1
|
Goldmann O, Medina E. Revisiting Pathogen Exploitation of Clathrin-Independent Endocytosis: Mechanisms and Implications. Cells 2025; 14:731. [PMID: 40422234 DOI: 10.3390/cells14100731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Revised: 05/04/2025] [Accepted: 05/13/2025] [Indexed: 05/28/2025] Open
Abstract
Endocytosis is a specialized transport mechanism in which the cell membrane folds inward to enclose large molecules, fluids, or particles, forming vesicles that are transported within the cell. It plays a crucial role in nutrient uptake, immune responses, and cellular communication. However, many pathogens exploit the endocytic pathway to invade and survive within host cells, allowing them to evade the immune system and establish infection. Endocytosis can be classified as clathrin-mediated (CME) or clathrin-independent (CIE), based on the mechanism of vesicle formation. Unlike CME, which involves the formation of clathrin-coated vesicles that bud from the plasma membrane, CIE does not rely on clathrin-coated vesicles. Instead, other mechanisms facilitate membrane invagination and vesicle formation. CIE encompasses a variety of pathways, including caveolin-mediated, Arf6-dependent, and flotillin-dependent pathways. In this review, we discuss key features of CIE pathways, including cargo selection, vesicle formation, routes taken by internalized cargo, and the regulatory mechanisms governing CIE. Many viruses and bacteria hijack host cell CIE mechanisms to facilitate intracellular trafficking and persistence. We also revisit the exploitation of CIE by bacterial and viral pathogens, highlighting recent discoveries in entry mechanisms, intracellular fate, and host-pathogen interactions. Understanding how pathogens manipulate CIE in host cells can inform the development of novel antimicrobial and immunomodulatory interventions, offering new avenues for disease prevention and treatment.
Collapse
Affiliation(s)
- Oliver Goldmann
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Eva Medina
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| |
Collapse
|
2
|
Zhang Q, Qu Y, Zhao H, Chen S, Liu Z, Li J, Li Y, Li J, Sun D. A Magnetically Driven Biodegradable Microsphere with Mass Production Capability for Subunit Vaccine Delivery and Enhanced Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:50344-50359. [PMID: 39265074 DOI: 10.1021/acsami.4c10301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/14/2024]
Abstract
Subunit vaccines have emerged as a promising strategy in immunotherapy for combating viral infections and cancer. Nevertheless, the clinical application of subunit vaccines is hindered by limitations in antigen delivery efficiency, characterized by rapid clearance and inadequate cellular uptake. Here, a novel subunit vaccine delivery system utilizing ovalbumin@magnetic nanoparticles (OVA@MNPs) encapsulated within biodegradable gelatin methacryloyl (GelMA) microspheres was proposed to enhance the efficacy of antigen delivery. OVA@MNPs-loaded GelMA microspheres, denoted as OMGMs, can be navigated through magnetic fields to deliver subunit vaccines into the lymphatic system efficiently. Moreover, the biodegradable OMGMs enabled the sustained release of subunit vaccines, concentrating OVA around lymph nodes and enhancing the efficacy of induced immune response. OMGMs were produced through a microfluidic droplet generation technique, enabling mass production. In murine models, OMGMs successfully accumulated antigens in lymph nodes abundant in antigen-presenting cells, leading to enhanced cellular and humoral immunity and pronounced antitumor effects with a single booster immunization. In conclusion, these findings highlight the promise of OMGMs as a practical subunit vaccination approach, thus addressing the limitations associated with antigen delivery efficiency and paving the way for advanced immunotherapeutic strategies.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Biomedical Engineering, City University of Hong Kong, 999077 Hong Kong, SAR, China
| | - Yun Qu
- Department of Biomedical Engineering, City University of Hong Kong, 999077 Hong Kong, SAR, China
| | - Han Zhao
- Department of Biomedical Engineering, City University of Hong Kong, 999077 Hong Kong, SAR, China
| | - Shuxun Chen
- Department of Biomedical Engineering, City University of Hong Kong, 999077 Hong Kong, SAR, China
| | - Zhen Liu
- Department of Biomedical Engineering, City University of Hong Kong, 999077 Hong Kong, SAR, China
| | - Jianing Li
- Department of Biomedical Engineering, City University of Hong Kong, 999077 Hong Kong, SAR, China
| | - Yanfang Li
- Department of Biomedical Engineering, City University of Hong Kong, 999077 Hong Kong, SAR, China
| | - Junyang Li
- Department of Electronic Engineering, Ocean University of China, Qingdao 266000, China
| | - Dong Sun
- Department of Biomedical Engineering, City University of Hong Kong, 999077 Hong Kong, SAR, China
| |
Collapse
|
3
|
Zheng Y, Luo S, Xu M, He Q, Xie J, Wu J, Huang Y. Transepithelial transport of nanoparticles in oral drug delivery: From the perspective of surface and holistic property modulation. Acta Pharm Sin B 2024; 14:3876-3900. [PMID: 39309496 PMCID: PMC11413706 DOI: 10.1016/j.apsb.2024.06.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/23/2024] [Accepted: 05/25/2024] [Indexed: 09/25/2024] Open
Abstract
Despite the promising prospects of nanoparticles in oral drug delivery, the process of oral administration involves a complex transportation pathway that includes cellular uptake, intracellular trafficking, and exocytosis by intestinal epithelial cells, which are necessary steps for nanoparticles to enter the bloodstream and exert therapeutic effects. Current researchers have identified several crucial factors that regulate the interaction between nanoparticles and intestinal epithelial cells, including surface properties such as ligand modification, surface charge, hydrophilicity/hydrophobicity, intestinal protein corona formation, as well as holistic properties like particle size, shape, and rigidity. Understanding these properties is essential for enhancing transepithelial transport efficiency and designing effective oral drug delivery systems. Therefore, this review provides a comprehensive overview of the surface and holistic properties that influence the transepithelial transport of nanoparticles, elucidating the underlying principles governing their impact on transepithelial transport. The review also outlines the chosen of parameters to be considered for the subsequent design of oral drug delivery systems.
Collapse
Affiliation(s)
- Yaxian Zheng
- Department of Pharmacy, the Third People's Hospital of Chengdu, the Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Shiqin Luo
- Department of Pharmacy, the Third People's Hospital of Chengdu, the Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Min Xu
- Department of Pharmacy, the Third People's Hospital of Chengdu, the Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Qin He
- Department of Pharmacy, the Third People's Hospital of Chengdu, the Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Jiang Xie
- Department of Pharmacy, the Third People's Hospital of Chengdu, the Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Jiawei Wu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Yuan Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
4
|
Kulma M, Šakanović A, Bedina-Zavec A, Caserman S, Omersa N, Šolinc G, Orehek S, Hafner-Bratkovič I, Kuhar U, Slavec B, Krapež U, Ocepek M, Kobayashi T, Kwiatkowska K, Jerala R, Podobnik M, Anderluh G. Sequestration of membrane cholesterol by cholesterol-binding proteins inhibits SARS-CoV-2 entry into Vero E6 cells. Biochem Biophys Res Commun 2024; 716:149954. [PMID: 38704887 DOI: 10.1016/j.bbrc.2024.149954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 03/26/2024] [Accepted: 04/15/2024] [Indexed: 05/07/2024]
Abstract
Membrane lipids and proteins form dynamic domains crucial for physiological and pathophysiological processes, including viral infection. Many plasma membrane proteins, residing within membrane domains enriched with cholesterol (CHOL) and sphingomyelin (SM), serve as receptors for attachment and entry of viruses into the host cell. Among these, human coronaviruses, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), use proteins associated with membrane domains for initial binding and internalization. We hypothesized that the interaction of lipid-binding proteins with CHOL in plasma membrane could sequestrate lipids and thus affect the efficiency of virus entry into host cells, preventing the initial steps of viral infection. We have prepared CHOL-binding proteins with high affinities for lipids in the plasma membrane of mammalian cells. Binding of the perfringolysin O domain four (D4) and its variant D4E458L to membrane CHOL impaired the internalization of the receptor-binding domain of the SARS-CoV-2 spike protein and the pseudovirus complemented with the SARS-CoV-2 spike protein. SARS-CoV-2 replication in Vero E6 cells was also decreased. Overall, our results demonstrate that the integrity of CHOL-rich membrane domains and the accessibility of CHOL in the membrane play an essential role in SARS-CoV-2 cell entry.
Collapse
Affiliation(s)
- Magdalena Kulma
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Aleksandra Šakanović
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Apolonija Bedina-Zavec
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Simon Caserman
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Neža Omersa
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Gašper Šolinc
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Sara Orehek
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
| | - Iva Hafner-Bratkovič
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia; EN-FIST Centre of Excellence, Trg Osvobodilne Fronte 13, 1000, Ljubljana, Slovenia
| | - Urška Kuhar
- Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000, Ljubljana, Slovenia
| | - Brigita Slavec
- Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000, Ljubljana, Slovenia
| | - Uroš Krapež
- Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000, Ljubljana, Slovenia
| | - Matjaž Ocepek
- Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000, Ljubljana, Slovenia
| | - Toshihide Kobayashi
- Lipid Biology Laboratory, RIKEN, 2-1, Hirosawa, Wako-shi, Saitama, 351-0198, Japan; UMR 7021 CNRS, Université de Strasbourg, F-67401, Illkirch, France
| | - Katarzyna Kwiatkowska
- Laboratory of Molecular Membrane Biology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, 3 Pasteur St., 02-093, Warsaw, Poland
| | - Roman Jerala
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia; EN-FIST Centre of Excellence, Trg Osvobodilne Fronte 13, 1000, Ljubljana, Slovenia
| | - Marjetka Podobnik
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Gregor Anderluh
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia.
| |
Collapse
|
5
|
Pedreañez A, Carrero Y, Vargas R, Hernández-Fonseca JP, Mosquera JA. Role of angiotensin II in cellular entry and replication of dengue virus. Arch Virol 2024; 169:121. [PMID: 38753119 DOI: 10.1007/s00705-024-06040-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 02/19/2024] [Indexed: 05/21/2024]
Abstract
Previous studies have demonstrated the relevance of several soluble molecules in the pathogenesis of dengue. In this regard, a possible role for angiotensin II (Ang II) in the pathophysiology of dengue has been suggested by the observation of a blockade of Ang II in patients with dengue, increased expression of molecules related to Ang II production in the plasma of dengue patients, increased expression of circulating cytokines and soluble molecules related to the action of Ang II, and an apparent relationship between DENV, Ang II effects, and miRNAs. In addition, in ex vivo experiments, the blockade of Ang II AT1 receptor and ACE-1 (angiotensin converting enzyme 1), both of which are involved in Ang II production and its function, inhibits infection of macrophages by DENV, suggesting a role of Ang II in viral entry or in intracellular viral replication of the virus. Here, we discuss the possible mechanisms of Ang II in the entry and replication of DENV. Ang II has the functions of increasing the expression of DENV entry receptors, creation of clathrin-coated vesicles, and increasing phagocytosis, all of which are involved in DENV entry. This hormone also modulates the expression of the Rab5 and Rab7 proteins, which are important in the endosomal processing of DENV during viral replication. This review summarizes the data related to the possible involvement of Ang II in the entry of DENV into cells and its replication.
