1
|
Skrtic M, Yusuf B, Patel S, Reddy EC, Ting KKY, Cybulsky MI, Freeman SA, Robinson LA. The neurorepellent SLIT2 inhibits LPS-induced proinflammatory signaling in macrophages. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:141-152. [PMID: 40073268 PMCID: PMC11844144 DOI: 10.1093/jimmun/vkae009] [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: 11/20/2023] [Accepted: 10/30/2024] [Indexed: 03/14/2025]
Abstract
Macrophages are important mediators of immune responses with critical roles in the recognition and clearance of pathogens, as well as in the resolution of inflammation and wound healing. The neuronal guidance cue SLIT2 has been widely studied for its effects on immune cell functions, most notably directional cell migration. Recently, SLIT2 has been shown to directly enhance bacterial killing by macrophages, but the effects of SLIT2 on inflammatory activation of macrophages are less known. Using RNA sequencing analysis, quantitative polymerase chain reaction, and enzyme-linked immunosorbent assay, we determined that in murine bone marrow-derived macrophages challenged with the potent proinflammatory mediator lipopolysaccharide (LPS), exposure to the bioactive N-terminal fragment of SLIT2 (NSLIT2) suppressed production of proinflammatory cytokines interleukin (IL)-6 and IL-12 and concurrently increased the anti-inflammatory cytokine IL-10. We found that NSLIT2 inhibited LPS-induced MyD88- and TRIF-mediated signaling cascades and did not inhibit LPS-induced internalization of Toll-like receptor 4 (TLR4), but instead inhibited LPS-induced upregulation of macropinocytosis. Inhibition of macropinocytosis in macrophages attenuated LPS-induced production of proinflammatory IL-6 and IL-12 and concurrently enhanced anti-inflammatory IL-10. Taken together, our results indicate that SLIT2 can selectively modulate macrophage response to potent proinflammatory stimuli, such as LPS, by attenuating proinflammatory activation and simultaneously enhancing anti-inflammatory activity. Our results highlight the role of macropinocytosis in proinflammatory activation of macrophages exposed to LPS. Given that LPS-producing bacteria cause host illness through synergistic direct bacterial infection and excessive LPS-induced systemic inflammation, our work suggests a novel therapeutic role for SLIT2 in combatting the significant morbidity and mortality of patients with Gram-negative bacterial sepsis.
Collapse
Affiliation(s)
- Marko Skrtic
- Division of Nephrology, Kingston Health Sciences Centre, Queen’s University, Kingston, ON, Canada
| | - Bushra Yusuf
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Sajedabanu Patel
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Emily C Reddy
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Kenneth K Y Ting
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Myron I Cybulsky
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Spencer A Freeman
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Lisa A Robinson
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
- Department of Paediatrics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
2
|
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
|
3
|
Wollman J, Wanniarachchi K, Pradhan B, Huang L, Kerkvliet JG, Hoppe AD, Thiex NW. Mannose receptor (MRC1) mediates uptake of dextran in macrophages via receptor-mediated endocytosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.13.607841. [PMID: 39211167 PMCID: PMC11360935 DOI: 10.1101/2024.08.13.607841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Macrophages maintain surveillance of their environment using receptor-mediated endocytosis and pinocytosis. Receptor-mediated endocytosis allows macrophages to recognize and internalize specific ligands whereas macropinocytosis non-selectively internalizes extracellular fluids and solutes. Here, 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 endocytic mannose receptor c-type 1 ( Mrc1 , also known as CD206) was a top hit in the screen. Targeted gene disruptions of Mrc1 reduced dextran uptake but had little effect on uptake of Lucifer yellow, a fluid-phase marker. Other screen hits also differentially affected the uptake of dextran and Lucifer yellow, indicating the solutes are internalized by different mechanisms. We further deduced that BMDMs take up dextran via MRC1-mediated endocytosis by showing that competition with mannan, a ligand of MRC1, as well as treatment with Dyngo-4a, a dynamin inhibitor, reduced dextran uptake. Finally, we observed that IL4-treated BMDM internalize more dextran than untreated BMDM by upregulating MRC1 expression. These results demonstrate that dextran is not an effective marker for the bulk uptake of fluids and solutes by macropinocytosis since it is internalized by both macropinocytosis and receptor-mediated endocytosis in cells expressing MRC1. This report identifies numerous genes that regulate dextran internalization in primary murine macrophages and predicts cellular pathways and processes regulating MRC1. This work lays the groundwork for identifying specific genes and regulatory networks that regulate MRC1 expression and MRC1-mediated endocytosis in macrophages. Significance Statement Macrophages constantly survey and clear tissues by specifically and non-specifically internalizing debris and solutes. However, the molecular mechanisms and modes of regulation of these endocytic and macropinocytic processes are not well understood. Here, CRISPR/Cas9 whole genome screens were used to identify genes regulating uptake of dextran, a sugar polymer that is frequently used as a marker macropinocytosis, and compared with Lucifer yellow, a fluorescent dye with no known receptors. The authors identified the mannose receptor as well as other proteins regulating expression of the mannose receptor as top hits in the screen. Targeted disruption of Mrc1 , the gene that encodes mannose receptor, greatly diminished dextran uptake but had no effect on cellular uptake of Lucifer yellow. Furthermore, exposure to the cytokine IL4 upregulated mannose receptor expression on the cell surface and increased uptake of dextran with little effect on Lucifer yellow uptake. Studies seeking to understand regulation of macropinocytosis in macrophages will be confounded by the use of dextran as a fluid-phase marker. MRC1 is a marker of alternatively activated/anti-inflammatory macrophages and is a potential target for delivery of therapeutics to macrophages. This work provides the basis for mechanistic underpinning of how MRC1 contributes to the receptor-mediated uptake of carbohydrates and glycoproteins from the tissue milieu and distinguishes genes regulating receptor-mediated endocytosis from those regulating the bona fide fluid-phase uptake of fluids and solutes by macropinocytosis.
Collapse
|
4
|
Kraus A, Kratzer B, Sehgal ANA, Trapin D, Khan M, Boucheron N, Pickl WF. Macropinocytosis Is the Principal Uptake Mechanism of Antigen-Presenting Cells for Allergen-Specific Virus-like Nanoparticles. Vaccines (Basel) 2024; 12:797. [PMID: 39066435 PMCID: PMC11281386 DOI: 10.3390/vaccines12070797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/29/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Virus-like nanoparticles (VNP) are regarded as efficient vaccination platforms and have proven to be useful for the non-anaphylactogenic delivery of allergen-specific immunotherapy in preclinical models previously. Herein, we sought to determine the mode of VNP uptake by antigen presenting cells (APC). Accordingly, we screened a collection of substances known to inhibit different uptake pathways by APC. The human leukemia monocytic cell line THP-1 and the murine dendritic cell line DC 2.4 were examined for the uptake of fluorescently labelled VNP in the presence or absence of inhibitors. The inhibitory effect of candidate substances that blocked VNP uptake in APC lines was subsequently evaluated in studies with primary APC present in splenocyte and lung cell homogenates in vitro and upon intratracheal application of VNP in vivo. The uptake of allergen-specific VNP in vitro and in vivo was mainly observed by macrophages and CD103+ dendritic cells and was sensitive to inhibitors that block macropinocytosis, such as hyperosmolarity induced by sucrose or the polyphenol compound Rottlerin at low micromolar concentrations but not by other inhibitors. Also, T-cell proliferation induced by allergen-specific VNP was significantly reduced by both substances. In contrast, substances that stimulate macropinocytosis, such as Heparin and phorbol myristate acetate (PMA), increased VNP-uptake and may, thus, help modulate allergen-specific T-cell responses. We have identified macropinocytosis as the principal uptake mechanism of APC for allergen-specific VNP in vitro and in vivo, paving the way for further improvement of VNP-based therapies, especially those that can be used for tolerance induction in allergy, in the future.
Collapse
Affiliation(s)
- Armin Kraus
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Bernhard Kratzer
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Al Nasar Ahmed Sehgal
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Doris Trapin
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Matarr Khan
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Nicole Boucheron
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Winfried F. Pickl
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
- Karl Landsteiner University of Health Sciences, 3500 Krems, Austria
| |
Collapse
|
5
|
Nakamura J, Shiohama Y, Röth D, Haruta T, Yamashita Y, Mitsuzono T, Mochizuki C, Kalkum M, Nakamura M. Size and Surface Properties of Functionalized Organosilica Particles Impact Cell-Particle Interactions Including Mitochondrial Activity. ACS APPLIED MATERIALS & INTERFACES 2024; 16:30980-30996. [PMID: 38857433 DOI: 10.1021/acsami.4c06455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
Understanding of the interactions between macrophages and multifunctional nanoparticles is important for development of novel macrophage-based immunotherapies. Here, we investigated the effects of fluorescent thiol-organosilica particle size and surface properties on cell-particle interactions, including mitochondrial activity, using the mouse macrophage cell line J774A.1. Three different sizes of thiol-organosilica particles (150, 400, and 680 nm in diameter) containing fluorescein (OS/F150, OS/F400, and OS/F680) and particles surface functionalized with polyethylenimine (PEI) (OS/F150PEI, OS/F400PEI, and OS/F680PEI) were prepared. Flow cytometric analysis, time-lapse imaging, and single-cell analysis of particle uptake and mitochondrial activity of J774A.1 cells demonstrated variations in uptake and kinetics depending on the particle size and surface as well as on each individual cell. Cells treated with OS/F150 and OS/F150PEI showed higher uptake and mitochondrial activity than those treated with other particles. The interaction between endosomes and mitochondria was observed using 3D fluorescent imaging and was characterized by the involvement of iron transport into mitochondria by iron-containing proteins adsorbed on the particle surface. Scanning electron microscopy of the cells treated with the particles revealed alterations in cell membrane morphology, depending on particle size and surface. We performed correlative light and electron microscopy combined with time-lapse and 3D imaging to develop an integrated correlation analysis of particle uptake, mitochondrial activity, and cell membrane morphology in single macrophages. These cell-specific characteristics of macrophages against functional particles and their evaluation methods are crucial for understanding the immunological functions of individual macrophages and developing novel immunotherapies.
