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
|
Castellano M, Blanco V, Li Calzi M, Costa B, Witwer K, Hill M, Cayota A, Segovia M, Tosar JP. Ribonuclease activity undermines immune sensing of naked extracellular RNA. CELL GENOMICS 2025; 5:100874. [PMID: 40334662 DOI: 10.1016/j.xgen.2025.100874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 02/26/2025] [Accepted: 04/10/2025] [Indexed: 05/09/2025]
Abstract
Cell membranes are thought of as barriers to extracellular RNA (exRNA) uptake. While naked exRNAs can be spontaneously internalized by certain cells, functional cytosolic delivery has been rarely observed. Here, we show that extracellular ribonucleases (RNases)-primarily from cell culture supplements-have obscured the study of exRNA functionality. When ribonuclease inhibitor (RI) is added to cell cultures, naked exRNAs can trigger pro-inflammatory responses in dendritic cells and macrophages, largely via endosomal Toll-like receptors (TLRs). Moreover, naked exRNAs can escape endosomes, engaging cytosolic RNA sensors. In addition, naked extracellular mRNAs can be spontaneously internalized and translated by various cell types in an RI-dependent manner. In vivo, RI co-injection amplifies naked-RNA-induced activation of splenic lymphocytes and myeloid leukocytes. Furthermore, naked RNA is inherently pro-inflammatory in RNase-poor compartments like the peritoneal cavity. These findings demonstrate that naked RNA is bioactive without requiring vesicular encapsulation, making a case for nonvesicular-exRNA-mediated intercellular communication.
Collapse
Affiliation(s)
- Mauricio Castellano
- Functional Genomics Laboratory, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay; Immunoregulation and Inflammation Laboratory, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay; Analytical Biochemistry Unit, School of Science, Universidad de la República, Montevideo 11400, Uruguay
| | - Valentina Blanco
- Functional Genomics Laboratory, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
| | - Marco Li Calzi
- Functional Genomics Laboratory, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
| | - Bruno Costa
- Functional Genomics Laboratory, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay; Analytical Biochemistry Unit, School of Science, Universidad de la República, Montevideo 11400, Uruguay
| | - Kenneth Witwer
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; EV Core Facility "EXCEL," Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Marcelo Hill
- Immunoregulation and Inflammation Laboratory, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay; Academic Unit of Immunobiology, School of Medicine, Universidad de la República, Montevideo 11800, Uruguay
| | - Alfonso Cayota
- Functional Genomics Laboratory, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay; Hospital de Clínicas, Universidad de la República, Montevideo 11600, Uruguay
| | - Mercedes Segovia
- Immunoregulation and Inflammation Laboratory, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay; Academic Unit of Immunobiology, School of Medicine, Universidad de la República, Montevideo 11800, Uruguay.
| | - Juan Pablo Tosar
- Functional Genomics Laboratory, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay; Analytical Biochemistry Unit, School of Science, Universidad de la República, Montevideo 11400, Uruguay.
| |
Collapse
|
3
|
Woo TG, Han J, Kim Y, Hwang YJ, Lee M, Kang SM, Park S, Ji Y, Chung YH, Baek S, Shin E, Minju-Kim, Jang H, Shin YJ, Kwon Y, Kim BH, Park BJ. Inhibition of SOD1 trimerization is a novel drug target for ALS disease. Transl Neurodegener 2025; 14:21. [PMID: 40350531 PMCID: PMC12067741 DOI: 10.1186/s40035-025-00483-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 03/28/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease that begins with motor neuron death in the spinal cord and cerebral cortex, ultimately resulting in death from respiratory distress (breathing failure). About 90% of ALS cases are sporadic, and 10% of ALS cases are of the inherited type with a genetic cause. About 150 different gene mutations have been reported so far. SOD1 is a well-identified gene associated with ALS. Indeed, SOD1 aggregation has been reported in ALS patients, but the mechanism of SOD1 aggregation remains unclear. Our previous work showed that inhibiting SOD1 aggregation with a hit compound (PRG-A-01) could reduce the SOD1-induced cytotoxicity and extend the lifespan of ALS mouse model (SOD1G93A-Tg). However, the low bioavailability and rapid degradation of the compound in vivo necessitates the development of a more effective candidate. We generated different derivatives and finally obtained the most potential drug candidate, PRG-A-04. METHODS Neuronal cell lines were transfected with the mutant SOD1 expression vector and incubated with PRG-A-04. SOD1 aggregation was examined by SOD1 oligomerization assay, immunofluorescence and dot blot assay. The interaction between GST-conjugated SOD1 recombinant proteins and PRG-A-04 was identified using LC-MS/MS and GST pull-down assay. To check the in vivo therapeutic effect of PRG-A-04, SOD1G93A-Tg mice were injected with PRG-A-04; then behavioral test, histological analysis and microarray were performed. RESULTS PRG-A-04 demonstrated favorable pharmacokinetics including high bioavailability and significant blood-brain barrier penetration. Indeed, oral administration of PRG-A-04 in ALS mouse model inhibited the aggregation of SOD1 in the spinal cord, protected against neuronal loss, and extended the lifespan of ALS mice by up to 3 weeks. In vitro, PRG-A-04 selectively bound to the mutant form of SOD1, but not the wild type, and efficiently inhibited the aggregation caused by SOD1-G147P (a SOD1 trimer stabilizer). CONCLUSIONS Our findings underscore the potential of targeting trimeric SOD1 in ALS treatment, positioning PRG-A-04 as a strong drug candidate for both familial and sporadic ALS.
Collapse
Affiliation(s)
- Tae-Gyun Woo
- Institute of Rare Genetic Disease, PRG S&Tech Co., LTD, Busan, 46274, Republic of Korea
| | - Jin Han
- Institute of Rare Genetic Disease, PRG S&Tech Co., LTD, Busan, 46274, Republic of Korea
| | - Yuju Kim
- Institute of Rare Genetic Disease, PRG S&Tech Co., LTD, Busan, 46274, Republic of Korea
| | - Young Jun Hwang
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, 46241, Republic of Korea
| | - Mua Lee
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, 46241, Republic of Korea
| | - So-Mi Kang
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, 46241, Republic of Korea
| | - Soyoung Park
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, 46241, Republic of Korea
| | - Yeongseon Ji
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, 46241, Republic of Korea
| | - Yeon-Ho Chung
- Institute of Rare Genetic Disease, PRG S&Tech Co., LTD, Busan, 46274, Republic of Korea
| | - Songyoung Baek
- Institute of Rare Genetic Disease, PRG S&Tech Co., LTD, Busan, 46274, Republic of Korea
| | - Eunbyeol Shin
- Institute of Rare Genetic Disease, PRG S&Tech Co., LTD, Busan, 46274, Republic of Korea
| | - Minju-Kim
- Institute of Rare Genetic Disease, PRG S&Tech Co., LTD, Busan, 46274, Republic of Korea
| | - Hyewon Jang
- Department of Agricultural Biotechnology, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yun-Jeong Shin
- Department of Agricultural Biotechnology, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yonghoon Kwon
- Department of Agricultural Biotechnology, Seoul National University, Seoul, 08826, Republic of Korea
| | - Bae-Hoon Kim
- Institute of Rare Genetic Disease, PRG S&Tech Co., LTD, Busan, 46274, Republic of Korea
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, 46241, Republic of Korea
| | - Bum-Joon Park
- Institute of Rare Genetic Disease, PRG S&Tech Co., LTD, Busan, 46274, Republic of Korea.
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, 46241, Republic of Korea.
| |
Collapse
|
4
|
Parui R, Roy H, Meher N, Ghosh SS, Iyer PK. Mechanistic investigation on cellular internalization triggering structure-induced conformational modulation of boron-nitrogen luminogens. Chem Sci 2025; 16:6023-6034. [PMID: 40070468 PMCID: PMC11891783 DOI: 10.1039/d4sc08296f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Exploring the effects of electron donor (D) and acceptor (A) functional groups in tuning the condensed state properties has been a challenging yet efficient approach to reveal promising materials for cutting-edge applications. Herein, a series of boron-nitrogen (BN) incorporated organic congeners (NBNMe2, NBOMe, NBF, NBCl, NBBr, NBCN, NBPy) appended with functional groups having various degrees of D/A characteristics were developed and their potential in controlling supramolecular assembly and condensed state luminescence features (>90 nm redshift in λ em,max) was explored. Despite the minor structural engineering in BN-based small molecules, they effectively modulated conformational orientation and molecular packing, leading to the directed growth of distinct and highly ordered self-assembly patterns, i.e., nanosheets, nanospheres, nanowires, and nanorods. The structure-property correlation investigation also highlighted the time-dependent fluorescence enhancement for NBPy owing to morphological growth via the fusion of nanospheres into nanowire conformation. Further, these nano-architectures with distinct conformations were employed to examine the mechanistic aspects as well as the influence of morphologies in cellular uptake and imaging, where all the nano-aggregates exhibited lysosomal localization following multiple endocytosis pathways and the nanorods possessed the highest uptakes (CTCF4h/0.5h = 3.11) with respect to other conformations. The in-depth inspection of the structural impact in single crystal X-ray diffraction (SCXRD) analysis disclosed the decisive role of boron atoms and functional group tuning that built a conceptual correlation between the molecular architecture and their photophysical characteristics, supramolecular assembly, and cellular internalization process, offering key insights on the development of rapid and effective drug delivery techniques.
Collapse
Affiliation(s)
- Retwik Parui
- Department of Chemistry, Indian Institute of Technology Guwahati Guwahati-781039 Assam India
| | - Hirakjyoti Roy
- Centre for Nanotechnology, Indian Institute of Technology Guwahati Guwahati-781039 Assam India
| | - Niranjan Meher
- Department of Chemistry, Indian Institute of Technology Guwahati Guwahati-781039 Assam India
| | - Siddhartha Sankar Ghosh
- Centre for Nanotechnology, Indian Institute of Technology Guwahati Guwahati-781039 Assam India
- Department of Bioscience and Bioengineering, Indian Institute of Technology Guwahati Guwahati-781039 Assam India
| | - Parameswar Krishnan Iyer
- Department of Chemistry, Indian Institute of Technology Guwahati Guwahati-781039 Assam India
- Centre for Nanotechnology, Indian Institute of Technology Guwahati Guwahati-781039 Assam India
| |
Collapse
|
5
|
Bolanle IO, de Liedekerke Beaufort GC, Weinberg PD. Transcytosis of LDL Across Arterial Endothelium: Mechanisms and Therapeutic Targets. Arterioscler Thromb Vasc Biol 2025; 45:468-480. [PMID: 40013359 PMCID: PMC11936472 DOI: 10.1161/atvbaha.124.321549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/11/2025] [Indexed: 02/28/2025]
Abstract
Transport of LDL (low-density lipoprotein) from plasma to arterial intima is thought to be rate limiting in the development of atherosclerosis. Its variation likely determines where lesions develop within arteries and might account for some of the currently unexplained difference in disease susceptibility between individuals. It may also be critical in the development of lipid-rich, unstable plaques. Mechanisms have been controversial but recent evidence suggests that caveolar transcytosis across endothelial cells is the dominant pathway. Receptors involved are LDLR (LDL receptor), SR-B1 (scavenger receptor class B type 1), and ALK1 (activin receptor-like kinase 1). The role of LDLR is influenced by IL-1β (interleukin-1β); the role of SR-B1 by HDL (high-density lipoprotein), DOCK4 (dedicator of cytokinesis 4), GPER (G-protein-coupled estrogen receptor), and HMGB1 (high mobility group box 1); and the role of ALK1 by BMP (bone morphogenetic protein) 9. Additionally, BMP4 stimulates transcytosis and FSTL1 (follistatin-like 1 protein) inhibits it. Fundamental transcytotic mechanisms include caveola formation, undocking, trafficking, and docking; they are influenced by cholesterol-lowering agents, MYDGF (myeloid-derived growth factor), MFSD2a (major facilitator superfamily domain containing 2a) in the blood-brain barrier, and inhibitors of dynamin-2 and tubulin polymerization. The relative merits of different therapeutic approaches are discussed, with statins, colchicine, benzimidazoles, and metformin being existing drugs that might be repurposed and salidroside and glycyrrhizic acid being nutraceuticals worth investigating. Finally, we discuss evidence against the ferry-boat model of transcytosis, the contributions of receptor-mediated, fluid-phase, and active transcytosis, and where inhibition of transcytosis might be most beneficial.
Collapse
Affiliation(s)
- Israel O. Bolanle
- Department of Bioengineering, Imperial College London, United Kingdom
| | | | - Peter D. Weinberg
- Department of Bioengineering, Imperial College London, United Kingdom
| |
Collapse
|
6
|
Furman VV, Semenova SB. Caco-2 Cellular Model to Study In Vitro the Mechanisms of Intestinal Barrier Permeability. CELL AND TISSUE BIOLOGY 2025; 19:125-129. [DOI: 10.1134/s1990519x24600704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 04/08/2025]
|
7
|
Masete KV, Massarani AS, Schulzke JD, Epple HJ, Hering NA. Tumour necrosis factor-α induces macromolecule translocation in HIV-derived duodenal organoids. Front Immunol 2025; 16:1563702. [PMID: 40170863 PMCID: PMC11959035 DOI: 10.3389/fimmu.2025.1563702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 02/26/2025] [Indexed: 04/03/2025] Open
Abstract
Background Disease progression from human immunodeficiency virus (HIV) infection to acquired immunodeficiency syndrome (AIDS) is marked by chronic immune activation, partly due to increased translocation of gut-derived microbial antigens. Elevated mucosal tumour necrosis factor-α (TNF-α) and resulting epithelial cell apoptosis may be the etiology. However, studies using carcinoma cell lines have failed to find a causal link, possibly due to cellular abnormalities related to the malignant transformation of these immortal cell lines. Methods We established intestinal organoid monolayers from healthy controls and HIV-infected adults and characterized their growth dynamics and cellular composition. We then examined the effects of HIV-associated cytokines on transepithelial resistance (TER), apoptosis and macromolecule translocation. Results Organoid monolayers from HIV-infected patients grew similarly to healthy controls, forming confluent monolayers within one to two weeks containing enterocytes, Paneth, goblet and stem cells. IFN-γ synergized with TNF-α, allowing TNF-α to cause caspase-mediated apoptosis and TER reduction within 5 ± 3 hours, reflecting patient sample heterogeneity. This led to paracellular passage of 4 kDa Dextran and transcytosis of 44 kDa horse radish peroxidase, both of which could be blocked by pan-caspase inhibitor, Q-VD-Oph. Conclusion Our study confirms that intestinal organoid monolayers from biopsies of HIV-infected individuals can be used to study apoptosis-related epithelial barrier dysfunction and macromolecular translocation. We provide direct evidence that TNF-α-induced apoptosis triggered two pathways of macromolecular translocation: paracellular passage via apoptotic leaks and transcytosis. Therapies targeting apoptosis may be useful in preventing disease progression from HIV to AIDS.
Collapse
Affiliation(s)
- Kopano Valerie Masete
- Department of Gastroenterology, Rheumatology and Infectious Diseases, Clinical Physiology/Nutritional Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Alain S. Massarani
- Department of Gastroenterology, Rheumatology and Infectious Diseases, Clinical Physiology/Nutritional Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Jörg-Dieter Schulzke
- Department of Gastroenterology, Rheumatology and Infectious Diseases, Clinical Physiology/Nutritional Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Hans-Jörg Epple
- Department of Gastroenterology, Rheumatology and Infectious Diseases, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Antibiotic Stewardship Team, Medical Directorate, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Nina A. Hering
- Department of General and Visceral Surgery, Charité – Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
8
|
Candor K, Ding L, Balchand S, Hammonds JE, Spearman P. The CLIC/GEEC pathway regulates particle uptake and formation of the virus-containing compartment (VCC) in HIV-1-infected macrophages. PLoS Pathog 2025; 21:e1012564. [PMID: 40067817 PMCID: PMC11925468 DOI: 10.1371/journal.ppat.1012564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 03/20/2025] [Accepted: 02/12/2025] [Indexed: 03/22/2025] Open
Abstract
HIV-1 particles are captured by the immunoglobulin superfamily member Siglec-1 on the surface of macrophages and dendritic cells, leading to particle internalization and facilitating trans-infection of CD4+ T cells. HIV-1-infected macrophages develop a unique intracellular compartment termed the virus-containing compartment (VCC) that exhibits characteristic markers of the late endosome and is enriched in components of the plasma membrane (PM). The VCC has been proposed as the major site of particle assembly in macrophages. Depleting Siglec-1 from macrophages significantly reduces VCC formation, implying a link between the capture and uptake of external HIV-1 particles and the development of VCCs within HIV-infected cells. We found that internalization of particles to the VCC was independent of clathrin, but required dynamin-2. CD98 and CD44, classical markers of the CLIC/GEEC pathway, colocalized with Siglec-1 and HIV-1 particles within the VCC. Virus-like particles (VLPs) were taken up within CD98 and Siglec-1-enriched tubular membranes that migrated centripetally over time to form VCC-like structures. Inhibition of CLIC/GEEC-mediated endocytosis resulted in the arrest of captured HIV-1 particles on the macrophage cell surface, prevented VCC formation, and significantly reduced the efficiency of trans-infection of T cells. These findings indicate that following capture of virus by Siglec-1, particles follow an endocytic route to the VCC that requires both the CLIC/GEEC pathway and dynamin-2. We propose a model in which internalization of HIV-1 particles together with CLIC/GEEC membranes leads to the formation of the VCC in HIV-infected macrophages, creating an intracellular platform that facilitates further particle assembly and budding.