Collapse
Affiliation(s)
- Adriana Pedreañez
- Cátedra de Inmunología, Escuela de Bioanálisis, Facultad de Medicina, Universidad del Zulia, Maracaibo, Venezuela
| | - Yenddy Carrero
- Instituto de Investigaciones Clínicas "Dr. Américo Negrette", Facultad de Medicina, Universidad del Zulia, Maracaibo, Venezuela
| | - Renata Vargas
- Instituto de Investigaciones Clínicas "Dr. Américo Negrette", Facultad de Medicina, Universidad del Zulia, Maracaibo, Venezuela
| | - Juan P Hernández-Fonseca
- Instituto de Investigaciones Clínicas "Dr. Américo Negrette", Facultad de Medicina, Universidad del Zulia, Maracaibo, Venezuela
- Servicio de Microscopia Electrónica del Centro Nacional de Biotecnología (CNB- CSIC), Madrid, España
| | - Jesús Alberto Mosquera
- Instituto de Investigaciones Clínicas "Dr. Américo Negrette", Facultad de Medicina, Universidad del Zulia, Maracaibo, Venezuela.
| |
Collapse
|
6
|
Tyagi K, Rai P, Gautam A, Kaur H, Kapoor S, Suttee A, Jaiswal PK, Sharma A, Singh G, Barnwal RP. Neurological manifestations of SARS-CoV-2: complexity, mechanism and associated disorders. Eur J Med Res 2023; 28:307. [PMID: 37649125 PMCID: PMC10469568 DOI: 10.1186/s40001-023-01293-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 08/16/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Coronaviruses such as Severe Acute Respiratory Syndrome coronavirus (SARS), Middle Eastern Respiratory Syndrome (MERS) and Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) are associated with critical illnesses, including severe respiratory disorders. SARS-CoV-2 is the causative agent of the deadly COVID-19 illness, which has spread globally as a pandemic. SARS-CoV-2 may enter the human body through olfactory lobes and interact with the angiotensin-converting enzyme2 (ACE2) receptor, further facilitating cell binding and entry into the cells. Reports have shown that the virus can pass through the blood-brain barrier (BBB) and enter the central nervous system (CNS), resulting in various disorders. Cell entry by SARS-CoV-2 largely relies on TMPRSS2 and cathepsin L, which activate S protein. TMPRSS2 is found on the cell surface of respiratory, gastrointestinal and urogenital epithelium, while cathepsin-L is a part of endosomes. AIM The current review aims to provide information on how SARS-CoV-2 infection affects brain function.. Furthermore, CNS disorders associated with SARS-CoV-2 infection, including ischemic stroke, cerebral venous thrombosis, Guillain-Barré syndrome, multiple sclerosis, meningitis, and encephalitis, are discussed. The many probable mechanisms and paths involved in developing cerebrovascular problems in COVID patients are thoroughly detailed. MAIN BODY There have been reports that the SARS-CoV-2 virus can cross the blood-brain barrier (BBB) and enter the central nervous system (CNS), where it could cause a various illnesses. Patients suffering from COVID-19 experience a range of neurological complications, including sleep disorders, viral encephalitis, headaches, dysgeusia, and cognitive impairment. The presence of SARS-CoV-2 in the cerebrospinal fluid (CSF) of COVID-19 patients has been reported. Health experts also reported its presence in cortical neurons and human brain organoids. The possible mechanism of virus infiltration into the brain can be neurotropic, direct infiltration and cytokine storm-based pathways. The olfactory lobes could also be the primary pathway for the entrance of SARS-CoV-2 into the brain. CONCLUSIONS SARS-CoV-2 can lead to neurological complications, such as cerebrovascular manifestations, motor movement complications, and cognitive decline. COVID-19 infection can result in cerebrovascular symptoms and diseases, such as strokes and thrombosis. The virus can affect the neural system, disrupt cognitive function and cause neurological disorders. To combat the epidemic, it is crucial to repurpose drugs currently in use quickly and develop novel therapeutics.
Collapse
Affiliation(s)
- Kritika Tyagi
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Prachi Rai
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Anuj Gautam
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Harjeet Kaur
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Sumeet Kapoor
- Centre for Biomedical Engineering, Indian Institute of Technology, New Delhi, India
| | - Ashish Suttee
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Pradeep Kumar Jaiswal
- Department of Biochemistry and Biophysics, Texas A & M University, College Station, TX, 77843, USA
| | - Akanksha Sharma
- Department of Biophysics, Panjab University, Chandigarh, India.
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India.
| | - Gurpal Singh
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India.
| | | |
Collapse
|
7
|
Mott L, Hancock M, Grulke EA, Pack DW. Polymer/Nanoceria Hybrid Polyplexes for Gene and Antioxidant Delivery. ACS APPLIED BIO MATERIALS 2023; 6:3166-3175. [PMID: 37493016 DOI: 10.1021/acsabm.3c00295] [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
Various diseases, including cancers and inflammatory diseases, are characterized by a disruption of redox homeostasis, suggesting the need for synergistic treatments involving co-delivery of gene therapies and free radical scavengers. In this report, polyethylenimine (PEI), nanoceria (NC), and DNA were complexed to form nanoparticles providing simultaneous delivery of a gene and an antioxidant. NC was coated in citric acid to provide stable, 4 nm particles that electrostatically bound PEI/DNA polyplexes. The resulting ternary particles transfected HeLa cells with similar efficiency to that of ternary polyplexes comprising 15 kDa poly-l-α-glutamic acid/PEI/DNA while providing smaller particle sizes by more than 100 nm. NC/PEI/DNA polyplexes exhibited enhanced radical-scavenging activity compared to free NC, and oxidative stress from the superoxide-generating agent, menadione, could be completely reversed by the delivery of NC/PEI/DNA polyplexes. Transfection by NC/PEI/DNA polyplexes was demonstrated to occur efficiently through caveolin-mediated endocytosis and macropinocytosis. Co-delivery of genes encoding reactive oxygen species-scavenging proteins, transcription factors, growth factors, tumor suppressors, or anti-inflammatory genes with NC, therefore, may be a promising strategy in synergistic therapeutics.
Collapse
Affiliation(s)
- Landon Mott
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Matthew Hancock
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Eric A Grulke
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Daniel W Pack
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, Kentucky 40506, United States
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky 40536, United States
| |
Collapse
|
8
|
Rational synthesis of carbon dots with phosphate ester group for direct mapping of endogenous alkaline phosphatase and polarity monitoring in living cells. J Colloid Interface Sci 2023; 640:626-636. [PMID: 36889060 DOI: 10.1016/j.jcis.2023.02.133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 02/02/2023] [Accepted: 02/25/2023] [Indexed: 03/04/2023]
Abstract
Carbon dots (CDs) have been extensively employed in biomolecule imaging. However, the imaging of biological enzymes with CDs has not been reported, which greatly limits their application in biological imaging. Herein, for the first time, a new type of fluorescent CDs is elaborately designed to realize the direct mapping of alkaline phosphatase (ALP) in cells. The obtained phosphorus and nitrogen co-doped CDs (P, N-CDs) generate specific structures including xanthene oxide and phosphate ester, thereby enabling P, N-CDs to be exclusively cleaved by ALP without auxiliary media. The fluorescence intensity of P, N-CDs can be specifically turned on in the presence of ALP, making them powerful probes for sensitive sensing of ALP activity with a detection limit of 1.27 U·L-1. Meanwhile, P, N-CDs possessing electron deficiency structure fulfill sensitive responding to polarity variations. The excellent photo-bleaching resistance and biocompatibility of the P, N-CDs are taken for directly mapping the intracellular endogenous ALP via turned-on fluorescence imaging, as well as real-time monitoring the polarity fluctuation in cells through ratiometric fluorescence imaging. The present work offers a new way to design and synthesize functional CDs for direct imaging of intracellular enzymes.
Collapse
|
9
|
Jia Y, Agbayani G, Chandan V, Iqbal U, Dudani R, Qian H, Jakubek Z, Chan K, Harrison B, Deschatelets L, Akache B, McCluskie MJ. Evaluation of Adjuvant Activity and Bio-Distribution of Archaeosomes Prepared Using Microfluidic Technology. Pharmaceutics 2022; 14:2291. [PMID: 36365110 PMCID: PMC9697222 DOI: 10.3390/pharmaceutics14112291] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/18/2022] [Accepted: 10/20/2022] [Indexed: 09/24/2023] Open
Abstract
Archaeosomes, composed of sulfated lactosyl archaeol (SLA) glycolipids, have been proven to be an effective vaccine adjuvant in multiple preclinical models of infectious disease or cancer. They have classically been prepared using a thin-film hydration method with an average particle size of 100-200 nm. In this study, we developed methods to generate SLA archaeosomes at different sizes, i.e., 30 nm and 100 nm, via microfluidic mixing technology and evaluated their physicochemical characteristics, as well as adjuvant activity and in vivo biodistribution in mice. Archaeosomes, prepared using thin-film and microfluidic mixing techniques, had similar nanostructures and physicochemical characteristics, with both appearing stable during the course of this study when stored at 4 °C or 37 °C. They also demonstrated similar adjuvant activity when admixed with ovalbumin antigen and used to immunize mice, generating equivalent antigen-specific immune responses. Archaeosomes, labeled with CellVueTM NIR815, had an equivalent biodistribution with both sizes, namely the highest signal at the injection site at 24 h post injection, followed by liver, spleen and inguinal lymph node. The presence of SLA archaeosomes of either size helped to retain OVA antigen (OVA-Cy5.5) longer at the injection site than unadjuvanted OVA. Overall, archaeosomes of two sizes (30 nm and 100 nm) prepared using microfluidic mixing maintained similar physicochemical properties, adjuvant activity and biodistribution of antigen, in comparison to those compared by the conventional thin film hydration method. This suggests that microfluidics based approaches could be applied to generate consistently sized archaeosomes for use as a vaccine adjuvant.
Collapse
Affiliation(s)
- Yimei Jia
- Human Health Therapeutics, National Research Council Canada, Ottawa, ON K1A0R6, Canada
| | - Gerard Agbayani
- Human Health Therapeutics, National Research Council Canada, Ottawa, ON K1A0R6, Canada
| | - Vandana Chandan
- Human Health Therapeutics, National Research Council Canada, Ottawa, ON K1A0R6, Canada
| | - Umar Iqbal
- Human Health Therapeutics, National Research Council Canada, Ottawa, ON K1A0R6, Canada
| | - Renu Dudani
- Human Health Therapeutics, National Research Council Canada, Ottawa, ON K1A0R6, Canada
| | - Hui Qian
- Nanotechnology Research Centre, National Research Council Canada, Edmonton, AB T6G2M9, Canada
| | - Zygmunt Jakubek
- Metrology Research Centre, National Research Council Canada, Ottawa, ON K1A0R6, Canada
| | - Kenneth Chan
- Human Health Therapeutics, National Research Council Canada, Ottawa, ON K1A0R6, Canada
| | - Blair Harrison
- Human Health Therapeutics, National Research Council Canada, Ottawa, ON K1A0R6, Canada
| | - Lise Deschatelets
- Human Health Therapeutics, National Research Council Canada, Ottawa, ON K1A0R6, Canada
| | - Bassel Akache
- Human Health Therapeutics, National Research Council Canada, Ottawa, ON K1A0R6, Canada
| | - Michael J. McCluskie
- Human Health Therapeutics, National Research Council Canada, Ottawa, ON K1A0R6, Canada
| |
Collapse
|
10
|
Ou YH, Liang J, Chng WH, Muthuramalingam RPK, Ng ZX, Lee CK, Neupane YR, Yau JNN, Zhang S, Lou CKL, Huang C, Wang JW, Pastorin G. Investigations on Cellular Uptake Mechanisms and Immunogenicity Profile of Novel Bio-Hybrid Nanovesicles. Pharmaceutics 2022; 14:1738. [PMID: 36015364 PMCID: PMC9413569 DOI: 10.3390/pharmaceutics14081738] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/12/2022] [Accepted: 08/17/2022] [Indexed: 12/19/2022] Open
Abstract
In drug delivery, the development of nanovesicles that combine both synthetic and cellular components provides added biocompatibility and targeting specificity in comparison to conventional synthetic carriers such as liposomes. Produced through the fusion of U937 monocytes' membranes and synthetic lipids, our nano-cell vesicle technology systems (nCVTs) showed promising results as targeted cancer treatment. However, no investigation has been conducted yet on the immunogenic profile and the uptake mechanisms of nCVTs. Hence, this study was aimed at exploring the potential cytotoxicity and immune cells' activation by nCVTs, as well as the routes through which cells internalize these biohybrid systems. The endocytic pathways were selectively inhibited to establish if the presence of cellular components in nCVTs affected the internalization route in comparison to both liposomes (made up of synthetic lipids only) and nano-cellular membranes (made up of biological material only). As a result, nCVTs showed an 8-to-40-fold higher cellular internalization than liposomes within the first hour, mainly through receptor-mediated processes (i.e., clathrin- and caveolae-mediated endocytosis), and low immunostimulatory potential (as indicated by the level of IL-1α, IL-6, and TNF-α cytokines) both in vitro and in vivo. These data confirmed that nCVTs preserved surface cues from their parent U937 cells and can be rationally engineered to incorporate ligands that enhance the selective uptake and delivery toward target cells and tissues.