Collapse
Affiliation(s)
- Junna Nakamura
- Department of Organ Anatomy and Nanomedicine, Yamaguchi University Graduate School of Medicine, 1-1-1 minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
- Core Clusters for Research Initiatives of Yamaguchi University, 1-1-1 minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
- Research Institute for Cell Design Medical Science, Yamaguchi University, 1-1-1 minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Yasuo Shiohama
- Department of Organ Anatomy and Nanomedicine, Yamaguchi University Graduate School of Medicine, 1-1-1 minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
- Core Clusters for Research Initiatives of Yamaguchi University, 1-1-1 minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
- Research Institute for Cell Design Medical Science, Yamaguchi University, 1-1-1 minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Daniel Röth
- Department of Department of Immunology & Theranostics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, California 91010, United States
| | - Tomohiro Haruta
- EM application group, EM business unit, JEOL Ltd., Akishima, Tokyo JP 196-8558, Japan
| | - Yukari Yamashita
- Department of Organ Anatomy and Nanomedicine, School of Medicine, Facuelty of Medicine and Health Sciences, Yamaguchi University, 1-1-1 minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Tomohiro Mitsuzono
- Department of Organ Anatomy and Nanomedicine, School of Medicine, Facuelty of Medicine and Health Sciences, Yamaguchi University, 1-1-1 minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Chihiro Mochizuki
- Department of Organ Anatomy and Nanomedicine, Yamaguchi University Graduate School of Medicine, 1-1-1 minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
- Core Clusters for Research Initiatives of Yamaguchi University, 1-1-1 minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
- Research Institute for Cell Design Medical Science, Yamaguchi University, 1-1-1 minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Markus Kalkum
- Department of Department of Immunology & Theranostics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, California 91010, United States
| | - Michihiro Nakamura
- Department of Organ Anatomy and Nanomedicine, Yamaguchi University Graduate School of Medicine, 1-1-1 minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
- Core Clusters for Research Initiatives of Yamaguchi University, 1-1-1 minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
- Research Institute for Cell Design Medical Science, Yamaguchi University, 1-1-1 minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| |
Collapse
|
6
|
Zhao J, Lu N, Qu Y, Liu W, Zhong H, Tang N, Li J, Wang L, Xi D, He F. Calcium-sensing receptor-mediated macrophage polarization improves myocardial remodeling in spontaneously hypertensive rats. Exp Biol Med (Maywood) 2024; 249:10112. [PMID: 38715976 PMCID: PMC11075494 DOI: 10.3389/ebm.2024.10112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 11/13/2023] [Indexed: 06/04/2024] Open
Abstract
Chronic inflammation is a key element in the progression of essential hypertension (EH). Calcium plays a key role in inflammation, so its receptor, the calcium-sensing receptor (CaSR), is an essential mediator of the inflammatory process. Compelling evidence suggests that CaSR mediates inflammation in tissues and immune cells, where it mediates their activity and chemotaxis. Macrophages (Mφs) play a major role in the inflammatory response process. This study provided convincing evidence that R568, a positive regulator of CaSR, was effective in lowering blood pressure in spontaneously hypertensive rats (SHRs), improving cardiac function by alleviating cardiac hypertrophy and fibrosis. R568 can increase the content of CaSR and M2 macrophages (M2Mφs, exert an anti-inflammatory effect) in myocardial tissue, reduce M1 macrophages (M1Mφs), which have a pro-inflammatory effect in this process. In contrast, NPS2143, a negative state regulator of CaSR, exerted the opposite effect in all of the above experiments. Following this study, R568 increased CaSR content in SHR myocardial tissue, lowered blood pressure, promoted macrophages to M2Mφs and improved myocardial fibrosis, but interestingly, both M1Mφs and M2Mφs were increased in the peritoneal cavity of SHRs, the number of M2Mφs remained lower than M1Mφs. In vitro, R568 increased CaSR content in RAW264.7 cells (a macrophage cell line), regulating intracellular Ca2+ ([Ca2+]i) inhibited NOD-like receptor family protein 3 (NLRP3) inflammasome activation and ultimately prevented its conversion to M1Mφs. The results showed that a decrease in CaSR in hypertensive rats causes further development of hypertension and cardiac damage. EH myocardial remodeling can be improved by CaSR overexpression by suppressing NLRP3 inflammasome activation and macrophage polarization toward M1Mφs and increasing M2Mφs.
Collapse
Affiliation(s)
- Jiaqi Zhao
- Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, NHC Key Laboratory for Prevention and Treatment of Central Asia High Incidence Diseases, Department of Pathophysiology, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Ning Lu
- School of Medicine, Tarim University, Alaer, Xinjiang, China
| | - Yuanyuan Qu
- Department of Respiratory Medicine, The First Affiliated Hospital of Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Wei Liu
- Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, NHC Key Laboratory for Prevention and Treatment of Central Asia High Incidence Diseases, Department of Pathophysiology, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Hua Zhong
- Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, NHC Key Laboratory for Prevention and Treatment of Central Asia High Incidence Diseases, Department of Pathophysiology, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Na Tang
- Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, NHC Key Laboratory for Prevention and Treatment of Central Asia High Incidence Diseases, Department of Pathophysiology, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Jiayi Li
- Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, NHC Key Laboratory for Prevention and Treatment of Central Asia High Incidence Diseases, Department of Pathophysiology, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Lamei Wang
- Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, NHC Key Laboratory for Prevention and Treatment of Central Asia High Incidence Diseases, Department of Pathophysiology, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Dongmei Xi
- Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, NHC Key Laboratory for Prevention and Treatment of Central Asia High Incidence Diseases, Department of Pathophysiology, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Fang He
- Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, NHC Key Laboratory for Prevention and Treatment of Central Asia High Incidence Diseases, Department of Pathophysiology, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| |
Collapse
|
7
|
Chamberland É, Moravveji S, Doyon N, Duchesne S. A computational model of Alzheimer's disease at the nano, micro, and macroscales. Front Neuroinform 2024; 18:1348113. [PMID: 38586183 PMCID: PMC10995318 DOI: 10.3389/fninf.2024.1348113] [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: 12/01/2023] [Accepted: 02/26/2024] [Indexed: 04/09/2024] Open
Abstract
Introduction Mathematical models play a crucial role in investigating complex biological systems, enabling a comprehensive understanding of interactions among various components and facilitating in silico testing of intervention strategies. Alzheimer's disease (AD) is characterized by multifactorial causes and intricate interactions among biological entities, necessitating a personalized approach due to the lack of effective treatments. Therefore, mathematical models offer promise as indispensable tools in combating AD. However, existing models in this emerging field often suffer from limitations such as inadequate validation or a narrow focus on single proteins or pathways. Methods In this paper, we present a multiscale mathematical model that describes the progression of AD through a system of 19 ordinary differential equations. The equations describe the evolution of proteins (nanoscale), cell populations (microscale), and organ-level structures (macroscale) over a 50-year lifespan, as they relate to amyloid and tau accumulation, inflammation, and neuronal death. Results Distinguishing our model is a robust foundation in biological principles, ensuring improved justification for the included equations, and rigorous parameter justification derived from published experimental literature. Conclusion This model represents an essential initial step toward constructing a predictive framework, which holds significant potential for identifying effective therapeutic targets in the fight against AD.
Collapse
Affiliation(s)
- Éléonore Chamberland
- Centre de Recherche CERVO, Institut Universitaire de Santé Mentale de Québec, Québec, QC, Canada
- Département de Mathématiques et de Statistique, Québec, QC, Canada
| | - Seyedadel Moravveji
- Centre de Recherche CERVO, Institut Universitaire de Santé Mentale de Québec, Québec, QC, Canada
- Département de Mathématiques et de Statistique, Québec, QC, Canada
| | - Nicolas Doyon
- Centre de Recherche CERVO, Institut Universitaire de Santé Mentale de Québec, Québec, QC, Canada
- Département de Mathématiques et de Statistique, Québec, QC, Canada
| | - Simon Duchesne
- Centre de Recherche CERVO, Institut Universitaire de Santé Mentale de Québec, Québec, QC, Canada
- Département de Radiologie et Médecine Nucléaire, Université Laval, Québec, QC, Canada
- Centre de Recherche de l'Institut Universitaire en Cardiologie et Pneumologie de Québec, Québec, QC, Canada
| |
Collapse
|
8
|
Ramzy A, Soliman AH, Hassanein SI, Sebak AA. Multitarget, multiagent PLGA nanoparticles for simultaneous tumor eradication and TME remodeling in a melanoma mouse model. Drug Deliv Transl Res 2024; 14:491-509. [PMID: 37612575 PMCID: PMC10761550 DOI: 10.1007/s13346-023-01413-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2023] [Indexed: 08/25/2023]
Abstract
Despite the fact that chemoimmunotherapy has emerged as a key component in the era of cancer immunotherapy, it is challenged by the complex tumor microenvironment (TME) that is jam-packed with cellular and non-cellular immunosuppressive components. The aim of this study was to design a nanoparticulate system capable of sufficiently accumulating in the tumor and spleen to mediate local and systemic immune responses, respectively. The study also aimed to remodel the immunosuppressive TME. For such reasons, multi-functional polylactic-co-glycolic acid (PLGA) nanoparticles (NPs) were engineered to simultaneously eradicate the cancer cells, silence the tumor-associated fibroblasts (TAFs), and re-educate the tumor-associated macrophages (TAMs) using doxorubicin, losartan, and metformin, respectively. These agents were also selected for their ability to tip the balance of the splenic immune cells towards immunostimulatory phenotypes. To establish TAM and TAF cultures, normal macrophages and fibroblasts were incubated with B16F10 melanoma cell (Mel)-derived secretome. Drug-loaded PLGA NPs were prepared, characterized, and tested in the target cell types. Organ distribution of fluorescein-loaded PLGA NPs was evaluated in a mouse model of melanoma. Finally, the local and systemic effects of different combination therapy programs were portrayed. The in vitro studies showed that the drug-loaded PLGA NPs could significantly ablate the immunosuppressive nature of Mel and skew TAMs and TAFs towards more favorable phenotypes. While in vivo, PLGA NPs were proven to exhibit long blood circulation time and to localize preferentially in the tumor and the spleen. The combination of either metformin or losartan with doxorubicin was superior to the monotherapy, both locally and systemically. However, the three-agent combo produced detrimental effects in the form of compromised well-being, immune depletion, and metastasis. These findings indicate the potential of TME remodeling as means to prime the tumors for successful chemoimmunotherapy. In addition, they shed light on the importance of the careful use of combination therapies and the necessity of employing dose-reduction strategies. D-NPs doxorubicin-loaded NPs, M-NPs metformin-loaded NPs, L-NPs losartan-loaded NPs, TAMs tumor-associated macrophages, TAFs tumor-associated fibroblasts, PD-L1 programmed death ligand 1, TNF-α tumor necrosis factor alpha, TGF-β transforming growth factor beta, CD206/40/86 cluster of differentiation 206/40/86, α-SMA alpha-smooth muscle actin, MMPs matrix metalloproteases.
