, Deng X. Cell membrane vesicles derived from hBMSCs and hUVECs enhance bone regeneration. Bone Res 2024; 12:23. [PMID: 38594236 PMCID: PMC11003965 DOI: 10.1038/s41413-024-00325-9] [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: 04/12/2023] [Revised: 02/04/2024] [Accepted: 03/08/2024] [Indexed: 04/11/2024] Open
Abstract
Bone tissue renewal can be enhanced through co-transplantation of bone mesenchymal stem cells (BMSCs) and vascular endothelial cells (ECs). However, there are apparent limitations in stem cell-based therapy which hinder its clinic translation. Hence, we investigated the potential of alternative stem cell substitutes for facilitating bone regeneration. In this study, we successfully prepared cell membrane vesicles (CMVs) from BMSCs and ECs. The results showed that BMSC-derived cell membrane vesicles (BMSC-CMVs) possessed membrane receptors involved in juxtacrine signaling and growth factors derived from their parental cells. EC-derived cell membrane vesicles (EC-CMVs) also contained BMP2 and VEGF derived from their parental cells. BMSC-CMVs enhanced tube formation and migration ability of hUVECs, while EC-CMVs promoted the osteogenic differentiation of hBMSCs in vitro. Using a rat skull defect model, we found that co-transplantation of BMSC-CMVs and EC-CMVs could stimulate angiogenesis and bone formation in vivo. Therefore, our research might provide an innovative and feasible approach for cell-free therapy in bone tissue regeneration.
Collapse
Affiliation(s)
- Dandan Wang
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Yaru Guo
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, P. R. China
| | - Boon Chin Heng
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, P. R. China
| | - Xuehui Zhang
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, P. R. China
| | - Yan Wei
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, P. R. China
| | - Ying He
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, P. R. China
| | - Mingming Xu
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, P. R. China
| | - Bin Xia
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China.
| | - Xuliang Deng
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, P. R. China.
| |
Collapse
|
16
|
Lee Y, Fracassi A, Devaraj NK. Control of giant vesicle assemblies by stimuli-responsive lipids. Chem Commun (Camb) 2024; 60:3930-3933. [PMID: 38497420 DOI: 10.1039/d4cc00322e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
We describe a bottom-up synthesis of giant vesicles (GVs) utilizing an artificial stimuli-responsive diazobenzene lipid building block. Controlled by light, the GVs can exhibit dynamic behaviors, including reversible formation, the generation of highly multilamellar assemblies, and vesicle capturing and releasing events.
Collapse
Affiliation(s)
- Youngjun Lee
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, USA.
| | - Alessandro Fracassi
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, USA.
| | - Neal K Devaraj
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, USA.
| |
Collapse
|
17
|
Yu F, Xu J, Chen H, Song S, Nie C, Hao K, Zhao Z. Proprotein convertase cleavage of Ictalurid herpesvirus 1 spike-like protein ORF46 is modulated by N-glycosylation. Virology 2024; 592:110008. [PMID: 38335866 DOI: 10.1016/j.virol.2024.110008] [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: 10/08/2023] [Revised: 01/05/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024]
Abstract
Viral spike proteins undergo a special maturation process that enables host cell receptor recognition, membrane fusion, and viral entry, facilitating effective virus infection. Here, we investigated the protease cleavage features of ORF46, a spike-like protein in Ictalurid herpesvirus 1 (IcHV-1) sharing similarity with spikes of Nidovirales members. We noted that during cleavage, full-length ORF46 is cleaved into ∼55-kDa and ∼100-kDa subunits. Moreover, truncation or site-directed mutagenesis at the recognition sites of proprotein convertases (PCs) abolishes this spike cleavage, highlighting the crucial role of Arg506/Arg507 and Arg668/Arg671 for the cleavage modification. ORF46 cleavage was suppressed by specific N-glycosylation inhibitors or mutation of its specific N-glycosylation sites (N192, etc.), suggesting that glycoprotein ORF46 cleavage is modulated by N-glycosylation. Notably, PCs and N-glycosylation inhibitors exhibited potent antiviral effects in host cells. Our findings, therefore, suggested that PCs cleavage of ORF46, modulated by N-glycosylation, is a potent antiviral target for fish herpesviruses.
Collapse
Affiliation(s)
- Fei Yu
- Jiangsu Province Engineering Research Center for Marine Bio-resources Sustainable Utilization, Hohai University, Nanjing 210024, China
| | - Jiehua Xu
- Jiangsu Province Engineering Research Center for Marine Bio-resources Sustainable Utilization, Hohai University, Nanjing 210024, China
| | - Hongxun Chen
- Jiangsu Province Engineering Research Center for Marine Bio-resources Sustainable Utilization, Hohai University, Nanjing 210024, China
| | - Siyang Song
- Jiangsu Province Engineering Research Center for Marine Bio-resources Sustainable Utilization, Hohai University, Nanjing 210024, China
| | - Chunlan Nie
- Jiangsu Province Engineering Research Center for Marine Bio-resources Sustainable Utilization, Hohai University, Nanjing 210024, China
| | - Kai Hao
- Jiangsu Province Engineering Research Center for Marine Bio-resources Sustainable Utilization, Hohai University, Nanjing 210024, China
| | - Zhe Zhao
- Jiangsu Province Engineering Research Center for Marine Bio-resources Sustainable Utilization, Hohai University, Nanjing 210024, China.
| |
Collapse
|
18
|
Xia Y, Wu K, Liu C, Zhao X, Wang J, Cao J, Chen Z, Fang M, Yu J, Zhu C, Zhang X, Wang Z. Filamentous-Actin-Mimicking Nanoplatform for Enhanced Cytosolic Protein Delivery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305600. [PMID: 38152963 PMCID: PMC10933650 DOI: 10.1002/advs.202305600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 11/29/2023] [Indexed: 12/29/2023]
Abstract
Despite the potential of protein therapeutics, the cytosolic delivery of proteins with high efficiency and bioactivity remains a significant challenge owing to exocytosis and lysosomal degradation after endocytosis. Therefore, it is important to develop a safe and efficient strategy to bypass endocytosis. Inspired by the extraordinary capability of filamentous-actin (F-actin) to promote cell membrane fusion, a cyanine dye assembly-containing nanoplatform mimicking the structure of natural F-actin is developed. The nanoplatform exhibits fast membrane fusion to cell membrane mimics and thus enters live cells through membrane fusion and bypasses endocytosis. Moreover, it is found to efficiently deliver protein cargos into live cells and quickly release them into the cytosol, leading to high protein cargo transfection efficiency and bioactivity. The nanoplatform also results in the superior inhibition of tumor cells when loaded with anti-tumor proteins. These results demonstrate that this fusogenic nanoplatform can be valuable for cytosolic protein delivery and tumor treatment.
Collapse
Affiliation(s)
- Yuqiong Xia
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & NeuroimagingMinistry of EducationSchool of Life Science and TechnologyXidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and TreatmentXi'anShaanxi710126P. R. China
- Guangzhou Institute of TechnologyXidian UniversityGuangzhouGuangdong510555P. R. China
| | - Keyun Wu
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & NeuroimagingMinistry of EducationSchool of Life Science and TechnologyXidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and TreatmentXi'anShaanxi710126P. R. China
- Guangzhou Institute of TechnologyXidian UniversityGuangzhouGuangdong510555P. R. China
| | - Chang Liu
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & NeuroimagingMinistry of EducationSchool of Life Science and TechnologyXidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and TreatmentXi'anShaanxi710126P. R. China
- Guangzhou Institute of TechnologyXidian UniversityGuangzhouGuangdong510555P. R. China
| | - Xuejuan Zhao
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & NeuroimagingMinistry of EducationSchool of Life Science and TechnologyXidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and TreatmentXi'anShaanxi710126P. R. China
- Guangzhou Institute of TechnologyXidian UniversityGuangzhouGuangdong510555P. R. China
| | - Jun Wang
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & NeuroimagingMinistry of EducationSchool of Life Science and TechnologyXidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and TreatmentXi'anShaanxi710126P. R. China
| | - Jianxia Cao
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & NeuroimagingMinistry of EducationSchool of Life Science and TechnologyXidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and TreatmentXi'anShaanxi710126P. R. China
| | - Zhaoxu Chen
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & NeuroimagingMinistry of EducationSchool of Life Science and TechnologyXidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and TreatmentXi'anShaanxi710126P. R. China
| | - Minchao Fang
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular StructuresSchool of Life SciencesTianjin University92 Weijin Road, Nankai DistrictTianjin300072P. R. China
| | - Jie Yu
- School of Biology and EngineeringGuizhou Medical UniversityGuizhouGuiyang550025P. R. China
| | - Cheng Zhu
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular StructuresSchool of Life SciencesTianjin University92 Weijin Road, Nankai DistrictTianjin300072P. R. China
| | - Xianghan Zhang
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & NeuroimagingMinistry of EducationSchool of Life Science and TechnologyXidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and TreatmentXi'anShaanxi710126P. R. China
- Guangzhou Institute of TechnologyXidian UniversityGuangzhouGuangdong510555P. R. China
| | - Zhongliang Wang
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & NeuroimagingMinistry of EducationSchool of Life Science and TechnologyXidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and TreatmentXi'anShaanxi710126P. R. China
| |
Collapse
|
19
|
Andrä J, Aisenbrey C, Sudheendra US, Prudhon M, Brezesinski G, Zschech C, Willumeit-Römer R, Leippe M, Gutsmann T, Bechinger B. Structural analysis of the NK-lysin-derived peptide NK-2 upon interaction with bacterial membrane mimetics consisting of phosphatidylethanolamine and phosphatidylglycerol. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2024; 1866:184267. [PMID: 38159877 DOI: 10.1016/j.bbamem.2023.184267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 01/03/2024]
Abstract
NK-2 is an antimicrobial peptide derived from helices 3 and 4 of the pore-forming protein of natural killer cells, NK-lysin. It has potent activities against Gram-negative and Gram-positive bacteria, fungi and protozoan parasites without being toxic to healthy human cells. In biophysical assays its membrane activities were found to require phosphatidylglycerol (PG) and phosphatidylethanolamine (PE), lipids which dominate the composition of bacterial membranes. Here the structure and activities of NK-2 in binary mixtures of different PE/PG composition were investigated. CD spectroscopy reveals that a threshold concentration of 50 % PG is needed for efficient membrane association of NK-2 concomitant with a random coil - helix transition. Association with PE occurs but is qualitatively different when compared to PG membranes. Oriented solid-state NMR spectroscopy of NK-2 specifically labelled with 15N indicates that the NK-2 helices are oriented parallel to the PG bilayer surface. Upon reduction of the PG content to 20 mol% interactions are weaker and/or an in average more tilted orientation is observed. Fluorescence spectroscopy of differently labelled lipids is in agreement of an interfacial localisation of both helices where the C-terminal end is in a less hydrophobic environment. By inserting into the membrane interface and interacting differently with PE and PG the peptides probably induce high curvature strain which result in membrane openings and rupture.