Collapse
Affiliation(s)
- Yi-Hsuan Ou
- Department of Pharmacy, National University of Singapore, Singapore 117543, Singapore
| | - Jeremy Liang
- Department of Pharmacy, National University of Singapore, Singapore 117543, Singapore
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Wei Heng Chng
- Department of Pharmacy, National University of Singapore, Singapore 117543, Singapore
- Integrative Sciences and Engineering Programme, NUS Graduate School, National University of Singapore, Singapore 119077, Singapore
| | | | - Zi Xiu Ng
- Department of Pharmacy, National University of Singapore, Singapore 117543, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore 117558, Singapore
| | - Choon Keong Lee
- Department of Pharmacy, National University of Singapore, Singapore 117543, Singapore
| | - Yub Raj Neupane
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, The University of Iowa, Iowa City, IA 52242, USA
| | - Jia Ning Nicolette Yau
- Department of Pharmacy, National University of Singapore, Singapore 117543, Singapore
- Integrative Sciences and Engineering Programme, NUS Graduate School, National University of Singapore, Singapore 119077, Singapore
| | - Sitong Zhang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Nanomedicine Translational Research Programme, Centre for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117609, Singapore
| | - Charles Kang Liang Lou
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Nanomedicine Translational Research Programme, Centre for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117609, Singapore
| | - Chenyuan Huang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Cardiovascular Research Institute, National University Heart Centre, Singapore 117599, Singapore
| | - Jiong-Wei Wang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Nanomedicine Translational Research Programme, Centre for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117609, Singapore
- Cardiovascular Research Institute, National University Heart Centre, Singapore 117599, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
| | - Giorgia Pastorin
- Department of Pharmacy, National University of Singapore, Singapore 117543, Singapore
- Integrative Sciences and Engineering Programme, NUS Graduate School, National University of Singapore, Singapore 119077, Singapore
| |
Collapse
|
11
|
Assies L, Mercier V, López‐Andarias J, Roux A, Sakai N, Matile S. The Dynamic Range of Acidity: Tracking Rules for the Unidirectional Penetration of Cellular Compartments. Chembiochem 2022; 23:e202200192. [PMID: 35535626 PMCID: PMC9400975 DOI: 10.1002/cbic.202200192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/09/2022] [Indexed: 12/03/2022]
Abstract
Labeled ammonium cations with pKa ∼7.4 accumulate in acidic organelles because they can be neutralized transiently to cross the membrane at cytosolic pH 7.2 but not at their internal pH<5.5. Retention in early endosomes with less acidic internal pH was achieved recently using weaker acids of up to pKa 9.8. We report here that primary ammonium cations with higher pKa 10.6, label early endosomes more efficiently. This maximized early endosome tracking coincides with increasing labeling of Golgi networks with similarly weak internal acidity. Guanidinium cations with pKa 13.5 cannot cross the plasma membrane in monomeric form and label the plasma membrane with selectivity for vesicles embarking into endocytosis. Self-assembled into micelles, guanidinium cations enter cells like arginine-rich cell-penetrating peptides and, driven by their membrane potential, penetrate mitochondria unidirectionally despite their high inner pH. The resulting tracking rules with an approximated dynamic range of pKa change ∼3.5 are expected to be generally valid, thus enabling the design of chemistry tools for biology research in the broadest sense. From a practical point of view, most relevant are two complementary fluorescent flipper probes that can be used to image the mechanics at the very beginning of endocytosis.
Collapse
Affiliation(s)
- Lea Assies
- School of Chemistry and BiochemistryNCCR Chemical BiologyUniversity of Geneva1211GenevaSwitzerland
| | - Vincent Mercier
- School of Chemistry and BiochemistryNCCR Chemical BiologyUniversity of Geneva1211GenevaSwitzerland
| | - Javier López‐Andarias
- School of Chemistry and BiochemistryNCCR Chemical BiologyUniversity of Geneva1211GenevaSwitzerland
| | - Aurelien Roux
- School of Chemistry and BiochemistryNCCR Chemical BiologyUniversity of Geneva1211GenevaSwitzerland
| | - Naomi Sakai
- School of Chemistry and BiochemistryNCCR Chemical BiologyUniversity of Geneva1211GenevaSwitzerland
| | - Stefan Matile
- School of Chemistry and BiochemistryNCCR Chemical BiologyUniversity of Geneva1211GenevaSwitzerland
| |
Collapse
|
12
|
Ginini L, Billan S, Fridman E, Gil Z. Insight into Extracellular Vesicle-Cell Communication: From Cell Recognition to Intracellular Fate. Cells 2022; 11:1375. [PMID: 35563681 PMCID: PMC9101098 DOI: 10.3390/cells11091375] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 01/27/2023] Open
Abstract
Extracellular vesicles (EVs) are heterogamous lipid bilayer-enclosed membranous structures secreted by cells. They are comprised of apoptotic bodies, microvesicles, and exosomes, and carry a range of nucleic acids and proteins that are necessary for cell-to-cell communication via interaction on the cells surface. They initiate intracellular signaling pathways or the transference of cargo molecules, which elicit pleiotropic responses in recipient cells in physiological processes, as well as pathological processes, such as cancer. It is therefore important to understand the molecular means by which EVs are taken up into cells. Accordingly, this review summarizes the underlying mechanisms involved in EV targeting and uptake. The primary method of entry by EVs appears to be endocytosis, where clathrin-mediated, caveolae-dependent, macropinocytotic, phagocytotic, and lipid raft-mediated uptake have been variously described as being prevalent. EV uptake mechanisms may depend on proteins and lipids found on the surfaces of both vesicles and target cells. As EVs have been shown to contribute to cancer growth and progression, further exploration and targeting of the gateways utilized by EVs to internalize into tumor cells may assist in the prevention or deceleration of cancer pathogenesis.
Collapse
Affiliation(s)
- Lana Ginini
- Rappaport Family Institute for Research in the Medical Sciences, Technion–Israel Institute of Technology, Haifa 31096, Israel; (L.G.); (E.F.)
| | - Salem Billan
- Head and Neck Institute, The Holy Family Hospital Nazareth, Nazareth 1641100, Israel;
- Medical Oncology and Radiation Therapy Program, Oncology Section, Rambam Health Care Campus, HaAliya HaShniya Street 8, Haifa 3109601, Israel
| | - Eran Fridman
- Rappaport Family Institute for Research in the Medical Sciences, Technion–Israel Institute of Technology, Haifa 31096, Israel; (L.G.); (E.F.)
| | - Ziv Gil
- Head and Neck Institute, The Holy Family Hospital Nazareth, Nazareth 1641100, Israel;
| |
Collapse
|
13
|
Dual Drug Loaded pH-sensitive Micelles for Efficient Bacterial Infection Treatment. Pharm Res 2022; 39:1165-1180. [DOI: 10.1007/s11095-022-03182-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 01/26/2022] [Indexed: 12/20/2022]
|
14
|
Song Y, Wu Y, Xu L, Jiang T, Tang C, Yin C. Caveolae-Mediated Endocytosis Drives Robust siRNA Delivery of Polymeric Nanoparticles to Macrophages. ACS NANO 2021; 15:8267-8282. [PMID: 33915044 DOI: 10.1021/acsnano.0c08596] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Cytosolic delivery of small interfering RNA (siRNA) remains challenging, and a profound understanding of the cellular uptake and intracellular processing of siRNA delivery systems could greatly improve the development of siRNA-based therapeutics. Here, we show that caveolae-mediated endocytosis (CvME) accounts for the robust siRNA delivery of mannose-modified trimethyl chitosan-cysteine/tripolyphosphate nanoparticles (MTC/TPP NPs) to macrophages by circumventing lysosomes. We show that the Golgi complex and ER are key organelles required for the efficient delivery of siRNA to macrophages in which the siRNA accumulation positively correlates with its silencing efficiency (r = 0.94). We also identify syntaxin6 and Niemann-Pick type C1 (NPC1) as indispensable regulators for MTC/TPP NPs-delivered siRNA into macrophages both in vitro and in vivo. Syntaxin6 and NPC1 knockout substantially decrease the cellular uptake and gene silencing of the siRNA delivered in MTC/TPP NPs in macrophages, which result in poor therapeutic outcomes for mice bearing acute hepatic injury. Our results suggest that highly efficient siRNA delivery can be achieved via CvME, which would give ideas for designing optimal delivery vectors to facilitate the clinical translation of siRNA drugs.
Collapse
Affiliation(s)
- Yudong Song
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, P.R. China
| | - Yanhua Wu
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200438, P.R. China
| | - Lu Xu
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, P.R. China
| | - Ting Jiang
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, P.R. China
| | - Cui Tang
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, P.R. China
| | - Chunhua Yin
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, P.R. China
| |
Collapse
|
15
|
Sanders DW, Jumper CC, Ackerman PJ, Bracha D, Donlic A, Kim H, Kenney D, Castello-Serrano I, Suzuki S, Tamura T, Tavares AH, Saeed M, Holehouse AS, Ploss A, Levental I, Douam F, Padera RF, Levy BD, Brangwynne CP. SARS-CoV-2 requires cholesterol for viral entry and pathological syncytia formation. eLife 2021; 10:e65962. [PMID: 33890572 PMCID: PMC8104966 DOI: 10.7554/elife.65962] [Citation(s) in RCA: 152] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/01/2021] [Indexed: 12/27/2022] Open
Abstract
Many enveloped viruses induce multinucleated cells (syncytia), reflective of membrane fusion events caused by the same machinery that underlies viral entry. These syncytia are thought to facilitate replication and evasion of the host immune response. Here, we report that co-culture of human cells expressing the receptor ACE2 with cells expressing SARS-CoV-2 spike, results in synapse-like intercellular contacts that initiate cell-cell fusion, producing syncytia resembling those we identify in lungs of COVID-19 patients. To assess the mechanism of spike/ACE2-driven membrane fusion, we developed a microscopy-based, cell-cell fusion assay to screen ~6000 drugs and >30 spike variants. Together with quantitative cell biology approaches, the screen reveals an essential role for biophysical aspects of the membrane, particularly cholesterol-rich regions, in spike-mediated fusion, which extends to replication-competent SARS-CoV-2 isolates. Our findings potentially provide a molecular basis for positive outcomes reported in COVID-19 patients taking statins and suggest new strategies for therapeutics targeting the membrane of SARS-CoV-2 and other fusogenic viruses.
Collapse
Affiliation(s)
- David W Sanders
- Department of Chemical and Biological Engineering, Princeton University, Princeton, United States
| | - Chanelle C Jumper
- Department of Chemical and Biological Engineering, Princeton University, Princeton, United States
| | - Paul J Ackerman
- Department of Chemical and Biological Engineering, Princeton University, Princeton, United States
| | - Dan Bracha
- Department of Chemical and Biological Engineering, Princeton University, Princeton, United States
| | - Anita Donlic
- Department of Chemical and Biological Engineering, Princeton University, Princeton, United States
| | - Hahn Kim
- Princeton University Small Molecule Screening Center, Princeton University, Princeton, United States
- Department of Chemistry, Princeton University, Princeton, United States
| | - Devin Kenney
- Department of Microbiology, Boston University School of Medicine, Boston, United States
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, United States
| | - Ivan Castello-Serrano
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, United States
| | - Saori Suzuki
- Department of Molecular Biology, Princeton University, Princeton, United States
| | - Tomokazu Tamura
- Department of Molecular Biology, Princeton University, Princeton, United States
| | - Alexander H Tavares
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, United States
- Department of Biochemistry, Boston University School of Medicine, Boston, United States
| | - Mohsan Saeed
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, United States
- Department of Biochemistry, Boston University School of Medicine, Boston, United States
| | - Alex S Holehouse
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, United States
| | - Alexander Ploss
- Department of Molecular Biology, Princeton University, Princeton, United States
| | - Ilya Levental
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, United States
| | - Florian Douam
- Department of Microbiology, Boston University School of Medicine, Boston, United States
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, United States
| | - Robert F Padera
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, United States
| | - Bruce D Levy
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, United States
| | - Clifford P Brangwynne
- Department of Chemical and Biological Engineering, Princeton University, Princeton, United States
- Howard Hughes Medical Institute, Princeton, United States
| |
Collapse
|
16
|
Winiarczyk D, Winiarczyk M, Winiarczyk S, Michalak K, Adaszek Ł. Proteomic Analysis of Tear Film Obtained from Diabetic Dogs. Animals (Basel) 2020; 10:ani10122416. [PMID: 33348610 PMCID: PMC7766195 DOI: 10.3390/ani10122416] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Canine diabetes is a serious disease, which can lead to severe complications, eventually even death. Currently, all the diagnostic procedures are the invasive ones, with blood collection remaining as a golden standard for both initial diagnosis, and later follow-up. Tears can be obtained in a non-invasive manner, which makes them a perfect candidate for a screening tool in canine diabetes. In this study we aimed to analyze the protein composition of the tears collected from the healthy animals and compared it to the diabetic group. There are significant differences between these two groups, and we believe that the identified proteins hold promise as a potential diagnostic tool, which can be later on used both in clinical practice, and for better understanding of the disease. Abstract Canine diabetes mellitus is a significant health burden, followed with numerous systemic complications, including diabetic cataracts and retinopathy, leading to blindness. Diabetes should be considered as a disease damaging all the body organs, including gastrointestinal tract, through a complex combination of vascular and metabolic pathologies, leading to impaired gut function. Tear film can be obtained in a non-invasive way, which makes it a feasible biomarker source. In this study we compared proteomic changes ongoing in tear film of diabetic dogs. The study group consisted of 15 diabetic dogs, and 13 dogs served as a control group. After obtaining tear film with Schirmer strips, we performed 2-dimensional electrophoresis, followed by Delta2D software analysis, which allowed to select statistically significant differentially expressed proteins. After their identification with MALDI-TOF (matrix assisted laser desorption and ionisation time of flight) spectrometry we found one up-regulated protein in tear film of diabetic dogs—SRC kinase signaling inhibitor 1 (SRCIN1). Eight proteins were down-regulated: phosphatidylinositol-4 kinase type 2 alpha (PI4KIIα), Pro-melanin concentrating hormone (Pro-MCH), Flotillin-1, Protein mono-ADP ribosyltransferase, GRIP and coiled coil domain containing protein 2, tetratricopeptide repeat protein 36, serpin, and Prelamin A/C. Identified proteins were analyzed by Panther Gene Ontology software, and their possible connections with diabetic etiopathology were discussed. We believe that this is the first study to target tear film proteome in canine diabetes. We believe that combined with traditional examination, the tear film proteomic analysis can be a new source of biomarkers both for clinical practice, and experimental research.