Collapse
Affiliation(s)
- Asmaa Ramzy
- Department of Pharmaceutical Technology, Faculty of Pharmacy & Biotechnology, the German University in Cairo, New Cairo, 11511, Egypt
| | - Aya H Soliman
- Department of Pharmaceutical Biology, Faculty of Pharmacy & Biotechnology, the German University in Cairo, New Cairo, 11511, Egypt
| | - Sally I Hassanein
- Department of Biochemistry, Faculty of Pharmacy & Biotechnology, the German University in Cairo, New Cairo, 11511, Egypt
| | - Aya A Sebak
- Department of Pharmaceutical Technology, Faculty of Pharmacy & Biotechnology, the German University in Cairo, New Cairo, 11511, Egypt.
| |
Collapse
|
9
|
Ahn W, Burnett FN, Pandey A, Ghoshal P, Singla B, Simon AB, Derella CC, A. Addo S, Harris RA, Lucas R, Csányi G. SARS-CoV-2 Spike Protein Stimulates Macropinocytosis in Murine and Human Macrophages via PKC-NADPH Oxidase Signaling. Antioxidants (Basel) 2024; 13:175. [PMID: 38397773 PMCID: PMC10885885 DOI: 10.3390/antiox13020175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 02/25/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) is an infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). While recent studies have demonstrated that SARS-CoV-2 may enter kidney and colon epithelial cells by inducing receptor-independent macropinocytosis, it remains unknown whether this process also occurs in cell types directly relevant to SARS-CoV-2-associated lung pneumonia, such as alveolar epithelial cells and macrophages. The goal of our study was to investigate the ability of SARS-CoV-2 spike protein subunits to stimulate macropinocytosis in human alveolar epithelial cells and primary human and murine macrophages. Flow cytometry analysis of fluid-phase marker internalization demonstrated that SARS-CoV-2 spike protein subunits S1, the receptor-binding domain (RBD) of S1, and S2 stimulate macropinocytosis in both human and murine macrophages in an angiotensin-converting enzyme 2 (ACE2)-independent manner. Pharmacological and genetic inhibition of macropinocytosis substantially decreased spike-protein-induced fluid-phase marker internalization in macrophages both in vitro and in vivo. High-resolution scanning electron microscopy (SEM) imaging confirmed that spike protein subunits promote the formation of membrane ruffles on the dorsal surface of macrophages. Mechanistic studies demonstrated that SARS-CoV-2 spike protein stimulated macropinocytosis via NADPH oxidase 2 (Nox2)-derived reactive oxygen species (ROS) generation. In addition, inhibition of protein kinase C (PKC) and phosphoinositide 3-kinase (PI3K) in macrophages blocked SARS-CoV-2 spike-protein-induced macropinocytosis. To our knowledge, these results demonstrate for the first time that SARS-CoV-2 spike protein subunits stimulate macropinocytosis in macrophages. These results may contribute to a better understanding of SARS-CoV-2 infection and COVID-19 pathogenesis.
Collapse
Affiliation(s)
- WonMo Ahn
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (W.A.); (F.N.B.); (A.P.); (B.S.); (S.A.A.); (R.L.)
| | - Faith N. Burnett
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (W.A.); (F.N.B.); (A.P.); (B.S.); (S.A.A.); (R.L.)
| | - Ajay Pandey
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (W.A.); (F.N.B.); (A.P.); (B.S.); (S.A.A.); (R.L.)
| | - Pushpankur Ghoshal
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (W.A.); (F.N.B.); (A.P.); (B.S.); (S.A.A.); (R.L.)
| | - Bhupesh Singla
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (W.A.); (F.N.B.); (A.P.); (B.S.); (S.A.A.); (R.L.)
| | - Abigayle B. Simon
- Georgia Prevention Institute, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (A.B.S.); (C.C.D.); (R.A.H.)
| | - Cassandra C. Derella
- Georgia Prevention Institute, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (A.B.S.); (C.C.D.); (R.A.H.)
| | - Stephen A. Addo
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (W.A.); (F.N.B.); (A.P.); (B.S.); (S.A.A.); (R.L.)
| | - Ryan A. Harris
- Georgia Prevention Institute, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (A.B.S.); (C.C.D.); (R.A.H.)
| | - Rudolf Lucas
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (W.A.); (F.N.B.); (A.P.); (B.S.); (S.A.A.); (R.L.)
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Gábor Csányi
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (W.A.); (F.N.B.); (A.P.); (B.S.); (S.A.A.); (R.L.)
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
10
|
Mangione MC, Wen J, Cao DJ. Mechanistic target of rapamycin in regulating macrophage function in inflammatory cardiovascular diseases. J Mol Cell Cardiol 2024; 186:111-124. [PMID: 38039845 PMCID: PMC10843805 DOI: 10.1016/j.yjmcc.2023.10.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 10/14/2023] [Accepted: 10/18/2023] [Indexed: 12/03/2023]
Abstract
The mechanistic target of rapamycin (mTOR) is evolutionarily conserved from yeast to humans and is one of the most fundamental pathways of living organisms. Since its discovery three decades ago, mTOR has been recognized as the center of nutrient sensing and growth, homeostasis, metabolism, life span, and aging. The role of dysregulated mTOR in common diseases, especially cancer, has been extensively studied and reported. Emerging evidence supports that mTOR critically regulates innate immune responses that govern the pathogenesis of various cardiovascular diseases. This review discusses the regulatory role of mTOR in macrophage functions in acute inflammation triggered by ischemia and in atherosclerotic cardiovascular disease (ASCVD) and heart failure with preserved ejection fraction (HFpEF), in which chronic inflammation plays critical roles. Specifically, we discuss the role of mTOR in trained immunity, immune senescence, and clonal hematopoiesis. In addition, this review includes a discussion on the architecture of mTOR, the function of its regulatory complexes, and the dual-arm signals required for mTOR activation to reflect the current knowledge state. We emphasize future research directions necessary to understand better the powerful pathway to take advantage of the mTOR inhibitors for innovative applications in patients with cardiovascular diseases associated with aging and inflammation.
Collapse
Affiliation(s)
- MariaSanta C Mangione
- Department of Internal Medicine, Cardiology Division, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jinhua Wen
- Department of Internal Medicine, Cardiology Division, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Dian J Cao
- Department of Internal Medicine, Cardiology Division, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; VA North Texas Health Care System, Dallas TX 75216, USA.
| |
Collapse
|
11
|
Church KA, Cardona AE, Hopp SC. Roles in Innate Immunity. ADVANCES IN NEUROBIOLOGY 2024; 37:263-286. [PMID: 39207697 DOI: 10.1007/978-3-031-55529-9_15] [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] [Indexed: 09/04/2024]
Abstract
Microglia are best known as the resident phagocytes of the central nervous system (CNS). As a resident brain immune cell population, microglia play key roles during the initiation, propagation, and resolution of inflammation. The discovery of resident adaptive immune cells in the CNS has unveiled a relationship between microglia and adaptive immune cells for CNS immune-surveillance during health and disease. The interaction of microglia with elements of the peripheral immune system and other CNS resident cells mediates a fine balance between neuroprotection and tissue damage. In this chapter, we highlight the innate immune properties of microglia, with a focus on how pattern recognition receptors, inflammatory signaling cascades, phagocytosis, and the interaction between microglia and adaptive immune cells regulate events that initiate an inflammatory or neuroprotective response within the CNS that modulates immune-mediated disease exacerbation or resolution.
Collapse
Affiliation(s)
- Kaira A Church
- Department of Molecular Microbiology & Immunology, The University of Texas at San Antonio, San Antonio, TX, USA
- South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Astrid E Cardona
- Department of Molecular Microbiology & Immunology, The University of Texas at San Antonio, San Antonio, TX, USA
- South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Sarah C Hopp
- Department of Pharmacology, Biggs Institute for Alzheimer's and Neurodegenerative Disease, The University of Texas Health Science Center San Antonio, San Antonio, TX, USA.
| |
Collapse
|
12
|
Salloum G, Bresnick AR, Backer JM. Macropinocytosis: mechanisms and regulation. Biochem J 2023; 480:335-362. [PMID: 36920093 DOI: 10.1042/bcj20210584] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/22/2023] [Accepted: 02/27/2023] [Indexed: 03/16/2023]
Abstract
Macropinocytosis is defined as an actin-dependent but coat- and dynamin-independent endocytic uptake process, which generates large intracellular vesicles (macropinosomes) containing a non-selective sampling of extracellular fluid. Macropinocytosis provides an important mechanism of immune surveillance by dendritic cells and macrophages, but also serves as an essential nutrient uptake pathway for unicellular organisms and tumor cells. This review examines the cell biological mechanisms that drive macropinocytosis, as well as the complex signaling pathways - GTPases, lipid and protein kinases and phosphatases, and actin regulatory proteins - that regulate macropinosome formation, internalization, and disposition.