Collapse
Affiliation(s)
- Jörg Andrä
- Department of Biotechnology, Faculty of Life Sciences, Hamburg University of Applied Sciences, Hamburg, Germany.
| | | | - U S Sudheendra
- University of Strasbourg / CNRS, UMR7177, Chemistry Institute, Strasbourg, France
| | - Marc Prudhon
- University of Strasbourg / CNRS, UMR7177, Chemistry Institute, Strasbourg, France
| | - Gerald Brezesinski
- Department of Physics, TU Darmstadt, Darmstadt, Germany; Department of Interfaces, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Claudia Zschech
- Department of Interfaces, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | | | - Matthias Leippe
- Comparative Immunobiology, Zoological Institute, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Thomas Gutsmann
- Research Center Borstel, Leibniz Lung Center, Borstel, Germany; Centre for Structural Systems Biology, Hamburg, Germany
| | - Burkhard Bechinger
- University of Strasbourg / CNRS, UMR7177, Chemistry Institute, Strasbourg, France; Institut Universitaire de France, 75005 Paris, France.
| |
Collapse
|
20
|
Jiménez-López D, Xoconostle-Cázares B, Calderón-Pérez B, Vargas-Hernández BY, Núñez-Muñoz LA, Ramírez-Pool JA, Ruiz-Medrano R. Evolutionary and Structural Analysis of PP16 in Viridiplantae. Int J Mol Sci 2024; 25:2839. [PMID: 38474088 DOI: 10.3390/ijms25052839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 02/23/2024] [Accepted: 02/25/2024] [Indexed: 03/14/2024] Open
Abstract
Members of the phloem protein 16 (PP16) gene family are induced by elicitors in rice and the corresponding proteins from cucurbits, which display RNA binding and intercellular transport activities, are accumulated in phloem sap. These proteins facilitate the movement of protein complexes through the phloem translocation flow and may be involved in the response to water deficit, among other functions. However, there is scant information regarding their function in other plants, including the identification of paralog genes in non-vascular plants and chlorophytes. In the present work, an evolutionary and structural analysis of the PP16 family in green plants (Viridiplantae) was carried out. Data mining in different databases indicated that PP16 likely originated from a larger gene present in an ancestral lineage that gave rise to chlorophytes and multicellular plants. This gene encodes a protein related to synaptotagmin, which is involved in vesicular transport in animal systems, although other members of this family play a role in lipid turnover in endomembranes and organelles. These proteins contain a membrane-binding C2 domain shared with PP16 proteins in vascular plants. In silico analysis of the predicted structure of the PP16 protein family identified several β-sheets, one α-helix, and intrinsically disordered regions. PP16 may have been originally involved in vesicular trafficking and/or membrane maintenance but specialized in long-distance signaling during the emergence of the plant vascular system.
Collapse
Affiliation(s)
- Domingo Jiménez-López
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico
| | - Beatriz Xoconostle-Cázares
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico
| | - Berenice Calderón-Pérez
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico
| | - Brenda Yazmín Vargas-Hernández
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico
| | - Leandro Alberto Núñez-Muñoz
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico
| | - José Abrahán Ramírez-Pool
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico
| | - Roberto Ruiz-Medrano
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico
| |
Collapse
|
21
|
He J, Xiu F, Chen Y, Yang Y, Liu H, Xi Y, Liu L, Li X, Wu Y, Luo H, Chen L, Ding N, Hu J, Chen E, You X. Aerobic glycolysis of bronchial epithelial cells rewires Mycoplasma pneumoniae pneumonia and promotes bacterial elimination. Infect Immun 2024; 92:e0024823. [PMID: 38205952 PMCID: PMC10863416 DOI: 10.1128/iai.00248-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 12/14/2023] [Indexed: 01/12/2024] Open
Abstract
The immune response to Mycoplasma pneumoniae infection plays a key role in clinical symptoms. Previous investigations focused on the pro-inflammatory effects of leukocytes and the pivotal role of epithelial cell metabolic status in finely modulating the inflammatory response have been neglected. Herein, we examined how glycolysis in airway epithelial cells is affected by M. pneumoniae infection in an in vitro model. Additionally, we investigated the contribution of ATP to pulmonary inflammation. Metabolic analysis revealed a marked metabolic shift in bronchial epithelial cells during M. pneumoniae infection, characterized by increased glucose uptake, enhanced aerobic glycolysis, and augmented ATP synthesis. Notably, these metabolic alterations are orchestrated by adaptor proteins, MyD88 and TRAM. The resulting synthesized ATP is released into the extracellular milieu via vesicular exocytosis and pannexin protein channels, leading to a substantial increase in extracellular ATP levels. The conditioned medium supernatant from M. pneumoniae-infected epithelial cells enhances the secretion of both interleukin (IL)-1β and IL-18 by peripheral blood mononuclear cells, partially mediated by the P2X7 purine receptor (P2X7R). In vivo experiments confirm that addition of a conditioned medium exacerbates pulmonary inflammation, which can be attenuated by pre-treatment with a P2X7R inhibitor. Collectively, these findings highlight the significance of airway epithelial aerobic glycolysis in enhancing the pulmonary inflammatory response and aiding pathogen clearance.
Collapse
Affiliation(s)
- Jun He
- Department of Clinical Laboratory, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| | - Feichen Xiu
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| | - Yiwen Chen
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| | - Yan Yang
- Department of Clinical Laboratory, Shanghai Putuo People's Hospital, Tongji University, Shanghai, China
| | - Hongwei Liu
- Department of Epidemiology and Health Statistics, School of Public Health, University of South China, Hengyang, China
| | - Yixuan Xi
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| | - Lu Liu
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| | - Xinru Li
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| | - Yueyue Wu
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| | - Haodang Luo
- Department of Clinical Laboratory, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Liesong Chen
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| | - Nan Ding
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| | - Jun Hu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - En Chen
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Xiaoxing You
- Department of Clinical Laboratory, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| |
Collapse
|
22
|
Aslam S, Zulfiqar F, Hameed W, Qureshi S, Zaroon, Bashir H. Fusion proteins development strategies and their role as cancer therapeutic agents. Biotechnol Appl Biochem 2024; 71:81-95. [PMID: 37822167 DOI: 10.1002/bab.2523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 10/01/2023] [Indexed: 10/13/2023]
Abstract
Cancer continues to be leading cause of morbidity and mortality despite decades of research and advancement in chemotherapy. Most tumors can be reduced via standard oncology treatments, such as chemotherapy, radiotherapy, and surgical resection, and they frequently recur. Significant progress has been made since targeted cancer therapy inception in creation of medications that exhibit improved tumor-selective action. Particularly in preclinical and clinical investigations, fusion proteins have shown strong activity and improved treatment outcomes for a number of human cancers. Synergistically combining many proteins into one complex allows the creation of synthetic fusion proteins with enhanced characteristics or new capabilities. Signal transduction pathways are important for onset, development, and spread of cancer. As result, signaling molecules are desirable targets for cancer therapies, and significant effort has been made into developing fusion proteins that would act as inhibitors of these pathways. A wide range of biotechnological and medicinal applications are made possible by fusion of protein domains that improves bioactivities or creates new functional combinations. Such proteins may function as immune effectors cell recruiters to tumors or as decoy receptors for various ligands. In this review article, we have outlined the standard methods for creating fusion proteins and covered the applications of fusion proteins in treatment of cancer. This article also highlights the role of fusion proteins in targeting the signaling pathways involved in cancer for effective treatment.
Collapse
Affiliation(s)
- Shakira Aslam
- Center for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| | | | - Warda Hameed
- King Edward Medical University, Lahore, Pakistan
| | - Shahnila Qureshi
- Center for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Zaroon
- Center for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Hamid Bashir
- Center for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| |
Collapse
|
23
|
Chan WJ, Li H. Recent advances in nano/micro systems for improved circulation stability, enhanced tumor targeting, penetration, and intracellular drug delivery: a review. Biomed Phys Eng Express 2024; 10:022001. [PMID: 38086099 DOI: 10.1088/2057-1976/ad14f0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 12/12/2023] [Indexed: 01/17/2024]
Abstract
In recent years, nanoparticles (NPs) have been extensively developed as drug carriers to overcome the limitations of cancer therapeutics. However, there are several biological barriers to nanomedicines, which include the lack of stability in circulation, limited target specificity, low penetration into tumors and insufficient cellular uptake, restricting the active targeting toward tumors of nanomedicines. To address these challenges, a variety of promising strategies were developed recently, as they can be designed to improve NP accumulation and penetration in tumor tissues, circulation stability, tumor targeting, and intracellular uptake. In this Review, we summarized nanomaterials developed in recent three years that could be utilized to improve drug delivery for cancer treatments.