Collapse
Affiliation(s)
- Kathleen Candor
- Immunology Graduate Program, University of Cincinnati, and Infectious Diseases Division, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Infectious Diseases Division, Cincinnati Children’s Hospital Medical Center and University of Cincinnati, Cincinnati, Ohio United States of America
| | - Lingmei Ding
- Infectious Diseases Division, Cincinnati Children’s Hospital Medical Center and University of Cincinnati, Cincinnati, Ohio United States of America
| | - Sai Balchand
- Infectious Diseases Division, Cincinnati Children’s Hospital Medical Center and University of Cincinnati, Cincinnati, Ohio United States of America
| | - Jason E. Hammonds
- Infectious Diseases Division, Cincinnati Children’s Hospital Medical Center and University of Cincinnati, Cincinnati, Ohio United States of America
| | - Paul Spearman
- Infectious Diseases Division, Cincinnati Children’s Hospital Medical Center and University of Cincinnati, Cincinnati, Ohio United States of America
| |
Collapse
|
9
|
Wong TH, Khater IM, Hallgrimson C, Li YL, Hamarneh G, Nabi IR. SuperResNET - single-molecule network analysis detects changes to clathrin structure induced by small-molecule inhibitors. J Cell Sci 2025; 138:JCS263570. [PMID: 39865933 DOI: 10.1242/jcs.263570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 01/17/2025] [Indexed: 01/28/2025] Open
Abstract
SuperResNET is a network analysis pipeline for the analysis of point cloud data generated by single-molecule localization microscopy (SMLM). Here, we applied SuperResNET network analysis of SMLM direct stochastic optical reconstruction microscopy (dSTORM) data to determine how the clathrin endocytosis inhibitors pitstop 2, dynasore and latrunculin A (LatA) alter the morphology of clathrin-coated pits. SuperResNET analysis of HeLa and Cos7 cells identified three classes of clathrin structures: small oligomers (class I), pits and vesicles (class II), and larger clusters corresponding to fused pits or clathrin plaques (class III). Pitstop 2 and dynasore treatment induced distinct homogeneous populations of class II structures in HeLa cells, suggesting that they arrest endocytosis at different stages. Inhibition of endocytosis was not via actin depolymerization, as the actin-depolymerizing agent LatA induced large, heterogeneous clathrin structures. Ternary analysis of SuperResNET shape features presented a distinct more planar profile for blobs from pitstop 2-treated cells, which aligned with clathrin pits identified with high-resolution minimal photon fluxes (MINFLUX) microscopy, whereas control structures resembled MINFLUX clathrin vesicles. SuperResNET analysis therefore showed that pitstop 2 arrests clathrin pit maturation at early stages of pit formation, representing an approach to detect the effect of small molecules on target structures in situ in the cell from SMLM datasets.
Collapse
Affiliation(s)
- Timothy H Wong
- Department of Cellular & Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Ismail M Khater
- School of Computing Science, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
- Department of Electrical and Computer Engineering, Faculty of Engineering and Technology, Birzeit University, Birzeit P627, Palestine
| | | | - Y Lydia Li
- Department of Cellular & Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Ghassan Hamarneh
- School of Computing Science, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Ivan R Nabi
- Department of Cellular & Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
10
|
Grouzdev D, Pales Espinosa E, Tettelbach S, Tanguy A, Boutet I, Tobi H, Allam B. Survival of the fittest: genomic investigations of the bay scallop reveal a shift in population structure through a summer mortality event. BMC Genomics 2025; 26:146. [PMID: 39955531 PMCID: PMC11829576 DOI: 10.1186/s12864-025-11337-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 02/10/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Understanding the genetic basis of resilience in marine organisms is critical for conservation and management, particularly in the face of escalating environmental stress and disease outbreaks. The bay scallop Argopecten irradians is a commercially and recreationally important shellfish species found in estuarine and coastal environments of the United States from New England to the Gulf of Mexico. In New York, adult bay scallop populations have been decimated every summer since 2019 leading to the collapse of their fishery. These mortality events were associated with annual outbreaks of an undescribed apicomplexan parasite recently named Bay Scallop Marosporida (BSM) that disrupts scallop kidneys. RESULTS This study investigates host-pathogen interactions and assesses changes in population structure during BSM-associated mortality events. The research compared wild and aquacultured scallops used for stock enhancement in New York, revealing significant change in population structures throughout the mortality outbreak. The results underscore the selective pressures exerted by BSM infection and environmental stressors, as evidenced by shifts in genetic divergence and allele frequencies particularly in genes associated with kidney function, stress and infection response. Through a detailed genomic and population genetic approach, this research represents a unique case study highlighting the impact of disease on marine biodiversity and advances our understanding of the impact of summer mortality events on the scallop population in NY. CONCLUSIONS This study highlights changes in the genomic structure of bay scallops during a BSM-associated mortality event. Identified mutations (such as the one in the nephrocystin-3-like gene) represent prime candidates for specific targeted investigations to link genotypes to phenotypes. By integrating genomic and epidemiological data, the research provides a basis for understanding the impact of disease on scallop biodiversity. These findings may help guide conservation strategies for sustainable fisheries in the face of environmental change and disease outbreaks.
Collapse
Affiliation(s)
- Denis Grouzdev
- School of Marine and Atmospheric Sciences, Stony Brook University, Stony Brook, NY, 11794-5000, USA.
| | | | - Stephen Tettelbach
- Cornell Cooperative Extension of Suffolk County, Southold, NY, 11971, USA
| | - Arnaud Tanguy
- Station Biologique de Roscoff, CNRS/Sorbonne Université, Place Georges Teissier, 29680, Roscoff, France
| | - Isabelle Boutet
- Station Biologique de Roscoff, CNRS/Sorbonne Université, Place Georges Teissier, 29680, Roscoff, France
| | - Harrison Tobi
- Cornell Cooperative Extension of Suffolk County, Southold, NY, 11971, USA
| | - Bassem Allam
- School of Marine and Atmospheric Sciences, Stony Brook University, Stony Brook, NY, 11794-5000, USA.
| |
Collapse
|
11
|
Alvarez-Olmedo D, Kamaliddin C, Verhey TB, Ho M, De Vinney R, Chaconas G. Transendothelial migration of the Lyme disease spirochete involves spirochete internalization as an intermediate step through a transcellular pathway that involves Cdc42 and Rac1. Microbiol Spectr 2025; 13:e0222124. [PMID: 39727396 PMCID: PMC11792520 DOI: 10.1128/spectrum.02221-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/22/2024] [Indexed: 12/28/2024] Open
Abstract
Despite its importance in pathogenesis, the hematogenous dissemination pathway of Borrelia burgdorferi is still largely uncharacterized. To probe the molecular details of transendothelial migration more easily, we studied this process using cultured primary or telomerase-immortalized human microvascular endothelial cells in a medium that maintains both the human cells and the spirochetes. In B. burgdorferi-infected monolayers, we observed ~55% of wild-type spirochetes crossing the monolayer. Microscopic characterization revealed entrance points across the cellular surface rather than at cellular junctions, supporting a transcellular route. In support of this pathway, locking the endothelial junctions using a vascular endothelial protein tyrosine phosphatase (VE-PTP) inhibitor did not reduce transendothelial migration. We also used inhibitors to block the most common endocytic pathways to elucidate effectors that might be involved in B. burgdorferi uptake and/or transmigration. Directly inhibiting Cdc42 reduced spirochete transmigration by impeding internalization. However, blocking Rac1 alone dramatically reduced transmigration by ~84% and resulted in a concomitant doubling in spirochete accumulation in the cell. Our combined results support that B. burgdorferi internalization is an intermediate step in the transendothelial migration process, which requires both Cdc42 and Rac1; Cdc42 is needed for spirochete internalization, while Rac1 is required for cellular egress. These are the first two host proteins implicated in B. burgdorferi transmigration across endothelial cells.IMPORTANCELyme borreliosis is caused by Borrelia burgdorferi and related bacteria. It is the most common tick-transmitted illness in the Northern Hemisphere. The ability of this pathogen to spread to a wide variety of locations results in a diverse set of clinical manifestations, yet little is known regarding vascular escape of the spirochete, an important pathway for dissemination. Our current work has studied the traversal of B. burgdorferi across a monolayer of microvascular endothelial cells grown using a new culture system. We show that this occurs by passage of the spirochetes directly through cells rather than at cellular junctions and that internalization of B. burgdorferi is an intermediate step in transmigration. We also identify the first two host proteins, Cdc42 and Rac1, that are used by the spirochetes to promote traversal of the cellular monolayer. Our new experimental system also provides a new avenue for further studies of this important process.
Collapse
Affiliation(s)
- Daiana Alvarez-Olmedo
- Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, Alberta, Canada
- Calvin, Phoebe & Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Claire Kamaliddin
- Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, Alberta, Canada
- Calvin, Phoebe & Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, The University of Calgary, Calgary, Alberta, Canada
| | - Theodore B. Verhey
- Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - May Ho
- Calvin, Phoebe & Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, The University of Calgary, Calgary, Alberta, Canada
| | - Rebekah De Vinney
- Calvin, Phoebe & Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, The University of Calgary, Calgary, Alberta, Canada
| | - George Chaconas
- Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, Alberta, Canada
- Calvin, Phoebe & Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, The University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
12
|
Jose S, Sharma H, Insan J, Sharma K, Arora V, Puranapanda S, Dhamija S, Eid N, Menon MB. Kinase Inhibitor-Induced Cell-Type Specific Vacuole Formation in the Absence of Canonical ATG5-Dependent Autophagy Initiation Pathway. Mol Cell Biol 2025; 45:99-115. [PMID: 39895059 DOI: 10.1080/10985549.2025.2454421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 11/20/2024] [Accepted: 01/10/2025] [Indexed: 02/04/2025] Open
Abstract
Pyridinyl-imidazole class p38 MAPKα/β (MAPK14/MAPK11) inhibitors including SB202190 have been shown to induce cell-type specific defective autophagy resulting in micron-scale vacuole formation, cell death, and tumor suppression. We had earlier shown that this is an off-target effect of SB202190. Here we provide evidence that this vacuole formation is independent of ATG5-mediated canonical autophagosome initiation. While SB202190 interferes with autophagic flux in many cell lines parallel to vacuolation, autophagy-deficient DU-145 cells and CRISPR/Cas9 gene-edited ATG5-knockout A549 cells also undergo vacuolation upon SB202190 treatment. Late-endosomal GTPase RAB7 colocalizes with these compartments and RAB7 GTP-binding is essential for SB202190-induced vacuolation. A screen for modulators of SB202190-induced vacuolation revealed molecules including multi-kinase inhibitor sorafenib as inhibitors of vacuolation and sorafenib co-treatment enhanced cytotoxicity of SB202190. Moreover, VE-821, an ATR inhibitor was found to phenocopy the cell-type specific vacuolation response of SB202190. To identify the factors determining the cell-type specificity of vacuolation induced by SB-compounds and VE-821, we compared the transcriptomics data from vacuole-forming and non-vacuole-forming cancer cell lines and identified a gene expression signature that may define sensitivity of cells to these small-molecules. Further analyses using small molecule tools and the gene signature discovered here, could reveal novel mechanisms regulating this interesting anti-cancer phenotype.
Collapse
Affiliation(s)
- Susan Jose
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
| | - Himanshi Sharma
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
| | - Janki Insan
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
| | - Khushboo Sharma
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
| | - Varun Arora
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
| | | | - Sonam Dhamija
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Nabil Eid
- Department of Anatomy, Division of Human Biology, School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| | - Manoj B Menon
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
| |
Collapse
|
13
|
Shimosaka M, Kondo J, Sonoda M, Kawaguchi R, Noda E, Nishikori K, Ogata A, Takamatsu S, Sasai K, Akita H, Eguchi H, Kamada Y, Okamoto S, Miyoshi E. Invasion of pancreatic ductal epithelial cells by Enterococcus faecalis is mediated by fibronectin and enterococcal fibronectin-binding protein A. Sci Rep 2025; 15:2585. [PMID: 39833342 PMCID: PMC11747100 DOI: 10.1038/s41598-025-86531-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025] Open
Abstract
The poor prognosis of pancreatic cancer is often attributed to difficulties of early detection due to a lack of appropriate risk factors. Previously, we demonstrated the presence of Enterococcus faecalis (E. faecalis) in pancreatic juice and tissues obtained from patients with cancers of the duodeno-pancreato-biliary region, suggesting the possible involvement of this bacterial species in chronic and malignant pancreatic diseases. However, it remains unclear if and how E. faecalis can infect pancreatic ductal cells. In this study, we used immortalized normal human pancreatic ductal epithelial cells (iPDECs) and pancreatic ductal cancer cell lines to demonstrate that E. faecalis adheres to and invades pancreatic ductal lineage epithelial cells. Inhibitors of micropinocytosis or clathrin- or caveolae-mediated endocytosis suppressed iPDEC invasion by E. faecalis. Mechanistically, bacterial expression of enterococcal fibronectin-binding protein A (EfbA) was correlated with adhesive potential of E. faecalis strains. Knockout of fibronectin 1, a binding partner of EfbA, in iPDECs resulted in suppressed E. faecalis adhesion and invasion, suggesting the importance of the EfbA-fibronectin axis in infection of pancreatic ductal epithelial lineage cells. Overall, these results suggest that E. faecalis can colonize pancreatic tissue by infecting iPDECs, at least in part, via the expression of the cell adhesion factor EfbA.
Collapse
Affiliation(s)
- Munefumi Shimosaka
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Jumpei Kondo
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Mamika Sonoda
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Rui Kawaguchi
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Emika Noda
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kaho Nishikori
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Asuka Ogata
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Shinji Takamatsu
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Ken Sasai
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hirofumi Akita
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoshihiro Kamada
- Department of Advanced Metabolic Hepatology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Shigefumi Okamoto
- Laboratory of Medical Microbiology and Microbiome, Department of Clinical Laboratory and Biomedical Sciences, Division of Health Sciences, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Eiji Miyoshi
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.
| |
Collapse
|
14
|
Kwaku GN, Jensen KN, Simaku P, Floyd DJ, Saelens JW, Reardon CM, Ward RA, Basham KJ, Hepworth OW, Vyas TD, Zamith-Miranda D, Nosanchuk JD, Vyas JM, Harding HB. Extracellular vesicles from diverse fungal pathogens induce species-specific and endocytosis-dependent immunomodulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.03.631181. [PMID: 39803513 PMCID: PMC11722428 DOI: 10.1101/2025.01.03.631181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2025]
Abstract
Microbial pathogens generate extracellular vesicles (EVs) for intercellular communication and quorum sensing. Microbial EVs also induce inflammatory pathways within host innate immune cells. We previously demonstrated that EVs secreted by Candida albicans trigger type I interferon signaling in host cells specifically via the cGAS-STING innate immune signaling pathway. Here, we show that despite sharing similar properties of morphology and internal DNA content, the interactions between EVs and the innate immune system differ according to the parental fungal species. EVs secreted by C. albicans, Saccharomyces cerevisiae, Cryptococcus neoformans, and Aspergillus fumigatus are endocytosed at different rates by murine macrophages triggering varied cytokine responses, innate immune signaling, and subsequent immune cell recruitment. Notably, cell wall constituents that decorate C. neoformans and A. fumigatus EVs inhibit efficient internalization by macrophages and dampen innate immune activation. Our data uncover the transcriptional and functional consequences of the internalization of diverse fungal EVs by immune cells and reveal novel insights into the early innate immune response to distinct clinically significant fungal pathogens.