Collapse
Affiliation(s)
- Dagmara Winiarczyk
- Department of Internal Diseases of Small Animals, University of Life Sciences of Lublin, 20-950 Lublin, Poland;
| | - Mateusz Winiarczyk
- Department of Vitreoretinal Surgery, Medical University of Lublin, 20-950 Lublin, Poland;
| | - Stanisław Winiarczyk
- Department of Epizootiology, University of Life Sciences of Lublin, 20-950 Lublin, Poland; (S.W.); (K.M.)
| | - Katarzyna Michalak
- Department of Epizootiology, University of Life Sciences of Lublin, 20-950 Lublin, Poland; (S.W.); (K.M.)
| | - Łukasz Adaszek
- Department of Epizootiology, University of Life Sciences of Lublin, 20-950 Lublin, Poland; (S.W.); (K.M.)
- Correspondence:
| |
Collapse
|
17
|
Fecchi K, Anticoli S, Peruzzu D, Iessi E, Gagliardi MC, Matarrese P, Ruggieri A. Coronavirus Interplay With Lipid Rafts and Autophagy Unveils Promising Therapeutic Targets. Front Microbiol 2020; 11:1821. [PMID: 32849425 PMCID: PMC7431668 DOI: 10.3389/fmicb.2020.01821] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/10/2020] [Indexed: 12/20/2022] Open
Abstract
Coronaviruses are enveloped, single-stranded, positive-sense RNA viruses that can infect animal and human hosts. The infection induces mild or sometimes severe acute respiratory diseases. Nowadays, the appearance of a new, highly pathogenic and lethal coronavirus variant, SARS-CoV-2, responsible for a pandemic (COVID-19), represents a global problem for human health. Unfortunately, only limited approaches are available to treat coronavirus infections and a vaccine against this new coronavirus variant is not yet available. The plasma membrane microdomain lipid rafts have been found by researchers to be involved in the replication cycle of numerous viruses, including coronaviruses. Indeed, some pathogen recognition receptors for coronaviruses as for other viruses cluster into lipid rafts, and it is therefore conceivable that the first contact between virus and host cells occurs into these specialized regions, representing a port of cell entry for viruses. Recent data highlighted the peculiar pro-viral or anti-viral role played by autophagy in the host immune responses to viral infections. Coronaviruses, like other viruses, were reported to be able to exploit the autophagic machinery to increase their replication or to inhibit the degradation of viral products. Agents known to disrupt lipid rafts, such as metil-β-cyclodextrins or statins, as well as autophagy inhibitor agents, were shown to have an anti-viral role. In this review, we briefly describe the involvement of lipid rafts and autophagy in coronavirus infection and replication. We also hint how lipid rafts and autophagy may represent a potential therapeutic target to be investigated for the treatment of coronavirus infections.
Collapse
Affiliation(s)
- Katia Fecchi
- Reference Center for Gender Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Simona Anticoli
- Reference Center for Gender Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Daniela Peruzzu
- Reference Center for Gender Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Elisabetta Iessi
- Reference Center for Gender Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | - Paola Matarrese
- Reference Center for Gender Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Anna Ruggieri
- Reference Center for Gender Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
18
|
Carmona-Ribeiro AM, Pérez-Betancourt Y. Cationic Nanostructures for Vaccines Design. Biomimetics (Basel) 2020; 5:biomimetics5030032. [PMID: 32645946 PMCID: PMC7560170 DOI: 10.3390/biomimetics5030032] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 12/20/2022] Open
Abstract
Subunit vaccines rely on adjuvants carrying one or a few molecular antigens from the pathogen in order to guarantee an improved immune response. However, to be effective, the vaccine formulation usually consists of several components: an antigen carrier, the antigen, a stimulator of cellular immunity such as a Toll-like Receptors (TLRs) ligand, and a stimulator of humoral response such as an inflammasome activator. Most antigens are negatively charged and combine well with oppositely charged adjuvants. This explains the paramount importance of studying a variety of cationic supramolecular assemblies aiming at the optimal activity in vivo associated with adjuvant simplicity, positive charge, nanometric size, and colloidal stability. In this review, we discuss the use of several antigen/adjuvant cationic combinations. The discussion involves antigen assembled to 1) cationic lipids, 2) cationic polymers, 3) cationic lipid/polymer nanostructures, and 4) cationic polymer/biocompatible polymer nanostructures. Some of these cationic assemblies revealed good yet poorly explored perspectives as general adjuvants for vaccine design.
Collapse
|
19
|
Kim CH. SARS-CoV-2 Evolutionary Adaptation toward Host Entry and Recognition of Receptor O-Acetyl Sialylation in Virus-Host Interaction. Int J Mol Sci 2020; 21:4549. [PMID: 32604730 PMCID: PMC7352545 DOI: 10.3390/ijms21124549] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/15/2020] [Accepted: 06/22/2020] [Indexed: 02/06/2023] Open
Abstract
The recently emerged SARS-CoV-2 is the cause of the global health crisis of the coronavirus disease 2019 (COVID-19) pandemic. No evidence is yet available for CoV infection into hosts upon zoonotic disease outbreak, although the CoV epidemy resembles influenza viruses, which use sialic acid (SA). Currently, information on SARS-CoV-2 and its receptors is limited. O-acetylated SAs interact with the lectin-like spike glycoprotein of SARS CoV-2 for the initial attachment of viruses to enter into the host cells. SARS-CoV-2 hemagglutinin-esterase (HE) acts as the classical glycan-binding lectin and receptor-degrading enzyme. Most β-CoVs recognize 9-O-acetyl-SAs but switched to recognizing the 4-O-acetyl-SA form during evolution of CoVs. Type I HE is specific for the 9-O-Ac-SAs and type II HE is specific for 4-O-Ac-SAs. The SA-binding shift proceeds through quasi-synchronous adaptations of the SA-recognition sites of the lectin and esterase domains. The molecular switching of HE acquisition of 4-O-acetyl binding from 9-O-acetyl SA binding is caused by protein-carbohydrate interaction (PCI) or lectin-carbohydrate interaction (LCI). The HE gene was transmitted to a β-CoV lineage A progenitor by horizontal gene transfer from a 9-O-Ac-SA-specific HEF, as in influenza virus C/D. HE acquisition, and expansion takes place by cross-species transmission over HE evolution. This reflects viral evolutionary adaptation to host SA-containing glycans. Therefore, CoV HE receptor switching precedes virus evolution driven by the SA-glycan diversity of the hosts. The PCI or LCI stereochemistry potentiates the SA-ligand switch by a simple conformational shift of the lectin and esterase domains. Therefore, examination of new emerging viruses can lead to better understanding of virus evolution toward transitional host tropism. A clear example of HE gene transfer is found in the BCoV HE, which prefers 7,9-di-O-Ac-SAs, which is also known to be a target of the bovine torovirus HE. A more exciting case of such a switching event occurs in the murine CoVs, with the example of the β-CoV lineage A type binding with two different subtypes of the typical 9-O-Ac-SA (type I) and the exclusive 4-O-Ac-SA (type II) attachment factors. The protein structure data for type II HE also imply the virus switching to binding 4-O acetyl SA from 9-O acetyl SA. Principles of the protein-glycan interaction and PCI stereochemistry potentiate the SA-ligand switch via simple conformational shifts of the lectin and esterase domains. Thus, our understanding of natural adaptation can be specified to how carbohydrate/glycan-recognizing proteins/molecules contribute to virus evolution toward host tropism. Under the current circumstances where reliable antiviral therapeutics or vaccination tools are lacking, several trials are underway to examine viral agents. As expected, structural and non-structural proteins of SARS-CoV-2 are currently being targeted for viral therapeutic designation and development. However, the modern global society needs SARS-CoV-2 preventive and therapeutic drugs for infected patients. In this review, the structure and sialobiology of SARS-CoV-2 are discussed in order to encourage and activate public research on glycan-specific interaction-based drug creation in the near future.
Collapse
Affiliation(s)
- Cheorl-Ho Kim
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Korea;
- Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul 06351, Korea
| |
Collapse
|
20
|
Hong X, Zhong X, Du G, Hou Y, Zhang Y, Zhang Z, Gong T, Zhang L, Sun X. The pore size of mesoporous silica nanoparticles regulates their antigen delivery efficiency. SCIENCE ADVANCES 2020; 6:eaaz4462. [PMID: 32596445 PMCID: PMC7304990 DOI: 10.1126/sciadv.aaz4462] [Citation(s) in RCA: 160] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 05/08/2020] [Indexed: 05/19/2023]
Abstract
Subunit vaccines generally proceed through a 4-step in vivo cascade-the DUMP cascade-to generate potent cell-mediated immune responses: (1) drainage to lymph nodes; (2) uptake by dendritic cells (DCs); (3) maturation of DCs; and (4) Presentation of peptide-MHC I complexes to CD8+ T cells. How the physical properties of vaccine carriers such as mesoporous silica nanoparticles (MSNs) influence this cascade is unclear. We fabricated 80-nm MSNs with different pore sizes (7.8 nm, 10.3 nm, and 12.9 nm) and loaded them with ovalbumin antigen. Results demonstrated these MSNs with different pore sizes were equally effective in the first three steps of the DUMP cascade, but those with larger pores showed higher cross-presentation efficiency (step 4). Consistently, large-pore MSNs loaded with B16F10 tumor antigens yielded the strongest antitumor effects. These results demonstrate the promise of our lymph node-targeting large-pore MSNs as vaccine-delivery vehicles for immune activation and cancer vaccination.
Collapse
Affiliation(s)
- Xiaoyu Hong
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xiaofang Zhong
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Guangsheng Du
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yingying Hou
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yunting Zhang
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Tao Gong
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Ling Zhang
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Xun Sun
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
21
|
Ivanova MM, Dao J, Kasaci N, Adewale B, Fikry J, Goker-Alpan O. Rapid Clathrin-Mediated Uptake of Recombinant α-Gal-A to Lysosome Activates Autophagy. Biomolecules 2020; 10:E837. [PMID: 32486191 PMCID: PMC7356514 DOI: 10.3390/biom10060837] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 05/25/2020] [Accepted: 05/28/2020] [Indexed: 02/08/2023] Open
Abstract
Enzyme replacement therapy (ERT) with recombinant alpha-galactosidase A (rh-α-Gal A) is the standard treatment for Fabry disease (FD). ERT has shown a significant impact on patients; however, there is still morbidity and mortality in FD, resulting in progressive cardiac, renal, and cerebrovascular pathology. The main pathway for delivery of rh-α-Gal A to lysosome is cation-independent mannose-6-phosphate receptor (CI-M6PR) endocytosis, also known as insulin-like growth factor 2 receptor (IGF2R) endocytosis. This study aims to investigate the mechanisms of uptake of rh-α-Gal-A in different cell types, with the exploration of clathrin-dependent and caveolin assisted receptor-mediated endocytosis and the dynamics of autophagy-lysosomal functions. rh-α-Gal-A uptake was evaluated in primary fibroblasts, urine originated kidney epithelial cells, and peripheral blood mononuclear cells derived from Fabry patients and healthy controls, and in cell lines HEK293, HTP1, and HUVEC. Uptake of rh-α-Gal-A was more efficient in the cells with the lowest endogenous enzyme activity. Chloroquine and monensin significantly blocked the uptake of rh-α-Gal-A, indicating that the clathrin-mediated endocytosis is involved in recombinant enzyme delivery. Alternative caveolae-mediated endocytosis coexists with clathrin-mediated endocytosis. However, clathrin-dependent endocytosis is a dominant mechanism for enzyme uptake in all cell lines. These results show that the uptake of rh-α-Gal-A occurs rapidly and activates the autophagy-lysosomal pathway.
Collapse
Affiliation(s)
- Margarita M. Ivanova
- Lysosomal and Rare Disorders Research and Treatment Center, Fairfax, VA 22030, USA; (J.D.); (N.K.); (B.A.); (J.F.); (O.G.-A.)
| | | | | | | | | | | |
Collapse
|
22
|
Brodwolf R, Volz-Rakebrand P, Stellmacher J, Wolff C, Unbehauen M, Haag R, Schäfer-Korting M, Zoschke C, Alexiev U. Faster, sharper, more precise: Automated Cluster-FLIM in preclinical testing directly identifies the intracellular fate of theranostics in live cells and tissue. Theranostics 2020; 10:6322-6336. [PMID: 32483455 PMCID: PMC7255044 DOI: 10.7150/thno.42581] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 04/01/2020] [Indexed: 12/25/2022] Open
Abstract
Fluorescence microscopy is widely used for high content screening in 2D cell cultures and 3D models. In particular, 3D tissue models are gaining major relevance in modern drug development. Enabling direct multiparametric evaluation of complex samples, fluorescence lifetime imaging (FLIM) adds a further level to intensity imaging by the sensitivity of the fluorescence lifetime to the microenvironment. However, the use of FLIM is limited amongst others by the acquisition of sufficient photon numbers without phototoxic effects in live cells. Herein, we developed a new cluster-based analysis method to enhance insight, and significantly speed up analysis and measurement time for the accurate translation of fluorescence lifetime information into pharmacological pathways. Methods: We applied a fluorescently-labeled dendritic core-multishell nanocarrier and its cargo Bodipy as molecules of interest (MOI) to human cells and reconstructed human tissue. Following the sensitivity and specificity assessment of the fitting-free Cluster-FLIM analysis of data in silico and in vitro, we evaluated the dynamics of cellular molecule uptake and intracellular interactions. For 3D live tissue investigations, we applied multiphoton (mp) FLIM. Owing to Cluster-FLIM's statistics-based fitting-free analysis, we utilized this approach for automatization. Results: To discriminate the fluorescence lifetime signatures of 5 different fluorescence species in a single color channel, the Cluster-FLIM method requires only 170, respectively, 90 counts per pixel to obtain 95% sensitivity (hit rate) and 95% specificity (correct rejection rate). Cluster-FLIM revealed cellular interactions of MOIs, representing their spatiotemporal intracellular fate. In a setting of an automated workflow, the assessment of lysosomal trapping of the MOI revealed relevant differences between normal and tumor cells, as well as between 2D and 3D models. Conclusion: The automated Cluster-FLIM tool is fitting-free, providing images with enhanced information, contrast, and spatial resolution at short exposure times and low fluorophore concentrations. Thereby, Cluster-FLIM increases the applicability of FLIM in high content analysis of target molecules in drug development and beyond.