Collapse
Affiliation(s)
- Gilbert Salloum
- Department of Molecular Pharamacology, Albert Einstein College of Medicine, Bronx, NY, U.S.A
| | - Anne R Bresnick
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, U.S.A
| | - Jonathan M Backer
- Department of Molecular Pharamacology, Albert Einstein College of Medicine, Bronx, NY, U.S.A
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, U.S.A
| |
Collapse
|
13
|
The role of the osmosensitive transcription factor NFAT5 in corneal edema resorption after injury. Exp Mol Med 2023; 55:565-573. [PMID: 36869067 PMCID: PMC10073147 DOI: 10.1038/s12276-023-00954-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 12/13/2022] [Accepted: 12/20/2022] [Indexed: 03/05/2023] Open
Abstract
The osmosensitive transcription factor nuclear factor of activated T cells 5 (NFAT5; or tonicity-responsive enhancer binding protein; TonEBP) plays a key role in macrophage-driven regulation of cutaneous salt and water balance. In the immune-privileged and transparent cornea, disturbances in fluid balance and pathological edema result in corneal transparency loss, which is one of the main causes of blindness worldwide. The role of NFAT5 in the cornea has not yet been investigated. We analyzed the expression and function of NFAT5 in naive corneas and in an established mouse model of perforating corneal injury (PCI), which causes acute corneal edema and transparency loss. In uninjured corneas, NFAT5 was mainly expressed in corneal fibroblasts. In contrast, after PCI, NFAT5 expression was highly upregulated in recruited corneal macrophages. NFAT5 deficiency did not alter corneal thickness in steady state; however, loss of NFAT5 led to accelerated resorption of corneal edema after PCI. Mechanistically, we found that myeloid cell-derived NFAT5 is crucial for controlling corneal edema, as edema resorption after PCI was significantly enhanced in mice with conditional loss of NFAT5 in the myeloid cell lineage, presumably due to increased pinocytosis of corneal macrophages. Collectively, we uncovered a suppressive role for NFAT5 in corneal edema resorption, thereby identifying a novel therapeutic target to combat edema-induced corneal blindness.
Collapse
|
14
|
Insights of Endocytosis Signaling in Health and Disease. Int J Mol Sci 2023; 24:ijms24032971. [PMID: 36769293 PMCID: PMC9918140 DOI: 10.3390/ijms24032971] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/20/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Endocytosis in mammalian cells is a fundamental cellular machinery that regulates vital physiological processes, such as the absorption of metabolites, release of neurotransmitters, uptake of hormone cellular defense, and delivery of biomolecules across the plasma membrane. A remarkable characteristic of the endocytic machinery is the sequential assembly of the complex proteins at the plasma membrane, followed by internalization and fusion of various biomolecules to different cellular compartments. In all eukaryotic cells, functional characterization of endocytic pathways is based on dynamics of the protein complex and signal transduction modules. To coordinate the assembly and functions of the numerous parts of the endocytic machinery, the endocytic proteins interact significantly within and between the modules. Clathrin-dependent and -independent endocytosis, caveolar pathway, and receptor mediated endocytosis have been attributed to a greater variety of physiological and pathophysiological roles such as, autophagy, metabolism, cell division, apoptosis, cellular defense, and intestinal permeabilization. Notably, any defect or alteration in the endocytic machinery results in the development of pathological consequences associated with human diseases such as cancer, cardiovascular diseases, neurological diseases, and inflammatory diseases. In this review, an in-depth endeavor has been made to illustrate the process of endocytosis, and associated mechanisms describing pathological manifestation associated with dysregulated endocytosis machinery.
Collapse
|
15
|
Santos CIM, Cicuéndez M, Gonçalves G, Rodríguez-Pérez L, Portolés MT, Faustino MAF, Herranz MÁ, Neves MGPMS, Martinho JMG, Maçôas EMS, Martín N. Safety assessment of new nanodiamonds@corrole hybrids addressed by the response of RAW-264.7 macrophages. J Mater Chem B 2023; 11:675-686. [PMID: 36562480 DOI: 10.1039/d2tb01863b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Safety assessment of carbon nanomaterials is of paramount importance since they are on the frontline for applications in sensing, bioimaging and drug delivery. The biocompatibility and safety of functionalized nanodiamonds (NDs) are here addressed through the study of the pro-inflammatory response of RAW-264.7 macrophages exposed to new nanodiamonds@corrole hybrids. The corrole unit selected is as a prototype for a hydrophobic organic molecule that can function as a NIR fluorophore reporter, an optical sensor, a photodynamic therapy agent or a photocatalyst. The new functional nanohybrids containing detonated nanodiamonds (NDs) were obtained through esterification using carboxylated NDs and glycol corroles. The success of the covalent functionalization via carbodiimide activation was confirmed through X-ray photoelectron spectroscopy (XPS), Raman and Fourier transform infrared (FTIR) spectroscopy. The UV-vis absorption and emission spectra of the hybrids are additive with respect to the corrole features. The cellular uptake, localization, cell viability and effects on immune cell activation of the new hybrids and of the precursors were carefully investigated using RAW-264.7 macrophages. Overall results showed that the ND@corrole hybrids had no pro-inflammatory effects on the RAW-264.7 macrophage cell line, making them an ideal candidate for a wide range of biomedical applications.
Collapse
Affiliation(s)
- Carla I M Santos
- CQE, Centro de Química Estrutural, Instituto Superior Técnico, Av. Rovisco Pais, 1049-001 Lisboa, Portugal.,LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Mónica Cicuéndez
- Chemistry Department, Faculty of Pharmacy, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), E-28040, Madrid, Spain
| | - Gil Gonçalves
- TEMA-Nanotechnology Research Group, Mechanical Engineering Department, University of Aveiro, Campus Universitario de Santiago, 3810-193 Aveiro, Portugal.,Intelligent Systems Associate Laboratory (LASI), Portugal
| | - Laura Rodríguez-Pérez
- Department of Organic Chemistry, Faculty of Chemistry, Universidad Complutense de Madrid, E-28040 Madrid, Spain.
| | - M Teresa Portolés
- Department of Biochemistry and Molecular Biology, Faculty of Chemistry, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain, E-28040 Madrid, Spain.,CIBER de Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, ISCIII, 28040 Madrid, Spain
| | - M Amparo F Faustino
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - M Ángeles Herranz
- Department of Organic Chemistry, Faculty of Chemistry, Universidad Complutense de Madrid, E-28040 Madrid, Spain.
| | - M Graça P M S Neves
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - José M G Martinho
- CQE, Centro de Química Estrutural, Instituto Superior Técnico, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Ermelinda M S Maçôas
- CQE, Centro de Química Estrutural, Instituto Superior Técnico, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Nazario Martín
- Department of Organic Chemistry, Faculty of Chemistry, Universidad Complutense de Madrid, E-28040 Madrid, Spain.
| |
Collapse
|
16
|
Maekawa M, Natsume R, Arita M. Functional significance of ion channels during macropinosome resolution in immune cells. Front Physiol 2022; 13:1037758. [DOI: 10.3389/fphys.2022.1037758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/10/2022] [Indexed: 11/13/2022] Open
Abstract
Macropinocytosis is a unique type of endocytosis accompanied by membrane ruffle formation. Closure of membrane ruffles leads to the uptake of large volumes of fluid phase and, subsequently, the formation of large vacuoles termed macropinosomes. Immune cells, such as dendritic cells, T cells, and macrophages, endocytose the surrounding amino acids and pathogens via macropinocytosis either constitutively or in a stimulus-dependent fashion. This process is critical for cell migration, mammalian target of rapamycin complex 1 (mTORC1) activation, and antigen presentation. Large vacuoles are fragmented into tubules and smaller vesicles during the progression and maturation of macropinosomes in immune cells. This process is called “macropinosome resolution” and requires osmotically driven shrinkage of macropinosomes, which is controlled by ion channels present in them. The crenation of membranes on shrunken macropinosomes is recognized by curvature-sensing proteins and results in intracellular membrane trafficking. In this mini review, we highlight the recent progress in research on macropinosome resolution in macrophages, with a focus on ion channels (TPC1/2 for Na+ and TMEM206 for Cl−) that is required for macropinosome resolution. We also discuss the potential contribution of membrane lipids to this process.
Collapse
|
17
|
Odfalk KF, Bieniek KF, Hopp SC. Microglia: Friend and foe in tauopathy. Prog Neurobiol 2022; 216:102306. [PMID: 35714860 PMCID: PMC9378545 DOI: 10.1016/j.pneurobio.2022.102306] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 04/24/2022] [Accepted: 06/10/2022] [Indexed: 12/16/2022]
Abstract
Aggregation of misfolded microtubule associated protein tau into abnormal intracellular inclusions defines a class of neurodegenerative diseases known as tauopathies. The consistent spatiotemporal progression of tau pathology in Alzheimer's disease (AD) led to the hypothesis that tau aggregates spread in the brain via bioactive tau "seeds" underlying advancing disease course. Recent studies implicate microglia, the resident immune cells of the central nervous system, in both negative and positive regulation of tau pathology. Polymorphisms in genes that alter microglial function are associated with the development of AD and other tauopathies. Experimental manipulation of microglia function can alter tau pathology and microglia-mediated neuroinflammatory cascades can exacerbate tau pathology. Microglia also exert protective functions by mitigating tau spread: microglia internalize tau seeds and have the capacity to degrade them. However, when microglia fail to degrade these tau seeds there are deleterious consequences, including secretion of exosomes containing tau that can spread to neurons. This review explores the intersection of microglia and tau from the perspective of neuropathology, neuroimaging, genetics, transcriptomics, and molecular biology. As tau-targeted therapies such as anti-tau antibodies advance through clinical trials, it is critical to understand the interaction between tau and microglia.