Collapse
Affiliation(s)
- Geneva N Kwaku
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Kirstine Nolling Jensen
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Patricia Simaku
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Daniel J Floyd
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Joseph W Saelens
- Pfizer Worldwide Research Development and Medical, Machine Learning and Computational Sciences, Cambridge, MA, USA
| | - Christopher M Reardon
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Rebecca A Ward
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Kyle J Basham
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Olivia W Hepworth
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Tammy D Vyas
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Daniel Zamith-Miranda
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Joshua D Nosanchuk
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jatin M Vyas
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Infectious Disease, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Hannah Brown Harding
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
15
|
Chen X, Kang Y, Tang C, Zhang L, Guo L. TLR4 promotes smooth muscle cell-derived foam cells formation by inducing receptor-independent macropinocytosis. Biosci Biotechnol Biochem 2024; 89:22-32. [PMID: 39455413 DOI: 10.1093/bbb/zbae153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024]
Abstract
Foam cells are primarily formed through scavenger receptors that mediate the uptake of various modified low-density lipoproteins (LDL) into cells. In addition to the receptor-dependent pathway, macropinocytosis is an essential nonreceptor endocytic pathway for vascular smooth muscle cells (VSMCs) to take up lipids. However, the molecular mechanisms underlying this process remain unclear. Primary cultured VSMCs were stimulated with 200 ng/mL lipopolysaccharide (LPS) and 200 µg/mL native LDL (nLDL). We observed a significant increase in Toll-like receptor 4 (TLR4) protein expression and a significant activation of macropinocytosis, which correlated with the highest uptake of nLDL and intracellular lipid deposition in WT VSMCs. However, macropinocytosis was inhibited and lipid accumulation decreased after treatment with macropinocytosis inhibitors and Syk inhibitors in WT VSMCs. Consistently, TLR4 knockout significantly suppressed macropinocytosis and lipid droplets accumulation in VSMCs. Taken together, our findings suggest a critical role of TLR4/Syk signaling in promoting receptor-independent macropinocytosis leading to VSMC-derived foam cells formation.
Collapse
MESH Headings
- Toll-Like Receptor 4/metabolism
- Pinocytosis/drug effects
- Animals
- Foam Cells/metabolism
- Foam Cells/cytology
- Foam Cells/drug effects
- Syk Kinase/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Signal Transduction
- Lipopolysaccharides/pharmacology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/cytology
- Mice
- Lipoproteins, LDL/metabolism
- Lipoproteins, LDL/pharmacology
- Cells, Cultured
- Mice, Knockout
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Xue Chen
- Department of Rheumatology and Clinical Immunology, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Yulai Kang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Chunhua Tang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Lili Zhang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Lu Guo
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| |
Collapse
|
16
|
Salgado L, Cifuentes-Delgado PC, Orozco JC, Muñoz-Camargo C, Reyes LH, Quezada V, Cruz JC. Evaluating the impact of cell-penetrating motif position on the cellular uptake of magnetite nanoparticles. Front Bioeng Biotechnol 2024; 12:1450694. [PMID: 39687269 PMCID: PMC11646778 DOI: 10.3389/fbioe.2024.1450694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
Cell-penetrating peptides (CPPs) have been employed to enhance the cellular uptake and intracellular delivery of various nanocarriers. Among them, nanoparticles (NPs) have been used as suitable vehicles for delivering different bioactive molecules in the treatment of a diverse range of diseases. Given the pivotal role of the conjugation method of CPPs, this study aims to evaluate the impact of the position of a cell-penetrating motif (LFVCR) on the biocompatibility, cellular uptake, and endosomal escape of magnetite NPs. The designed peptide's physicochemical properties suggest they are well-suited for efficient cell penetration with minimal cytotoxicity. The resulting designed nanoconjugates were characterized using Fourier transform infrared spectroscopy (FTIR), thermogravimetric analysis (TGA), dynamic light scattering (DLS), and transmission electron microscopy (TEM). The results indicate that motif position significantly impacts the cellular uptake and endosomal escape of the designed nanobioconjugates. Key findings suggest that motif exposure enhances endocytosis-mediated cell internalization and improves endosomal escape efficiency. These results were compared with nanobioconjugates displaying previously reported CPPs. The selected nanobioconjugate demonstrated superior performance in endosomal escape and comparable cell uptake to the reference nanobioconjugates. These results, along with the nanobioconjugate's physicochemical characteristics and high biocompatibility, position the nanocarrier as a suitable candidate for delivering diverse bioactive molecules.
Collapse
Affiliation(s)
- Laura Salgado
- Department of Biomedical Engineering, Universidad de Los Andes, Bogotá, Colombia
| | | | - Juan Camilo Orozco
- Center for Microscopy (MicroCore), Vice Presidency for Research and Creation, Universidad de Los Andes, Bogotá, Colombia
| | | | - Luis H. Reyes
- Product and Process Design Group (GDPP), Department of Chemical and Food Engineering, Universidad de Los Andes, Bogotá, Colombia
| | - Valentina Quezada
- Department of Biomedical Engineering, Universidad de Los Andes, Bogotá, Colombia
| | - Juan C. Cruz
- Department of Biomedical Engineering, Universidad de Los Andes, Bogotá, Colombia
| |
Collapse
|
17
|
Chen S, Zeng N, Liu GY, Wang H, Lin T, Tai Y, Chen C, Fang Y, Chuang Y, Kao C, Cheng H, Wu B, Sun P, Bayansan O, Chiu Y, Shih C, Chung W, Yang J, Wang LH, Chiang P, Chen C, Wagner OI, Wang Y, Lin Y. Precise Control of Intracellular Trafficking and Receptor-Mediated Endocytosis in Living Cells and Behaving Animals. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405568. [PMID: 39401410 PMCID: PMC11615828 DOI: 10.1002/advs.202405568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/15/2024] [Indexed: 12/06/2024]
Abstract
Intracellular trafficking, an extremely complex network, dynamically orchestrates nearly all cellular activities. A versatile method that enables the manipulation of target transport pathways with high spatiotemporal accuracy in vitro and in vivo is required to study how this network coordinates its functions. Here, a new method called RIVET (Rapid Immobilization of target Vesicles on Engaged Tracks) is presented. Utilizing inducible dimerization between target vesicles and selective cytoskeletons, RIVET can spatiotemporally halt numerous intracellular trafficking pathways within seconds in a reversible manner. Its highly specific perturbations allow for the real-time dissection of the dynamic relationships among different trafficking pathways. Moreover, RIVET is capable of inhibiting receptor-mediated endocytosis. This versatile system can be applied from the cellular level to whole organisms. RIVET opens up new avenues for studying intracellular trafficking under various physiological and pathological conditions and offers potential strategies for treating trafficking-related disorders.
Collapse
Affiliation(s)
- Shiau‐Chi Chen
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchu300044Taiwan
| | - Neng‐Jie Zeng
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchu300044Taiwan
| | - Grace Y. Liu
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchu300044Taiwan
| | - Hsien‐Chu Wang
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchu300044Taiwan
| | - Tzu‐Ying Lin
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchu300044Taiwan
| | - Yi‐Ling Tai
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchu300044Taiwan
| | - Chiao‐Yun Chen
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchu300044Taiwan
| | - Yin Fang
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchu300044Taiwan
| | - Yi‐Chien Chuang
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchu300044Taiwan
| | - Ching‐Lin Kao
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchu300044Taiwan
| | - Hsuan Cheng
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchu300044Taiwan
| | - Bing‐Huang Wu
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchu300044Taiwan
| | - Pin‐Chiao Sun
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchu300044Taiwan
| | - Odvogmed Bayansan
- Institute of Molecular and Cellular BiologyNational Tsing Hua UniversityHsinchu300044Taiwan
| | - Yu‐Ting Chiu
- Institute of Molecular and Cellular BiologyNational Tsing Hua UniversityHsinchu300044Taiwan
| | - Chi‐Hsuan Shih
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchu300044Taiwan
| | - Wen‐Hong Chung
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchu300044Taiwan
| | - Jia‐Bin Yang
- Institute of Molecular and Cellular BiologyNational Taiwan UniversityTaipei106319Taiwan
| | - Lily Hui‐Ching Wang
- Institute of Molecular and Cellular BiologyNational Tsing Hua UniversityHsinchu300044Taiwan
- School of MedicineNational Tsing Hua UniversityHsinChu300044Taiwan
- Department of Medical ScienceNational Tsing Hua UniversityHsinchu300044Taiwan
| | - Po‐Han Chiang
- Institute of Biomedical EngineeringNational Yang Ming Chiao Tung UniversityHsinchu300093Taiwan
| | - Chun‐Hao Chen
- Institute of Molecular and Cellular BiologyNational Taiwan UniversityTaipei106319Taiwan
| | - Oliver I. Wagner
- Institute of Molecular and Cellular BiologyNational Tsing Hua UniversityHsinchu300044Taiwan
| | - Yi‐Ching Wang
- Department of PharmacologyCollege of MedicineNational Cheng Kung UniversityTainan701401Taiwan
| | - Yu‐Chun Lin
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchu300044Taiwan
- Department of Medical ScienceNational Tsing Hua UniversityHsinchu300044Taiwan
| |
Collapse
|
18
|
Wollman J, Wanniarachchi K, Pradhan B, Huang L, Kerkvliet JG, Hoppe AD, Thiex NW. Mannose receptor (MRC1) mediates uptake of dextran by bone marrow-derived macrophages. Mol Biol Cell 2024; 35:ar153. [PMID: 39504444 PMCID: PMC11656472 DOI: 10.1091/mbc.e24-08-0355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/29/2024] [Accepted: 11/01/2024] [Indexed: 11/08/2024] Open
Abstract
Macrophages survey their environment using receptor-mediated endocytosis and pinocytosis. Receptor-mediated endocytosis allows internalization of specific ligands, whereas pinocytosis nonselectively internalizes extracellular fluids and solutes. CRISPR/Cas9 whole-genome screens were used to identify genes regulating constitutive and growth factor-stimulated dextran uptake in murine bone marrow-derived macrophages (BMDM). The mannose receptor c-type 1 (MRC1/CD206) was a top hit in the screen. Targeted gene disruptions of Mrc1 reduced dextran uptake but had little effect on fluid-phase uptake of Lucifer yellow. Other screen hits also differentially affected the uptake of dextran and Lucifer yellow, indicating internalization by separate mechanisms. Visualization of dextran and Lucifer yellow uptake by microscopy showed enrichment of dextran in small puncta, which was inhibitable by mannan, a ligand of MRC1. In contrast, Lucifer yellow predominantly was internalized in larger macropinosomes. In addition, IL4-treated BMDMs internalized more dextran than untreated BMDM correlating with increased MRC1 expression. Therefore, dextran is not an effective marker for pinocytosis in BMDMs since it is internalized by receptor-mediated process. Numerous genes that regulate dextran internalization in primary murine macrophages were identified in the whole-genome screens, which can inform understanding of the regulation of MRC1 expression and MRC1-mediated uptake in macrophages.
Collapse
Affiliation(s)
- Jared Wollman
- Biology and Microbiology Department, South Dakota State University, Brookings, SD 57007
| | - Kevin Wanniarachchi
- Biology and Microbiology Department, South Dakota State University, Brookings, SD 57007
| | - Bijaya Pradhan
- Biology and Microbiology Department, South Dakota State University, Brookings, SD 57007
| | - Lu Huang
- Biology and Microbiology Department, South Dakota State University, Brookings, SD 57007
| | - Jason G Kerkvliet
- Biology and Microbiology Department, South Dakota State University, Brookings, SD 57007
- Chemistry, Biochemistry and Physics Department, South Dakota State University, Brookings, SD 57007
| | - Adam D Hoppe
- Chemistry, Biochemistry and Physics Department, South Dakota State University, Brookings, SD 57007
| | - Natalie W Thiex
- Biology and Microbiology Department, South Dakota State University, Brookings, SD 57007
| |
Collapse
|
19
|
Al Nasr IS, Koko WS, Khan TA, Schobert R, Biersack B. Antiparasitic Activities of Acyl Hydrazones from Cinnamaldehydes and Structurally Related Fragrances. Antibiotics (Basel) 2024; 13:1114. [PMID: 39766505 PMCID: PMC11672724 DOI: 10.3390/antibiotics13121114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/16/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025] Open
Abstract
Background: New drugs for the treatment of protozoal parasite infections such as toxoplasmosis and leishmaniasis are required. Cinnamaldehyde and its derivatives appear to be promising antiparasitic drug candidates. Methods: Acyl hydrazones of cinnamaldehyde, 4-dimethylaminocinnamaldehyde, and of the synthetic fragrances silvialTM and florhydralTM were prepared and tested for activity against Toxoplasma gondii (T. gondii) and Leishmania major (L. major) parasites. Results: Three cinnamaldehyde acyl hydrazones (3-hydroxy-2-naphthoyl 2a and the salicyloyls 2c and 2d) showed good activity against T. gondii, and two compounds derived from cinnamaldehyde and florhydralTM (3-hydroxy-2-naphthoyls 2a and 4a) exhibited moderate activity against L. major promastigotes. Conclusions: In particular, the identified antitoxoplasmal activities are promising and might lead to the development of new potent and cost-effective drug candidates for the therapy of toxoplasmosis.
Collapse
Affiliation(s)
- Ibrahim S. Al Nasr
- Department of Biology, College of Science, Qassim University, Qassim 51452, Saudi Arabia; (I.S.A.N.); (W.S.K.)
| | - Waleed S. Koko
- Department of Biology, College of Science, Qassim University, Qassim 51452, Saudi Arabia; (I.S.A.N.); (W.S.K.)
| | - Tariq A. Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Qassim 51452, Saudi Arabia;
| | - Rainer Schobert
- Organic Chemistry Laboratory, University Bayreuth, Universitätsstrasse 30, 95440 Bayreuth, Germany;
| | - Bernhard Biersack
- Organic Chemistry Laboratory, University Bayreuth, Universitätsstrasse 30, 95440 Bayreuth, Germany;
| |
Collapse
|
20
|
Thomas BJ, Awan SZ, Joshi T, Daniels MA, Porciani D, Burke DH. Anti-EGFR aptamer exhibits direct anti-cancer effects in NSCLC cells harboring EGFR L858R mutations. NPJ Precis Oncol 2024; 8:271. [PMID: 39572699 PMCID: PMC11582725 DOI: 10.1038/s41698-024-00758-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 11/08/2024] [Indexed: 11/24/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) adenocarcinoma (LUAD) is a leading cause of death worldwide. Activating mutations in the tyrosine kinase domain of the oncogene epidermal growth factor receptor (EGFR) are responsible for ~10-50% of all LUAD cases. Although tyrosine kinase inhibitors (TKIs) have been effective in prolonging patient survival and quality of life, acquired resistance and disease progression are inevitable, presenting a clear unmet need for alternative or adjuvant therapeutics. Here we show that an anti-EGFR aptamer (EGFRapt) decreases viability and tumor growth of LUAD cell lines harboring the L858R ± T790M mutation in EGFR. Additionally, we elucidate the mechanism by which EGFRapt exerts these effects by monitoring cellular processes associated with kinase-dependent and kinase-independent mechanisms. Overall, these data establish that EGFRapt has direct anti-cancer activity in mutant EGFR positive LUAD via targetable mechanisms that are independent of existing approaches, and they provide a foundation for further development of nucleic acid-based therapies that target EGFR.
Collapse
Affiliation(s)
- Brian J Thomas
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO, USA
- Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Sania Z Awan
- MU Institute for Data Science and Informatics, University of Missouri, Columbia, MO, USA
| | - Trupti Joshi
- Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
- MU Institute for Data Science and Informatics, University of Missouri, Columbia, MO, USA
- Department of Biomedical Informatics, Biostatistics and Medical Epidemiology (BBME), University of Missouri School of Medicine, Columbia, MO, USA
| | - Mark A Daniels
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO, USA
- NextGen Precision Health, University of Missouri, Columbia, MO, USA
| | - David Porciani
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO, USA.
- Bond Life Sciences Center, University of Missouri, Columbia, MO, USA.
- Standard BioTools Inc./SomaLogic Inc., Boulder, CO, USA.
| | - Donald H Burke
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO, USA.