Collapse
Affiliation(s)
- Wei-Jen Chan
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, United States of America
| | - Huatian Li
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, United States of America
| |
Collapse
|
24
|
Keating PM, Schifano NP, Wei X, Kong MY, Lee J. pH-dependent conformational change within the Lassa virus transmembrane domain elicits efficient membrane fusion. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2024; 1866:184233. [PMID: 37734457 DOI: 10.1016/j.bbamem.2023.184233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/05/2023] [Accepted: 09/16/2023] [Indexed: 09/23/2023]
Abstract
Lassa virus (LASV) is the most prevalent member of the arenavirus family and the causative agent of Lassa fever, a viral hemorrhagic fever. Although there are annual outbreaks in West Africa, and recently isolated cases worldwide, there are no current therapeutics or vaccines, which poses LASV as a significant global public health threat. One of the key steps in LASV infection is the delivery of its genetic material by fusing its viral membrane with the host cell membrane. This process is facilitated by significant conformational changes within glycoprotein 2 (GP2), yielding distinct prefusion and postfusion structural states. However, structural information is missing to understand the changes that occur in the transmembrane domain during the fusion process. Here, we used CD and NMR spectroscopy to show that the transmembrane domain has pH-dependent conformational changes that result in an extension of the alpha helix at the N-terminal end. Proline mutants of key residues in that region prevent the helical extension, as seen in CD and NMR. We developed a modified lipid mixing assay to study the importance of this extension on the function of GP2. Our assay shows that membrane fusion efficiency is optimal at low pH values but introducing the proline mutants results in lower fusion efficiency. These results indicate that these pH-dependent conformational changes are important to the fusion mechanism. This information can be used to design therapeutics to combat Lassa virus infections and prevent its potential spread.
Collapse
Affiliation(s)
- Patrick M Keating
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742, USA
| | - Nicholas P Schifano
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742, USA
| | - Xinrui Wei
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742, USA
| | - Matthew Y Kong
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742, USA
| | - Jinwoo Lee
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
25
|
Karageorgos S, Platt AS, Bassiri H. Genetics of Primary Hemophagocytic Lymphohistiocytosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1448:75-101. [PMID: 39117809 DOI: 10.1007/978-3-031-59815-9_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Hemophagocytic lymphohistiocytosis (HLH) constitutes a rare, potentially life-threatening hyperinflammatory immune dysregulation syndrome that can present with a variety of clinical signs and symptoms, including fever, hepatosplenomegaly, and abnormal laboratory and immunological findings such as cytopenias, hyperferritinemia, hypofibrinogenemia, hypertriglyceridemia, elevated blood levels of soluble CD25 (interleukin (IL)-2 receptor α-chain), or diminished natural killer (NK)-cell cytotoxicity (reviewed in detail in Chapter 11 of this book). While HLH can be triggered by an inciting event (e.g., infections), certain monogenic causes have been associated with a significantly elevated risk of development of HLH, or recurrence of HLH in patients who have recovered from their disease episode. These monogenic predisposition syndromes are variably referred to as "familial" (FHL) or "primary" HLH (henceforth referred to as "pHLH") and are the focus of this chapter. Conversely, secondary HLH (sHLH) often occurs in the absence of monogenic etiologies that are commonly associated with pHLH and can be triggered by infections, malignancies, or rheumatological diseases; these triggers and the genetics associated with sHLH are discussed in more detail in other chapters in this book.
Collapse
Affiliation(s)
- Spyridon Karageorgos
- First Department of Pediatrics, "Aghia Sophia" Children's Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Anna S Platt
- Roberts Individualized Medical Genetics Center and Immune Dysregulation Program, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Hamid Bassiri
- Immune Dysregulation Program and Division of Infectious Diseases, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| |
Collapse
|
26
|
Ijabi J, Ijabi R, Roozehdar P, Kaminsky ZA, Moradi-Sardareh H, Tehranian N, Ahmed N. Tumor Targeting via siRNA-COG3 to Suppress Tumor Progression in Mice and Inhibit Cancer Metastasis and Angiogenesis in Ovarian Cancer Cell Lines. Microrna 2024; 13:140-154. [PMID: 38243930 DOI: 10.2174/0122115366275856240101083442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 12/03/2023] [Accepted: 12/15/2023] [Indexed: 01/22/2024]
Abstract
BACKGROUND The COG complex is implicated in the tethering of retrograde intra-Golgi vesicles, which involves vesicular tethering and SNAREs. SNARE complexes mediate the invasion and metastasis of cancer cells through MMPs which activate growth factors for ECM fragments by binding to integrin receptors. Increasing MMPs is in line with YKL40 since YKL40 is linked to promoting angiogenesis through VEGF and can increase ovarian cancer (OC) resistance to chemotropic and cell migration. OBJECTIVE The aim of this study is an assessment of siRNA-COG3 on proliferation, invasion, and apoptosis of OC cells. In addition, siRNA-COG3 may prevent the growth of OC cancer in mice with tumors. METHODS Primary OC cell lines will be treated with siRNA-COG3 to assay YKL40 and identified angiogenesis by Tube-like structure formation in HOMECs. The Golgi morphology was analyzed using Immunofluorescence microscopy. Furthermore, the effects of siRNA-COG3 on the proliferation and apoptosis of cells were evaluated using MTT and TUNEL assays. Clones of the HOSEpiC OC cell line were subcutaneously implanted in FVB/N mice. Mice were treated after two weeks of injection of cells using siRNA-COG3. Tumor development suppression was detected by D-luciferin. RT-PCR and western blotting analyses were applied to determine COG3, MT1- MMP, SNAP23, and YKL40 expression to investigate the effects of COG3 gene knockdown. RESULTS siRNA-COG3 exhibited a substantial effect in suppressing tumor growth in mice. It dramatically reduced OC cell proliferation and triggered apoptosis (all p < 0.01). Inhibition of COG3, YKL-40, and MT1-MPP led to suppression of angiogenesis and reduction of microvessel density through SNAP23 in OC cells. CONCLUSION Overall, by knockdown of the COG3 gene, MT1-MMP and YKL40 were dropped, leading to suppressed angiogenesis along with decreasing migration and proliferation. SiRNACOG3 may be an ideal agent to consider for clinical trial assessment therapy for OC, especially when an antiangiogenic SNAR-pathway targeting drug.
Collapse
Affiliation(s)
- Janat Ijabi
- Department of Hematology, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Hematology, Faculty of Medicine and Health Sciences, University Sains Malaysia, Kubang Kerian, 15200 Kota Bharu, Kelantan, Malaysia
| | - Roghayeh Ijabi
- Faculty of Reproductive Health, Golestan University of Medical Sciences, Gorgan, Iran
- Department of Pharmacology, Faculty of Medicine and Health Sciences, University Putra Malaysia, 43400 Serdang, Selangor Darul Ehsan, Malaysia
| | - Parisa Roozehdar
- Department of Medical Veterinary, Azad University, Garmsar Branch, Garmsar, Iran
| | - Zachary A Kaminsky
- Faculty of Medicine, Department of Cellular and Molecular Medicine, Royal's Institute of Mental Health Research, University of Ottawa, Ottawa, Canada
| | - Hemen Moradi-Sardareh
- Department of Basic Sciences, Asadabad School of Medical Sciences, Hamedan University of Medical Sciences, Hamedan, Iran
| | - Najmeh Tehranian
- Department of Midwifery & Reproductive Health, Faculty of Medical Sciences, Tarbiat Modares University, Jalal Ale Ahmad Highway, Tehran, Iran
| | - Naveed Ahmed
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, 16150, Malaysia
| |
Collapse
|
27
|
Sieler M, Dittmar T. Cell Fusion and Syncytia Formation in Cancer. Results Probl Cell Differ 2024; 71:433-465. [PMID: 37996689 DOI: 10.1007/978-3-031-37936-9_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2023]
Abstract
The natural phenomenon of cell-cell fusion does not only take place in physiological processes, such as placentation, myogenesis, or osteoclastogenesis, but also in pathophysiological processes, such as cancer. More than a century ago postulated, today the hypothesis that the fusion of cancer cells with normal cells leads to the formation of cancer hybrid cells with altered properties is in scientific consensus. Some studies that have investigated the mechanisms and conditions for the fusion of cancer cells with other cells, as well as studies that have characterized the resulting cancer hybrid cells, are presented in this review. Hypoxia and the cytokine TNFα, for example, have been found to promote cell fusion. In addition, it has been found that both the protein Syncytin-1, which normally plays a role in placentation, and phosphatidylserine signaling on the cell membrane are involved in the fusion of cancer cells with other cells. In human cancer, cancer hybrid cells were detected not only in the primary tumor, but also in the circulation of patients as so-called circulating hybrid cells, where they often correlated with a worse outcome. Although some data are available, the questions of how and especially why cancer cells fuse with other cells are still not fully answered.
Collapse
Affiliation(s)
- Mareike Sieler
- Institute of Immunology, Center for Biomedical Education and Research (ZBAF), University of Witten/Herdecke, Witten, Germany.
| | - Thomas Dittmar
- Institute of Immunology, Center for Biomedical Education and Research (ZBAF), University of Witten/Herdecke, Witten, Germany
| |
Collapse
|
28
|
Mathew L, Kapoor S. Methods for Studying Fusion of Bacterial Extracellular Vesicles with Intact Bacteria and Host Cells. Methods Mol Biol 2024; 2843:119-136. [PMID: 39141297 DOI: 10.1007/978-1-0716-4055-5_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Bacterial extracellular vesicles (BEVs) are nano- or micrometer-sized membrane-bound lipid vesicles released from both Gram-negative and Gram-positive bacteria. Cellular transport, communication, pathogenesis, and host-pathogen interactions are some of the major biological processes impacted by BEVs. Among these, host-pathogen interactions and bacterial pathogenesis are emerging as highly important targetable avenues underlined by the issues of antimicrobial resistance, thus demanding novel targets and approaches to treat bacterial infections. In this aspect, the study of the interaction of BEVs with bacteria and/or host cells becomes imperative and brings the membrane fusion process to the forefront. Furthermore, membrane fusion also underscores the performance of BEVs as nano-therapeutic delivery platforms. Here, we report methods to study fusion kinetics between mycobacteria-derived extracellular vesicles, which we refer to as MEVs, and intact mycobacteria or MEVs themselves. We also discuss the isolation of MEVs and their characterization. We outline critical factors that affect fusion kinetics by MEVs. The same principle can be extended for studying fusion between BEVs and mammalian host cells important for understanding how BEVs influence host-pathogen crosstalk.
Collapse
Affiliation(s)
- Lydia Mathew
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai, Maharashtra, India
| | - Shobhna Kapoor
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai, Maharashtra, India.