Collapse
Affiliation(s)
- Kristian F Odfalk
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA; Department of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Kevin F Bieniek
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA; Department of Pathology and Laboratory Medicine, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Sarah C Hopp
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA; Department of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, TX, USA.
| |
Collapse
|
18
|
Griffiths G, Gruenberg J, Marsh M, Wohlmann J, Jones AT, Parton RG. Nanoparticle entry into cells; the cell biology weak link. Adv Drug Deliv Rev 2022; 188:114403. [PMID: 35777667 DOI: 10.1016/j.addr.2022.114403] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 12/22/2022]
Abstract
Nanoparticles (NP) are attractive options for the therapeutic delivery of active pharmaceutical drugs, proteins and nucleic acids into cells, tissues and organs. Research into the development and application of NP most often starts with a diverse group of scientists, including chemists, bioengineers and material and pharmaceutical scientists, who design, fabricate and characterize NP in vitro (Stage 1). The next step (Stage 2) generally investigates cell toxicity as well as the processes by which NP bind, are internalized and deliver their cargo to appropriate model tissue culture cells. Subsequently, in Stage 3, selected NP are tested in animal systems, mostly mouse. Whereas the chemistry-based development and analysis in Stage 1 is increasingly sophisticated, the investigations in Stage 2 are not what could be regarded as 'state-of-the-art' for the cell biology field and the quality of research into NP interactions with cells is often sub-standard. In this review we describe our current understanding of the mechanisms by which particles gain entry into mammalian cells via endocytosis. We summarize the most important areas for concern, highlight some of the most common mis-conceptions, and identify areas where NP scientists could engage with trained cell biologists. Our survey of the different mechanisms of uptake into cells makes us suspect that claims for roles for caveolae, as well as macropinocytosis, in NP uptake into cells have been exaggerated, whereas phagocytosis has been under-appreciated.
Collapse
Affiliation(s)
- Gareth Griffiths
- Department Biosciences, University of Oslo, Blindernveien 31, PO Box 1041, 0316 Oslo, Norway.
| | - Jean Gruenberg
- Department of Biochemistry, University of Geneva, 30 quai E. Ansermet, 1211-Geneva-4, Switzerland
| | - Mark Marsh
- Laboratory for Molecular Cell Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Jens Wohlmann
- Department Biosciences, University of Oslo, Blindernveien 31, PO Box 1041, 0316 Oslo, Norway
| | - Arwyn T Jones
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, Cardiff, Wales CF103NB, UK
| | - Robert G Parton
- Institute for Molecular Bioscience and Centre for Microscopy and Microanalysis, The University of Queensland, Qld 4072, Australia
| |
Collapse
|
19
|
Miyazaki T. Pinocytotic engulfment of lipoproteins by macrophages. Front Cardiovasc Med 2022; 9:957897. [PMID: 36105534 PMCID: PMC9464914 DOI: 10.3389/fcvm.2022.957897] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/11/2022] [Indexed: 11/22/2022] Open
Abstract
Atherosclerosis is a major cause of acute coronary syndrome and stroke. Foam cell formation in macrophages is involved in controlling plaque stability and the pathogenesis of atherosclerosis. Accordingly, many studies have examined the processes of lipid incorporation, such as scavenger receptor-mediated uptake of oxidized low-density lipoprotein, in cells. In addition to receptor-mediated machinery, growing evidence has suggested that pinocytosis, which is a receptor-independent endocytic pathway, is associated with foam cell formation when a sufficient number of lipoproteins is accumulated around cells. Pinocytotic engulfment of nanoparticles is initiated by plasma membrane ruffling in a phosphatidylinositol-3 kinase-dependent manner. Subsequent to pinosome closure, the majority of pinosomes are internalized through endocytic processes, and they can be recycled into the plasma membrane. These pinocytotic processes are modulated by small GTPases and their cytoskeletal rearrangement. Moreover, pinocytotic abilities may vary between immunological subsets in cells. Accordingly, macrophages may show diverse pinocytotic abilities depending on the surrounding microenvironment. This review summarizes the current understanding of pinocytotic engulfment of lipoprotein in macrophages, and discusses how this endocytic process is governed under hypercholesterolemic conditions.
Collapse
|
20
|
The Delivery of mRNA Vaccines for Therapeutics. LIFE (BASEL, SWITZERLAND) 2022; 12:life12081254. [PMID: 36013433 PMCID: PMC9410089 DOI: 10.3390/life12081254] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/05/2022] [Accepted: 08/15/2022] [Indexed: 12/12/2022]
Abstract
mRNA vaccines have been revolutionary in combating the COVID-19 pandemic in the past two years. They have also become a versatile tool for the prevention of infectious diseases and treatment of cancers. For effective vaccination, mRNA formulation, delivery method and composition of the mRNA carrier play an important role. mRNA vaccines can be delivered using lipid nanoparticles, polymers, peptides or naked mRNA. The vaccine efficacy is influenced by the appropriate delivery materials, formulation methods and selection of a proper administration route. In addition, co-delivery of several mRNAs could also be beneficial and enhance immunity against various variants of an infectious pathogen or several pathogens altogether. Here, we review the recent progress in the delivery methods, modes of delivery and patentable mRNA vaccine technologies.
Collapse
|
21
|
Gundu C, Arruri VK, Yadav P, Navik U, Kumar A, Amalkar VS, Vikram A, Gaddam RR. Dynamin-Independent Mechanisms of Endocytosis and Receptor Trafficking. Cells 2022; 11:cells11162557. [PMID: 36010634 PMCID: PMC9406725 DOI: 10.3390/cells11162557] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/03/2022] [Accepted: 08/13/2022] [Indexed: 11/16/2022] Open
Abstract
Endocytosis is a fundamental mechanism by which cells perform housekeeping functions. It occurs via a variety of mechanisms and involves many regulatory proteins. The GTPase dynamin acts as a “molecular scissor” to form endocytic vesicles and is a critical regulator among the proteins involved in endocytosis. Some GTPases (e.g., Cdc42, arf6, RhoA), membrane proteins (e.g., flotillins, tetraspanins), and secondary messengers (e.g., calcium) mediate dynamin-independent endocytosis. These pathways may be convergent, as multiple pathways exist in a single cell. However, what determines the specific path of endocytosis is complex and challenging to comprehend. This review summarizes the mechanisms of dynamin-independent endocytosis, the involvement of microRNAs, and factors that contribute to the cellular decision about the specific route of endocytosis.
Collapse
Affiliation(s)
- Chayanika Gundu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, Telangana, India
| | - Vijay Kumar Arruri
- Department of Neurological Surgery, University of Wisconsin, Madison, WI 53792, USA
| | - Poonam Yadav
- Department of Pharmacology, Central University of Punjab, Bathinda 151001, Punjab, India
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Bathinda 151001, Punjab, India
| | - Ashutosh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata 700054, West Bengal, India
| | - Veda Sudhir Amalkar
- Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, IA 52242, USA
| | - Ajit Vikram
- Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, IA 52242, USA
| | - Ravinder Reddy Gaddam
- Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, IA 52242, USA
- Correspondence:
| |
Collapse
|
22
|
Qiu Z, Liu W, Zhu Q, Ke K, Zhu Q, Jin W, Yu S, Yang Z, Li L, Sun X, Ren S, Liu Y, Zhu Z, Zeng J, Huang X, Huang Y, Wei L, Ma M, Lu J, Chen X, Mou Y, Xie T, Sui X. The Role and Therapeutic Potential of Macropinocytosis in Cancer. Front Pharmacol 2022; 13:919819. [PMID: 36046825 PMCID: PMC9421435 DOI: 10.3389/fphar.2022.919819] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/17/2022] [Indexed: 11/20/2022] Open
Abstract
Macropinocytosis, a unique endocytosis pathway characterized by nonspecific internalization, has a vital role in the uptake of extracellular substances and antigen presentation. It is known to have dual effects on cancer cells, depending on cancer type and certain microenvironmental conditions. It helps cancer cells survive in nutrient-deficient environments, enhances resistance to anticancer drugs, and promotes invasion and metastasis. Conversely, overexpression of the RAS gene alongside drug treatment can lead to methuosis, a novel mode of cell death. The survival and proliferation of cancer cells is closely related to macropinocytosis in the tumor microenvironment (TME), but identifying how these cells interface with the TME is crucial for creating drugs that can limit cancer progression and metastasis. Substantial progress has been made in recent years on designing anticancer therapies that utilize the effects of macropinocytosis. Both the induction and inhibition of macropinocytosis are useful strategies for combating cancer cells. This article systematically reviews the general mechanisms of macropinocytosis, its specific functions in tumor cells, its occurrence in nontumor cells in the TME, and its application in tumor therapies. The aim is to elucidate the role and therapeutic potential of macropinocytosis in cancer treatment.
Collapse
Affiliation(s)
- Zejing Qiu
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Wencheng Liu
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Qianru Zhu
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Kun Ke
- Department of Gastrointestinal-Pancreatic Surgery, General Surgery, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Qicong Zhu
- Department of Gastrointestinal-Pancreatic Surgery, General Surgery, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Weiwei Jin
- Department of Gastrointestinal-Pancreatic Surgery, General Surgery, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Shuxian Yu
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Zuyi Yang
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Lin Li
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Xiaochen Sun
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Shuyi Ren
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Yanfen Liu
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Zhiyu Zhu
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Jiangping Zeng
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Xiaoyu Huang
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Yan Huang
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Lu Wei
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Mengmeng Ma
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Jun Lu
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Xiaoyang Chen
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Yiping Mou
- Department of Gastrointestinal-Pancreatic Surgery, General Surgery, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
- *Correspondence: Yiping Mou, ; Tian Xie, ; Xinbing Sui,
| | - Tian Xie
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
- *Correspondence: Yiping Mou, ; Tian Xie, ; Xinbing Sui,
| | - Xinbing Sui
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
- *Correspondence: Yiping Mou, ; Tian Xie, ; Xinbing Sui,
| |
Collapse
|
23
|
Xiang P, Blanchard V, Francis GA. Smooth Muscle Cell—Macrophage Interactions Leading to Foam Cell Formation in Atherosclerosis: Location, Location, Location. Front Physiol 2022; 13:921597. [PMID: 35795646 PMCID: PMC9251363 DOI: 10.3389/fphys.2022.921597] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Cholesterol-overloaded cells or “foam cells” in the artery wall are the biochemical hallmark of atherosclerosis, and are responsible for much of the growth, inflammation and susceptibility to rupture of atherosclerotic lesions. While it has previously been thought that macrophages are the main contributor to the foam cell population, recent evidence indicates arterial smooth muscle cells (SMCs) are the source of the majority of foam cells in both human and murine atherosclerosis. This review outlines the timeline, site of appearance and proximity of SMCs and macrophages with lipids in human and mouse atherosclerosis, and likely interactions between SMCs and macrophages that promote foam cell formation and removal by both cell types. An understanding of these SMC-macrophage interactions in foam cell formation and regression is expected to provide new therapeutic targets to reduce the burden of atherosclerosis for the prevention of coronary heart disease, stroke and peripheral vascular disease.