- Bond Life Sciences Center, University of Missouri, Columbia, MO, USA.
- Department of Biochemistry, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
21
|
Johnson D, Colijn S, Richee J, Yano J, Burns M, Davis AE, Pham VN, Saric A, Jain A, Yin Y, Castranova D, Melani M, Fujita M, Grainger S, Bonifacino JS, Weinstein BM, Stratman AN. Angiogenesis is limited by LIC1-mediated lysosomal trafficking. Angiogenesis 2024; 27:943-962. [PMID: 39356418 PMCID: PMC11653708 DOI: 10.1007/s10456-024-09951-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 09/25/2024] [Indexed: 10/03/2024]
Abstract
Dynein cytoplasmic 1 light intermediate chain 1 (LIC1, DYNC1LI1) is a core subunit of the dynein motor complex. The LIC1 subunit also interacts with various cargo adaptors to regulate Rab-mediated endosomal recycling and lysosomal degradation. Defects in this gene are predicted to alter dynein motor function, Rab binding capabilities, and cytoplasmic cargo trafficking. Here, we have identified a dync1li1 zebrafish mutant, harboring a premature stop codon at the exon 12/13 splice acceptor site, that displays increased angiogenesis. In vitro, LIC1-deficient human endothelial cells display increases in cell surface levels of the pro-angiogenic receptor VEGFR2, SRC phosphorylation, and Rab11-mediated endosomal recycling. In vivo, endothelial-specific expression of constitutively active Rab11a leads to excessive angiogenesis, similar to the dync1li1 mutants. Increased angiogenesis is also evident in zebrafish harboring mutations in rilpl1/2, the adaptor proteins that promote Rab docking to Lic1 to mediate lysosomal targeting. These findings suggest that LIC1 and the Rab-adaptor proteins RILPL1 and 2 restrict angiogenesis by promoting degradation of VEGFR2-containing recycling endosomes. Disruption of LIC1- and RILPL1/2-mediated lysosomal targeting increases Rab11-mediated recycling endosome activity, promoting excessive SRC signaling and angiogenesis.
Collapse
Affiliation(s)
- Dymonn Johnson
- Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO, 63110, USA
| | - Sarah Colijn
- Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO, 63110, USA
| | - Jahmiera Richee
- Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO, 63110, USA
| | - Joseph Yano
- Division of Developmental Biology, Section on Vertebrate Organogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
- Cell and Molecular Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Margaret Burns
- Division of Developmental Biology, Section on Vertebrate Organogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Andrew E Davis
- Division of Developmental Biology, Section on Vertebrate Organogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Van N Pham
- Division of Developmental Biology, Section on Vertebrate Organogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Amra Saric
- Section On Intracellular Protein Trafficking, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
- Neurosciences and Mental Health Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
| | - Akansha Jain
- Section On Intracellular Protein Trafficking, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ying Yin
- Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO, 63110, USA
| | - Daniel Castranova
- Division of Developmental Biology, Section on Vertebrate Organogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Mariana Melani
- Division of Developmental Biology, Section on Vertebrate Organogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
- Fundación Instituto Leloir, Buenos Aires, Argentina
- Consejo Nacional De Investigaciones Científicas Y Técnicas (CONICET), Buenos Aires, Argentina
- Departamento De Fisiología, Biología Molecular Y Celular, Facultad De Ciencias Exactas Y Naturales, Universidad De Buenos Aires, Buenos Aires, Argentina
| | - Misato Fujita
- Division of Developmental Biology, Section on Vertebrate Organogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
- Kanagawa University, Kanagawa, 221-8686, Japan
| | - Stephanie Grainger
- Department of Cell Biology, Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - Juan S Bonifacino
- Section On Intracellular Protein Trafficking, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Brant M Weinstein
- Division of Developmental Biology, Section on Vertebrate Organogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Amber N Stratman
- Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
22
|
Wang S, Cheng P, Guo K, Ren S, Tadele BA, Liang Z, Sun Y, Yin X, Wang X. Lumpy skin disease virus enters into host cells via dynamin-mediated endocytosis and macropinocytosis. Vet Microbiol 2024; 298:110254. [PMID: 39307114 DOI: 10.1016/j.vetmic.2024.110254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/13/2024] [Accepted: 09/14/2024] [Indexed: 11/19/2024]
Abstract
Lumpy skin disease virus (LSDV), a ruminant poxvirus of the Capripoxvirus genus, is the etiologic agent of an economically important cattle disease categorized as a notifiable disease by the World Organization for Animal Health. However, the endocytic pathway and their regulatory molecules have not been characterized for LSDV. In the present study, specific pharmacological inhibitors were used to analyze the mechanism of LSDV entry into Mardin-Darby Bovine Kidney cell (MDBK) and bovine mammary epithelial cell (BMEC). The results showed that LSDV entered MDBK and BMEC cells depends on low-pH conditions and dynamin. However, the inhibitor of caveolae- and clathrin-mediated endocytosis cann't inhibit LSDV entry into MDBK and BMEC cells. Furthermore, treatment with specific inhibitors demonstrated that LSDV entry into MDBK and BMEC cells via macropinocytosis depended on the Na1/H1 exchanger (NHE) but not phosphatidylinositol 3-kinase (PI3K). In addition, results demonstrated that these inhibitors inhibited LSDV entry but did not have effect on LSDV binding. Taken together, our study demonstrated that LSDV enters MDBK and BMEC cells through macropinocytosis pathway in a low-PH- and dynamin-dependent manner while independent on PI3K. Results presented in this study potentially provides insight into the entry mechanisms of LSDV, and it may facilitate the development of therapeutic interventions.
Collapse
Affiliation(s)
- Shasha Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Pengyuan Cheng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Ke Guo
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Shanhui Ren
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Berihun Afera Tadele
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China; Mekelle University College of Veterinary Sciences, Mekelle, Tigray, Ethiopia
| | - Zhengji Liang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yuefeng Sun
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xiangping Yin
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xiangwei Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.
| |
Collapse
|
23
|
Pavlova S, Fab L, Dzarieva F, Ryabova A, Revishchin A, Panteleev D, Antipova O, Usachev D, Kopylov A, Pavlova G. Unite and Conquer: Association of Two G-Quadruplex Aptamers Provides Antiproliferative and Antimigration Activity for Cells from High-Grade Glioma Patients. Pharmaceuticals (Basel) 2024; 17:1435. [PMID: 39598347 PMCID: PMC11597096 DOI: 10.3390/ph17111435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024] Open
Abstract
Background: High-grade gliomas remain a virtually incurable form of brain cancer. Current therapies are unable to completely eradicate the tumor, and the tumor cells that survive chemotherapy or radiation therapy often become more aggressive and resistant to further treatment, leading to inevitable relapses. While the antiproliferative effects of new therapeutic molecules are typically the primary focus of research, less attention is given to their influence on tumor cell migratory activity, which can play a significant role in recurrence. A potential solution may lie in the synergistic effects of multiple drugs on the tumor. Objectives: In this study, we investigated the effect of combined exposure to bi-(AID-1-T), an anti-proliferative aptamer, and its analog bi-(AID-1-C), on the migratory activity of human GBM cells. Results: We examined the effects of various sequences of adding bi-(AID-1-T) and bi-(AID-1-C) on five human GBM cell cultures. Our findings indicate that certain sequences significantly reduced the ability of tumor cells to migrate and proliferate. Additionally, the expression of Nestin, PARP1, L1CAM, Caveolin-1, and c-Myc was downregulated in human GBM cells that survived exposure, suggesting that the treatment had a persistent antitumor effect on these cells.
Collapse
Affiliation(s)
- Svetlana Pavlova
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 117485 Moscow, Russia
- Institution N. N. Burdenko National Medical Research Center of Neurosurgery of the Ministry of Health of the Russian Federation, 125047 Moscow, Russia
| | - Lika Fab
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 117485 Moscow, Russia
| | - Fatima Dzarieva
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 117485 Moscow, Russia
- Institution N. N. Burdenko National Medical Research Center of Neurosurgery of the Ministry of Health of the Russian Federation, 125047 Moscow, Russia
| | - Anastasia Ryabova
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 119991 Moscow, Russia
| | - Alexander Revishchin
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 117485 Moscow, Russia
| | - Dmitriy Panteleev
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 117485 Moscow, Russia
| | - Olga Antipova
- Institution N. N. Burdenko National Medical Research Center of Neurosurgery of the Ministry of Health of the Russian Federation, 125047 Moscow, Russia
- Belozersky Research Institute of Physical Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Dmitry Usachev
- Institution N. N. Burdenko National Medical Research Center of Neurosurgery of the Ministry of Health of the Russian Federation, 125047 Moscow, Russia
| | - Alexey Kopylov
- Institution N. N. Burdenko National Medical Research Center of Neurosurgery of the Ministry of Health of the Russian Federation, 125047 Moscow, Russia
- Belozersky Research Institute of Physical Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Galina Pavlova
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 117485 Moscow, Russia
- Institution N. N. Burdenko National Medical Research Center of Neurosurgery of the Ministry of Health of the Russian Federation, 125047 Moscow, Russia
| |
Collapse
|
24
|
Bjørnestad SA, Solbakken MH, Krokene P, Thiede B, Hylland K, Jakobsen KS, Jentoft S, Bakke O, Progida C. The Atlantic Cod MHC I compartment has the properties needed for cross-presentation in the absence of MHC II. Sci Rep 2024; 14:25404. [PMID: 39455705 PMCID: PMC11511864 DOI: 10.1038/s41598-024-76225-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Atlantic cod has a peculiar immune system, characterized by the loss of Major Histocompatibility Complex (MHC) class II pathway, and an extreme expansion of the MHC class I gene repertoire. This has led to the hypothesis that some of the MHC I variants have replaced MHC II by presenting exogenous-peptides in a process similar to cross-presentation. In mammals, MHC I loads endogenous antigens in the endoplasmic reticulum, but we recently found that different Atlantic cod MHC I gene variants traffic to endolysosomes. There, they colocalize with Tapasin and other components of the peptide-loading complex, indicating a plausible peptide-loading system outside the endoplasmic reticulum. In this study, we further characterize the identity of the Atlantic cod MHC I compartment (cMIC). We found that, similarly to mammalian MHC II compartment, cMIC contains late endosomal markers such as Rab7, LAMP1 and CD63. Furthermore, we identified Hsp90b1 (also known as grp94) and LRP1 (also known as CD91) as interactors of MHC I by mass spectrometry. As these two proteins are involved in cross-presentation in mammals, this further suggests that Atlantic cod MHC I might use a similar mechanism to present exogenous peptides, thus, compensating for the absence of MHC II.
Collapse
Affiliation(s)
| | - Monica Hongrø Solbakken
- Department of Biosciences, University of Oslo, Oslo, Norway
- Norwegian University of Life Sciences, Ås, Norway
| | - Pia Krokene
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Bernd Thiede
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Ketil Hylland
- Department of Biosciences, University of Oslo, Oslo, Norway
| | | | - Sissel Jentoft
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Oddmund Bakke
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Cinzia Progida
- Department of Biosciences, University of Oslo, Oslo, Norway.
| |
Collapse
|
25
|
Martínez-Carrasco R, Fini ME. Dynasore modulates store-operated calcium entry and mitochondrial calcium release in corneal epithelial cells. Exp Eye Res 2024; 247:110029. [PMID: 39127237 PMCID: PMC11413707 DOI: 10.1016/j.exer.2024.110029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/15/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024]
Abstract
Dysregulation of calcium homeostasis can precipitate a cascade of pathological events that lead to tissue damage and cell death. Dynasore is a small molecule that inhibits endocytosis by targeting classic dynamins. In a previous study, we showed that dynasore can protect human corneal epithelial cells from damage due to tert-butyl hydroperoxide (tBHP) exposure by restoring cellular calcium (Ca2+) homeostasis. Here we report results of a follow-up study aimed at identifying the source of the damaging Ca2+. Store-operated Ca2+ entry (SOCE) is a cellular mechanism to restore intracellular calcium stores from the extracellular milieu. We found that dynasore effectively blocks SOCE in cells treated with thapsigargin (TG), a small molecule that inhibits pumping of Ca2+ into the endoplasmic reticulum (ER). Unlike dynasore however, SOCE inhibitor YM-58483 did not interfere with the cytosolic Ca2+ overload caused by tBHP exposure. We also found that dynasore effectively blocks Ca2+ release from internal sources. The inefficacy of inhibitors of ER Ca2+ channels suggested that this compartment was not the source of the Ca2+ surge caused by tBHP exposure. However, using a Ca2+-measuring organelle-entrapped protein indicator (CEPIA) reporter targeted to mitochondria, we found that dynasore can block mitochondrial Ca2+ release due to tBHP exposure. Our results suggest that dynasore exerts multiple effects on cellular Ca2+ homeostasis, with inhibition of mitochondrial Ca2+ release playing a key role in protection of corneal epithelial cells against oxidative stress due to tBHP exposure.
Collapse
Affiliation(s)
- Rafael Martínez-Carrasco
- New England Eye Center, Tufts Medical Center and Department of Ophthalmology, Tufts University School of Medicine, 800 Washington St, Boston, MA, 02111, USA
| | - M Elizabeth Fini
- New England Eye Center, Tufts Medical Center and Department of Ophthalmology, Tufts University School of Medicine, 800 Washington St, Boston, MA, 02111, USA; Program in Genetics, Molecular & Cellular Biology, Tufts Graduate School of Biomedical Sciences, Tufts University, 136 Harrison Ave, Boston, MA, USA; Program in Pharmacology & Drug Development, Tufts Graduate School of Biomedical Sciences, 136 Harrison Ave, Tufts University, Boston, MA, USA.
| |
Collapse
|
26
|
Gopaldass N, Chen KE, Collins B, Mayer A. Assembly and fission of tubular carriers mediating protein sorting in endosomes. Nat Rev Mol Cell Biol 2024; 25:765-783. [PMID: 38886588 DOI: 10.1038/s41580-024-00746-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2024] [Indexed: 06/20/2024]
Abstract
Endosomes are central protein-sorting stations at the crossroads of numerous membrane trafficking pathways in all eukaryotes. They have a key role in protein homeostasis and cellular signalling and are involved in the pathogenesis of numerous diseases. Endosome-associated protein assemblies or coats collect transmembrane cargo proteins and concentrate them into retrieval domains. These domains can extend into tubular carriers, which then pinch off from the endosomal membrane and deliver the cargoes to appropriate subcellular compartments. Here we discuss novel insights into the structure of a number of tubular membrane coats that mediate the recruitment of cargoes into these carriers, focusing on sorting nexin-based coats such as Retromer, Commander and ESCPE-1. We summarize current and emerging views of how selective tubular endosomal carriers form and detach from endosomes by fission, highlighting structural aspects, conceptual challenges and open questions.
Collapse
Affiliation(s)
- Navin Gopaldass
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland.
| | - Kai-En Chen
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland, Australia
| | - Brett Collins
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland, Australia
| | - Andreas Mayer
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland.
| |
Collapse
|
27
|
Hanafy MS, Sandoval MA, Dao HM, Williams RO, Stachowiak JC, Cui Z. Functional dry powders of connexin-containing extracellular vesicles. Int J Pharm 2024; 663:124576. [PMID: 39134288 DOI: 10.1016/j.ijpharm.2024.124576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 08/21/2024]
Abstract
Extracellular vesicles (EVs) have emerged as a promising drug delivery system. Connectosomes are a specialized type of EVs that contain connexins in their membranes. Connexin is a surface transmembrane protein that forms connexin hemichannels. When a connexin hemichannel on a connectosome docks with another connexin hemichannel of a target cell, they form a gap junction that allows direct intracellular delivery of therapeutic cargos from within the connectosome to the cytoplasm of the recipient cell. In the present study, we tested the feasibility of converting connectosomes into dry powders by (thin-film) freeze-drying to enable their potential storage in temperatures higher than the recommended -80 °C, while maintaining their activity. Connectosomes were isolated from a genetically engineered HeLa cell line that overexpressing connexin-43 subunit protein tagged with red fluorescence protein. To facilitate the testing of the function of the connectosomes, they were loaded with calcein green dye. Calcein green-loaded connectosomes were thin-film freeze-dried with trehalose alone or trehalose and a polyvinylpyrrolidone polymer as lyoprotectant(s) to produce amorphous powders with high glass transition temperatures (>100 °C). Thin-film freeze-drying did not significantly change the morphology and structure of the connectosomes, nor their particle size distribution. Based on data from confocal microscopy, flow cytometry, and fluorescence spectrometry, the connexin hemichannels in the connectosomes reconstituted from the thin-film freeze-dried powder remained functional, allowing the passage of calcein green through the hemichannels and the release of the calcein green from the connectosomes when the channels were opened by chelating calcium in the reconstituted medium. The function of connectosomes was assessed after one month storage at different temperatures. The connexin hemichannels in connectosomes in liquid lost their function when stored at -19.5 ± 2.2 °C or 6.0 ± 0.5 °C for a month, while those in dry powder form remained functional under the same storage conditions. Finally, using doxorubicin-loaded connectosomes, we showed that the connectosomes reconstituted from thin-film freeze-dried powder remained pharmacologically active. These findings demonstrate that (thin-film) freeze-drying represents a viable method to prepare stable and functional powders of EVs that contain connexins in their membranes.