Collapse
|
23
|
Kotzé LA, Young C, Leukes VN, John V, Fang Z, Walzl G, Lutz MB, du Plessis N. Mycobacterium tuberculosis and myeloid-derived suppressor cells: Insights into caveolin rich lipid rafts. EBioMedicine 2020; 53:102670. [PMID: 32113158 PMCID: PMC7047144 DOI: 10.1016/j.ebiom.2020.102670] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/18/2019] [Accepted: 12/02/2019] [Indexed: 02/06/2023] Open
Abstract
Mycobacterium tuberculosis (M.tb) is likely the most successful human pathogen, capable of evading protective host immune responses and driving metabolic changes to support its own survival and growth. Ineffective innate and adaptive immune responses inhibit effective clearance of the bacteria from the human host, resulting in the progression to active TB disease. Many regulatory mechanisms exist to prevent immunopathology, however, chronic infections result in the overproduction of regulatory myeloid cells, like myeloid-derived suppressor cells (MDSC), which actively suppress protective host T lymphocyte responses among other immunosuppressive mechanisms. The mechanisms of M.tb internalization by MDSC and the involvement of host-derived lipid acquisition, have not been fully elucidated. Targeted research aimed at investigating MDSC impact on phagocytic control of M.tb, would be advantageous to our collective anti-TB arsenal. In this review we propose a mechanism by which M.tb may be internalized by MDSC and survive via the manipulation of host-derived lipid sources.
Collapse
Affiliation(s)
- Leigh A Kotzé
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medical and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Carly Young
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medical and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Vinzeigh N Leukes
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medical and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Vini John
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Zhuo Fang
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medical and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Gerhard Walzl
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medical and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Manfred B Lutz
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Nelita du Plessis
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medical and Health Sciences, Stellenbosch University, Cape Town, South Africa.
| |
Collapse
|
24
|
Simple Nanoparticles from the Assembly of Cationic Polymer and Antigen as Immunoadjuvants. Vaccines (Basel) 2020; 8:vaccines8010105. [PMID: 32121174 PMCID: PMC7157673 DOI: 10.3390/vaccines8010105] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 02/19/2020] [Accepted: 02/24/2020] [Indexed: 01/05/2023] Open
Abstract
Since antigens are negatively charged, they combine well with positively charged adjuvants. Here, ovalbumin (OVA) (0.1 mg·mL-1) and poly (diallyldimethylammonium chloride) (PDDA) (0.01 mg·mL-1) yielded PDDA/OVA assemblies characterized by dynamic light scattering (DLS) and scanning electron microscopy (SEM) as spherical nanoparticles (NPs) of 170 ± 4 nm hydrodynamic diameter, 30 ± 2 mV of zeta-potential and 0.11 ± 0.01 of polydispersity. Mice immunization with the NPs elicited high OVA-specific IgG1 and low OVA-specific IgG2a production, indicating a Th-2 response. Delayed-type hypersensitivity reaction (DTH) was low and comparable to the one elicited by Al(OH)3/OVA, suggesting again a Th-2 response. PDDA advantages as an adjuvant were simplicity (a single-component adjuvant), low concentration needed (0.01 mg·mL-1 PDDA) combined with antigen yielding neglectable cytotoxicity, and high stability of PDDA/OVA dispersions. The NPs elicited much higher OVA-specific antibodies production than Al(OH)3/OVA. In vivo, the nano-metric size possibly assured antigen presentation by antigen-presenting cells (APC) at the lymph nodes, in contrast to the location of Al(OH)3/OVA microparticles at the site of injection for longer periods with stimulation of local dendritic cells. In the future, it will be interesting to evaluate combinations of the antigen with NPs carrying both PDDA and elicitors of the Th-1 response.
Collapse
|
25
|
Amini MA, Talebi SS, Karimi J. Reactive Oxygen Species Modulator 1 (ROMO1), a New Potential Target for Cancer Diagnosis and Treatment. Chonnam Med J 2019; 55:136-143. [PMID: 31598470 PMCID: PMC6769249 DOI: 10.4068/cmj.2019.55.3.136] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 06/14/2019] [Accepted: 06/19/2019] [Indexed: 02/01/2023] Open
Abstract
Today, the incidence of cancer in the world is rising, and it is expected that in the next several decades, the number of people suffering from cancer or (the cancer rate) will double. Cancer is defined as the excessive and uncontrolled growth of cells; of course (in simple terms), cancer is considered to be a set of other diseases that ultimately causes normal cells to be transformed into neoplastic cells. One of the most important causes of the onset and exacerbation of cancer is excessive oxidative stress. One of the most important proteins in the inner membrane of mitochondria is Reactive Oxygen Species (ROS) Modulator 1 (ROMO1) that interferes with the production of ROS, and with increasing the rate of this protein, oxidative stress will increase, which ultimately leads to some diseases, especially cancer. In this overview, we use some global databases to provide information about ROMO1 cellular signaling pathways, their related proteins and molecules, and some of the diseases associated with the mitochondrial protein, especially cancer.
Collapse
Affiliation(s)
- Mohammad Amin Amini
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Seyed Saman Talebi
- Department of Internal Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Jamshid Karimi
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
26
|
Chiu PC, Hsieh PY, Kang JW, Chang PH, Shen LJ. Study of the intracellular delivery mechanism of a pH-sensitive peptide modified with enhanced green fluorescent protein. J Drug Target 2019; 28:408-418. [PMID: 31524004 DOI: 10.1080/1061186x.2019.1669041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The targeted delivery of therapeutic agents is a promising approach to enhance the efficacy and reduce the toxicity of cancer treatments. Understanding the intracellular endocytic mechanisms of a cell penetrating peptide (CPP) in an acidic environment is important for targeted delivery of macromolecules to tumours. In this study, we constructed a pH-sensitive CPP-based delivery system for the intracellular delivery of macromolecules. A pH-sensitive CPP, HBHAc, was fused with a model protein, enhanced green fluorescent protein (EGFP), through recombinant DNA technology. We found that is essential that negatively charged proteoglycans on the cell surface interact with HBHAc-EGFP prior to the cellular uptake of HBHAc-EGFP. The uptake was significantly restricted at 4 °C under pH conditions of both 6.5 and 7.5. The increased positive charge of HBHAc-EGFP under the acidic condition leads to a pH-dependent cellular uptake, and we observed that the internalisation of HBHAc-EGFP was significantly higher at pH 6.5 than at pH 7.5 (p < .05). Thus, with pH-sensitive activity, HBHAc is expected to improve tumour-targeted intracellular protein delivery. Moreover, our findings provide a new insight that the endocytic pathway may change under different pH conditions and suggest that this unique phenomenon benefits pH-sensitive drug delivery for tumour therapy.
Collapse
Affiliation(s)
- Po-Chuan Chiu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Pei-Yu Hsieh
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Jyun-Wei Kang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Po-Hsun Chang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Li-Jiuan Shen
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Clinical Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Pharmacy, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
27
|
Reid LM, Verma CS, Essex JW. The role of molecular simulations in understanding the mechanisms of cell-penetrating peptides. Drug Discov Today 2019; 24:1821-1835. [DOI: 10.1016/j.drudis.2019.06.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/12/2019] [Accepted: 06/17/2019] [Indexed: 01/06/2023]
|
28
|
Hajavi J, Hashemi M, Sankian M. Evaluation of size and dose effects of rChe a 3 allergen loaded PLGA nanoparticles on modulation of Th2 immune responses by sublingual immunotherapy in mouse model of rhinitis allergic. Int J Pharm 2019; 563:282-292. [DOI: 10.1016/j.ijpharm.2019.03.040] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 03/11/2019] [Accepted: 03/18/2019] [Indexed: 01/08/2023]
|
29
|
Vetten M, Gulumian M. Differences in uptake of 14 nm PEG-liganded gold nanoparticles into BEAS-2B cells is dependent on their functional groups. Toxicol Appl Pharmacol 2019; 363:131-141. [DOI: 10.1016/j.taap.2018.11.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/26/2018] [Accepted: 11/28/2018] [Indexed: 01/30/2023]
|
30
|
Huang Q, Zhong W, Hu Z, Tang X. A review of the role of cav-1 in neuropathology and neural recovery after ischemic stroke. J Neuroinflammation 2018; 15:348. [PMID: 30572925 PMCID: PMC6302517 DOI: 10.1186/s12974-018-1387-y] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 11/29/2018] [Indexed: 12/12/2022] Open
Abstract
Ischemic stroke starts a series of pathophysiological processes that cause brain injury. Caveolin-1 (cav-1) is an integrated protein and locates at the caveolar membrane. It has been demonstrated that cav-1 can protect blood–brain barrier (BBB) integrity by inhibiting matrix metalloproteases (MMPs) which degrade tight junction proteins. This article reviews recent developments in understanding the mechanisms underlying BBB dysfunction, neuroinflammation, and oxidative stress after ischemic stroke, and focuses on how cav-1 modulates a series of activities after ischemic stroke. In general, cav-1 reduces BBB permeability mainly by downregulating MMP9, reduces neuroinflammation through influencing cytokines and inflammatory cells, promotes nerve regeneration and angiogenesis via cav-1/VEGF pathway, reduces apoptosis, and reduces the damage mediated by oxidative stress. In addition, we also summarize some experimental results that are contrary to the above and explore possible reasons for these differences.
Collapse
Affiliation(s)
- Qianyi Huang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Renmin Road 139#, Changsha, 410011, Hunan, China
| | - Wei Zhong
- Department of Neurology, The Second Xiangya Hospital, Central South University, Renmin Road 139#, Changsha, 410011, Hunan, China
| | - Zhiping Hu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Renmin Road 139#, Changsha, 410011, Hunan, China
| | - Xiangqi Tang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Renmin Road 139#, Changsha, 410011, Hunan, China.
| |
Collapse
|
31
|
Verma DK, Gupta D, Lal SK. Host Lipid Rafts Play a Major Role in Binding and Endocytosis of Influenza A Virus. Viruses 2018; 10:v10110650. [PMID: 30453689 PMCID: PMC6266268 DOI: 10.3390/v10110650] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 10/08/2018] [Accepted: 10/13/2018] [Indexed: 12/14/2022] Open
Abstract
Influenza still remains one of the most challenging diseases, posing a significant threat to public health. Host lipid rafts play a critical role in influenza A virus (IAV) assembly and budding, however, their role in polyvalent IAV host binding and endocytosis had remained elusive until now. In the present study, we observed co-localization of IAV with a lipid raft marker ganglioside, GM1, on the host surface. Further, we isolated the lipid raft micro-domains from IAV infected cells and detected IAV protein in the raft fraction. Finally, raft disruption using Methyl-β-Cyclodextrin revealed significant reduction in IAV host binding, suggesting utilization of host rafts for polyvalent binding on the host cell surface. In addition to this, cyclodextrin mediated inhibition of raft-dependent endocytosis showed significantly reduced IAV internalization. Interestingly, exposure of cells to cyclodextrin two hours post-IAV binding showed no such reduction in IAV entry, indicating use of raft-dependent endocytosis for host entry. In summary, this study demonstrates that host lipid rafts are selected by IAV as a host attachment factors for multivalent binding, and IAV utilizes these micro-domains to exploit raft-dependent endocytosis for host internalization, a virus entry route previously unknown for IAV.
Collapse
Affiliation(s)
- Dileep Kumar Verma
- International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India.
| | - Dinesh Gupta
- International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India.
| | - Sunil Kumar Lal
- International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India.