Collapse
|
24
|
Delgado MG, Rivera CA, Lennon-Duménil AM. Macropinocytosis and Cell Migration: Don't Drink and Drive…. Subcell Biochem 2022; 98:85-102. [PMID: 35378704 DOI: 10.1007/978-3-030-94004-1_5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Macropinocytosis is a nonspecific mechanism by which cells compulsively "drink" the surrounding extracellular fluids in order to feed themselves or sample the molecules therein, hence gaining information about their environment. This process is cell-intrinsically incompatible with the migration of many cells, implying that the two functions are antagonistic. The migrating cell uses a molecular switch to stop and explore its surrounding fluid by macropinocytosis, after which it employs the same molecular machinery to start migrating again to examine another location. This cycle of migration/macropinocytosis allows cells to explore tissues, and it is key to a range of physiological processes. Evidence of this evolutionarily conserved antagonism between the two processes can be found in several cell types-immune cells, for example, being particularly adept-and ancient organisms (e.g., the social amoeba Dictyostelium discoideum). How macropinocytosis and migration are negatively coupled is the subject of this chapter.
Collapse
|
25
|
Abstract
Phagocytes play critical roles in the maintenance of organismal homeostasis and immunity. Central to their role is their ability to take up and process exogenous material via the related processes of phagocytosis and macropinocytosis. The mechanisms and functions underlying macropinocytosis have remained severely understudied relative to phagocytosis. In recent years, however, there has been a renaissance in macropinocytosis research. Phagocytes can engage in various forms of macropinocytosis including an "induced" form and a "constitutive" form. This chapter, however, will focus on constitutive macropinocytosis and its role in the maintenance of immunity. Functions previously attributed to macropinocytosis, including antigen presentation and immune surveillance, will be revisited in light of recent revelations and emerging concepts will be highlighted.
Collapse
Affiliation(s)
- Johnathan Canton
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
26
|
Means N, Elechalawar CK, Chen WR, Bhattacharya R, Mukherjee P. Revealing macropinocytosis using nanoparticles. Mol Aspects Med 2022; 83:100993. [PMID: 34281720 PMCID: PMC8761201 DOI: 10.1016/j.mam.2021.100993] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/12/2021] [Indexed: 02/03/2023]
Abstract
Endocytosis mechanisms are one of the methods that cells use to interact with their environments. Endocytosis mechanisms vary from the clathrin-mediated endocytosis to the receptor independent macropinocytosis. Macropinocytosis is a niche of endocytosis that is quickly becoming more relevant in various fields of research since its discovery in the 1930s. Macropinocytosis has several distinguishing factors from other receptor-mediated forms of endocytosis, including: types of extracellular material for uptake, signaling cascade, and niche uses between cell types. Nanoparticles (NPs) are an important tool for various applications, including drug delivery and disease treatment. However, surface engineering of NPs could be tailored to target them inside the cells exploiting different endocytosis pathways, such as endocytosis versus macropinocytosis. Such surface engineering of NPs mainly, size, charge, shape and the core material will allow identification of new adapter molecules regulating different endocytosis process and provide further insight into how cells tweak these pathways to meet their physiological need. In this review, we focus on the description of macropinocytosis, a lesser studied endocytosis mechanism than the conventional receptor mediated endocytosis. Additionally, we will discuss nanoparticle endocytosis (including macropinocytosis), and how the physio-chemical properties of the NP (size, charge, and surface coating) affect their intracellular uptake and exploiting them as tools to identify new adapter molecules regulating these processes.
Collapse
Affiliation(s)
- Nicolas Means
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | - Wei R Chen
- Stephenson School of Biomedical Engineering, Gallogly College of Engineering, University of Oklahoma, Norman, OK, 73019, USA
| | - Resham Bhattacharya
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Priyabrata Mukherjee
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
27
|
Antonioli L, Pacher P, Haskó G. Adenosine and inflammation: it's time to (re)solve the problem. Trends Pharmacol Sci 2022; 43:43-55. [PMID: 34776241 DOI: 10.1016/j.tips.2021.10.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/14/2021] [Accepted: 10/20/2021] [Indexed: 02/07/2023]
Abstract
Resolution of inflammation requires proresolving molecular pathways triggered as part of the host response during the inflammatory phase. Adenosine and its receptors, which are collectively called the adenosine system, shape inflammatory cell activity during the active phase of inflammation, leading these immune cells toward a functional repolarization, thus contributing to the onset of resolution. Strategies based on the resolution of inflammation have shaped a new area of pharmacology referred to as 'resolution pharmacology' and in this regard, the adenosine system represents an interesting target to design novel pharmacological tools to 'resolve' the inflammatory process. In this review, we outline the role of the adenosine system in driving the events required for an effective transition from the proinflammatory phase to the onset and establishment of resolution.
Collapse
Affiliation(s)
- Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Pál Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health/NIAAA, Bethesda, MD 20892, USA
| | - György Haskó
- Department of Anesthesiology, Columbia University, New York, NY, 10032, USA.
| |
Collapse
|
28
|
Zeng C, Zhang C, Walker PG, Dong Y. Formulation and Delivery Technologies for mRNA Vaccines. Curr Top Microbiol Immunol 2022; 440:71-110. [PMID: 32483657 PMCID: PMC8195316 DOI: 10.1007/82_2020_217] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
mRNA vaccines have become a versatile technology for the prevention of infectious diseases and the treatment of cancers. In the vaccination process, mRNA formulation and delivery strategies facilitate effective expression and presentation of antigens, and immune stimulation. mRNA vaccines have been delivered in various formats: encapsulation by delivery carriers, such as lipid nanoparticles, polymers, peptides, free mRNA in solution, and ex vivo through dendritic cells. Appropriate delivery materials and formulation methods often boost the vaccine efficacy which is also influenced by the selection of a proper administration route. Co-delivery of multiple mRNAs enables synergistic effects and further enhances immunity in some cases. In this chapter, we overview the recent progress and existing challenges in the formulation and delivery technologies of mRNA vaccines with perspectives for future development.
Collapse
Affiliation(s)
- Chunxi Zeng
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, 43210, Columbus, OH, USA
| | - Chengxiang Zhang
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, 43210, Columbus, OH, USA
| | - Patrick G Walker
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Yizhou Dong
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, 43210, Columbus, OH, USA. .,The Center for Clinical and Translational Science, The Ohio State University, Columbus, OH, 43210, USA. .,The Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA. .,Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University, 43210, Columbus, OH, USA. .,Department of Radiation Oncology, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
29
|
Porges E, Jenner D, Taylor AW, Harrison JS, De Grazia A, Hailes AR, Wright KM, Whelan AO, Norville IH, Prior JL, Mahajan S, Rowland CA, Newman TA, Evans ND. Antibiotic-Loaded Polymersomes for Clearance of Intracellular Burkholderia thailandensis. ACS NANO 2021; 15:19284-19297. [PMID: 34739227 PMCID: PMC7612142 DOI: 10.1021/acsnano.1c05309] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Melioidosis caused by the facultative intracellular pathogen Burkholderia pseudomallei is difficult to treat due to poor intracellular bioavailability of antibiotics and antibiotic resistance. In the absence of novel compounds, polymersome (PM) encapsulation may increase the efficacy of existing antibiotics and reduce antibiotic resistance by promoting targeted, infection-specific intracellular uptake. In this study, we developed PMs composed of widely available poly(ethylene oxide)-polycaprolactone block copolymers and demonstrated their delivery to intracellular B. thailandensis infection using multispectral imaging flow cytometry (IFC) and coherent anti-Stokes Raman scattering microscopy. Antibiotics were tightly sequestered in PMs and did not inhibit the growth of free-living B. thailandensis. However, on uptake of antibiotic-loaded PMs by infected macrophages, IFC demonstrated PM colocalization with intracellular B. thailandensis and a significant inhibition of their growth. We conclude that PMs are a viable approach for the targeted antibiotic treatment of persistent intracellular Burkholderia infection.