Collapse
Affiliation(s)
- Mahmoud S Hanafy
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, TX, United States
| | - Michael A Sandoval
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, TX, United States
| | - Huy M Dao
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, TX, United States
| | - Robert O Williams
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, TX, United States
| | - Jeanne C Stachowiak
- Department of Biomedical Engineering, Cockrell College of Engineering, The University of Texas at Austin, Austin, TX, United States
| | - Zhengrong Cui
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, TX, United States.
| |
Collapse
|
28
|
Kirby J, Standfest M, Binkley J, Barnes C, Brown E, Cairncross T, Cartwright A, Dadisman D, Mowat C, Wilmot D, Houseman T, Murphy C, Engelsman C, Haller J, Jones D. The dynamin inhibitor, dynasore, prevents zoledronate-induced viability loss in human gingival fibroblasts by partially blocking zoledronate uptake and inhibiting endosomal acidification. J Appl Oral Sci 2024; 32:e20240224. [PMID: 39356951 PMCID: PMC11464084 DOI: 10.1590/1678-7757-2024-0224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/21/2024] [Accepted: 08/13/2024] [Indexed: 10/04/2024] Open
Abstract
OBJECTIVE For treatment of medication-related osteonecrosis of the jaw, one proposed approach is the use of a topical agent to block entry of these medications in oral soft tissues. We tested the ability of phosphonoformic acid (PFA), an inhibitor of bisphosphonate entry through certain sodium-dependent phosphate contransporters (SLC20A1, 20A2, 34A1-3) as well as Dynasore, a macropinocytosis inhibitor, for their abilities to prevent zoledronate-induced (ZOL) death in human gingival fibroblasts (HGFs). METHODOLOGY MTT assay dose-response curves were performed to determine non-cytotoxic levels of both PFA and Dynasore. In the presence of 50 μM ZOL, optimized PFA and Dynasore doses were tested for their ability to restore HGF viability. To determine SLC expression in HGFs, total HGF RNA was subjected to quantitative real-time RT-PCR. Confocal fluorescence microscopy was employed to see if Dynasore inhibited macropinocytotic HGF entry of AF647-ZOL. Endosomal acidification in the presence of Dynasore was measured by live cell imaging utilizing LysoSensor Green DND-189. As a further test of Dynasore's ability to interfere with ZOL-containing endosomal maturation, perinuclear localization of mature endosomes containing AF647-ZOL or TRITC-dextran as a control were assessed via confocal fluorescence microscopy with CellProfiler™ software analysis of the resulting photomicrographs. RESULTS 0.5 mM PFA did not rescue HGFs from ZOL-induced viability loss at 72 hours while 10 and 30 μM geranylgeraniol did partially rescue. HGFs did not express the SLC transporters as compared to the expression in positive control tissues. 10 μM Dynasore completely prevented ZOL-induced viability loss. In the presence of Dynasore, AF647-ZOL and FITC-dextran co-localized in endosomes. Endosomal acidification was inhibited by Dynasore and perinuclear localization of both TRITC-dextran- and AF647-ZOL-containing endosomes was inhibited by 30 μM Dynasore. CONCLUSION Dynasore prevents ZOL-induced viability loss in HGFs by partially interfering with macropinocytosis and by inhibiting the endosomal maturation pathway thought to be needed for ZOL delivery to the cytoplasm.
Collapse
Affiliation(s)
- Jacob Kirby
- Indiana Wesleyan University, Division of Natural Sciences, Indiana, United States
| | - Makayla Standfest
- Indiana Wesleyan University, Division of Natural Sciences, Indiana, United States
| | - Jessica Binkley
- Indiana Wesleyan University, Division of Natural Sciences, Indiana, United States
| | - Charles Barnes
- Indiana Wesleyan University, Division of Natural Sciences, Indiana, United States
| | - Evan Brown
- Indiana Wesleyan University, Division of Natural Sciences, Indiana, United States
| | - Tyler Cairncross
- Indiana Wesleyan University, Division of Natural Sciences, Indiana, United States
| | - Alex Cartwright
- Indiana Wesleyan University, Division of Natural Sciences, Indiana, United States
| | - Danielle Dadisman
- Indiana Wesleyan University, Division of Natural Sciences, Indiana, United States
| | - Colten Mowat
- Indiana Wesleyan University, Division of Natural Sciences, Indiana, United States
| | - Daniel Wilmot
- Indiana Wesleyan University, Division of Natural Sciences, Indiana, United States
| | - Theodore Houseman
- Indiana Wesleyan University, Division of Natural Sciences, Indiana, United States
| | - Conner Murphy
- Indiana Wesleyan University, Division of Natural Sciences, Indiana, United States
| | - Caleb Engelsman
- Indiana Wesleyan University, Division of Natural Sciences, Indiana, United States
| | - Josh Haller
- Indiana Wesleyan University, Division of Natural Sciences, Indiana, United States
| | - Daniel Jones
- Indiana Wesleyan University, Division of Natural Sciences, Indiana, United States
| |
Collapse
|
29
|
Andreu S, Ripa I, López-Guerrero JA, Bello-Morales R. Human Coronavirus 229E Uses Clathrin-Mediated Endocytosis as a Route of Entry in Huh-7 Cells. Biomolecules 2024; 14:1232. [PMID: 39456165 PMCID: PMC11505773 DOI: 10.3390/biom14101232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/17/2024] [Accepted: 09/26/2024] [Indexed: 10/28/2024] Open
Abstract
Human coronavirus 229E (HCoV-229E) is an endemic coronavirus responsible for approximately one-third of "common cold" cases. To infect target cells, HCoV-229E first binds to its receptor on the cell surface and then can follow different pathways, entering by direct fusion or by taking advantage of host cell mechanisms such as endocytosis. Based on the role of clathrin, the process can be classified into clathrin-dependent or -independent endocytosis. This study characterizes the role of clathrin-mediated endocytosis (CME) in HCoV-229E infection of the human hepatoma cell line Huh-7. Using specific CME inhibitory drugs, we demonstrated that blocking CME significantly reduces HCoV-229E infection. Additionally, CRISPR/Cas9-mediated knockout of the µ subunit of adaptor protein complex 2 (AP-2) further corroborated the role of CME, as KOs showed over a 50% reduction in viral infection. AP-2 plays an important role in clathrin recruitment and the maturation of clathrin-coated vesicles. Our study also confirmed that in Huh-7 cells, HCoV-229E requires endosomal acidification for successful entry, as viral entry decreased when treated with lysomotropic agents. Furthermore, the colocalization of HCoV-229E with early endosome antigen 1 (EEA-1), only present in early endosomes, suggested that the virus uses an endosomal route for entry. These findings highlight, for the first time, the role of CME in HCoV-229E infection and confirm previous data of the use of the endosomal route at a low pH in the experimental cell model Huh-7. Our results provide new insights into the mechanisms of entry of HCoV-229E and provide a new basis for the development of targeted antiviral therapies.
Collapse
Affiliation(s)
- Sabina Andreu
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas), 28049 Madrid, Spain
| | - Inés Ripa
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas), 28049 Madrid, Spain
| | - José Antonio López-Guerrero
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas), 28049 Madrid, Spain
| | - Raquel Bello-Morales
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas), 28049 Madrid, Spain
| |
Collapse
|
30
|
Brown DW, Wee P, Bhandari P, Bukhari A, Grin L, Vega H, Hejazi M, Sosnowski D, Ablack J, Clancy EK, Pink D, Kumar J, Solis Ares MP, Lamb S, Quevedo R, Rawal B, Elian F, Rana N, Morales L, Govindasamy N, Todd B, Delmage A, Gupta S, McMullen N, MacKenzie D, Beatty PH, Garcia H, Parmar M, Gyoba J, McAllister C, Scholz M, Duncan R, Raturi A, Lewis JD. Safe and effective in vivo delivery of DNA and RNA using proteolipid vehicles. Cell 2024; 187:5357-5375.e24. [PMID: 39260374 DOI: 10.1016/j.cell.2024.07.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 05/08/2024] [Accepted: 07/12/2024] [Indexed: 09/13/2024]
Abstract
Genetic medicines show promise for treating various diseases, yet clinical success has been limited by tolerability, scalability, and immunogenicity issues of current delivery platforms. To overcome these, we developed a proteolipid vehicle (PLV) by combining features from viral and non-viral approaches. PLVs incorporate fusion-associated small transmembrane (FAST) proteins isolated from fusogenic orthoreoviruses into a well-tolerated lipid formulation, using scalable microfluidic mixing. Screening a FAST protein library, we identified a chimeric FAST protein with enhanced membrane fusion activity that improved gene expression from an optimized lipid formulation. Systemically administered FAST-PLVs showed broad biodistribution and effective mRNA and DNA delivery in mouse and non-human primate models. FAST-PLVs show low immunogenicity and maintain activity upon repeat dosing. Systemic administration of follistatin DNA gene therapy with FAST-PLVs raised circulating follistatin levels and significantly increased muscle mass and grip strength. These results demonstrate the promising potential of FAST-PLVs for redosable gene therapies and genetic medicines.
Collapse
Affiliation(s)
- Douglas W Brown
- Department of Oncology, University of Alberta, Edmonton, AB T6G 2E1, Canada; Entos Pharmaceuticals, 10230 Jasper Avenue, Suite 4550, Edmonton, AB T5J 4P6, Canada
| | - Ping Wee
- Entos Pharmaceuticals, 10230 Jasper Avenue, Suite 4550, Edmonton, AB T5J 4P6, Canada
| | - Prakash Bhandari
- Entos Pharmaceuticals, 10230 Jasper Avenue, Suite 4550, Edmonton, AB T5J 4P6, Canada
| | - Amirali Bukhari
- Department of Oncology, University of Alberta, Edmonton, AB T6G 2E1, Canada; Entos Pharmaceuticals, 10230 Jasper Avenue, Suite 4550, Edmonton, AB T5J 4P6, Canada
| | - Liliya Grin
- Entos Pharmaceuticals, 10230 Jasper Avenue, Suite 4550, Edmonton, AB T5J 4P6, Canada
| | - Hector Vega
- Entos Pharmaceuticals, 10230 Jasper Avenue, Suite 4550, Edmonton, AB T5J 4P6, Canada
| | - Maryam Hejazi
- Department of Oncology, University of Alberta, Edmonton, AB T6G 2E1, Canada; Entos Pharmaceuticals, 10230 Jasper Avenue, Suite 4550, Edmonton, AB T5J 4P6, Canada
| | - Deborah Sosnowski
- Department of Oncology, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Jailal Ablack
- Entos Pharmaceuticals, 10230 Jasper Avenue, Suite 4550, Edmonton, AB T5J 4P6, Canada; OncoSenX, 701 Fifth Avenue, Suite 4200, Seattle, WA 98104, USA
| | - Eileen K Clancy
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Desmond Pink
- Department of Oncology, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Jitendra Kumar
- Entos Pharmaceuticals, 10230 Jasper Avenue, Suite 4550, Edmonton, AB T5J 4P6, Canada
| | | | - Suellen Lamb
- Department of Oncology, University of Alberta, Edmonton, AB T6G 2E1, Canada; Entos Pharmaceuticals, 10230 Jasper Avenue, Suite 4550, Edmonton, AB T5J 4P6, Canada
| | - Rodrigo Quevedo
- Entos Pharmaceuticals, 10230 Jasper Avenue, Suite 4550, Edmonton, AB T5J 4P6, Canada
| | - Bijal Rawal
- Entos Pharmaceuticals, 10230 Jasper Avenue, Suite 4550, Edmonton, AB T5J 4P6, Canada
| | - Fahed Elian
- Entos Pharmaceuticals, 10230 Jasper Avenue, Suite 4550, Edmonton, AB T5J 4P6, Canada
| | - Natasha Rana
- Entos Pharmaceuticals, 10230 Jasper Avenue, Suite 4550, Edmonton, AB T5J 4P6, Canada
| | - Luis Morales
- Entos Pharmaceuticals, 10230 Jasper Avenue, Suite 4550, Edmonton, AB T5J 4P6, Canada
| | - Natasha Govindasamy
- Entos Pharmaceuticals, 10230 Jasper Avenue, Suite 4550, Edmonton, AB T5J 4P6, Canada
| | - Brendan Todd
- Entos Pharmaceuticals, 10230 Jasper Avenue, Suite 4550, Edmonton, AB T5J 4P6, Canada
| | - Angela Delmage
- Entos Pharmaceuticals, 10230 Jasper Avenue, Suite 4550, Edmonton, AB T5J 4P6, Canada
| | - Somnath Gupta
- Entos Pharmaceuticals, 10230 Jasper Avenue, Suite 4550, Edmonton, AB T5J 4P6, Canada
| | - Nichole McMullen
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Duncan MacKenzie
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Perrin H Beatty
- Entos Pharmaceuticals, 10230 Jasper Avenue, Suite 4550, Edmonton, AB T5J 4P6, Canada
| | - Henry Garcia
- Oisin Biotechnologies, 701 Fifth Avenue, Suite 4200, Seattle, WA 98104, USA
| | - Manoj Parmar
- Entos Pharmaceuticals, 10230 Jasper Avenue, Suite 4550, Edmonton, AB T5J 4P6, Canada
| | - Jennifer Gyoba
- Entos Pharmaceuticals, 10230 Jasper Avenue, Suite 4550, Edmonton, AB T5J 4P6, Canada
| | - Chandra McAllister
- Entos Pharmaceuticals, 10230 Jasper Avenue, Suite 4550, Edmonton, AB T5J 4P6, Canada
| | - Matthew Scholz
- Oisin Biotechnologies, 701 Fifth Avenue, Suite 4200, Seattle, WA 98104, USA
| | - Roy Duncan
- Entos Pharmaceuticals, 10230 Jasper Avenue, Suite 4550, Edmonton, AB T5J 4P6, Canada; Department of Microbiology & Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Arun Raturi
- Department of Oncology, University of Alberta, Edmonton, AB T6G 2E1, Canada; Entos Pharmaceuticals, 10230 Jasper Avenue, Suite 4550, Edmonton, AB T5J 4P6, Canada.
| | - John D Lewis
- Department of Oncology, University of Alberta, Edmonton, AB T6G 2E1, Canada; Entos Pharmaceuticals, 10230 Jasper Avenue, Suite 4550, Edmonton, AB T5J 4P6, Canada; OncoSenX, 701 Fifth Avenue, Suite 4200, Seattle, WA 98104, USA; Oisin Biotechnologies, 701 Fifth Avenue, Suite 4200, Seattle, WA 98104, USA.
| |
Collapse
|
31
|
Chan ET, Kural C. Targeting endocytosis to sensitize cancer cells to programmed cell death. Biochem Soc Trans 2024; 52:1703-1713. [PMID: 39092762 PMCID: PMC11519968 DOI: 10.1042/bst20231332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/01/2024] [Accepted: 07/18/2024] [Indexed: 08/04/2024]
Abstract
Evading programmed cell death (PCD) is a hallmark of cancer that allows tumor cells to survive and proliferate unchecked. Endocytosis, the process by which cells internalize extracellular materials, has emerged as a key regulator of cell death pathways in cancer. Many tumor types exhibit dysregulated endocytic dynamics that fuel their metabolic demands, promote resistance to cytotoxic therapies, and facilitate immune evasion. This review examines the roles of endocytosis in apoptotic resistance and immune escape mechanisms utilized by cancer cells. We highlight how inhibiting endocytosis can sensitize malignant cells to therapeutic agents and restore susceptibility to PCD. Strategies to modulate endocytosis for enhanced cancer treatment are discussed, including targeting endocytic regulatory proteins, altering membrane biophysical properties, and inhibiting Rho-associated kinases. While promising, challenges remain regarding the specificity and selectivity of endocytosis-targeting agents. Nonetheless, harnessing endocytic pathways represents an attractive approach to overcome apoptotic resistance and could yield more effective therapies by rendering cancer cells vulnerable to PCD. Understanding the interplay between endocytosis and PCD regulation is crucial for developing novel anticancer strategies that selectively induce tumor cell death.