- School of Science and Tropical Medicine & Biology Platform, Monash University, Malaysia, Bandar Sunway, Selangor, DE 47500, Malaysia.
| |
Collapse
|
32
|
Zheng Y, Wu J, Shan W, Wu L, Zhou R, Liu M, Cui Y, Zhou M, Zhang Z, Huang Y. Multifunctional Nanoparticles Enable Efficient Oral Delivery of Biomacromolecules via Improving Payload Stability and Regulating the Transcytosis Pathway. ACS APPLIED MATERIALS & INTERFACES 2018; 10:34039-34049. [PMID: 30207680 DOI: 10.1021/acsami.8b13707] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
In oral delivery of biomacromolecules, ligand-modified nanoparticles (NPs) have emerged as a promising tool to improve the epithelial uptake of the loaded protein/peptide. Unfortunately, the stability and the transport mechanisms of the biotherapeutics during the intracellular transportation still remained unclear, leading to the poor transepithelial efficiency. Additionally, developing novel approaches to simultaneously monitor the payload bioactivity during the transport processes is veritably benefit for keeping their bioactivity. In the present study, EGP peptide (KRKKKGKGLGKKRDPCLRKYK), a ligand with high affinity to heparan sulfate proteoglycans (HSPGs), was found remarkably increasing the cellular uptake (4.5-fold) and also surprisingly achieving high transcytosis efficiency (4.2-fold) of poly(lactide- co-glycolide) NPs on Caco-2 cell monolayer. Compared with unmodified NPs (C NPs), EGP modified NPs (EGP NPs) exhibited more desirable colloidal stability within epithelia. In the subsequent study, the bioactivity of encapsulated insulin during the cellular transportation was innovatively monitored by a glucose consumption assay. Inspiringly, EGP NPs could mostly retain the bioactivity of loaded insulin whereas insulin from INS-C NPs was significantly degraded. Then the detailed mechanism study revealed that the binding of EGP to HSPGs played a vital role on NP transportation. Unlike C NPs being delivered in the endo/lysosomal pathway, EGP NPs were involved in caveolae-mediated transport, which contributes to the efficient avoidance of the lysosomal entrapment and sequentially facilitates the direct apical-to-basolateral transcytosis. The enhanced absorption of EGP NPs was confirmed in in situ intestinal loop models. Most importantly, oral administrated INS-EGP NPs generated a strong hypoglycemic response on diabetic rats with 10.2-fold and 2.6-fold increase in bioavailability compared with free insulin and INS-C NPs, respectively. The work provided an innovative strategy to monitor the payload bioactivity during the transport processes and proposed a novel aspect to increase oral bioavailability of biomacromolecules via improving payload stability and regulating the transcytosis pathway of nanocarriers.
Collapse
Affiliation(s)
- Yaxian Zheng
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy , Sichuan University , No. 17, Block 3, Southern Renmin Road , Chengdu 610041 , P.R. China
| | - Jiawei Wu
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy , Sichuan University , No. 17, Block 3, Southern Renmin Road , Chengdu 610041 , P.R. China
| | - Wei Shan
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy , Sichuan University , No. 17, Block 3, Southern Renmin Road , Chengdu 610041 , P.R. China
| | - Lei Wu
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy , Sichuan University , No. 17, Block 3, Southern Renmin Road , Chengdu 610041 , P.R. China
| | - Rui Zhou
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy , Sichuan University , No. 17, Block 3, Southern Renmin Road , Chengdu 610041 , P.R. China
| | - Min Liu
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy , Sichuan University , No. 17, Block 3, Southern Renmin Road , Chengdu 610041 , P.R. China
| | - Yi Cui
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy , Sichuan University , No. 17, Block 3, Southern Renmin Road , Chengdu 610041 , P.R. China
| | - Minglu Zhou
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy , Sichuan University , No. 17, Block 3, Southern Renmin Road , Chengdu 610041 , P.R. China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy , Sichuan University , No. 17, Block 3, Southern Renmin Road , Chengdu 610041 , P.R. China
| | - Yuan Huang
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy , Sichuan University , No. 17, Block 3, Southern Renmin Road , Chengdu 610041 , P.R. China
| |
Collapse
|
33
|
Kim CL, Jung MY, Kim YS, Jang JW, Lee GM. Improving the production of recombinant human bone morphogenetic protein-4 in Chinese hamster ovary cell cultures by inhibition of undesirable endocytosis. Biotechnol Bioeng 2018; 115:2565-2575. [PMID: 30011067 DOI: 10.1002/bit.26798] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 06/30/2018] [Accepted: 07/11/2018] [Indexed: 12/17/2023]
Abstract
Endocytic regulation serves a critical role in modulating the extracellular level of signaling molecules, such as bone morphogenetic proteins (BMPs). Unfortunately, endocytosis may result in poor yields of recombinant human BMP-4 (rhBMP-4) from Chinese hamster ovary (CHO) cell cultures. When rhBMP-4 was incubated with CHO cells, rhBMP-4 was actively internalized into cells. Cell surface bound heparan sulfate proteoglycans (HSPGs) served as the major receptors for rhBMP-4 internalization. Removal of cell surface heparan sulfate (HS) by heparinases or reduction of HSPG synthesis by knockdown of xylosyltransferase2 (xylt2) in CHO cells decreased internalization of rhBMP-4. In addition, treatment with endocytosis inhibitors (chlorpromazine, genistein, and dynasore) identified a clathrin- and dynamin-dependent endocytic pathway as the major route for rhBMP-4 internalization. To enhance product yield by minimizing rhBMP-4 internalization in recombinant CHO (rCHO) cell cultures, we have tested various strategies to reduce HSPG synthesis (knockdown of xylt2 and sodium chlorate treatment) or inhibit the binding of rhBMP-4 to cell-surface-bound HSPGs (supplementation with heparin or dextran sulfate [DS]). Among these approaches, DS, which is a linear anionic sulfated polysaccharide with similarity to HS chains, was the most effective in enhancing rhBMP-4 production in rCHO cell cultures. Compared with the control cultures, DS addition to the culture medium (1.0 g/L) resulted in 1.4-fold and 2.3-fold increases in maximum rhBMP-4 concentration in batch and fed-batch cultures, respectively. Taken together, the addition of DS, an effective competitor of HSPGs, improved rhBMP-4 production in rCHO cell cultures through blockage of rhBMP-4 internalization.
Collapse
Affiliation(s)
- Che Lin Kim
- Department of Biological Sciences, KAIST, Daejeon, Republic of Korea
| | - Mi Yeong Jung
- Institute of Biomaterial and Medical Engineering, Cellumed, Seoul, Republic of Korea
| | - Young Sik Kim
- Institute of Biomaterial and Medical Engineering, Cellumed, Seoul, Republic of Korea
| | - Ju Woong Jang
- Institute of Biomaterial and Medical Engineering, Cellumed, Seoul, Republic of Korea
| | - Gyun Min Lee
- Department of Biological Sciences, KAIST, Daejeon, Republic of Korea
| |
Collapse
|
34
|
Jia J, Zhang Y, Xin Y, Jiang C, Yan B, Zhai S. Interactions Between Nanoparticles and Dendritic Cells: From the Perspective of Cancer Immunotherapy. Front Oncol 2018; 8:404. [PMID: 30319969 PMCID: PMC6167641 DOI: 10.3389/fonc.2018.00404] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/04/2018] [Indexed: 02/02/2023] Open
Abstract
Dendritic cells (DCs) are the primary antigen-presenting cells and play key roles in the orchestration of the innate and adaptive immune system. Targeting DCs by nanotechnology stands as a promising strategy for cancer immunotherapy. The physicochemical properties of nanoparticles (NPs) influence their interactions with DCs, thus altering the immune outcome of DCs by changing their functions in the processes of maturation, homing, antigen processing and antigen presentation. In this review, we summarize the recent progress in targeting DCs using NPs as a drug delivery carrier in cancer immunotherapy, the recognition of NPs by DCs, and the ways the physicochemical properties of NPs affect DCs' functions. Finally, the molecular pathways in DCs that are affected by NPs are also discussed.
Collapse
Affiliation(s)
- Jianbo Jia
- Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Institute of Environmental Research at Greater Bay, Guangzhou University, Guangzhou, China
| | - Yi Zhang
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, China
| | - Yan Xin
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, China
| | - Cuijuan Jiang
- School of Environmental Science and Engineering, Shandong University, Jinan, China
| | - Bing Yan
- Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Institute of Environmental Research at Greater Bay, Guangzhou University, Guangzhou, China.,School of Environmental Science and Engineering, Shandong University, Jinan, China
| | - Shumei Zhai
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, China
| |
Collapse
|
35
|
E S, Mao QX, Yuan XL, Kong XL, Chen XW, Wang JH. Targeted imaging of the lysosome and endoplasmic reticulum and their pH monitoring with surface regulated carbon dots. NANOSCALE 2018; 10:12788-12796. [PMID: 29947397 DOI: 10.1039/c8nr03453b] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Organelles play crucial roles in cellular activities and the functions of organelles are related greatly to the pH values, therefore, the bio-imaging of targeted organelles and their related pH sensing is of great importance in biological assays. Herein we report the fluorescence imaging of specific organelles, i.e., lysosomes and endoplasmic reticulum, and their pH sensing with surface regulated carbon dots (CDs). Carbon dots functionalized with amine groups (ACDs) are first prepared by hydrothermal treatment of citric acid and urea, and then laurylamine functionalized CDs (LCDs) are obtained via the conjugation of laurylamine with ACDs. The as-prepared ACDs and LCDs provide clear and bright imaging results for the lysosome and endoplasmic reticulum, respectively. The subcellular targeting features of the two CDs are attributed to their surface chemistries and cellular uptake pathways. Moreover, both the CDs are pH responsive within a certain pH range, i.e., 4.0-5.4 for ACDs and 6.2-7.2 for LCDs. The ACDs and LCDs are thus successfully applied to visualize the pH fluctuations of the lysosome and endoplasmic reticulum in MCF-7 cells.
Collapse
Affiliation(s)
- Shuang E
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China.
| | | | | | | | | | | |
Collapse
|
36
|
Yu X, Noll RR, Romero Dueñas BP, Allgood SC, Barker K, Caplan JL, Machner MP, LaBaer J, Qiu J, Neunuebel MR. Legionella effector AnkX interacts with host nuclear protein PLEKHN1. BMC Microbiol 2018; 18:5. [PMID: 29433439 PMCID: PMC5809941 DOI: 10.1186/s12866-017-1147-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 12/21/2017] [Indexed: 11/13/2022] Open
Abstract
Background The intracellular bacterial pathogen Legionella pneumophila proliferates in human alveolar macrophages, resulting in a severe pneumonia termed Legionnaires’ disease. Throughout the course of infection, L. pneumophila remains enclosed in a specialized membrane compartment that evades fusion with lysosomes. The pathogen delivers over 300 effector proteins into the host cell, altering host pathways in a manner that sets the stage for efficient pathogen replication. The L. pneumophila effector protein AnkX targets host Rab GTPases and functions in preventing fusion of the Legionella-containing vacuole with lysosomes. However, the current understanding of AnkX’s interaction with host proteins and the means through which it exerts its cellular function is limited. Results Here, we investigated the protein interaction network of AnkX by using the nucleic acid programmable protein array (NAPPA), a high-density platform comprising 10,000 unique human ORFs. This approach facilitated the discovery of PLEKHN1 as a novel interaction partner of AnkX. We confirmed this interaction through multiple independent in vitro pull-down, co-immunoprecipitation, and cell-based assays. Structured illumination microscopy revealed that endogenous PLEKHN1 is found in the nucleus and on vesicular compartments, whereas ectopically produced AnkX co-localized with lipid rafts at the plasma membrane. In mammalian cells, HaloTag-AnkX co-localized with endogenous PLEKHN1 on vesicular compartments. A central fragment of AnkX (amino acids 491–809), containing eight ankyrin repeats, extensively co-localized with endogenous PLEKHN1, indicating that this region may harbor a new function. Further, we found that PLEKHN1 associated with multiple proteins involved in the inflammatory response. Conclusions Altogether, our study provides evidence that in addition to Rab GTPases, the L. pneumophila effector AnkX targets nuclear host proteins and suggests that AnkX may have novel functions related to manipulating the inflammatory response. Electronic supplementary material The online version of this article (10.1186/s12866-017-1147-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaobo Yu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Radiation Medicine, Beijing, 102206, China
| | - Rebecca R Noll
- Department of Biological Sciences, University of Delaware, 105 The Green, Newark, DE, 19716, USA
| | - Barbara P Romero Dueñas
- Department of Biological Sciences, University of Delaware, 105 The Green, Newark, DE, 19716, USA
| | - Samual C Allgood
- Department of Biological Sciences, University of Delaware, 105 The Green, Newark, DE, 19716, USA
| | - Kristi Barker
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA
| | - Jeffrey L Caplan
- Department of Biological Sciences, University of Delaware, 105 The Green, Newark, DE, 19716, USA.,Delaware Biotechnology Institute, University of Delaware, Newark, 19716, DE, USA
| | - Matthias P Machner
- Cell Biology and Neurobiology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Joshua LaBaer
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA
| | - Ji Qiu
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA.
| | - M Ramona Neunuebel
- Department of Biological Sciences, University of Delaware, 105 The Green, Newark, DE, 19716, USA.
| |
Collapse
|
37
|
Bagam P, Singh DP, Inda ME, Batra S. Unraveling the role of membrane microdomains during microbial infections. Cell Biol Toxicol 2017; 33:429-455. [PMID: 28275881 PMCID: PMC7088210 DOI: 10.1007/s10565-017-9386-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 02/06/2017] [Indexed: 01/06/2023]
Abstract
Infectious diseases pose major socioeconomic and health-related threats to millions of people across the globe. Strategies to combat infectious diseases derive from our understanding of the complex interactions between the host and specific bacterial, viral, and fungal pathogens. Lipid rafts are membrane microdomains that play important role in life cycle of microbes. Interaction of microbial pathogens with host membrane rafts influences not only their initial colonization but also their spread and the induction of inflammation. Therefore, intervention strategies aimed at modulating the assembly of membrane rafts and/or regulating raft-directed signaling pathways are attractive approaches for the. management of infectious diseases. The current review discusses the latest advances in terms of techniques used to study the role of membrane microdomains in various pathological conditions and provides updated information regarding the role of membrane rafts during bacterial, viral and fungal infections.