Collapse
Affiliation(s)
- Eleanor Porges
- Bioengineering Sciences Group, Faculty of Engineering and the Environment, University of Southampton, Highfield, Southampton, SO17 1BJ, United Kingdom
- Centre for Human Development, Stem Cells and Regeneration, Bone and Joint Research Group, University of Southampton Faculty of Medicine, Southampton, SO16 6YD,United Kingdom
- Clinical and Experimental Sciences, Faculty of Medicine, Institute for Life Sciences, University of Southampton, Highfield, Southampton, SO17 1BJ, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom
| | - Dominic Jenner
- Defence Science and Technology Laboratory, Chemical, Biological and Radiological Division, Porton Down, Salisbury, SP4 0JQ, United Kingdom
| | - Adam W. Taylor
- Defence Science and Technology Laboratory, Chemical, Biological and Radiological Division, Porton Down, Salisbury, SP4 0JQ, United Kingdom
- London School of Hygiene and Tropical Medicine, London, WC1E 7HT, United Kingdom
| | - James S.P. Harrison
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom
- School of Chemistry, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom
| | - Antonio De Grazia
- Bioengineering Sciences Group, Faculty of Engineering and the Environment, University of Southampton, Highfield, Southampton, SO17 1BJ, United Kingdom
- Clinical and Experimental Sciences, Faculty of Medicine, Institute for Life Sciences, University of Southampton, Highfield, Southampton, SO17 1BJ, United Kingdom
| | - Alethia R. Hailes
- Bioengineering Sciences Group, Faculty of Engineering and the Environment, University of Southampton, Highfield, Southampton, SO17 1BJ, United Kingdom
- Centre for Human Development, Stem Cells and Regeneration, Bone and Joint Research Group, University of Southampton Faculty of Medicine, Southampton, SO16 6YD,United Kingdom
- Clinical and Experimental Sciences, Faculty of Medicine, Institute for Life Sciences, University of Southampton, Highfield, Southampton, SO17 1BJ, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom
| | - Kimberley M. Wright
- Defence Science and Technology Laboratory, Chemical, Biological and Radiological Division, Porton Down, Salisbury, SP4 0JQ, United Kingdom
| | - Adam O. Whelan
- Defence Science and Technology Laboratory, Chemical, Biological and Radiological Division, Porton Down, Salisbury, SP4 0JQ, United Kingdom
| | - Isobel H. Norville
- Defence Science and Technology Laboratory, Chemical, Biological and Radiological Division, Porton Down, Salisbury, SP4 0JQ, United Kingdom
| | - Joann L. Prior
- Defence Science and Technology Laboratory, Chemical, Biological and Radiological Division, Porton Down, Salisbury, SP4 0JQ, United Kingdom
| | - Sumeet Mahajan
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom
- School of Chemistry, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom
| | - Caroline A. Rowland
- Defence Science and Technology Laboratory, Chemical, Biological and Radiological Division, Porton Down, Salisbury, SP4 0JQ, United Kingdom
| | - Tracey A. Newman
- Clinical and Experimental Sciences, Faculty of Medicine, Institute for Life Sciences, University of Southampton, Highfield, Southampton, SO17 1BJ, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom
| | - Nicholas D. Evans
- Bioengineering Sciences Group, Faculty of Engineering and the Environment, University of Southampton, Highfield, Southampton, SO17 1BJ, United Kingdom
- Centre for Human Development, Stem Cells and Regeneration, Bone and Joint Research Group, University of Southampton Faculty of Medicine, Southampton, SO16 6YD,United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom
| |
Collapse
|
30
|
Li A, Guo F, Pan Q, Chen S, Chen J, Liu HF, Pan Q. Mesenchymal Stem Cell Therapy: Hope for Patients With Systemic Lupus Erythematosus. Front Immunol 2021; 12:728190. [PMID: 34659214 PMCID: PMC8516390 DOI: 10.3389/fimmu.2021.728190] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 09/14/2021] [Indexed: 12/26/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease. Although previous studies have demonstrated that SLE is related to the imbalance of cells in the immune system, including B cells, T cells, and dendritic cells, etc., the mechanisms underlying SLE pathogenesis remain unclear. Therefore, effective and low side-effect therapies for SLE are lacking. Recently, mesenchymal stem cell (MSC) therapy for autoimmune diseases, particularly SLE, has gained increasing attention. This therapy can improve the signs and symptoms of refractory SLE by promoting the proliferation of Th2 and Treg cells and inhibiting the activity of Th1, Th17, and B cells, etc. However, MSC therapy is also reported ineffective in some patients with SLE, which may be related to MSC- or patient-derived factors. Therefore, the therapeutic effects of MSCs should be further confirmed. This review summarizes the status of MSC therapy in refractory SLE treatment and potential reasons for the ineffectiveness of MSC therapy from three perspectives. We propose various MSC modification methods that may be beneficial in enhancing the immunosuppression of MSCs in SLE. However, their safety and protective effects in patients with SLE still need to be confirmed by further experimental and clinical evidence.
Collapse
Affiliation(s)
- Aifen Li
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Fengbiao Guo
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Quanren Pan
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Shuxian Chen
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jiaxuan Chen
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Hua-Feng Liu
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Qingjun Pan
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
31
|
Quinn SE, Huang L, Kerkvliet JG, Swanson JA, Smith S, Hoppe AD, Anderson RB, Thiex NW, Scott BL. The structural dynamics of macropinosome formation and PI3-kinase-mediated sealing revealed by lattice light sheet microscopy. Nat Commun 2021. [PMID: 34376698 DOI: 10.1101/2020.12.01.390195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2023] Open
Abstract
Macropinosomes are formed by shaping actin-rich plasma membrane ruffles into large intracellular organelles in a phosphatidylinositol 3-kinase (PI3K)-coordinated manner. Here, we utilize lattice lightsheet microscopy and image visualization methods to map the three-dimensional structure and dynamics of macropinosome formation relative to PI3K activity. We show that multiple ruffling morphologies produce macropinosomes and that the majority form through collisions of adjacent PI3K-rich ruffles. By combining multiple volumetric representations of the plasma membrane structure and PI3K products, we show that PI3K activity begins early throughout the entire ruffle volume and continues to increase until peak activity concentrates at the base of the ruffle after the macropinosome closes. Additionally, areas of the plasma membrane rich in ruffling had increased PI3K activity and produced many macropinosomes of various sizes. Pharmacologic inhibition of PI3K activity had little effect on the rate and morphology of membrane ruffling, demonstrating that early production of 3'-phosphoinositides within ruffles plays a minor role in regulating their morphology. However, 3'-phosphoinositides are critical for the fusogenic activity that seals ruffles into macropinosomes. Taken together, these data indicate that local PI3K activity is amplified in ruffles and serves as a priming mechanism for closure and sealing of ruffles into macropinosomes.
Collapse
Affiliation(s)
- Shayne E Quinn
- Nanoscience and Nanoengineering, South Dakota School of Mines and Technology (South Dakota Mines), Rapid City, SD, USA
- BioSNTR, South Dakota Mines, Rapid City, SD, USA
| | - Lu Huang
- Department of Biology and Microbiology, South Dakota State University (SDSU), Brookings, SD, USA
- BioSNTR, SDSU, Brookings, SD, USA
| | - Jason G Kerkvliet
- BioSNTR, SDSU, Brookings, SD, USA
- Department of Chemistry and Biochemistry, SDSU, Brookings, SD, USA
| | - Joel A Swanson
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Steve Smith
- Nanoscience and Nanoengineering, South Dakota School of Mines and Technology (South Dakota Mines), Rapid City, SD, USA
- BioSNTR, South Dakota Mines, Rapid City, SD, USA
| | - Adam D Hoppe
- BioSNTR, SDSU, Brookings, SD, USA
- Department of Chemistry and Biochemistry, SDSU, Brookings, SD, USA
| | - Robert B Anderson
- Nanoscience and Nanoengineering, South Dakota School of Mines and Technology (South Dakota Mines), Rapid City, SD, USA.
- BioSNTR, South Dakota Mines, Rapid City, SD, USA.
| | - Natalie W Thiex
- Department of Biology and Microbiology, South Dakota State University (SDSU), Brookings, SD, USA.
- BioSNTR, SDSU, Brookings, SD, USA.
| | - Brandon L Scott
- Nanoscience and Nanoengineering, South Dakota School of Mines and Technology (South Dakota Mines), Rapid City, SD, USA.
- BioSNTR, South Dakota Mines, Rapid City, SD, USA.
| |
Collapse
|
32
|
Quinn SE, Huang L, Kerkvliet JG, Swanson JA, Smith S, Hoppe AD, Anderson RB, Thiex NW, Scott BL. The structural dynamics of macropinosome formation and PI3-kinase-mediated sealing revealed by lattice light sheet microscopy. Nat Commun 2021; 12:4838. [PMID: 34376698 PMCID: PMC8355319 DOI: 10.1038/s41467-021-25187-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 07/22/2021] [Indexed: 12/11/2022] Open
Abstract
Macropinosomes are formed by shaping actin-rich plasma membrane ruffles into large intracellular organelles in a phosphatidylinositol 3-kinase (PI3K)-coordinated manner. Here, we utilize lattice lightsheet microscopy and image visualization methods to map the three-dimensional structure and dynamics of macropinosome formation relative to PI3K activity. We show that multiple ruffling morphologies produce macropinosomes and that the majority form through collisions of adjacent PI3K-rich ruffles. By combining multiple volumetric representations of the plasma membrane structure and PI3K products, we show that PI3K activity begins early throughout the entire ruffle volume and continues to increase until peak activity concentrates at the base of the ruffle after the macropinosome closes. Additionally, areas of the plasma membrane rich in ruffling had increased PI3K activity and produced many macropinosomes of various sizes. Pharmacologic inhibition of PI3K activity had little effect on the rate and morphology of membrane ruffling, demonstrating that early production of 3'-phosphoinositides within ruffles plays a minor role in regulating their morphology. However, 3'-phosphoinositides are critical for the fusogenic activity that seals ruffles into macropinosomes. Taken together, these data indicate that local PI3K activity is amplified in ruffles and serves as a priming mechanism for closure and sealing of ruffles into macropinosomes.
Collapse
Affiliation(s)
- Shayne E Quinn
- Nanoscience and Nanoengineering, South Dakota School of Mines and Technology (South Dakota Mines), Rapid City, SD, USA
- BioSNTR, South Dakota Mines, Rapid City, SD, USA
| | - Lu Huang
- Department of Biology and Microbiology, South Dakota State University (SDSU), Brookings, SD, USA
- BioSNTR, SDSU, Brookings, SD, USA
| | - Jason G Kerkvliet
- BioSNTR, SDSU, Brookings, SD, USA
- Department of Chemistry and Biochemistry, SDSU, Brookings, SD, USA
| | - Joel A Swanson
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Steve Smith
- Nanoscience and Nanoengineering, South Dakota School of Mines and Technology (South Dakota Mines), Rapid City, SD, USA
- BioSNTR, South Dakota Mines, Rapid City, SD, USA
| | - Adam D Hoppe
- BioSNTR, SDSU, Brookings, SD, USA
- Department of Chemistry and Biochemistry, SDSU, Brookings, SD, USA
| | - Robert B Anderson
- Nanoscience and Nanoengineering, South Dakota School of Mines and Technology (South Dakota Mines), Rapid City, SD, USA.