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan.
| |
Collapse
|
29
|
Tror S, Jeon S, Nguyen HT, Huh E, Shin K. A Self-Regenerating Artificial Cell, that is One Step Closer to Living Cells: Challenges and Perspectives. SMALL METHODS 2023; 7:e2300182. [PMID: 37246263 DOI: 10.1002/smtd.202300182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 04/29/2023] [Indexed: 05/30/2023]
Abstract
Controllable, self-regenerating artificial cells (SRACs) can be a vital advancement in the field of synthetic biology, which seeks to create living cells by recombining various biological molecules in the lab. This represents, more importantly, the first step on a long journey toward creating reproductive cells from rather fragmentary biochemical mimics. However, it is still a difficult task to replicate the complex processes involved in cell regeneration, such as genetic material replication and cell membrane division, in artificially created spaces. This review highlights recent advances in the field of controllable, SRACs and the strategies to achieve the goal of creating such cells. Self-regenerating cells start by replicating DNA and transferring it to a location where proteins can be synthesized. Functional but essential proteins must be synthesized for sustained energy generation and survival needs and function in the same liposomal space. Finally, self-division and repeated cycling lead to autonomous, self-regenerating cells. The pursuit of controllable, SRACs will enable authors to make bold advances in understanding life at the cellular level, ultimately providing an opportunity to use this knowledge to understand the nature of life.
Collapse
Affiliation(s)
- Seangly Tror
- Department of Chemistry and Institute of Biological Interfaces, Sogang University, Seoul, 04107, Republic of Korea
| | - SeonMin Jeon
- Department of Chemistry and Institute of Biological Interfaces, Sogang University, Seoul, 04107, Republic of Korea
| | - Huong Thanh Nguyen
- Department of Chemistry and Institute of Biological Interfaces, Sogang University, Seoul, 04107, Republic of Korea
| | - Eunjin Huh
- Department of Chemistry and Institute of Biological Interfaces, Sogang University, Seoul, 04107, Republic of Korea
| | - Kwanwoo Shin
- Department of Chemistry and Institute of Biological Interfaces, Sogang University, Seoul, 04107, Republic of Korea
| |
Collapse
|
30
|
Gamage YI, Pan J. Elucidating the Influence of Lipid Composition on Bilayer Perturbations Induced by the N-terminal Region of the Huntingtin Protein. BIOPHYSICA 2023; 3:582-597. [PMID: 38737720 PMCID: PMC11087071 DOI: 10.3390/biophysica3040040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2024]
Abstract
Understanding the membrane interactions of the N-terminal 17 residues of the huntingtin protein (HttN) is essential for unraveling its role in cellular processes and its impact on huntingtin misfolding. In this study, we used atomic force microscopy (AFM) to examine the effects of lipid specificity in mediating bilayer perturbations induced by HttN. Across various lipid environments, the peptide consistently induced bilayer disruptions in the form of holes. Notably, our results unveiled that cholesterol enhanced bilayer perturbation induced by HttN, while phosphatidylethanolamine (PE) lipids suppressed hole formation. Furthermore, anionic phosphatidylglycerol (PG) and cardiolipin lipids, along with cholesterol at high concentrations, promoted the formation of double-bilayer patches. This unique structure suggests that the synergy among HttN, anionic lipids, and cholesterol can enhance bilayer fusion, potentially by facilitating lipid intermixing between adjacent bilayers. Additionally, our AFM-based force spectroscopy revealed that HttN enhanced the mechanical stability of lipid bilayers, as evidenced by an elevated bilayer puncture force. These findings illuminate the complex interplay between HttN and lipid membranes and provide useful insights into the role of lipid composition in modulating membrane interactions with the huntingtin protein.
Collapse
Affiliation(s)
| | - Jianjun Pan
- Department of Physics, University of South Florida, Tampa, FL 33620
| |
Collapse
|
31
|
Courtney KC, Mandal T, Mehta N, Wu L, Li Y, Das D, Cui Q, Chapman ER. Synaptotagmin-7 outperforms synaptotagmin-1 to promote the formation of large, stable fusion pores via robust membrane penetration. Nat Commun 2023; 14:7761. [PMID: 38012142 PMCID: PMC10681989 DOI: 10.1038/s41467-023-42497-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 10/11/2023] [Indexed: 11/29/2023] Open
Abstract
Synaptotagmin-1 and synaptotagmin-7 are two prominent calcium sensors that regulate exocytosis in neuronal and neuroendocrine cells. Upon binding calcium, both proteins partially penetrate lipid bilayers that bear anionic phospholipids, but the specific underlying mechanisms that enable them to trigger exocytosis remain controversial. Here, we examine the biophysical properties of these two synaptotagmin isoforms and compare their interactions with phospholipid membranes. We discover that synaptotagmin-1-membrane interactions are greatly influenced by membrane order; tight packing of phosphatidylserine inhibits binding due to impaired membrane penetration. In contrast, synaptotagmin-7 exhibits robust membrane binding and penetration activity regardless of phospholipid acyl chain structure. Thus, synaptotagmin-7 is a super-penetrator. We exploit these observations to specifically isolate and examine the role of membrane penetration in synaptotagmin function. Using nanodisc-black lipid membrane electrophysiology, we demonstrate that membrane penetration is a critical component that underlies how synaptotagmin proteins regulate reconstituted, exocytic fusion pores in response to calcium.
Collapse
Affiliation(s)
- Kevin C Courtney
- Howard Hughes Medical Institute and the Department of Neuroscience, University of Wisconsin, 1111 Highland Avenue, Madison, WI, 53705, USA
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Taraknath Mandal
- Department of Chemistry, Boston University, Boston, MA, 02215, USA
- Department of Physics, Indian Institute of Technology - Kanpur, Kanpur, 208016, India
| | - Nikunj Mehta
- Howard Hughes Medical Institute and the Department of Neuroscience, University of Wisconsin, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Lanxi Wu
- Howard Hughes Medical Institute and the Department of Neuroscience, University of Wisconsin, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Yueqi Li
- Howard Hughes Medical Institute and the Department of Neuroscience, University of Wisconsin, 1111 Highland Avenue, Madison, WI, 53705, USA
- Center for Bioanalytical Chemistry, University of Science and Technology of China, Hefei, 230026, China
| | - Debasis Das
- Howard Hughes Medical Institute and the Department of Neuroscience, University of Wisconsin, 1111 Highland Avenue, Madison, WI, 53705, USA
- Department of Biological Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Navy Nagar, Colaba, Mumbai, 400005, India
| | - Qiang Cui
- Department of Chemistry, Boston University, Boston, MA, 02215, USA
| | - Edwin R Chapman
- Howard Hughes Medical Institute and the Department of Neuroscience, University of Wisconsin, 1111 Highland Avenue, Madison, WI, 53705, USA.
| |
Collapse
|
32
|
Furukawa H, Kimura Y, Inaba H, Matsuura K. A supramolecular system mimicking the infection process of an enveloped virus through membrane fusion. Sci Rep 2023; 13:19934. [PMID: 37968508 PMCID: PMC10651892 DOI: 10.1038/s41598-023-47347-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 11/12/2023] [Indexed: 11/17/2023] Open
Abstract
Membrane fusion is an essential step for the entry of enveloped viruses, such as human immunodeficiency virus and influenza virus, into the host cell, often triggered by the binding of membrane proteins on the viral envelope to host cell membrane. Recently, external stimuli was shown to trigger membrane fusion in an artificial system. Direct observation of artificial membrane fusion using a giant unilamellar vesicle (GUV), which is similar in size to a cell, is useful as a biological model system. However, there are no model systems for studying membrane fusion of enveloped viruses with host cells. Here, we report a supramolecular model system for viral entry into a GUV or cell through membrane fusion. The system was constructed by complexing a cationic lipid bilayer on an anionic artificial viral capsid, self-assembled from viral β-annulus peptides. We demonstrate that the cationic enveloped artificial viral capsid electrostatically interacts with the anionic GUV or cell, and the capsid enters the GUV or cell through membrane fusion. The model system established in this study will be important for analyzing membrane fusion during infection of a natural virus.
Collapse
Affiliation(s)
- Hiroto Furukawa
- Department of Chemistry and Biotechnology, Graduate School of Engineering, Tottori University, 4-101 Koyama-Minami, Tottori, 680-8552, Japan
| | - Yuuna Kimura
- Department of Chemistry and Biotechnology, Graduate School of Engineering, Tottori University, 4-101 Koyama-Minami, Tottori, 680-8552, Japan
| | - Hiroshi Inaba
- Department of Chemistry and Biotechnology, Graduate School of Engineering, Tottori University, 4-101 Koyama-Minami, Tottori, 680-8552, Japan
- Center for Research on Green Sustainable Chemistry, Tottori University, 4-101 Koyama-Minami, Tottori, 680-8552, Japan
| | - Kazunori Matsuura
- Department of Chemistry and Biotechnology, Graduate School of Engineering, Tottori University, 4-101 Koyama-Minami, Tottori, 680-8552, Japan.
- Center for Research on Green Sustainable Chemistry, Tottori University, 4-101 Koyama-Minami, Tottori, 680-8552, Japan.
| |
Collapse
|
33
|
Yi K, Kong H, Zheng C, Zhuo C, Jin Y, Zhong Q, Mintz RL, Ju E, Wang H, Lv S, Lao YH, Tao Y, Li M. A LIGHTFUL nanomedicine overcomes EGFR-mediated drug resistance for enhanced tyrosine-kinase-inhibitor-based hepatocellular carcinoma therapy. Biomaterials 2023; 302:122349. [PMID: 37844429 DOI: 10.1016/j.biomaterials.2023.122349] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 09/18/2023] [Accepted: 10/06/2023] [Indexed: 10/18/2023]
Abstract
Targeting the activated epidermal growth factor receptor (EGFR) via clustered regularly interspaced short palindromic repeat (CRISPR) technology is appealing to overcome the drug resistance of hepatocellular carcinoma (HCC) towards tyrosine kinase inhibitor (TKI) therapy. However, combining these two distinct drugs using traditional liposomes results in a suboptimal synergistic anti-HCC effect due to the limited CRISPR/Cas9 delivery efficiency caused by lysosomal entrapment after endocytosis. Herein, we developed a liver-targeting gene-hybridizing-TKI fusogenic liposome (LIGHTFUL) that can achieve high CRISPR/Cas9 expression to reverse the EGFR-mediated drug resistance for enhanced TKI-based HCC therapy efficiently. Coated with a galactose-modified membrane-fusogenic lipid layer, LIGHTFUL reached the targeting liver site to fuse with HCC tumor cells, directly and efficiently transporting interior CDK5- and PLK1-targeting CRISPR/Cas9 plasmids (pXG333-CPs) into the HCC cell cytoplasm and then the cell nucleus for efficient expression. Such membrane-fusion-mediated pXG333-CP delivery resulted in effective downregulation of both CDK5 and PLK1, sufficiently inactivating EGFR to improve the anti-HCC effects of the co-delivered TKI, lenvatinib. This membrane-fusion-participant codelivery strategy optimized the synergetic effect of CRISPR/Cas9 and TKI combinational therapy as indicated by the 0.35 combination index in vitro and the dramatic reduction of subcutaneous and orthotopic TKI-insensitive HCC tumor growth in mice. Therefore, the established LIGHTFUL provides a unique co-delivery platform to combine gene editing and TKI therapies for enhanced synergetic therapy.