Collapse
Affiliation(s)
- Prathyusha Bagam
- Laboratory of Pulmonary Immuno-Toxicology, Department of Environmental Toxicology, Health Research Center, Southern University and A&M College, Baton Rouge, LA, 70813, USA
| | - Dhirendra P Singh
- Laboratory of Pulmonary Immuno-Toxicology, Department of Environmental Toxicology, Health Research Center, Southern University and A&M College, Baton Rouge, LA, 70813, USA
| | - Maria Eugenia Inda
- Departamento de Microbiología, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Suipacha, Rosario, Argentina
| | - Sanjay Batra
- Laboratory of Pulmonary Immuno-Toxicology, Department of Environmental Toxicology, Health Research Center, Southern University and A&M College, Baton Rouge, LA, 70813, USA.
| |
Collapse
|
38
|
Solomon M, Muro S. Lysosomal enzyme replacement therapies: Historical development, clinical outcomes, and future perspectives. Adv Drug Deliv Rev 2017; 118:109-134. [PMID: 28502768 PMCID: PMC5828774 DOI: 10.1016/j.addr.2017.05.004] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/26/2017] [Accepted: 05/08/2017] [Indexed: 01/06/2023]
Abstract
Lysosomes and lysosomal enzymes play a central role in numerous cellular processes, including cellular nutrition, recycling, signaling, defense, and cell death. Genetic deficiencies of lysosomal components, most commonly enzymes, are known as "lysosomal storage disorders" or "lysosomal diseases" (LDs) and lead to lysosomal dysfunction. LDs broadly affect peripheral organs and the central nervous system (CNS), debilitating patients and frequently causing fatality. Among other approaches, enzyme replacement therapy (ERT) has advanced to the clinic and represents a beneficial strategy for 8 out of the 50-60 known LDs. However, despite its value, current ERT suffers from several shortcomings, including various side effects, development of "resistance", and suboptimal delivery throughout the body, particularly to the CNS, lowering the therapeutic outcome and precluding the use of this strategy for a majority of LDs. This review offers an overview of the biomedical causes of LDs, their socio-medical relevance, treatment modalities and caveats, experimental alternatives, and future treatment perspectives.
Collapse
Affiliation(s)
- Melani Solomon
- Institute for Bioscience and Biotechnology Research, University Maryland, College Park, MD 20742, USA
| | - Silvia Muro
- Institute for Bioscience and Biotechnology Research, University Maryland, College Park, MD 20742, USA; Fischell Department of Bioengineering, University Maryland, College Park, MD 20742, USA.
| |
Collapse
|
39
|
Zhang L, Yang X, Lv Y, Xin X, Qin C, Han X, Yang L, He W, Yin L. Cytosolic co-delivery of miRNA-34a and docetaxel with core-shell nanocarriers via caveolae-mediated pathway for the treatment of metastatic breast cancer. Sci Rep 2017; 7:46186. [PMID: 28383524 PMCID: PMC5382875 DOI: 10.1038/srep46186] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 03/10/2017] [Indexed: 01/19/2023] Open
Abstract
Co-delivery of microRNAs and chemotherapeutic drugs into tumor cells is an attractive strategy for synergetic breast cancer therapy due to their complementary mechanisms. In this work, a core-shell nanocarrier coated by cationic albumin was developed to simultaneously deliver miRNA-34a and docetaxel (DTX) into breast cancer cells for improved therapeutic effect. The co-delivery nanocarriers showed a spherical morphology with an average particle size of 183.9 nm, and they efficiently protected miRNA-34a from degradation by RNase and serum. Importantly, the nanocarriers entered the cytosol via a caveolae-mediated pathway without entrapment in endosomes/lysosomes, thus improving the utilization of the cargo. In vitro, the co-delivery nanocarriers suppressed the expression of anti-apoptosis gene Bcl-2 at both transcription and protein levels, inhibited tumor cell migration and efficiently induced cell apoptosis and cytotoxicity. In vivo, the co-delivery nanocarriers prolonged the blood circulation of DTX, enhanced tumor accumulation of the cargo and significantly inhibited tumor growth and metastasis in 4T1-tumor bearing mice models. Taken together, the present nanocarrier co-loading with DTX and miRNA-34a is a new nanoplatform for the combination of insoluble drugs and gene/protein drugs and provides a promising strategy for the treatment of metastatic breast cancer.
Collapse
Affiliation(s)
- Li Zhang
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P.R. China.,Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Xin Yang
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P.R. China.,Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Yaqi Lv
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P.R. China.,Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Xiaofei Xin
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P.R. China.,Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Chao Qin
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P.R. China.,Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Xiaopeng Han
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P.R. China.,Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Lei Yang
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P.R. China.,Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Wei He
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P.R. China.,Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Lifang Yin
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P.R. China.,Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, P.R. China
| |
Collapse
|
40
|
Guo H, Huang M, Yuan Q, Wei Y, Gao Y, Mao L, Gu L, Tan YW, Zhong Y, Liu D, Sun S. The Important Role of Lipid Raft-Mediated Attachment in the Infection of Cultured Cells by Coronavirus Infectious Bronchitis Virus Beaudette Strain. PLoS One 2017; 12:e0170123. [PMID: 28081264 PMCID: PMC5231368 DOI: 10.1371/journal.pone.0170123] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 12/29/2016] [Indexed: 11/19/2022] Open
Abstract
Lipid raft is an important element for the cellular entry of some viruses, including coronavirus infectious bronchitis virus (IBV). However, the exact role of lipid rafts in the cellular membrane during the entry of IBV into host cells is still unknown. In this study, we biochemically fractionated IBV-infected cells via sucrose density gradient centrifugation after depleting plasma membrane cholesterol with methyl-β-cyclodextrin or Mevastatin. Our results demonstrated that unlike IBV non-structural proteins, IBV structural proteins co-localized with lipid raft marker caveolin-1. Infectivity assay results of Vero cells illustrated that the drug-induced disruption of lipid rafts significantly suppressed IBV infection. Further studies revealed that lipid rafts were not required for IBV genome replication or virion release at later stages. However, the drug-mediated depletion of lipid rafts in Vero cells before IBV attachment significantly reduced the expression of viral structural proteins, suggesting that drug treatment impaired the attachment of IBV to the cell surface. Our results indicated that lipid rafts serve as attachment factors during the early stages of IBV infection, especially during the attachment stage.
Collapse
Affiliation(s)
- Huichen Guo
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping, Lanzhou, Gansu, The P.R. China
| | - Mei Huang
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping, Lanzhou, Gansu, The P.R. China
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Quan Yuan
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Yanquan Wei
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping, Lanzhou, Gansu, The P.R. China
| | - Yuan Gao
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping, Lanzhou, Gansu, The P.R. China
| | - Lejiao Mao
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping, Lanzhou, Gansu, The P.R. China
| | - Lingjun Gu
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping, Lanzhou, Gansu, The P.R. China
- College of Animal Science, Yangtze University, Jingzhou, P.R. China
| | - Yong Wah Tan
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Yanxin Zhong
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Dingxiang Liu
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping, Lanzhou, Gansu, The P.R. China
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- * E-mail: (SS); (DL)
| | - Shiqi Sun
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping, Lanzhou, Gansu, The P.R. China
- * E-mail: (SS); (DL)
| |
Collapse
|
41
|
Synthetic Biodegradable Microparticle and Nanoparticle Vaccines against the Respiratory Syncytial Virus. Vaccines (Basel) 2016; 4:vaccines4040045. [PMID: 27918420 PMCID: PMC5192365 DOI: 10.3390/vaccines4040045] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 11/22/2016] [Accepted: 11/22/2016] [Indexed: 01/06/2023] Open
Abstract
Synthetic biodegradable microparticle and nanoparticle platform technology provides the opportunity to design particles varying in composition, size, shape and surface properties for application in vaccine development. The use of particle vaccine formulations allows improvement of antigen stability and immunogenicity while allowing targeted delivery and slow release. This technology has been design to develop novel vaccines against the respiratory syncytial virus (RSV), the leading cause of lower respiratory tract infection in infants. In the last decade, several nano- and micro-sized RSV vaccine candidates have been developed and tested in animal models showing promising results. This review provides an overview of recent advances in prophylactic particle vaccines for RSV and the multiple factors that can affect vaccine efficacy.
Collapse
|
42
|
Affiliation(s)
- Mingming Zhang
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, 236 Baidi Road, Nankai District, Tianjin 300192, China
| | - Yanhang Hong
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, 236 Baidi Road, Nankai District, Tianjin 300192, China
| | - Wenjuan Chen
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, 236 Baidi Road, Nankai District, Tianjin 300192, China
| | - Chun Wang
- Department
of Biomedical Engineering, University of Minnesota, 7-105 Hasselmo
Hall, 312 Church Street S. E., Minneapolis, Minnesota 55455, United States
| |
Collapse
|
43
|
O'Donoghue EJ, Krachler AM. Mechanisms of outer membrane vesicle entry into host cells. Cell Microbiol 2016; 18:1508-1517. [PMID: 27529760 PMCID: PMC5091637 DOI: 10.1111/cmi.12655] [Citation(s) in RCA: 229] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 08/05/2016] [Accepted: 08/12/2016] [Indexed: 01/18/2023]
Abstract
Bacterial outer membrane vesicles (OMVs) are nano‐sized compartments consisting of a lipid bilayer that encapsulates periplasm‐derived, luminal content. OMVs, which pinch off of Gram‐negative bacteria, are now recognized as a generalized secretion pathway which provides a means to transfer cargo to other bacterial cells as well as eukaryotic cells. Compared with other secretion systems, OMVs can transfer a chemically extremely diverse range of cargo, including small molecules, nucleic acids, proteins, and lipids to proximal cells. Although it is well recognized that OMVs can enter and release cargo inside host cells during infection, the mechanisms of host association and uptake are not well understood. This review highlights existing studies focusing on OMV‐host cell interactions and entry mechanisms, and how these entry routes affect cargo processing within the host. It further compares the wide range of methods currently used to dissect uptake mechanisms, and discusses potential sources of discrepancy regarding the mechanism of OMV uptake across different studies.
Collapse
Affiliation(s)
- Eloise J O'Donoghue
- University of Birmingham, Institute of Microbiology and Infection, School of Biosciences, Edgbaston, Birmingham, B15 2TT, UK
| | - Anne Marie Krachler
- University of Birmingham, Institute of Microbiology and Infection, School of Biosciences, Edgbaston, Birmingham, B15 2TT, UK.
| |
Collapse
|
44
|
Li Y, Gao L, Tan X, Li F, Zhao M, Peng S. Lipid rafts-mediated endocytosis and physiology-based cell membrane traffic models of doxorubicin liposomes. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1858:1801-1811. [PMID: 27117641 DOI: 10.1016/j.bbamem.2016.04.014] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 03/09/2016] [Accepted: 04/13/2016] [Indexed: 12/01/2022]
Abstract
The clathrin-mediated endocytosis is likely a major mechanism of liposomes' internalization. A kinetic approach was used to assess the internalization mechanism of doxorubicin (Dox) loaded cationic liposomes and to establish physiology-based cell membrane traffic mathematic models. Lipid rafts-mediated endocytosis, including dynamin-dependent or -independent endocytosis of noncaveolar structure, was a dominant process. The mathematic models divided Dox loaded liposomes binding lipid rafts (B) into saturable binding (SB) and nonsaturable binding (NSB) followed by energy-driven endocytosis. The intracellular trafficking demonstrated early endosome-late endosome-lysosome or early/late endosome-cytoplasm-nucleus pathways. The three properties of liposome structures, i.e., cationic lipid, fusogenic lipid, and pegylation, were investigated to compare their contributions to cell membrane and intracellular traffic. The results revealed great contribution of cationic lipid DOTAP and fusogenic lipid DOPE to cell membrane binding and internalization. The valid Dox in the nuclei of HepG2 and A375 cells treated with cationic liposomes containing 40mol% of DOPE were 1.2-fold and 1.5-fold higher than that in the nuclei of HepG2 and A375 cells treated with liposomes containing 20mol% of DOPE, respectively, suggesting the dependence of cell type. This tendency was proportional to the increase of cell-associated total liposomal Dox. The mathematic models would be useful to predict intracellular trafficking of liposomal Dox.