Collapse
Affiliation(s)
- Emily T. Chan
- Interdisciplinary Biophysics Graduate Program, The Ohio State University, Columbus, OH 43210, U.S.A
| | - Cömert Kural
- Interdisciplinary Biophysics Graduate Program, The Ohio State University, Columbus, OH 43210, U.S.A
- Department of Physics, The Ohio State University, Columbus, OH 43210, U.S.A
| |
Collapse
|
32
|
Sandvig K, Iversen TG, Skotland T. Entry of nanoparticles into cells and tissues: status and challenges. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2024; 15:1017-1029. [PMID: 39161463 PMCID: PMC11331539 DOI: 10.3762/bjnano.15.83] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/23/2024] [Indexed: 08/21/2024]
Abstract
In this article we discuss how nanoparticles (NPs) of different compositions may interact with and be internalized by cells, and the consequences of that for cellular functions. A large number of NPs are made with the intention to improve cancer treatment, the goal being to increase the fraction of injected drug delivered to the tumor and thereby improve the therapeutic effect and decrease side effects. Thus, we discuss how NPs are delivered to tumors and some challenges related to investigations of biodistribution, pharmacokinetics, and excretion. Finally, we discuss requirements for bringing NPs into clinical use and aspects when it comes to usage of complex and slowly degraded or nondegradable NPs.
Collapse
Affiliation(s)
- Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
- Centre for Cancer Cell Reprogramming, University of Oslo, 0379 Oslo, Norway
- Department of Biosciences, University of Oslo, 0316 Oslo, Norway
| | - Tore Geir Iversen
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
- Centre for Cancer Cell Reprogramming, University of Oslo, 0379 Oslo, Norway
| | - Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
- Centre for Cancer Cell Reprogramming, University of Oslo, 0379 Oslo, Norway
| |
Collapse
|
33
|
Lee YA, Shin MH. Dynamin 2-mediated endocytosis of BLT1 is required for IL-8 production in HMC-1 cells induced by Trichomonas vaginalis-derived secretory products. PARASITES, HOSTS AND DISEASES 2024; 62:281-293. [PMID: 39218627 PMCID: PMC11366542 DOI: 10.3347/phd.24049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024]
Abstract
We previously reported that leukotriene B4 (LTB4) contained in Trichomonas vaginalis-derived secretory products (TvSP) play an essential role in interleukin-8 (IL-8) production in human mast cell line (HMC-1 cells) via LTB4 receptor (BLT)-mediated Nuclear Factor-kappa B (NF-кB) activation. Dynamin, a GTPase, has been known to be involved in endocytosis of receptors for signaling of production of cytokine or chemokines. In the present study, we investigated the role of dynamin-mediated BLT1 endocytosis in TvSP-induced IL-8 production. When HMC-1 cells were transfected with BLT1 or BLT2 siRNA, TvSP-induced IL-8 production was significantly inhibited compared with that in cells transfected with control siRNA. In addition, pretreatment of HMC-1 cells with a dynamin inhibitor (Dynasore) reduced IL-8 production induced by TvSP or LTB4. TvSP- or LTB4- induced phosphorylation of NF-кB was also attenuated by pretreatment with Dynasore. After exposing HMC-1 cells to TvSP or LTB4, BLT1 was translocated from the intracellular compartments to the plasma membrane within 30 min. At 60 min after stimulation with TvSP or LTB4, BLT1 remigrated from the cell surface to intracellular areas. Pretreatment of HMC-1 cells with dynamin-2 siRNA blocked internalization of BLT1 induced by TvSP or LTB4. Co-immunoprecipitation experiments revealed that dynamin-2 strongly interacted with BLT1 60 min after stimulation with TvSP or LTB4. These results suggest that T. vaginalis-secreted LTB4 induces IL-8 production in HMC-1 cells via dynamin 2-mediated endocytosis of BLT1 and phosphorylation of NF-кB.
Collapse
Affiliation(s)
- Young Ah Lee
- Department of Tropical Medicine and Institute of Tropical Medicine, Yonsei University College of Medicine, Seoul 03722,
Korea
| | - Myeong Heon Shin
- Department of Tropical Medicine and Institute of Tropical Medicine, Yonsei University College of Medicine, Seoul 03722,
Korea
| |
Collapse
|
34
|
Castellano M, Blanco V, Calzi ML, Costa B, Witwer K, Hill M, Cayota A, Segovia M, Tosar JP. Ribonuclease activity undermines immune sensing of naked extracellular RNA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.23.590771. [PMID: 38712104 PMCID: PMC11071435 DOI: 10.1101/2024.04.23.590771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
The plasma membrane and the membrane of endosomal vesicles are considered physical barriers preventing extracellular RNA uptake. While naked RNA can be spontaneously internalized by certain cells types, functional delivery of naked RNA into the cytosol has been rarely observed. Here we show that extracellular ribonucleases, mainly derived from cell culture supplements, have so far hindered the study of extracellular RNA functionality. In the presence of active ribonuclease inhibitors (RI), naked bacterial RNA is pro-inflammatory when spiked in the media of dendritic cells and macrophages. In murine cells, this response mainly depends on the action of endosomal Toll-like receptors. However, we also show that naked RNA can perform endosomal escape and engage with cytosolic RNA sensors and ribosomes. For example, naked mRNAs encoding reporter proteins can be spontaneously internalized and translated by a variety of cell types, in an RI-dependent manner. In vivo, RI co-injection enhances the activation induced by naked extracellular RNA on splenic lymphocytes and myeloid-derived leukocytes. Furthermore, naked extracellular RNA is inherently pro-inflammatory in ribonuclease-poor compartments such as the peritoneal cavity. Overall, these results demonstrate that naked RNA is bioactive and does not need encapsulation inside synthetic or biological lipid vesicles for functional uptake, making a case for nonvesicular extracellular RNA-mediated intercellular communication.
Collapse
Affiliation(s)
- Mauricio Castellano
- Functional Genomics Laboratory, Institut Pasteur Montevideo, Uruguay
- Immunoregulation and Inflammation Laboratory, Institut Pasteur Montevideo, Uruguay
| | - Valentina Blanco
- Functional Genomics Laboratory, Institut Pasteur Montevideo, Uruguay
| | - Marco Li Calzi
- Functional Genomics Laboratory, Institut Pasteur Montevideo, Uruguay
| | - Bruno Costa
- Functional Genomics Laboratory, Institut Pasteur Montevideo, Uruguay
- Analytical Biochemistry Unit, School of Science, Universidad de la República, Uruguay
| | - Kenneth Witwer
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- EV Core Facility “EXCEL”, Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Richman Family Precision Medicine Center of Excellence in Alzheimer’s Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Marcelo Hill
- Immunoregulation and Inflammation Laboratory, Institut Pasteur Montevideo, Uruguay
- Academic Unit of Immunobiology, School of Medicine, Universidad de la República, Uruguay
| | - Alfonso Cayota
- Functional Genomics Laboratory, Institut Pasteur Montevideo, Uruguay
- Hospital de Clínicas, Universidad de la República, Uruguay
| | - Mercedes Segovia
- Immunoregulation and Inflammation Laboratory, Institut Pasteur Montevideo, Uruguay
- Academic Unit of Immunobiology, School of Medicine, Universidad de la República, Uruguay
| | - Juan Pablo Tosar
- Functional Genomics Laboratory, Institut Pasteur Montevideo, Uruguay
- Analytical Biochemistry Unit, School of Science, Universidad de la República, Uruguay
| |
Collapse
|
35
|
Johnson D, Colijn S, Richee J, Yano J, Burns M, Davis AE, Pham VN, Saric A, Jain A, Yin Y, Castranova D, Melani M, Fujita M, Grainger S, Bonifacino JS, Weinstein BM, Stratman AN. Regulation of angiogenesis by endocytic trafficking mediated by cytoplasmic dynein 1 light intermediate chain 1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.01.587559. [PMID: 38903077 PMCID: PMC11188074 DOI: 10.1101/2024.04.01.587559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
Dynein cytoplasmic 1 light intermediate chain 1 (LIC1, DYNC1LI1) is a core subunit of the dynein motor complex. The LIC1 subunit also interacts with various cargo adaptors to regulate Rab-mediated endosomal recycling and lysosomal degradation. Defects in this gene are predicted to alter dynein motor function, Rab binding capabilities, and cytoplasmic cargo trafficking. Here, we have identified a dync1li1 zebrafish mutant, harboring a premature stop codon at the exon 12/13 splice acceptor site, that displays increased angiogenesis. In vitro, LIC1-deficient human endothelial cells display increases in cell surface levels of the pro-angiogenic receptor VEGFR2, SRC phosphorylation, and Rab11-mediated endosomal recycling. In vivo, endothelial-specific expression of constitutively active Rab11a leads to excessive angiogenesis, similar to the dync1li1 mutants. Increased angiogenesis is also evident in zebrafish harboring mutations in rilpl1/2, the adaptor proteins that promote Rab docking to Lic1 to mediate lysosomal targeting. These findings suggest that LIC1 and the Rab-adaptor proteins RILPL1 and 2 restrict angiogenesis by promoting degradation of VEGFR2-containing recycling endosomes. Disruption of LIC1- and RILPL1/2-mediated lysosomal targeting increases Rab11-mediated recycling endosome activity, promoting excessive SRC signaling and angiogenesis.
Collapse
Affiliation(s)
- Dymonn Johnson
- Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110
| | - Sarah Colijn
- Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110
| | - Jahmiera Richee
- Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110
| | - Joseph Yano
- Section on Vertebrate Organogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892
- Cell and Molecular Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Margaret Burns
- Section on Vertebrate Organogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892
| | - Andrew E. Davis
- Section on Vertebrate Organogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892
| | - Van N. Pham
- Section on Vertebrate Organogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892
| | - Amra Saric
- Section on Intracellular Protein Trafficking, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892
- Neurosciences and Mental Health Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
| | - Akansha Jain
- Section on Intracellular Protein Trafficking, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892
| | - Ying Yin
- Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110
| | - Daniel Castranova
- Section on Vertebrate Organogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892
| | - Mariana Melani
- Section on Vertebrate Organogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892
- Fundación Instituto Leloir, Buenos Aires, Argentina
- Consejo Nacional De Investigaciones Científicas Y Técnicas (CONICET), Buenos Aires, Argentina
- Departamento De Fisiología, Biología Molecular Y Celular, Facultad De Ciencias Exactas Y Naturales, Universidad De Buenos Aires, Buenos Aires, Argentina
| | - Misato Fujita
- Section on Vertebrate Organogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892
- Kanagawa University, Kanagawa, 221-8686, Japan
| | - Stephanie Grainger
- Department of Cell Biology, Van Andel Institute, Grand Rapids, MI, 49503
| | - Juan S. Bonifacino
- Section on Intracellular Protein Trafficking, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892
| | - Brant M. Weinstein
- Section on Vertebrate Organogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892
| | - Amber N. Stratman
- Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110
| |
Collapse
|
36
|
Sakurai C, Yamashita N, Azuma K, Hatsuzawa K. VAMP5 promotes Fcγ receptor-mediated phagocytosis and regulates phagosome maturation in macrophages. Mol Biol Cell 2024; 35:ar44. [PMID: 38265888 PMCID: PMC10916865 DOI: 10.1091/mbc.e23-04-0149] [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: 05/01/2023] [Revised: 01/12/2024] [Accepted: 01/18/2024] [Indexed: 01/26/2024] Open
Abstract
Phagosome formation and maturation reportedly occur via sequential membrane fusion events mediated by synaptosomal-associated protein of 23 kDa (SNAP23), a plasma membrane-localized soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) family. Vesicle-associated membrane protein 5 (VAMP5), also a plasmalemma SNARE, interacts with SNAP23; however, its precise function in phagocytosis in macrophages remains elusive. To elucidate this aspect, we investigated the characteristics of macrophages in the presence of VAMP5 overexpression or knockdown and found that VAMP5 participates in Fcγ receptor-mediated phagosome formation, although not directly in phagosome maturation. Overexpressed VAMP5 was localized to the early phagosomal membrane but no longer localized to the lysosomal-associated membrane protein 1-positive maturing phagosomal membrane. Analyses using compound-based selective inhibitors demonstrated that VAMP5 dissociation from early phagosomes occurs in a clathrin- and dynamin-dependent manner and is indispensable for SNAP23 function in subsequent membrane fusion during phagosome maturation. Accordingly, to the best of our knowledge, we demonstrate, for the first time, that VAMP5 exerts an immunologically critical function during phagosome formation and maturation via SNARE-based membrane trafficking in macrophages.
Collapse
Affiliation(s)
- Chiye Sakurai
- Division of Molecular Biology, School of Life Sciences, Faculty of Medicine, Tottori University, Yonago, Tottori 683-8503, Japan
| | - Natsumi Yamashita
- Division of Molecular Biology, School of Life Sciences, Faculty of Medicine, Tottori University, Yonago, Tottori 683-8503, Japan
| | - Kento Azuma
- Division of Molecular Biology, School of Life Sciences, Faculty of Medicine, Tottori University, Yonago, Tottori 683-8503, Japan
| | - Kiyotaka Hatsuzawa
- Division of Molecular Biology, School of Life Sciences, Faculty of Medicine, Tottori University, Yonago, Tottori 683-8503, Japan
| |
Collapse
|
37
|
Cheng YW, Anzell AR, Morosky SA, Schwartze TA, Hinck CS, Hinck AP, Roman BL, Davidson LA. Shear Stress and Sub-Femtomolar Levels of Ligand Synergize to Activate ALK1 Signaling in Endothelial Cells. Cells 2024; 13:285. [PMID: 38334677 PMCID: PMC10854672 DOI: 10.3390/cells13030285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/17/2024] [Accepted: 01/26/2024] [Indexed: 02/10/2024] Open
Abstract
Endothelial cells (ECs) respond to concurrent stimulation by biochemical factors and wall shear stress (SS) exerted by blood flow. Disruptions in flow-induced responses can result in remodeling issues and cardiovascular diseases, but the detailed mechanisms linking flow-mechanical cues and biochemical signaling remain unclear. Activin receptor-like kinase 1 (ALK1) integrates SS and ALK1-ligand cues in ECs; ALK1 mutations cause hereditary hemorrhagic telangiectasia (HHT), marked by arteriovenous malformation (AVM) development. However, the mechanistic underpinnings of ALK1 signaling modulation by fluid flow and the link to AVMs remain uncertain. We recorded EC responses under varying SS magnitudes and ALK1 ligand concentrations by assaying pSMAD1/5/9 nuclear localization using a custom multi-SS microfluidic device and a custom image analysis pipeline. We extended the previously reported synergy between SS and BMP9 to include BMP10 and BMP9/10. Moreover, we demonstrated that this synergy is effective even at extremely low SS magnitudes (0.4 dyn/cm2) and ALK1 ligand range (femtogram/mL). The synergistic response to ALK1 ligands and SS requires the kinase activity of ALK1. Moreover, ALK1's basal activity and response to minimal ligand levels depend on endocytosis, distinct from cell-cell junctions, cytoskeleton-mediated mechanosensing, or cholesterol-enriched microdomains. However, an in-depth analysis of ALK1 receptor trafficking's molecular mechanisms requires further investigation.