Collapse
Affiliation(s)
- Ke Yi
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Huimin Kong
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Chunxiong Zheng
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Chenya Zhuo
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Yuanyuan Jin
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Qingguo Zhong
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Rachel L Mintz
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Enguo Ju
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Haixia Wang
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Shixian Lv
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Yeh-Hsing Lao
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China; Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, 510630, China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China; Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, 510630, China.
| |
Collapse
|
34
|
Nguyen H, Nguyen HL, Lan PD, Thai NQ, Sikora M, Li MS. Interaction of SARS-CoV-2 with host cells and antibodies: experiment and simulation. Chem Soc Rev 2023; 52:6497-6553. [PMID: 37650302 DOI: 10.1039/d1cs01170g] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the devastating global COVID-19 pandemic announced by WHO in March 2020. Through unprecedented scientific effort, several vaccines, drugs and antibodies have been developed, saving millions of lives, but the fight against COVID-19 continues as immune escape variants of concern such as Delta and Omicron emerge. To develop more effective treatments and to elucidate the side effects caused by vaccines and therapeutic agents, a deeper understanding of the molecular interactions of SARS-CoV-2 with them and human cells is required. With special interest in computational approaches, we will focus on the structure of SARS-CoV-2 and the interaction of its spike protein with human angiotensin-converting enzyme-2 (ACE2) as a prime entry point of the virus into host cells. In addition, other possible viral receptors will be considered. The fusion of viral and human membranes and the interaction of the spike protein with antibodies and nanobodies will be discussed, as well as the effect of SARS-CoV-2 on protein synthesis in host cells.
Collapse
Affiliation(s)
- Hung Nguyen
- Institute of Physics, Polish Academy of Sciences, al. Lotnikow 32/46, 02-668 Warsaw, Poland.
| | - Hoang Linh Nguyen
- Institute of Fundamental and Applied Sciences, Duy Tan University, Ho Chi Minh City 700000, Vietnam
- Faculty of Environmental and Natural Sciences, Duy Tan University, Da Nang 550000, Vietnam
| | - Pham Dang Lan
- Life Science Lab, Institute for Computational Science and Technology, Quang Trung Software City, Tan Chanh Hiep Ward, District 12, 729110 Ho Chi Minh City, Vietnam
- Faculty of Physics and Engineering Physics, VNUHCM-University of Science, 227, Nguyen Van Cu Street, District 5, 749000 Ho Chi Minh City, Vietnam
| | - Nguyen Quoc Thai
- Dong Thap University, 783 Pham Huu Lau Street, Ward 6, Cao Lanh City, Dong Thap, Vietnam
| | - Mateusz Sikora
- Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Mai Suan Li
- Institute of Physics, Polish Academy of Sciences, al. Lotnikow 32/46, 02-668 Warsaw, Poland.
| |
Collapse
|
35
|
Vlieghe A, Niort K, Fumat H, Guigner JM, Cohen MM, Tareste D. Role of Lipids and Divalent Cations in Membrane Fusion Mediated by the Heptad Repeat Domain 1 of Mitofusin. Biomolecules 2023; 13:1341. [PMID: 37759741 PMCID: PMC10527301 DOI: 10.3390/biom13091341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/21/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Mitochondria are highly dynamic organelles that constantly undergo fusion and fission events to maintain their shape, distribution and cellular function. Mitofusin 1 and 2 proteins are two dynamin-like GTPases involved in the fusion of outer mitochondrial membranes (OMM). Mitofusins are anchored to the OMM through their transmembrane domain and possess two heptad repeat domains (HR1 and HR2) in addition to their N-terminal GTPase domain. The HR1 domain was found to induce fusion via its amphipathic helix, which interacts with the lipid bilayer structure. The lipid composition of mitochondrial membranes can also impact fusion. However, the precise mode of action of lipids in mitochondrial fusion is not fully understood. In this study, we examined the role of the mitochondrial lipids phosphatidylethanolamine (PE), cardiolipin (CL) and phosphatidic acid (PA) in membrane fusion induced by the HR1 domain, both in the presence and absence of divalent cations (Ca2+ or Mg2+). Our results showed that PE, as well as PA in the presence of Ca2+, effectively stimulated HR1-mediated fusion, while CL had a slight inhibitory effect. By considering the biophysical properties of these lipids in the absence or presence of divalent cations, we inferred that the interplay between divalent cations and specific cone-shaped lipids creates regions with packing defects in the membrane, which provides a favorable environment for the amphipathic helix of HR1 to bind to the membrane and initiate fusion.
Collapse
Affiliation(s)
- Anaïs Vlieghe
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), Inserm UMR-S 1266, Team Membrane Traffic in Healthy & Diseased Brain, 75014 Paris, France
| | - Kristina Niort
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), Inserm UMR-S 1266, Team Membrane Traffic in Healthy & Diseased Brain, 75014 Paris, France
| | - Hugo Fumat
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), Inserm UMR-S 1266, Team Membrane Traffic in Healthy & Diseased Brain, 75014 Paris, France
| | - Jean-Michel Guigner
- Sorbonne Université, Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie (IMPMC), CNRS UMR 7590, MNHN, IRD UR 206, 75005 Paris, France
| | - Mickaël M. Cohen
- Sorbonne Université, Institut de Biologie Physico-Chimique (IBPC), CNRS UMR 8226, Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes, 75005 Paris, France
| | - David Tareste
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), Inserm UMR-S 1266, Team Membrane Traffic in Healthy & Diseased Brain, 75014 Paris, France
| |
Collapse
|
36
|
Huang F, Xue H, Fu Y, Ouyang Y, Chen D, Xia F, Willner I. Three Compartment Liposome Fusion: Functional Protocells for Biocatalytic Cascades and Operation of Dynamic DNA Machineries. ADVANCED FUNCTIONAL MATERIALS 2023; 33. [DOI: 10.1002/adfm.202302814] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Indexed: 01/06/2025]
Abstract
AbstractNucleic acid‐functionalized liposomes modified at their boundaries with o‐nitrobenzyl phosphate‐caged hairpin units and pH‐responsive C‐G·C+ triplex forming strands are used for the concomitant light and pH‐triggered fusion of three types of loaded liposomes. The fusion processes are followed by light‐scattering size enlargement measurements, optical methods, and biocatalytic cascades activated upon the mixing of the liposomes loaded with enzymes and their substrates and their fusion into the cell‐like containments. The fused liposomes act as functional protocells for the integration of biocatalytic machineries. This is exemplified by the operation of an autonomous polymerization/nickase machinery synthesizing a Mg2+‐ion‐dependent DNAzyme and of a transcription machinery yielding the Malachite Green‐RNA aptamer product.
Collapse
Affiliation(s)
- Fujian Huang
- State Key Laboratory of Biogeology and Environmental Geology Engineering Research Center of Nano‐Geomaterials of Ministry of Education Faculty of Materials Science and Chemistry China University of Geosciences Wuhan 430074 China
- Hefei National Research Center for Physical Sciences at the Microscale University of Science and Technology of China Hefei 230026 China
| | - Huiying Xue
- State Key Laboratory of Biogeology and Environmental Geology Engineering Research Center of Nano‐Geomaterials of Ministry of Education Faculty of Materials Science and Chemistry China University of Geosciences Wuhan 430074 China
| | - Yuzhe Fu
- State Key Laboratory of Biogeology and Environmental Geology Engineering Research Center of Nano‐Geomaterials of Ministry of Education Faculty of Materials Science and Chemistry China University of Geosciences Wuhan 430074 China
| | - Yu Ouyang
- Institute of Chemistry and Center for Nanoscience and Nanotechnology The Hebrew University of Jerusalem Jerusalem 91904 Israel
| | - Danlong Chen
- State Key Laboratory of Biogeology and Environmental Geology Engineering Research Center of Nano‐Geomaterials of Ministry of Education Faculty of Materials Science and Chemistry China University of Geosciences Wuhan 430074 China
| | - Fan Xia
- State Key Laboratory of Biogeology and Environmental Geology Engineering Research Center of Nano‐Geomaterials of Ministry of Education Faculty of Materials Science and Chemistry China University of Geosciences Wuhan 430074 China
| | - Itamar Willner
- Institute of Chemistry and Center for Nanoscience and Nanotechnology The Hebrew University of Jerusalem Jerusalem 91904 Israel
| |
Collapse
|
37
|
Chiang YP, Li Z, He M, Jones Q, Pan M, Han X, Jiang XC. Sphingomyelin synthase-related protein SMSr is a phosphatidylethanolamine phospholipase C that promotes nonalcoholic fatty liver disease. J Biol Chem 2023; 299:105162. [PMID: 37586586 PMCID: PMC10494463 DOI: 10.1016/j.jbc.2023.105162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/28/2023] [Accepted: 08/04/2023] [Indexed: 08/18/2023] Open
Abstract
Sphingomyelin synthase (SMS)-related protein (SMSr) is a phosphatidylethanolamine phospholipase C (PE-PLC) that is conserved and ubiquitous in mammals. However, its biological function is still not clear. We previously observed that SMS1 deficiency-mediated glucosylceramide accumulation caused nonalcoholic fatty liver diseases (NAFLD), including nonalcoholic steatohepatitis (NASH) and liver fibrosis. Here, first, we evaluated high-fat diet/fructose-induced NAFLD in Smsr KO and WT mice. Second, we evaluated whether SMSr deficiency can reverse SMS1 deficiency-mediated NAFLD, using Sms1/Sms2 double and Sms1/Sms2/Smsr triple KO mice. We found that SMSr/PE-PLC deficiency attenuated high-fat diet/fructose-induced fatty liver and NASH, and attenuated glucosylceramide accumulation-induced NASH, fibrosis, and tumor formation. Further, we found that SMSr/PE-PLC deficiency reduced the expression of many inflammatory cytokines and fibrosis-related factors, and PE supplementation in vitro or in vivo mimicked the condition of SMSr/PE-PLC deficiency. Furthermore, we demonstrated that SMSr/PE-PLC deficiency or PE supplementation effectively prevented membrane-bound β-catenin transfer to the nucleus, thereby preventing tumor-related gene expression. Finally, we observed that patients with NASH had higher SMSr protein levels in the liver, lower plasma PE levels, and lower plasma PE/phosphatidylcholine ratios, and that human plasma PE levels are negatively associated with tumor necrosis factor-α and transforming growth factor β1 levels. In conclusion, SMSr/PE-PLC deficiency causes PE accumulation, which can attenuate fatty liver, NASH, and fibrosis. These results suggest that SMSr/PE-PLC inhibition therapy may mitigate NAFLD.