Collapse
Affiliation(s)
- Yinghuan Li
- Beijing area major laboratory of peptide and small molecular drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, PR China
| | - Lei Gao
- School of Biomedical Engineering, Capital Medical University, 10 Xitoutiao, You An Men, Beijing 100069, PR China
| | - Xi Tan
- Beijing area major laboratory of peptide and small molecular drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, PR China
| | - Feiyang Li
- Beijing area major laboratory of peptide and small molecular drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, PR China
| | - Ming Zhao
- Beijing area major laboratory of peptide and small molecular drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, PR China; Faculty of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Shiqi Peng
- Beijing area major laboratory of peptide and small molecular drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, PR China.
| |
Collapse
|
45
|
Chaurasia M, Singh PK, Jaiswal AK, Kumar A, Pawar VK, Dube A, Paliwal SK, Chourasia MK. Bioinspired Calcium Phosphate Nanoparticles Featuring as Efficient Carrier and Prompter for Macrophage Intervention in Experimental Leishmaniasis. Pharm Res 2016; 33:2617-29. [DOI: 10.1007/s11095-016-1985-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 06/23/2016] [Indexed: 11/29/2022]
|
46
|
Singh S, Mittal A. Transmembrane Domain Lengths Serve as Signatures of Organismal Complexity and Viral Transport Mechanisms. Sci Rep 2016; 6:22352. [PMID: 26925972 PMCID: PMC4772119 DOI: 10.1038/srep22352] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 02/12/2016] [Indexed: 12/24/2022] Open
Abstract
It is known that membrane proteins are important in various secretory pathways, with
a possible role of their transmembrane domains (TMDs) as sorting determinant
factors. One key aspect of TMDs associated with various
“checkposts” (i.e. organelles) of intracellular trafficking
is their length. To explore possible linkages in organisms with varying
“complexity” and differences in TMD lengths of membrane
proteins associated with different organelles (such as Endoplasmic Reticulum, Golgi,
Endosomes, Nucleus, Plasma Membrane), we analyzed ~70000 membrane
protein sequences in over 300 genomes of fungi, plants, non-mammalian vertebrates
and mammals. We report that as we move from simpler to complex organisms, variation
in organellar TMD lengths decreases, especially compared to their respective plasma
membranes, with increasing organismal complexity. This suggests an evolutionary
pressure in modulating length of TMDs of membrane proteins with increasing
complexity of communication between sub-cellular compartments. We also report
functional applications of our findings by discovering remarkable distinctions in
TMD lengths of membrane proteins associated with different intracellular transport
pathways. Finally, we show that TMD lengths extracted from viral proteins can serve
as somewhat weak indicators of viral replication sites in plant cells but very
strong indicators of different entry pathways employed by animal viruses.
Collapse
Affiliation(s)
- Snigdha Singh
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Aditya Mittal
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| |
Collapse
|
47
|
Rammohan A, Mishra G, Mahaling B, Tayal L, Mukhopadhyay A, Gambhir S, Sharma A, Sivakumar S. PEGylated Carbon Nanocapsule: A Universal Reactor and Carrier for In Vivo Delivery of Hydrophobic and Hydrophilic Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2016; 8:350-362. [PMID: 26646711 DOI: 10.1021/acsami.5b08885] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
We have developed PEGylated mesoporous carbon nanocapsule as a universal nanoreactor and carrier for the delivery of highly crystalline hydrophobic/hydrophilic nanoparticles (NPs) which shows superior biocompatibility, dispersion in body fluids, good biodistribution and NPs independent cellular uptake mechanism. The hydrophobic/hydrophilic NPs without surface modification were synthesized in situ inside the cavities of mesoporous carbon capsules (200-850 nm). Stable and inert nature of carbon capsules in a wide range of reaction conditions like high temperature and harsh solvents, make it suitable for being used as nano/microreactors for the syntheses of a variety of NPs for bioimaging applications, such as NaYF4:Eu(3+)(5%), LaVO4:Eu(3+)(10%), GdVO4:Eu(3+)(10%), Y2O3:Eu(3+)(5%), GdF3:Tb(3+)(10%), Mo, Pt, Pd, Au, and Ag. Multiple types of NPs (Y2O3:Eu(3+)(5%) (hydrophobic) and GdF3:Tb(3+)(10%) (hydrophilic)) were coloaded inside the carbon capsules to create a multimodal agent for magneto-fluorescence imaging. Our in vivo study clearly suggests that carbon capsules have biodistribution in many organs including liver, heart, spleen, lungs, blood pool, and muscles.
Collapse
Affiliation(s)
- Amritha Rammohan
- Department of Chemical Engineering, Centre for Environmental Science & Engineering, Thematic Unit of Excellence in Soft Nanofabrication, Indian Institute of Technology , Kanpur, Uttar Pradesh-208016, India
| | - Gargi Mishra
- Department of Chemical Engineering, Centre for Environmental Science & Engineering, Thematic Unit of Excellence in Soft Nanofabrication, Indian Institute of Technology , Kanpur, Uttar Pradesh-208016, India
| | - Binapani Mahaling
- Department of Chemical Engineering, Centre for Environmental Science & Engineering, Thematic Unit of Excellence in Soft Nanofabrication, Indian Institute of Technology , Kanpur, Uttar Pradesh-208016, India
| | - Lokesh Tayal
- Department of Chemical Engineering, Centre for Environmental Science & Engineering, Thematic Unit of Excellence in Soft Nanofabrication, Indian Institute of Technology , Kanpur, Uttar Pradesh-208016, India
| | - Ahana Mukhopadhyay
- Department of Chemical Engineering, Centre for Environmental Science & Engineering, Thematic Unit of Excellence in Soft Nanofabrication, Indian Institute of Technology , Kanpur, Uttar Pradesh-208016, India
| | - Sanjay Gambhir
- Sanjay Gandhi Post Graduate Institute of Medical Sciences , Lucknow, Uttar Pradesh India
| | - Ashutosh Sharma
- Department of Chemical Engineering, Centre for Environmental Science & Engineering, Thematic Unit of Excellence in Soft Nanofabrication, Indian Institute of Technology , Kanpur, Uttar Pradesh-208016, India
| | - Sri Sivakumar
- Department of Chemical Engineering, Centre for Environmental Science & Engineering, Thematic Unit of Excellence in Soft Nanofabrication, Indian Institute of Technology , Kanpur, Uttar Pradesh-208016, India
- Materials Science Programme, Indian Institute of Technology , Kanpur, Uttar Pradesh-208016, India
| |
Collapse
|
48
|
Rappaport J, Manthe RL, Solomon M, Garnacho C, Muro S. A Comparative Study on the Alterations of Endocytic Pathways in Multiple Lysosomal Storage Disorders. Mol Pharm 2016; 13:357-368. [PMID: 26702793 DOI: 10.1021/acs.molpharmaceut.5b00542] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Many cellular activities and pharmaceutical interventions involve endocytosis and delivery to lysosomes for processing. Hence, lysosomal processing defects can cause cell and tissue damage, as in lysosomal storage diseases (LSDs) characterized by lysosomal accumulation of undegraded materials. This storage causes endocytic and trafficking alterations, which exacerbate disease and hinder treatment. However, there have been no systematic studies comparing different endocytic routes in LSDs. Here, we used genetic and pharmacological models of four LSDs (type A Niemann-Pick, type C Niemann-Pick, Fabry, and Gaucher diseases) and evaluated the pinocytic and receptor-mediated activity of the clathrin-, caveolae-, and macropinocytic routes. Bulk pinocytosis was diminished in all diseases, suggesting a generic endocytic alteration linked to lysosomal storage. Fluid-phase (dextran) and ligand (transferrin) uptake via the clathrin route were lower for all LSDs. Fluid-phase and ligand (cholera toxin B) uptake via the caveolar route were both affected but less acutely in Fabry or Gaucher diseases. Epidermal growth factor-induced macropinocytosis was altered in Niemann-Pick cells but not other LSDs. Intracellular trafficking of ligands was also distorted in LSD versus wild-type cells. The extent of these endocytic alterations paralleled the level of cholesterol storage in disease cell lines. Confirming this, pharmacological induction of cholesterol storage in wild-type cells disrupted endocytosis, and model therapeutics restored uptake in proportion to their efficacy in attenuating storage. This suggests a proportional and reversible relationship between endocytosis and lipid (cholesterol) storage. By analogy, the accumulation of biological material in other diseases, or foreign material from drugs or their carriers, may cause similar deficits, warranting further investigation.
Collapse
Affiliation(s)
- Jeff Rappaport
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742-4450, USA
| | - Rachel L Manthe
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742-4450, USA
| | - Melani Solomon
- Institute for Bioscience and Biotechnology Research, University of Maryland, 5115 Plant Sciences Building, College Park, MD 20742-4450, USA
| | - Carmen Garnacho
- Department of Normal and Pathological Histology and Cytology, University of Seville School of Medicine, Seville 41009, Spain
| | - Silvia Muro
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742-4450, USA.,Institute for Bioscience and Biotechnology Research, University of Maryland, 5115 Plant Sciences Building, College Park, MD 20742-4450, USA
| |
Collapse
|
49
|
Ehrlich M. Endocytosis and trafficking of BMP receptors: Regulatory mechanisms for fine-tuning the signaling response in different cellular contexts. Cytokine Growth Factor Rev 2015; 27:35-42. [PMID: 26776724 DOI: 10.1016/j.cytogfr.2015.12.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Signaling by bone morphogenetic protein (BMP) receptors is regulated at multiple levels in order to ensure proper interpretation of BMP stimuli in different cellular settings. As with other signaling receptors, regulation of the amount of exposed and signaling-competent BMP receptors at the plasma-membrane is predicted to be a key mechanism in governing their signaling output. Currently, the endocytosis of BMP receptors is thought to resemble that of the structurally related transforming growth factor-β (TGF-β) receptors, as BMP receptors are constitutively internalized (independently of ligand binding), with moderate kinetics, and mostly via clathrin-mediated endocytosis. Also similar to TGF-β receptors, BMP receptors are able to signal from the plasma membrane, while internalization to endosomes may have a signal modulating effect. When at the plasma membrane, BMP receptors localize to different membrane domains including cholesterol rich domains and caveolae, suggesting a complex interplay between membrane distribution and internalization. An additional layer of complexity stems from the putative regulatory influence on the signaling and trafficking of BMP receptors exerted by ligand traps and/or co-receptors. Furthermore, the trafficking and signaling of BMP receptors are subject to alterations in cellular context. For example, genetic diseases involving changes in the expression of auxiliary factors of endocytic pathways hamper retrograde BMP signals in neurons, and perturb the regulation of synapse formation. This review summarizes current understanding of the trafficking of BMP receptors and discusses the role of trafficking in regulation of BMP signals.
Collapse
Affiliation(s)
- Marcelo Ehrlich
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
50
|
Laval K, Favoreel HW, Van Cleemput J, Poelaert KCK, Brown IK, Verhasselt B, Nauwynck HJ. Entry of equid herpesvirus 1 into CD172a+ monocytic cells. J Gen Virol 2015; 97:733-746. [PMID: 26684016 DOI: 10.1099/jgv.0.000375] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Equid herpesvirus 1 (EHV-1) causes respiratory disease, abortion and neurological disorders in horses. Cells from the myeloid lineage (CD172a+) are one of the main target cells of EHV-1 during primary infection. Recently, we showed that EHV-1 restricts and delays its replication in CD172a+ cells as part of an immune-evasive strategy to disseminate to target organs. Here, we hypothesize that a low efficiency of EHV-1 binding to and entry in CD172a+ cells is responsible for this restriction. Thus, we characterized EHV-1 binding and entry into CD172a+ cells, and showed that EHV-1 only bound to 15-20 % of CD172a+ cells compared with 70 % of RK-13 control cells. Enzymic removal of heparan sulphate did not reduce EHV-1 infection, suggesting that EHV-1 does not use heparan sulphate to bind and enter CD172a+ cells. In contrast, we found that treatment of cells with neuraminidase (NA) reduced infection by 85-100 % compared with untreated cells, whilst NA treatment of virus had no effect on infection. This shows that sialic acid residues present on CD172a+ cells are essential in the initiation of EHV-1 infection. We found that αVβ3 integrins are involved in the post-binding stage of CD172a+ cell infection. Using pharmacological inhibitors, we showed that EHV-1 does not enter CD172a+ cells via a clathrin- or caveolae-dependent endocytic pathway, nor by macropinocytosis, but requires cholesterol, tyrosine kinase, actin, dynamin and endosomal acidification, pointing towards a phagocytic mechanism. Overall, these results show that the narrow tropism of EHV-1 amongst CD172a+ cells is determined by the presence of specific cellular receptors.
Collapse
Affiliation(s)
- Kathlyn Laval
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Herman W Favoreel
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Jolien Van Cleemput
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Katrien C K Poelaert
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Ivy K Brown
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Bruno Verhasselt
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, De Pintelaan 185, 9000 Gent, Belgium
| | - Hans J Nauwynck
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| |
Collapse
|