Collapse
Affiliation(s)
- Ya-Wen Cheng
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA;
| | - Anthony R. Anzell
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Stefanie A. Morosky
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Tristin A. Schwartze
- Department of Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Cynthia S. Hinck
- Department of Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Andrew P. Hinck
- Department of Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Beth L. Roman
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Lance A. Davidson
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA;
- Department of Developmental Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
38
|
Andreu S, Agúndez C, Ripa I, López-Guerrero JA, Bello-Morales R. Pseudorabies virus uses clathrin mediated endocytosis to enter PK15 swine cell line. Front Microbiol 2024; 15:1332175. [PMID: 38374920 PMCID: PMC10876092 DOI: 10.3389/fmicb.2024.1332175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 01/15/2024] [Indexed: 02/21/2024] Open
Abstract
Pseudorabies virus (PRV), a herpesvirus responsible for Aujeszky's disease, causes high mortality in swine populations. To develop effective and novel antiviral strategies, it is essential to understand the mechanism of entry used by PRV to infect its host. Viruses have different ways of entering host cells. Among others, they can use endocytosis, a fundamental cellular process by which substances from the external environment are internalized into the cell. This process is classified into clathrin-mediated endocytosis (CME) and clathrin-independent endocytosis (CIE), depending on the role of clathrin. Although the involvement of cholesterol-rich lipid rafts in the entry of PRV has already been described, the importance of other endocytic pathways involving clathrin remains unexplored to date. Here, we characterize the role of CME in PRV entry into the PK15 swine cell line. By using CME inhibitory drugs, we report a decrease in PRV infection when the CME pathway is blocked. We also perform the shRNA knockdown of the μ-subunit of the adaptor protein AP-2 (AP2M1), which plays an important role in the maturation of clathrin-coated vesicles, and the infection is greatly reduced when this subunit is knocked down. Furthermore, transmission electron microscopy images report PRV virions inside clathrin-coated vesicles. Overall, this study suggests for the first time that CME is a mechanism used by PRV to enter PK15 cells and provides valuable insights into its possible routes of entry.
Collapse
Affiliation(s)
- Sabina Andreu
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas), Madrid, Spain
| | - Carmen Agúndez
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain
| | - Inés Ripa
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas), Madrid, Spain
| | - José Antonio López-Guerrero
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas), Madrid, Spain
| | - Raquel Bello-Morales
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas), Madrid, Spain
| |
Collapse
|
39
|
Majer J, Kindermann M, Pinkas D, Chvatil D, Cigler P, Libusova L. Cellular uptake and fate of cationic polymer-coated nanodiamonds delivering siRNA: a mechanistic study. NANOSCALE 2024; 16:2490-2503. [PMID: 38197438 DOI: 10.1039/d3nr05738k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
Gene silencing using small interfering RNAs (siRNAs) is a selective and promising approach for treatment of numerous diseases. However, broad applications of siRNAs are compromised by their low stability in a biological environment and limited ability to penetrate cells. Nanodiamonds (NDs) coated with cationic polymers can enable cellular delivery of siRNAs. Recently, we developed a new type of ND coating based on a random copolymer consisting of (2-dimethylaminoethyl) methacrylate (DMAEMA) and N-(2-hydroxypropyl) methacrylamide (HPMA) monomers. These hybrid ND-polymer particles (Cop+-FND) provide near-infrared fluorescence, form stable complexes with siRNA in serum, show low toxicity, and effectively deliver siRNA into cells in vitro and in vivo. Here, we present data on the mechanism of cellular uptake and cell trafficking of Cop+-FND : siRNA complexes and their ability to selectively suppress mRNA levels, as well as their cytotoxicity, viability and colloidal stability. We identified clathrin-mediated endocytosis as the predominant entry mechanism for Cop+-FND : siRNA into U-2 OS human bone osteosarcoma cells, with a substantial fraction of Cop+-FND : siRNA following the lysosome pathway. Cop+-FND : siRNA potently inhibited the target GAPDH gene with negligible toxicity and sufficient colloidal stability. Based on our results, we suggest that Cop+-FND : siRNA can serve as a suitable in vivo delivery system for siRNA.
Collapse
Affiliation(s)
- Jan Majer
- Department of Cell Biology, Faculty of Science, Charles University, Vinicna 7, Prague 2, 128 00, Czechia.
- Institute of Organic Chemistry and Biochemistry of the CAS, Flemingovo namesti 2, 166 10 Prague 6, Czechia.
| | - Marek Kindermann
- Institute of Organic Chemistry and Biochemistry of the CAS, Flemingovo namesti 2, 166 10 Prague 6, Czechia.
- Department of Physical Chemistry, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague 6, Czechia
| | - Dominik Pinkas
- Electron Microscopy Core Facility of the Microscopy Centre, Institute of Molecular Genetics of the CAS, Videnska 1083, 142 20 Prague 4, Czechia
| | - David Chvatil
- Nuclear Physics Institute of the CAS, 250 68 Husinec-Rez 130, Czechia
| | - Petr Cigler
- Institute of Organic Chemistry and Biochemistry of the CAS, Flemingovo namesti 2, 166 10 Prague 6, Czechia.
| | - Lenka Libusova
- Department of Cell Biology, Faculty of Science, Charles University, Vinicna 7, Prague 2, 128 00, Czechia.
| |
Collapse
|
40
|
Markou A, Kitchen P, Aldabbagh A, Repici M, Salman MM, Bill RM, Balklava Z. Mechanisms of aquaporin-4 vesicular trafficking in mammalian cells. J Neurochem 2024; 168:100-114. [PMID: 38102893 PMCID: PMC10953025 DOI: 10.1111/jnc.16029] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 11/24/2023] [Accepted: 12/02/2023] [Indexed: 12/17/2023]
Abstract
The aquaporin-4 (AQP4) water channel is abundantly expressed in the glial cells of the central nervous system and facilitates brain swelling following diverse insults, such as traumatic injury or stroke. Lack of specific and therapeutic AQP4 inhibitors highlights the need to explore alternative routes to control the water permeability of glial cell membranes. The cell surface abundance of AQP4 in mammalian cells fluctuates rapidly in response to changes in oxygen levels and tonicity, suggesting a role for vesicular trafficking in its translocation to and from the cell surface. However, the molecular mechanisms of AQP4 trafficking are not fully elucidated. In this work, early and recycling endosomes were investigated as likely candidates of rapid AQP4 translocation together with changes in cytoskeletal dynamics. In transiently transfected HEK293 cells a significant amount of AQP-eGFP colocalised with mCherry-Rab5-positive early endosomes and mCherry-Rab11-positive recycling endosomes. When exposed to hypotonic conditions, AQP4-eGFP rapidly translocated from intracellular vesicles to the cell surface. Co-expression of dominant negative forms of the mCherry-Rab5 and -Rab11 with AQP4-eGFP prevented hypotonicity-induced AQP4-eGFP trafficking and led to concentration at the cell surface or intracellular vesicles respectively. Use of endocytosis inhibiting drugs indicated that AQP4 internalisation was dynamin-dependent. Cytoskeleton dynamics-modifying drugs also affected AQP4 translocation to and from the cell surface. AQP4 trafficking mechanisms were validated in primary human astrocytes, which express high levels of endogenous AQP4. The results highlight the role of early and recycling endosomes and cytoskeletal dynamics in AQP4 translocation in response to hypotonic and hypoxic stress and suggest continuous cycling of AQP4 between intracellular vesicles and the cell surface under physiological conditions.
Collapse
Affiliation(s)
- Andrea Markou
- College of Health and Life SciencesAston UniversityBirminghamUK
- School of Biosciences, Faculty of Health and Medical SciencesUniversity of SurreyGuildfordUK
| | - Philip Kitchen
- College of Health and Life SciencesAston UniversityBirminghamUK
| | - Ahmed Aldabbagh
- College of Health and Life SciencesAston UniversityBirminghamUK
| | | | - Mootaz M. Salman
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
- Kavli Institute for NanoScience DiscoveryUniversity of OxfordOxfordUK
| | - Roslyn M. Bill
- College of Health and Life SciencesAston UniversityBirminghamUK
| | - Zita Balklava
- College of Health and Life SciencesAston UniversityBirminghamUK
| |
Collapse
|
41
|
Beverley KM, Levitan I. Cholesterol regulation of mechanosensitive ion channels. Front Cell Dev Biol 2024; 12:1352259. [PMID: 38333595 PMCID: PMC10850386 DOI: 10.3389/fcell.2024.1352259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 01/17/2024] [Indexed: 02/10/2024] Open
Abstract
The purpose of this review is to evaluate the role of cholesterol in regulating mechanosensitive ion channels. Ion channels discussed in this review are sensitive to two types of mechanical signals, fluid shear stress and/or membrane stretch. Cholesterol regulates the channels primarily in two ways: 1) indirectly through localizing the channels into cholesterol-rich membrane domains where they interact with accessory proteins and/or 2) direct binding of cholesterol to the channel at specified putative binding sites. Cholesterol may also regulate channel function via changes of the biophysical properties of the membrane bilayer. Changes in cholesterol affect both mechanosensitivity and basal channel function. We focus on four mechanosensitive ion channels in this review Piezo, Kir2, TRPV4, and VRAC channels. Piezo channels were shown to be regulated by auxiliary proteins that enhance channel function in high cholesterol domains. The direct binding mechanism was shown in Kir2.1 and TRPV4 where cholesterol inhibits channel function. Finally, cholesterol regulation of VRAC was attributed to changes in the physical properties of lipid bilayer. Additional studies should be performed to determine the physiological implications of these sterol effects in complex cellular environments.
Collapse
Affiliation(s)
- Katie M. Beverley
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Irena Levitan
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
42
|
Wan Y, Zhao Y, Cao M, Wang J, Tran SV, Song Z, Hsueh BW, Wang SE. Lung Fibroblasts Take up Breast Cancer Cell-derived Extracellular Vesicles Partially Through MEK2-dependent Macropinocytosis. CANCER RESEARCH COMMUNICATIONS 2024; 4:170-181. [PMID: 38259097 PMCID: PMC10802141 DOI: 10.1158/2767-9764.crc-23-0316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/14/2023] [Accepted: 01/02/2024] [Indexed: 01/24/2024]
Abstract
Extracellular vesicles (EV) have emerged as critical effectors in the cross-talk between cancer and normal cells by transferring intracellular materials between adjacent or distant cells. Previous studies have begun to elucidate how cancer cells, by secreting EVs, adapt normal cells at a metastatic site to facilitate cancer cell metastasis. In this study, we utilized a high-content microscopic screening platform to investigate the mechanisms of EV uptake by primary lung fibroblasts. A selected library containing 90 FDA-approved anticancer drugs was screened for the effect on fibroblast uptake of EVs from MDA-MB-231 breast cancer cells. Among the drugs identified to inhibit EV uptake without exerting significant cytotoxicity, we validated the dose-dependent effect of Trametinib (a MEK1/2 inhibitor) and Copanlisib (a PI3K inhibitor). Trametinib suppressed macropinocytosis in lung fibroblasts and inhibited EV uptake with a higher potency comparing with Copanlisib. Gene knockdown and overexpression studies demonstrated that uptake of MDA-MB-231 EVs by lung fibroblasts required MEK2. These findings provide important insights into the mechanisms underlying lung fibroblast uptake of breast cancer cell-derived EVs, which could play a role in breast cancer metastasis to the lungs and suggest potential therapeutic targets for preventing or treating this deadly disease. SIGNIFICANCE Through a phenotypic screen, we found that MEK inhibitor Trametinib suppressed EV uptake and macropinocytosis in lung fibroblasts, and that EV uptake is mediated by MEK2 in these cells. Our results suggest that MEK2 inhibition could serve as a strategy to block cancer EV uptake by lung fibroblasts.
Collapse
Affiliation(s)
- Yuhao Wan
- Department of Pathology, University of California San Diego, La Jolla, California
| | - Yue Zhao
- Department of Pathology, University of California San Diego, La Jolla, California
| | - Minghui Cao
- Department of Pathology, University of California San Diego, La Jolla, California
| | - Jingyi Wang
- Department of Pathology, University of California San Diego, La Jolla, California
| | - Sheila V. Tran
- Department of Pathology, University of California San Diego, La Jolla, California
| | - Zhixuan Song
- Department of Pathology, University of California San Diego, La Jolla, California
| | - Brent W. Hsueh
- Department of Pathology, University of California San Diego, La Jolla, California
| | - Shizhen Emily Wang
- Department of Pathology, University of California San Diego, La Jolla, California
| |
Collapse
|
43
|
Azadi M, David AE. Enhancing Ocular Drug Delivery: The Effect of Physicochemical Properties of Nanoparticles on the Mechanism of Their Uptake by Human Cornea Epithelial Cells. ACS Biomater Sci Eng 2024; 10:429-441. [PMID: 38055935 DOI: 10.1021/acsbiomaterials.3c01144] [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: 12/08/2023]
Abstract
This study investigates the effect of nanoparticle size and surface chemistry on interactions of the nanoparticles with human cornea epithelial cells (HCECs). Poly(lactic-co-glycolic) acid (PLGA) nanoparticles were synthesized using the emulsion-solvent evaporation method and surface modified with mucoadhesive (alginate [ALG] and chitosan [CHS]) and mucopenetrative (polyethylene glycol [PEG]) polymers. Particles were found to be monodisperse (polydispersity index (PDI) below 0.2), spherical, and with size and zeta potential ranging from 100 to 250 nm and from -25 to +15 mV, respectively. Evaluation of cytotoxicity with the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay indicated that incubating cells with nanoparticles for 24 h at concentrations up to 100 μg/mL caused only mild toxicity (70-100% cell viability). Cellular uptake studies were conducted using an in vitro model developed with a monolayer of HCECs integrated with simulated mucosal solution. Evaluation of nanoparticle uptake revealed that energy-dependent endocytosis is the primary uptake mechanism. Among the different nanoparticles studied, 100 nm PLGA NPs and PEG-PLGA-150 NPs showed the highest levels of uptake by HCECs. Additionally, uptake studies in the presence of various inhibitors suggested that macropinocytosis and caveolae-mediated endocytosis are the dominant pathways. While clathrin-mediated endocytosis was found to also be partially responsible for nanoparticle uptake, phagocytosis did not play a role within the studied ranges of size and surface chemistries. These important findings could lead to improved nanoparticle-based formulations that could improve therapies for ocular diseases.
Collapse
Affiliation(s)
- Marjan Azadi
- Department of Chemical Engineering, Auburn University, Auburn, Alabama 36849, United States
| | - Allan E David
- Department of Chemical Engineering, Auburn University, Auburn, Alabama 36849, United States
| |
Collapse
|
44
|
Hacker C, Sendra K, Keisham P, Filipescu T, Lucocq J, Salimi F, Ferguson S, Bhella D, MacNeill SA, Embley M, Lucocq J. Biogenesis, inheritance, and 3D ultrastructure of the microsporidian mitosome. Life Sci Alliance 2024; 7:e202201635. [PMID: 37903625 PMCID: PMC10618108 DOI: 10.26508/lsa.202201635] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/02/2023] [Accepted: 10/03/2023] [Indexed: 11/01/2023] Open
Abstract
During the reductive evolution of obligate intracellular parasites called microsporidia, a tiny remnant mitochondrion (mitosome) lost its typical cristae, organellar genome, and most canonical functions. Here, we combine electron tomography, stereology, immunofluorescence microscopy, and bioinformatics to characterise mechanisms of growth, division, and inheritance of this minimal mitochondrion in two microsporidia species (grown within a mammalian RK13 culture-cell host). Mitosomes of Encephalitozoon cuniculi (2-12/cell) and Trachipleistophora hominis (14-18/nucleus) displayed incremental/non-phasic growth and division and were closely associated with an organelle identified as equivalent to the fungal microtubule-organising centre (microsporidian spindle pole body; mSPB). The mitosome-mSPB association was resistant to treatment with microtubule-depolymerising drugs nocodazole and albendazole. Dynamin inhibitors (dynasore and Mdivi-1) arrested mitosome division but not growth, whereas bioinformatics revealed putative dynamins Drp-1 and Vps-1, of which, Vps-1 rescued mitochondrial constriction in dynamin-deficient yeast (Schizosaccharomyces pombe). Thus, microsporidian mitosomes undergo incremental growth and dynamin-mediated division and are maintained through ordered inheritance, likely mediated via binding to the microsporidian centrosome (mSPB).