Collapse
Affiliation(s)
- Yeun-Po Chiang
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, New York, USA
| | - Zhiqiang Li
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, New York, USA
| | - Mulin He
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, New York, USA
| | - Quiana Jones
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, New York, USA
| | - Meixia Pan
- Lipidomics Core, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Xianlin Han
- Lipidomics Core, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Xian-Cheng Jiang
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, New York, USA; Molecular and Cellular Cardiology Program, VA New York Harbor Healthcare System, Brooklyn, New York, USA.
| |
Collapse
|
38
|
Li Z, Wang J, O’Hagan MP, Huang F, Xia F, Willner I. Dynamic Fusion of Nucleic Acid Functionalized Nano-/Micro-Cell-Like Containments: From Basic Concepts to Applications. ACS NANO 2023; 17:15308-15327. [PMID: 37549398 PMCID: PMC10448756 DOI: 10.1021/acsnano.3c04415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 08/01/2023] [Indexed: 08/09/2023]
Abstract
Membrane fusion processes play key roles in biological transformations, such as endocytosis/exocytosis, signal transduction, neurotransmission, or viral infections, and substantial research efforts have been directed to emulate these functions by artificial means. The recognition and dynamic reconfiguration properties of nucleic acids provide a versatile means to induce membrane fusion. Here we address recent advances in the functionalization of liposomes or membranes with structurally engineered lipidated nucleic acids guiding the fusion of cell-like containments, and the biophysical and chemical parameters controlling the fusion of the liposomes will be discussed. Intermembrane bridging by duplex or triplex nucleic acids and light-induced activation of membrane-associated nucleic acid constituents provide the means for spatiotemporal fusion of liposomes or nucleic acid modified liposome fusion with native cell membranes. The membrane fusion processes lead to exchange of loads in the fused containments and are a means to integrate functional assemblies. This is exemplified with the operation of biocatalytic cascades and dynamic DNA polymerization/nicking or transcription machineries in fused protocell systems. Membrane fusion processes of protocell assemblies are found to have important drug-delivery, therapeutic, sensing, and biocatalytic applications. The future challenges and perspectives of DNA-guided fused containments and membranes are addressed.
Collapse
Affiliation(s)
- Zhenzhen Li
- The
Institute of Chemistry, The Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Jianbang Wang
- The
Institute of Chemistry, The Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Michael P. O’Hagan
- The
Institute of Chemistry, The Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Fujian Huang
- State
Key Laboratory of Biogeology and Environmental Geology, Engineering
Research Center of Nano-Geomaterials of Ministry of Education, Faculty
of Materials Science and Chemistry, China
University of Geosciences, Wuhan 430074, People’s Republic of China
| | - Fan Xia
- State
Key Laboratory of Biogeology and Environmental Geology, Engineering
Research Center of Nano-Geomaterials of Ministry of Education, Faculty
of Materials Science and Chemistry, China
University of Geosciences, Wuhan 430074, People’s Republic of China
| | - Itamar Willner
- The
Institute of Chemistry, The Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| |
Collapse
|
39
|
Mathew L, Verma DK, Liang K, Duan M, Dadhich R, Kapoor S. Fusion Landscape of Mycobacterial Envelope-Derived Lipid Vesicles with Intact Bacteria Dictates High Intracellular Drug Retention. ACS APPLIED BIO MATERIALS 2023; 6:3066-3073. [PMID: 37493278 DOI: 10.1021/acsabm.3c00286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
Membrane vesicles are critical regulators of pathogenic diseases. In tubercular infections, the use of mycobacteria derived vesicles as delivery vehicles to overcome drug resistance and complex treatment regimens has never been attempted. Here, we first address how these vesicles interact with their target cells, especially via membrane fusion. Membrane fusion between alike mycobacterial outer and inner membrane layer-derived lipid vesicles is shown to be driven by the structural, geometrical, and biophysical attributes of constituent lipids. The increased fusion of outer-membrane-derived vesicles with intact bacteria ensures enhanced intracellular drug levels and is presented as a "natural" antitubercular drug delivery vehicle.
Collapse
Affiliation(s)
- Lydia Mathew
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Dheeraj Kumar Verma
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Kuan Liang
- National Centre for Magnetic Resonance in Wuhan, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, Hubei, China
| | - Mojie Duan
- National Centre for Magnetic Resonance in Wuhan, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, Hubei, China
| | - Ruchika Dadhich
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Shobhna Kapoor
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai 400076, India
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8528, Japan
| |
Collapse
|
40
|
Azimi FC, Dean TT, Minari K, Basso LGM, Vance TDR, Serrão VHB. A Frame-by-Frame Glance at Membrane Fusion Mechanisms: From Viral Infections to Fertilization. Biomolecules 2023; 13:1130. [PMID: 37509166 PMCID: PMC10377500 DOI: 10.3390/biom13071130] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/09/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Viral entry and fertilization are distinct biological processes that share a common mechanism: membrane fusion. In viral entry, enveloped viruses attach to the host cell membrane, triggering a series of conformational changes in the viral fusion proteins. This results in the exposure of a hydrophobic fusion peptide, which inserts into the host membrane and brings the viral and host membranes into close proximity. Subsequent structural rearrangements in opposing membranes lead to their fusion. Similarly, membrane fusion occurs when gametes merge during the fertilization process, though the exact mechanism remains unclear. Structural biology has played a pivotal role in elucidating the molecular mechanisms underlying membrane fusion. High-resolution structures of the viral and fertilization fusion-related proteins have provided valuable insights into the conformational changes that occur during this process. Understanding these mechanisms at a molecular level is essential for the development of antiviral therapeutics and tools to influence fertility. In this review, we will highlight the biological importance of membrane fusion and how protein structures have helped visualize both common elements and subtle divergences in the mechanisms behind fusion; in addition, we will examine the new tools that recent advances in structural biology provide researchers interested in a frame-by-frame understanding of membrane fusion.
Collapse
Affiliation(s)
- Farshad C. Azimi
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada;
| | - Trevor T. Dean
- Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL 60612, USA;
| | - Karine Minari
- Biomolecular Cryo-Electron Microscopy Facility, University of California-Santa Cruz, Santa Cruz, CA 95064, USA;
| | - Luis G. M. Basso
- Laboratório de Ciências Físicas, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, Rio de Janeiro 28013-602, Brazil;
| | - Tyler D. R. Vance
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada;
| | - Vitor Hugo B. Serrão
- Biomolecular Cryo-Electron Microscopy Facility, University of California-Santa Cruz, Santa Cruz, CA 95064, USA;
- Department of Chemistry and Biochemistry, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| |
Collapse
|
41
|
Liu F, Dong Z, Li M, Sun J, Hou Z, Younas A, Wan X, Shang H, Zhang N. A macrophage plasma membrane-coated and DNA structured nanomedicine targets to alleviate rheumatoid arthritis via dual inhibition to TNF-α and NF-κB. Int J Pharm 2023:123188. [PMID: 37394158 DOI: 10.1016/j.ijpharm.2023.123188] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/10/2023] [Accepted: 06/27/2023] [Indexed: 07/04/2023]
Abstract
High heterogenicity of rheumatoid arthritis (RA) leads to poor response in many patients. Combined therapies that simultaneously inhibit multiple proinflammatory targets may improve anti-RA efficacy. However, which monotherapies to combine and how to achieve the combination are critical issues. Here, we design a macrophage plasma membrane-coated and DNA structured nanomedicine to achieve a dual inhibitory therapy to Tumor necrosis factor alpha (TNF-α) and NF-κB. An anti-NF-κB decoy oligodeoxynucleotides (dODN) is first conjugated to a DNA cage with precise numbers and locations (Cage-dODN). Meanwhile, an anti-TNF-α siRNA is anchored to extracted macrophage plasma membrane (siRNA@M). Subsequently, siRNA@M is used to encapsulate Cage-dODN to fabricate siRNA@M(Cage-dODN) (siMCO). The size and zeta potential of siMCO are 63.1 ± 15.7 nm and -20.7 ± 3.8 mV respectively. siMCO shows increased intracellular uptake by inflamed macrophages and enhanced accumulation in inflamed mouse paws. siMCO also reduces pro-inflammatory factors at genetic and protein levels, alleviates arthritic symptoms, and shows no influence to major blood components. These results show that siMCO is a potential targeted, efficient, and safe dual inhibitory therapy for the treatment of inflammatory arthritis. The macrophage plasma membrane can be utilized to improve the targeting, stability, and efficacy of DNA structured nanomedicines.
Collapse
Affiliation(s)
- Fenfen Liu
- Department of Pharmaceutics and Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases of Henan Province, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P.R. China; School of Pharmaceutical Sciences, Zhengzhou University of Industrial Technology, Xinzheng City 451100, Henan, P.R.China.
| | - Zhuolin Dong
- Department of Pharmaceutics and Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases of Henan Province, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P.R. China.
| | - Mengru Li
- Department of Pharmaceutics and Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases of Henan Province, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P.R. China.
| | - Junhong Sun
- Department of Pharmaceutics and Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases of Henan Province, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P.R. China; Bayinguoleng Mongolian Autonomous Prefecture People's Hospital, Korla 841000, Xinjiang Uygur Autonomous Region, P.R. China.
| | - Ziye Hou
- Department of Pharmaceutics and Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases of Henan Province, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P.R. China.
| | - Ayesha Younas
- Department of Pharmaceutics and Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases of Henan Province, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P.R. China.
| | - Xiangling Wan
- Department of Pharmaceutics and Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases of Henan Province, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P.R. China.
| | - Hongtao Shang
- School of Sport Sciences (main campus), Zhengzhou University, Zhengzhou 450001, Henan, P.R. China.
| | - Nan Zhang
- Department of Pharmaceutics and Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases of Henan Province, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P.R. China.
| |
Collapse
|
42
|
Martínez-Mármol R, Giordano-Santini R, Kaulich E, Cho AN, Przybyla M, Riyadh MA, Robinson E, Chew KY, Amor R, Meunier FA, Balistreri G, Short KR, Ke YD, Ittner LM, Hilliard MA. SARS-CoV-2 infection and viral fusogens cause neuronal and glial fusion that compromises neuronal activity. SCIENCE ADVANCES 2023; 9:eadg2248. [PMID: 37285437 PMCID: PMC10246911 DOI: 10.1126/sciadv.adg2248] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 05/01/2023] [Indexed: 06/09/2023]
Abstract
Numerous viruses use specialized surface molecules called fusogens to enter host cells. Many of these viruses, including the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), can infect the brain and are associated with severe neurological symptoms through poorly understood mechanisms. We show that SARS-CoV-2 infection induces fusion between neurons and between neurons and glia in mouse and human brain organoids. We reveal that this is caused by the viral fusogen, as it is fully mimicked by the expression of the SARS-CoV-2 spike (S) protein or the unrelated fusogen p15 from the baboon orthoreovirus. We demonstrate that neuronal fusion is a progressive event, leads to the formation of multicellular syncytia, and causes the spread of large molecules and organelles. Last, using Ca2+ imaging, we show that fusion severely compromises neuronal activity. These results provide mechanistic insights into how SARS-CoV-2 and other viruses affect the nervous system, alter its function, and cause neuropathology.
Collapse
Affiliation(s)
- Ramón Martínez-Mármol
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Rosina Giordano-Santini
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Eva Kaulich
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Ann-Na Cho
- Dementia Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Magdalena Przybyla
- Dementia Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Md Asrafuzzaman Riyadh
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Emilija Robinson
- Dementia Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Keng Yih Chew
- School of Chemistry and Molecular Biosciences, Faculty of Science, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Rumelo Amor
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Frédéric A. Meunier
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Giuseppe Balistreri
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
- Department of Virology, Faculty of Medicine, University of Helsinki, Helsinki FIN-00014, Finland
| | - Kirsty R. Short
- School of Chemistry and Molecular Biosciences, Faculty of Science, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Yazi D. Ke
- Dementia Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Lars M. Ittner
- Dementia Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Massimo A. Hilliard
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
43
|
Amm I, Weberruss M, Hellwig A, Schwarz J, Tatarek-Nossol M, Lüchtenborg C, Kallas M, Brügger B, Hurt E, Antonin W. Distinct domains in Ndc1 mediate its interaction with the Nup84 complex and the nuclear membrane. J Cell Biol 2023; 222:e202210059. [PMID: 37154843 PMCID: PMC10165475 DOI: 10.1083/jcb.202210059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 01/31/2023] [Accepted: 03/17/2023] [Indexed: 05/10/2023] Open
Abstract
Nuclear pore complexes (NPCs) are embedded in the nuclear envelope and built from ∼30 different nucleoporins (Nups) in multiple copies, few are integral membrane proteins. One of these transmembrane nucleoporins, Ndc1, is thought to function in NPC assembly at the fused inner and outer nuclear membranes. Here, we show a direct interaction of Ndc1's transmembrane domain with Nup120 and Nup133, members of the pore membrane coating Y-complex. We identify an amphipathic helix in Ndc1's C-terminal domain binding highly curved liposomes. Upon overexpression, this amphipathic motif is toxic and dramatically alters the intracellular membrane organization in yeast. Ndc1's amphipathic motif functionally interacts with related motifs in the C-terminus of the nucleoporins Nup53 and Nup59, important for pore membrane binding and interconnecting NPC modules. The essential function of Ndc1 can be suppressed by deleting the amphipathic helix from Nup53. Our data indicate that nuclear membrane and presumably NPC biogenesis depends on a balanced ratio between amphipathic motifs in diverse nucleoporins.
Collapse
Affiliation(s)
- Ingo Amm
- Heidelberg University Biochemistry Center (BZH), University of Heidelberg, Heidelberg, Germany
| | - Marion Weberruss
- Institute of Biochemistry and Molecular Cell Biology, Medical School, RWTH Aachen University, Aachen, Germany
| | - Andrea Hellwig
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, Heidelberg, Germany
| | - Johannes Schwarz
- Heidelberg University Biochemistry Center (BZH), University of Heidelberg, Heidelberg, Germany
| | - Marianna Tatarek-Nossol
- Institute of Biochemistry and Molecular Cell Biology, Medical School, RWTH Aachen University, Aachen, Germany
| | - Christian Lüchtenborg
- Heidelberg University Biochemistry Center (BZH), University of Heidelberg, Heidelberg, Germany
| | - Martina Kallas
- Heidelberg University Biochemistry Center (BZH), University of Heidelberg, Heidelberg, Germany
| | - Britta Brügger
- Heidelberg University Biochemistry Center (BZH), University of Heidelberg, Heidelberg, Germany
| | - Ed Hurt
- Heidelberg University Biochemistry Center (BZH), University of Heidelberg, Heidelberg, Germany
| | - Wolfram Antonin
- Institute of Biochemistry and Molecular Cell Biology, Medical School, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
44
|
Romani A, Sergi D, Zauli E, Voltan R, Lodi G, Vaccarezza M, Caruso L, Previati M, Zauli G. Nutrients, herbal bioactive derivatives and commensal microbiota as tools to lower the risk of SARS-CoV-2 infection. Front Nutr 2023; 10:1152254. [PMID: 37324739 PMCID: PMC10267353 DOI: 10.3389/fnut.2023.1152254] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 05/15/2023] [Indexed: 06/17/2023] Open
Abstract
The SARS-CoV-2 outbreak has infected a vast population across the world, causing more than 664 million cases and 6.7 million deaths by January 2023. Vaccination has been effective in reducing the most critical aftermath of this infection, but some issues are still present regarding re-infection prevention, effectiveness against variants, vaccine hesitancy and worldwide accessibility. Moreover, although several old and new antiviral drugs have been tested, we still lack robust and specific treatment modalities. It appears of utmost importance, facing this continuously growing pandemic, to focus on alternative practices grounded on firm scientific bases. In this article, we aim to outline a rigorous scientific background and propose complementary nutritional tools useful toward containment, and ultimately control, of SARS-CoV-2 infection. In particular, we review the mechanisms of viral entry and discuss the role of polyunsaturated fatty acids derived from α-linolenic acid and other nutrients in preventing the interaction of SARS-CoV-2 with its entry gateways. In a similar way, we analyze in detail the role of herbal-derived pharmacological compounds and specific microbial strains or microbial-derived polypeptides in the prevention of SARS-CoV-2 entry. In addition, we highlight the role of probiotics, nutrients and herbal-derived compounds in stimulating the immunity response.
Collapse
Affiliation(s)
- Arianna Romani
- Department of Environmental and Prevention Sciences and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Domenico Sergi
- Department of Translational Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Enrico Zauli
- Department of Translational Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Rebecca Voltan
- Department of Environmental and Prevention Sciences and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Giada Lodi
- Department of Environmental and Prevention Sciences and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Mauro Vaccarezza
- Curtin Medical School & Curtin Health Innovation Research Institute (CHIRI), Faculty of Health Sciences, Curtin University, Perth, WA, Australia
| | - Lorenzo Caruso
- Department of Environmental and Prevention Sciences and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Maurizio Previati
- Department of Translational Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Giorgio Zauli
- Research Department, King Khaled Eye Specialistic Hospital, Riyadh, Saudi Arabia
| |
Collapse
|
45
|
Yesylevskyy S, Martinez-Seara H, Jungwirth P. Curvature Matters: Modeling Calcium Binding to Neutral and Anionic Phospholipid Bilayers. J Phys Chem B 2023; 127:4523-4531. [PMID: 37191140 DOI: 10.1021/acs.jpcb.3c01962] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
In this work, the influence of membrane curvature on the Ca2+ binding to phospholipid bilayers is investigated by means of molecular dynamics simulations. In particular, we compared Ca2+ binding to flat, elastically buckled, or uniformly bent zwitterionic and anionic phospholipid bilayers. We demonstrate that Ca2+ ions bind preferably to the concave membrane surfaces in both types of bilayers. We also show that the membrane curvature leads to pronounced changes in Ca2+ binding including differences in free ion concentrations, lipid coordination distributions, and the patterns of ion binding to different chemical groups of lipids. Moreover, these effects differ substantially for the concave and convex membrane monolayers. Comparison between force fields with either full or scaled charges indicates that charge scaling results in reduction of the Ca2+ binding to curved phosphatidylserine bilayers, while for phosphatidylcholine membranes, calcium binds only weakly for both force fields.