Collapse
Affiliation(s)
| | - Kacper Sendra
- Biosciences Institute, The Medical School, Catherine Cookson Building, Newcastle University, Newcastle upon Tyne, UK
| | | | | | - James Lucocq
- Department of Surgery, Dundee Medical School Ninewells Hospital, Dundee, UK
| | - Fatemeh Salimi
- School of Medicine, University of St Andrews, St Andrews, UK
| | - Sophie Ferguson
- School of Medicine, University of St Andrews, St Andrews, UK
| | - David Bhella
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | | | - Martin Embley
- Biosciences Institute, Centre for Bacterial Cell Biology, Baddiley-Clark Building, Newcastle University, Newcastle upon Tyne, UK
| | - John Lucocq
- School of Medicine, University of St Andrews, St Andrews, UK
| |
Collapse
|
45
|
Al Othman A, Bagrov D, Rozenberg JM, Glazova O, Skryabin G, Tchevkina E, Mezentsev A, Durymanov M. Retrovirus-like gag protein Arc/Arg3.1 is involved in extracellular-vesicle-mediated mRNA transfer between glioma cells. Biochim Biophys Acta Gen Subj 2024; 1868:130522. [PMID: 37995879 DOI: 10.1016/j.bbagen.2023.130522] [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: 04/26/2023] [Revised: 10/29/2023] [Accepted: 11/12/2023] [Indexed: 11/25/2023]
Abstract
BACKGROUND Activity-regulated cytoskeleton-associated (Arc) protein is predominantly expressed in excitatory glutamatergic neurons of vertebrates, where it plays a pivotal role in regulation of synaptic plasticity. Arc protein forms capsid-like particles, which can encapsulate and transfer mRNA in extracellular vesicles (EVs) between hippocampal neurons. Once glioma cell networks actively interact with neurons via paracrine signaling and formation of neurogliomal glutamatergic synapses, we predicted the involvement of Arc in a process of EV-mediated mRNA transfer between glioma cells. MATERIALS AND METHODS Arc expression in three human glioma cell lines was evaluated by WB and immunocytochemistry. The properties of Arc protein/mRNA-containing EVs produced by glioma cells were analyzed by RT-PCR, TEM, and WB. Flow cytometry, RT-PCR, and fluorescent microscopy were used to show the involvement of Arc in EV-mediated mRNA transfer between glioma cells. RESULTS It was found that human glioma cells can produce EVs containing Arc/Arg3.1 protein and Arc mRNA (or "Arc EVs"). Arc EVs from U87 glioma cells internalize and deliver Arc mRNA to recipient U87 cells, where it is translated into a protein. Arc overexpression significantly increases EV production, alters EV morphology, and enhances intercellular transfer of highly expressed mRNA in glioma cell culture. CONCLUSION These findings indicate involvement of Arc EVs into mRNA transfer between glioma cells that could contribute to tumor progression and affect synaptic plasticity in cancer patients.
Collapse
Affiliation(s)
- Aya Al Othman
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, Russia
| | - Dmitry Bagrov
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia; Department of Bioengineering, Faculty of Biology, M.V. Lomonosov Moscow State University, Leninskie Gory, Moscow, Russia
| | - Julian M Rozenberg
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, Russia
| | - Olga Glazova
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, Russia
| | - Gleb Skryabin
- Institute of Carcinogenesis, N.N. Blokhin National Medical Research Center of Oncology, Moscow, Russia
| | - Elena Tchevkina
- Institute of Carcinogenesis, N.N. Blokhin National Medical Research Center of Oncology, Moscow, Russia
| | - Alexandre Mezentsev
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, Russia
| | - Mikhail Durymanov
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, Russia; Department of Radiochemistry, Faculty of Chemistry, M.V. Lomonosov Moscow State University, Moscow, Russia.
| |
Collapse
|
46
|
Minns LA, Sausman KT, Brown AP, York RA, McCall JR. Karenia brevis Extract Induces Cellular Entry through Distinct Mechanisms in Phagocytic RAW 264.7 Macrophages versus Non-Phagocytic Vero Cells. Mar Drugs 2023; 22:4. [PMID: 38276642 PMCID: PMC10820030 DOI: 10.3390/md22010004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 01/27/2024] Open
Abstract
Marine algae extracts are an important area of potential drug discovery; however, nearly all studies to date have used non-fluorescent-based methods to determine changes in target cell activity. Many of the most robust immunological and cellular analyses rely on fluorescent probes and readouts, which can be problematic when the algae extract is fluorescent itself. In this study, we identified the fluorescent spectrum of an isolated extract from the marine dinoflagellate Karenia brevis, which included two fluorescing components: chlorophyll α and pheophytin α. When excited at 405 nm and 664 nm, the extract emitted fluorescence at 676 nm and 696 nm, respectively. The extract and its fluorescing components, chlorophyll α and pheophytin α, entered phagocytic RAW 264.7 macrophages and non-phagocytic Vero kidney cells through distinct mechanisms. When incubated with the extract and its main components, both the RAW 264.7 macrophages and the Vero cells accumulated fluorescence as early as 30 min and continued through 48 h. Vero kidney cells accumulated the K. brevis fluorescent extract through a dynamin-independent and acidified endosomal-dependent mechanism. RAW 264.7 macrophages accumulated fluorescent extract through a dynamin-independent, acidified endosomal-independent mechanism, which supports accumulation through phagocytosis. Furthermore, RAW 264.7 macrophages downregulated cell-surface expression of CD206 in response to extract stimulation indicating activation of phagocytic responses and potential immunosuppression of these immune cells. This study represents the first characterization of the cellular update of K. brevis extracts in phagocytic versus non-phagocytic cells. The data suggest the importance of understanding cellular uptake of fluorescing algae extracts and their mechanism of action for future drug discovery efforts.
Collapse
Affiliation(s)
- Laurie A. Minns
- School of Nursing, College of Health and Human Services, University of North Carolina Wilmington, 601 S. College Road, Wilmington, NC 28403, USA; (L.A.M.)
- Center for Marine Science, University of North Carolina Wilmington, 5600 Marvin K Moss Lane, Wilmington, NC 28409, USA
| | - Kathryn T. Sausman
- School of Nursing, College of Health and Human Services, University of North Carolina Wilmington, 601 S. College Road, Wilmington, NC 28403, USA; (L.A.M.)
- Center for Marine Science, University of North Carolina Wilmington, 5600 Marvin K Moss Lane, Wilmington, NC 28409, USA
| | - Ariel P. Brown
- School of Nursing, College of Health and Human Services, University of North Carolina Wilmington, 601 S. College Road, Wilmington, NC 28403, USA; (L.A.M.)
- Center for Marine Science, University of North Carolina Wilmington, 5600 Marvin K Moss Lane, Wilmington, NC 28409, USA
| | - Robert A. York
- Center for Marine Science, University of North Carolina Wilmington, 5600 Marvin K Moss Lane, Wilmington, NC 28409, USA
- Algal Resources Collection, University of North Carolina Wilmington, 5600 Marvin K Moss Lane, Wilmington, NC 28409, USA
| | - Jennifer R. McCall
- School of Nursing, College of Health and Human Services, University of North Carolina Wilmington, 601 S. College Road, Wilmington, NC 28403, USA; (L.A.M.)
- Center for Marine Science, University of North Carolina Wilmington, 5600 Marvin K Moss Lane, Wilmington, NC 28409, USA
| |
Collapse
|
47
|
Silva SV, Lima MA, Hodgson L, Freitas VM, Rodríguez-Manzaneque JC. ADAMTS-1 has nuclear localization in cells with epithelial origin and leads to decreased cell migration. Exp Cell Res 2023; 433:113852. [PMID: 37951335 PMCID: PMC10841765 DOI: 10.1016/j.yexcr.2023.113852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 11/02/2023] [Accepted: 11/03/2023] [Indexed: 11/13/2023]
Abstract
In the study of tumorigenesis, the involvement of molecules within the extracellular matrix (ECM) is crucial. ADAMTSs (A Disintegrin and Metalloproteinase with Thrombospondin motifs), a group of secreted proteases known for their role in ECM remodeling, were primarily considered to be extracellular proteases. However, our research specifically detected ADAMTS-1, a member of this family, predominantly within the nucleus of mammary cells. Our main objective was to understand the mechanism of ADAMTS-1 translocation to the nucleus and its functional significance in this cellular compartment. Our investigation uncovered that nuclear ADAMTS-1 was present in cells exhibiting an epithelial phenotype, while cells of mesenchymal origin contained the protease in the cytoplasm. Moreover, disruption of ADAMTS-1 secretion, induced by Monensin treatment, resulted in its accumulation in the cytoplasm. Notably, our research indicated that alterations in the secretory pathways could influence the protease's compartmentalization. Additionally, experiments with conditioned medium from cells containing nuclear ADAMTS-1 demonstrated its internalization into the nucleus by HT-1080 cells and fibroblasts. Furthermore, heightened levels of ADAMTS-1 within the ECM reduced the migratory potential of mesenchymal cells. This highlights the potential significance of nuclear ADAMTS-1 as a critical component within the tumor microenvironment due to its functional activity in this specific cellular compartment.
Collapse
Affiliation(s)
- Suély V Silva
- Department of Cell and Developmental Biology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil.
| | - Maíra A Lima
- Department of Cell and Developmental Biology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil; Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Louis Hodgson
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Vanessa M Freitas
- Department of Cell and Developmental Biology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil.
| | | |
Collapse
|
48
|
Zeng Y, Estapé Senti M, Labonia MCI, Papadopoulou P, Brans MAD, Dokter I, Fens MH, van Mil A, Sluijter JPG, Schiffelers RM, Vader P, Kros A. Fusogenic Coiled-Coil Peptides Enhance Lipid Nanoparticle-Mediated mRNA Delivery upon Intramyocardial Administration. ACS NANO 2023; 17:23466-23477. [PMID: 37982378 PMCID: PMC10722601 DOI: 10.1021/acsnano.3c05341] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 11/02/2023] [Accepted: 11/09/2023] [Indexed: 11/21/2023]
Abstract
Heart failure is a serious condition that results from the extensive loss of specialized cardiac muscle cells called cardiomyocytes (CMs), typically caused by myocardial infarction (MI). Messenger RNA (mRNA) therapeutics are emerging as a very promising gene medicine for regenerative cardiac therapy. To date, lipid nanoparticles (LNPs) represent the most clinically advanced mRNA delivery platform. Yet, their delivery efficiency has been limited by their endosomal entrapment after endocytosis. Previously, we demonstrated that a pair of complementary coiled-coil peptides (CPE4/CPK4) triggered efficient fusion between liposomes and cells, bypassing endosomal entrapment and resulting in efficient drug delivery. Here, we modified mRNA-LNPs with the fusogenic coiled-coil peptides and demonstrated efficient mRNA delivery to difficult-to-transfect induced pluripotent stem-cell-derived cardiomyocytes (iPSC-CMs). As proof of in vivo applicability of these fusogenic LNPs, local administration via intramyocardial injection led to significantly enhanced mRNA delivery and concomitant protein expression. This represents the successful application of the fusogenic coiled-coil peptides to improve mRNA-LNPs transfection in the heart and provides the potential for the advanced development of effective regenerative therapies for heart failure.
Collapse
Affiliation(s)
- Ye Zeng
- Department
of Supramolecular & Biomaterials Chemistry, Leiden Institute of
Chemistry, Leiden University, 2333 CC Leiden, The Netherlands
| | - Mariona Estapé Senti
- CDL
Research, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - M. Clara I. Labonia
- Department
of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Panagiota Papadopoulou
- Department
of Supramolecular & Biomaterials Chemistry, Leiden Institute of
Chemistry, Leiden University, 2333 CC Leiden, The Netherlands
| | - Maike A. D. Brans
- Department
of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Inge Dokter
- Department
of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Regenerative
Medicine Center Utrecht, University Utrecht,
University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands
| | - Marcel H. Fens
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CS Utrecht, The Netherlands
| | - Alain van Mil
- Department
of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Regenerative
Medicine Center Utrecht, University Utrecht,
University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands
| | - Joost P. G. Sluijter
- Department
of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Regenerative
Medicine Center Utrecht, University Utrecht,
University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands
| | | | - Pieter Vader
- CDL
Research, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Department
of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Alexander Kros
- Department
of Supramolecular & Biomaterials Chemistry, Leiden Institute of
Chemistry, Leiden University, 2333 CC Leiden, The Netherlands
| |
Collapse
|
49
|
Kyrrestad I, Larsen AK, Sánchez Romano J, Simón-Santamaría J, Li R, Sørensen KK. Infection of liver sinusoidal endothelial cells with Muromegalovirus muridbeta1 involves binding to neuropilin-1 and is dynamin-dependent. Front Cell Infect Microbiol 2023; 13:1249894. [PMID: 38029264 PMCID: PMC10665495 DOI: 10.3389/fcimb.2023.1249894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Liver sinusoidal endothelial cells (LSEC) are scavenger cells with a remarkably high capacity for clearance of several blood-borne macromolecules and nanoparticles, including some viruses. Endocytosis in LSEC is mainly via the clathrin-coated pit mediated route, which is dynamin-dependent. LSEC can also be a site of infection and latency of betaherpesvirus, but mode of virus entry into these cells has not yet been described. In this study we have investigated the role of dynamin in the early stage of muromegalovirus muridbeta1 (MuHV-1, murid betaherpesvirus 1, murine cytomegalovirus) infection in mouse LSECs. LSEC cultures were freshly prepared from C57Bl/6JRj mouse liver. We first examined dose- and time-dependent effects of two dynamin-inhibitors, dynasore and MitMAB, on cell viability, morphology, and endocytosis of model ligands via different LSEC scavenger receptors to establish a protocol for dynamin-inhibition studies in these primary cells. LSECs were challenged with MuHV-1 (MOI 0.2) ± dynamin inhibitors for 1h, then without inhibitors and virus for 11h, and nuclear expression of MuHV-1 immediate early antigen (IE1) measured by immune fluorescence. MuHV-1 efficiently infected LSECs in vitro. Infection was significantly and independently inhibited by dynasore and MitMAB, which block dynamin function via different mechanisms, suggesting that initial steps of MuHV-1 infection is dynamin-dependent in LSECs. Infection was also reduced in the presence of monensin which inhibits acidification of endosomes. Furthermore, competitive binding studies with a neuropilin-1 antibody blocked LSEC infection. This suggests that MuHV-1 infection in mouse LSECs involves virus binding to neuropilin-1 and occurs via endocytosis.
Collapse
Affiliation(s)
- Ingelin Kyrrestad
- Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | | | | | | | | | | |
Collapse
|
50
|
Debnath M, Forster J, Ramesh A, Kulkarni A. Protein Corona Formation on Lipid Nanoparticles Negatively Affects the NLRP3 Inflammasome Activation. Bioconjug Chem 2023; 34:1766-1779. [PMID: 37707953 DOI: 10.1021/acs.bioconjchem.3c00329] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
The interaction between lipid nanoparticles (LNPs) and serum proteins, giving rise to a unique identification in the form of the protein corona, has been shown to be associated with novel recognition by cell receptors. The presence of the corona enveloping the nanoparticle strongly affects the interplay with immune cells. The immune responses mediated by protein corona can affect nanoparticle toxicity and targeting capabilities. But the intracellular signaling of LNPs after corona formation resulting in the change of nanoparticles' ability to provoke immune responses remains unclear. Therefore, a more systematic and delineated approach must be considered to present the correlation between corona complexes and the shift in nanoparticle immunogenicity. Here, we studied and reported the inhibiting effect of the absorbed proteins on the LNPs on the NLRP3 inflammasome activation, a key intracellular protein complex that modulates several inflammatory responses. Ionizable lipid as a component of LNP was observed to play an important role in modulating the activation of NLRP3 inflammasome in serum-free conditions. However, in the presence of serum proteins, the corona layer on LNPs caused a significant reduction in the inflammasome activation. Reduction in the lysosomal rupture after treatment with corona-LNPs significantly reduced inflammasome activation. Furthermore, a strong reduction of cellular uptake in macrophages after the corona formation was observed. On inspecting the uptake mechanisms in macrophages using transport inhibitors, lipid formulation was found to play a critical role in determining the endocytic pathways for the LNPs in macrophages. This study highlights the need to critically analyze the protein interactions with nanomaterials and their concomitant adaptability with immune cells to evaluate nano-bio surfaces and successfully design nanomaterials for biological applications.
Collapse
Affiliation(s)
- Maharshi Debnath
- Department of Chemical Engineering, University of Massachusetts Amherst, 686 North Pleasant St., Amherst, Massachusetts 01003, United States
| | - James Forster
- Department of Chemical Engineering, University of Massachusetts Amherst, 686 North Pleasant St., Amherst, Massachusetts 01003, United States
| | - Anujan Ramesh
- Department of Biomedical Engineering, University of Massachusetts Amherst, 240 Thatcher Road, Amherst, Massachusetts 01003, United States
| | - Ashish Kulkarni
- Department of Chemical Engineering, University of Massachusetts Amherst, 686 North Pleasant St., Amherst, Massachusetts 01003, United States
- Department of Biomedical Engineering, University of Massachusetts Amherst, 240 Thatcher Road, Amherst, Massachusetts 01003, United States
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts Amherst, 240 Thatcher Road, Amherst, Massachusetts 01003, United States
| |
Collapse
|