Collapse
Affiliation(s)
- Semen Yesylevskyy
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Flemingovo náměstí 542/2, 160 00 Prague 6, Czech Republic
- Department of Physics of Biological Systems, Institute of Physics of the National Academy of Sciences of Ukraine, Nauky Avenue 46, 03038 Kyiv, Ukraine
- Receptor.AI Incorporated, 20-22 Wenlock Road, N1 7GU London, U.K
| | - Hector Martinez-Seara
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Flemingovo náměstí 542/2, 160 00 Prague 6, Czech Republic
| | - Pavel Jungwirth
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Flemingovo náměstí 542/2, 160 00 Prague 6, Czech Republic
| |
Collapse
|
46
|
Toyooka K, Goto Y, Hashimoto K, Wakazaki M, Sato M, Hirai MY. Endoplasmic Reticulum Bodies in the Lateral Root Cap Are Involved in the Direct Transport of Beta-Glucosidase to Vacuoles. PLANT & CELL PHYSIOLOGY 2023; 64:461-473. [PMID: 36617247 DOI: 10.1093/pcp/pcac177] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 12/22/2022] [Accepted: 01/05/2023] [Indexed: 05/17/2023]
Abstract
Programmed cell death (PCD) in lateral root caps (LRCs) is crucial for maintaining root cap functionality. Endoplasmic reticulum (ER) bodies play important roles in plant immunity and PCD. However, the distribution of ER bodies and their communication with vacuoles in the LRC remain elusive. In this study, we investigated the ultrastructure of LRC cells of wild-type and transgenic Arabidopsis lines using an auto-acquisition transmission electron microscope (TEM) system and high-pressure freezing. Gigapixel-scale high-resolution TEM imaging of the transverse and longitudinal sections of roots followed by three-dimensional imaging identified sausage-shaped structures budding from the ER. These were subsequently identified as ER bodies using GFPh transgenic lines expressing green fluorescent protein (GFP) fused with an ER retention signal (HDEL). Immunogold labeling using an anti-GFP antibody detected GFP signals in the ER bodies and vacuoles. The fusion of ER bodies with vacuoles in LRC cells was identified using correlative light and electron microscopy. Imaging of the root tips of a GFPh transgenic line with a PYK10 promoter revealed the localization of PYK10, a member of the β-glucosidase family with an ER retention signal, in the ER bodies in the inner layer along with a fusion of ER bodies with vacuoles in the middle layer and collapse of vacuoles in the outer layer of the LRC. These findings suggest that ER bodies in LRC directly transport β-glucosidases to the vacuoles, and that a subsequent vacuolar collapse triggered by an unknown mechanism releases protective substances to the growing root tip to protect it from the invaders.
Collapse
Affiliation(s)
- Kiminori Toyooka
- RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045 Japan
| | - Yumi Goto
- RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045 Japan
| | - Kei Hashimoto
- RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045 Japan
| | - Mayumi Wakazaki
- RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045 Japan
| | - Mayuko Sato
- RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045 Japan
| | - Masami Yokota Hirai
- RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045 Japan
- Department of Applied Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8601 Japan
| |
Collapse
|
47
|
Hindi SM, Petrany MJ, Greenfeld E, Focke LC, Cramer AAW, Whitt MA, Khairallah RJ, Ward CW, Chamberlain JS, Podbilewicz B, Prasad V, Millay DP. Enveloped viruses pseudotyped with mammalian myogenic cell fusogens target skeletal muscle for gene delivery. Cell 2023; 186:2062-2077.e17. [PMID: 37075755 PMCID: PMC11181154 DOI: 10.1016/j.cell.2023.03.033] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 02/08/2023] [Accepted: 03/28/2023] [Indexed: 04/21/2023]
Abstract
Entry of enveloped viruses into cells is mediated by viral fusogenic proteins that drive membrane rearrangements needed for fusion between viral and target membranes. Skeletal muscle development also requires membrane fusion events between progenitor cells to form multinucleated myofibers. Myomaker and Myomerger are muscle-specific cell fusogens but do not structurally or functionally resemble classical viral fusogens. We asked whether the muscle fusogens could functionally substitute for viral fusogens, despite their structural distinctiveness, and fuse viruses to cells. We report that engineering of Myomaker and Myomerger on the membrane of enveloped viruses leads to specific transduction of skeletal muscle. We also demonstrate that locally and systemically injected virions pseudotyped with the muscle fusogens can deliver μDystrophin to skeletal muscle of a mouse model of Duchenne muscular dystrophy and alleviate pathology. Through harnessing the intrinsic properties of myogenic membranes, we establish a platform for delivery of therapeutic material to skeletal muscle.
Collapse
Affiliation(s)
- Sajedah M Hindi
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Michael J Petrany
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Elena Greenfeld
- Department of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Leah C Focke
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Alyssa A W Cramer
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Michael A Whitt
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA
| | | | - Christopher W Ward
- Department of Orthopedics and Center for Biomedical Engineering and Technology (BioMET), University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jeffrey S Chamberlain
- Departments of Neurology, Medicine and Biochemistry, Wellstone Muscular Dystrophy Specialized Research Center, University of Washington School of Medicine, Seattle, WA, USA
| | | | - Vikram Prasad
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Douglas P Millay
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
48
|
Sakuragi T, Nagata S. Regulation of phospholipid distribution in the lipid bilayer by flippases and scramblases. Nat Rev Mol Cell Biol 2023:10.1038/s41580-023-00604-z. [PMID: 37106071 PMCID: PMC10134735 DOI: 10.1038/s41580-023-00604-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2023] [Indexed: 04/29/2023]
Abstract
Cellular membranes function as permeability barriers that separate cells from the external environment or partition cells into distinct compartments. These membranes are lipid bilayers composed of glycerophospholipids, sphingolipids and cholesterol, in which proteins are embedded. Glycerophospholipids and sphingolipids freely move laterally, whereas transverse movement between lipid bilayers is limited. Phospholipids are asymmetrically distributed between membrane leaflets but change their location in biological processes, serving as signalling molecules or enzyme activators. Designated proteins - flippases and scramblases - mediate this lipid movement between the bilayers. Flippases mediate the confined localization of specific phospholipids (phosphatidylserine (PtdSer) and phosphatidylethanolamine) to the cytoplasmic leaflet. Scramblases randomly scramble phospholipids between leaflets and facilitate the exposure of PtdSer on the cell surface, which serves as an important signalling molecule and as an 'eat me' signal for phagocytes. Defects in flippases and scramblases cause various human diseases. We herein review the recent research on the structure of flippases and scramblases and their physiological roles. Although still poorly understood, we address the mechanisms by which they translocate phospholipids between lipid bilayers and how defects cause human diseases.
Collapse
Affiliation(s)
- Takaharu Sakuragi
- Biochemistry & Immunology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Shigekazu Nagata
- Biochemistry & Immunology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.
| |
Collapse
|
49
|
Milogrodzka I, Nguyen Pham DT, Sama GR, Samadian H, Zhai J, de Campo L, Kirby NM, Scott TF, Banaszak Holl MM, van 't Hag L. Effect of Cholesterol on Biomimetic Membrane Curvature and Coronavirus Fusion Peptide Encapsulation. ACS NANO 2023; 17:8598-8612. [PMID: 37078604 DOI: 10.1021/acsnano.3c01095] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Biomimetic cubic phases can be used for protein encapsulation in a variety of applications such as biosensors and drug delivery. Cubic phases with a high concentration of cholesterol and phospholipids were obtained herein. It is shown that the cubic phase structure can be maintained with a higher concentration of biomimetic membrane additives than has been reported previously. Opposing effects on the curvature of the membrane were observed upon the addition of phospholipids and cholesterol. Furthermore, the coronavirus fusion peptide significantly increased the negative curvature of the biomimetic membrane with cholesterol. We show that the viral fusion peptide can undergo structural changes leading to the formation of hydrophobic α-helices that insert into the lipid bilayer. This is of high importance, as a fusion peptide that induces increased negative curvature as shown by the formation of inverse hexagonal phases allows for greater contact area between two membranes, which is required for viral fusion to occur. The cytotoxicity assay showed that the toxicity toward HeLa cells was dramatically decreased when the cholesterol or peptide level in the nanoparticles increased. This suggests that the addition of cholesterol can improve the biocompatibility of the cubic phase nanoparticles, making them safer for use in biomedical applications. As the results, this work improves the potential for the biomedical end-use applications of the nonlamellar lipid nanoparticles and shows the need of systematic formulation studies due to the complex interplay of all components.
Collapse
Affiliation(s)
- Izabela Milogrodzka
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Duy Tue Nguyen Pham
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Gopal R Sama
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Hajar Samadian
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Jiali Zhai
- School of Science, STEM College, RMIT University, Melbourne, VIC 3000, Australia
| | - Liliana de Campo
- Australian Centre for Neutron Scattering, Australian Nuclear Science and Technology Organisation, Kirrawee, NSW 2234, Australia
| | - Nigel M Kirby
- Australian Synchrotron, 800 Blackburn Road, Clayton, VIC 3168, Australia
| | - Timothy F Scott
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia
- Department of Materials Science and Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Mark M Banaszak Holl
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia
- Department of Mechanical and Materials Engineering, University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Leonie van 't Hag
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia
| |
Collapse
|
50
|
Bach H, Lorenzo-Leal AC. Use of niosomes for the treatment of intracellular pathogens infecting the lungs. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023:e1891. [PMID: 37032602 DOI: 10.1002/wnan.1891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 02/24/2023] [Accepted: 03/14/2023] [Indexed: 06/19/2023]
Abstract
The delivery of drugs in an encapsulated environment is designed to precisely target specific tissues, avoiding a systemic circulation of the drug. Lungs are organs exposed to the environment with multiple defense barriers. However, many pathogens can still colonize and infect the airways bypassing the hostile environment of the lungs. In more complicated situations, some pathogens have developed strategies to multiply and survive within macrophages, one of the first immune cell responses to clearing infections in mammals. Niosomes are artificial vesicles that can be loaded with drugs, offering an alternative strategy to treat intracellular pathogens as nanocarriers. Members of the mycobacteria genus are intracellular pathogens that have evolved to escape the immunological response, specifically in macrophages, the white cells responsible for the clearance of pathogens. This review analyzed the state-of-the-art niosome synthesis aimed at tackling the problem of intracellular pathogen therapy. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Horacio Bach
- Department of Medicine, Division of Infectious Diseases, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ana C Lorenzo-Leal
- Department of Medicine, Division of Infectious Diseases, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|