- BioSNTR, South Dakota Mines, Rapid City, SD, USA.
| | - Natalie W Thiex
- Department of Biology and Microbiology, South Dakota State University (SDSU), Brookings, SD, USA.
- BioSNTR, SDSU, Brookings, SD, USA.
| | - Brandon L Scott
- Nanoscience and Nanoengineering, South Dakota School of Mines and Technology (South Dakota Mines), Rapid City, SD, USA.
- BioSNTR, South Dakota Mines, Rapid City, SD, USA.
| |
Collapse
|
33
|
ARHGEF26 enhances Salmonella invasion and inflammation in cells and mice. PLoS Pathog 2021; 17:e1009713. [PMID: 34242364 PMCID: PMC8294491 DOI: 10.1371/journal.ppat.1009713] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 07/21/2021] [Accepted: 06/14/2021] [Indexed: 11/25/2022] Open
Abstract
Salmonella hijack host machinery in order to invade cells and establish infection. While considerable work has described the role of host proteins in invasion, much less is known regarding how natural variation in these invasion-associated host proteins affects Salmonella pathogenesis. Here we leveraged a candidate cellular GWAS screen to identify natural genetic variation in the ARHGEF26 (Rho Guanine Nucleotide Exchange Factor 26) gene that renders lymphoblastoid cells susceptible to Salmonella Typhi and Typhimurium invasion. Experimental follow-up redefined ARHGEF26’s role in Salmonella epithelial cell infection. Specifically, we identified complex serovar-by-host interactions whereby ARHGEF26 stimulation of S. Typhi and S. Typhimurium invasion into host cells varied in magnitude and effector-dependence based on host cell type. While ARHGEF26 regulated SopB- and SopE-mediated S. Typhi (but not S. Typhimurium) infection of HeLa cells, the largest effect of ARHGEF26 was observed with S. Typhimurium in polarized MDCK cells through a SopB- and SopE2-independent mechanism. In both cell types, knockdown of the ARHGEF26-associated protein DLG1 resulted in a similar phenotype and serovar specificity. Importantly, we show that ARHGEF26 plays a critical role in S. Typhimurium pathogenesis by contributing to bacterial burden in the enteric fever murine model, as well as inflammation in the colitis infection model. In the enteric fever model, SopB and SopE2 are required for the effects of Arhgef26 deletion on bacterial burden, and the impact of sopB and sopE2 deletion in turn required ARHGEF26. In contrast, SopB and SopE2 were not required for the impacts of Arhgef26 deletion on colitis. A role for ARHGEF26 on inflammation was also seen in cells, as knockdown reduced IL-8 production in HeLa cells. Together, these data reveal pleiotropic roles for ARHGEF26 during infection and highlight that many of the interactions that occur during infection that are thought to be well understood likely have underappreciated complexity. During infection, Salmonella manipulates host cells into engulfing the bacteria and establishing an intracellular niche. While many studies have identified genes involved in different stages of this Salmonella invasion process, few studies have examined how differences between human hosts contribute to infection susceptibility. Here we leveraged a candidate genetic screen to identify natural genetic variation in the human ARHGEF26 gene that correlates with Salmonella invasion. Springboarding from this result, we experimentally tested and redefined ARHGEF26’s role in Salmonella invasion, discovered a new role for ARHGEF26 in regulating inflammation during Salmonella disease, and demonstrated the relevance of these findings in mouse models. Building on how ARHGEF26 functions in other contexts, we implicated two ARHGEF26-interacting host proteins as contributors to Salmonella pathobiology. Collectively, these results identify a potential source of inter-person diversity in susceptibility to Salmonella disease and expand our molecular understanding of Salmonella infection to include a multifaceted role for ARHGEF26. They further identify important future directions in understanding how Salmonella recruit and manipulate ARHGEF26 as well as how ARHGEF26 is able to drive Salmonella-beneficial processes.
Collapse
|
34
|
Mylvaganam S, Freeman SA, Grinstein S. The cytoskeleton in phagocytosis and macropinocytosis. Curr Biol 2021; 31:R619-R632. [PMID: 34033794 DOI: 10.1016/j.cub.2021.01.036] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cells of the innate immune system, notably macrophages, neutrophils and dendritic cells, perform essential antimicrobial and homeostatic functions. These functions rely on the dynamic surveillance of the environment supported by the formation of elaborate membrane protrusions. Such protrusions - pseudopodia, lamellipodia and filopodia - facilitate the sampling of the surrounding fluid by macropinocytosis, as well as the engulfment of particulates by phagocytosis. Both processes entail extreme plasma membrane deformations that require the coordinated rearrangement of cytoskeletal polymers, which exert protrusive force and drive membrane coalescence and scission. The resulting vacuolar compartments undergo pronounced remodeling and ultimate resolution by mechanisms that also involve the cytoskeleton. Here, we describe the regulation and functions of cytoskeletal assembly and remodeling during macropinocytosis and phagocytosis.
Collapse
Affiliation(s)
- Sivakami Mylvaganam
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Spencer A Freeman
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Sergio Grinstein
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
35
|
Pandya AD, Iversen TG, Moestue S, Grinde MT, Mørch Ý, Snipstad S, Åslund AKO, Øy GF, Kildal W, Engebråten O, Sandvig K, Skotland T, Mælandsmo GM. Biodistribution of Poly(alkyl cyanoacrylate) Nanoparticles in Mice and Effect on Tumor Infiltration of Macrophages into a Patient-Derived Breast Cancer Xenograft. NANOMATERIALS 2021; 11:nano11051140. [PMID: 33924869 PMCID: PMC8145722 DOI: 10.3390/nano11051140] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 11/18/2022]
Abstract
We have investigated the biodistribution and tumor macrophage infiltration after intravenous injection of the poly(alkyl cyanoacrylate) nanoparticles (NPs): PEBCA (poly(2-ethyl-butyl cyanoacrylate), PBCA (poly(n-butyl cyanoacrylate), and POCA (poly(octyl cyanoacrylate), in mice. These NPs are structurally similar, have similar PEGylation, and have previously been shown to give large variations in cellular responses in vitro. The PEBCA NPs had the highest uptake both in the patient-derived breast cancer xenograft MAS98.12 and in lymph nodes, and therefore, they are the most promising of these NPs for delivery of cancer drugs. High-resolution magic angle spinning magnetic resonance (HR MAS MR) spectroscopy did not reveal any differences in the metabolic profiles of tumors following injection of the NPs, but the PEBCA NPs resulted in higher tumor infiltration of the anti-tumorigenic M1 macrophages than obtained with the two other NPs. The PEBCA NPs also increased the ratio of M1/M2 (anti-tumorigenic/pro-tumorigenic) macrophages in the tumors, suggesting that these NPs might be used both as a vehicle for drug delivery and to modulate the immune response in favor of enhanced therapeutic effects.
Collapse
Affiliation(s)
- Abhilash D. Pandya
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, 0379 Oslo, Norway; (A.D.P.); (G.F.Ø.); (O.E.)
| | - Tore-Geir Iversen
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, 0379 Oslo, Norway; (T.-G.I.); (K.S.)
| | - Siver Moestue
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, 7491 Trondheim, Norway;
| | - Maria T. Grinde
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, 7491 Trondheim, Norway;
| | - Ýrr Mørch
- SINTEF AS, Department of Biotechnology and Nanomedicine, 7034 Trondheim, Norway; (Ý.M.); (S.S.); (A.K.O.Å.)
| | - Sofie Snipstad
- SINTEF AS, Department of Biotechnology and Nanomedicine, 7034 Trondheim, Norway; (Ý.M.); (S.S.); (A.K.O.Å.)
- Department of Physics, Norwegian University of Science and Technology, 7491 Trondheim, Norway
- Cancer Clinic, St. Olav’s Hospital, 7030 Trondheim, Norway
| | - Andreas K. O. Åslund
- SINTEF AS, Department of Biotechnology and Nanomedicine, 7034 Trondheim, Norway; (Ý.M.); (S.S.); (A.K.O.Å.)
| | - Geir F. Øy
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, 0379 Oslo, Norway; (A.D.P.); (G.F.Ø.); (O.E.)
| | - Wanja Kildal
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, The Norwegian Radium Hospital, 0379 Oslo, Norway;
| | - Olav Engebråten
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, 0379 Oslo, Norway; (A.D.P.); (G.F.Ø.); (O.E.)
- Department of Oncology, Oslo University Hospital, 0450 Oslo, Norway
- Institute of Clinical Medicine, Medical Faculty, University of Oslo, 0315 Oslo, Norway
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, 0379 Oslo, Norway; (T.-G.I.); (K.S.)
- Department of Biosciences, University of Oslo, 0315 Oslo, Norway
| | - Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, 0379 Oslo, Norway; (T.-G.I.); (K.S.)
- Correspondence: (T.S.); (G.M.M.)
| | - Gunhild M. Mælandsmo
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, 0379 Oslo, Norway; (A.D.P.); (G.F.Ø.); (O.E.)
- Department of Medical Biology, University of Tromsø, 9019 Tromsø, Norway
- Correspondence: (T.S.); (G.M.M.)
| |
Collapse
|
36
|
Arlauckas S, Oh N, Li R, Weissleder R, Miller MA. Macrophage imaging and subset analysis using single-cell RNA sequencing. Nanotheranostics 2021; 5:36-56. [PMID: 33391974 PMCID: PMC7738942 DOI: 10.7150/ntno.50185] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/19/2020] [Indexed: 12/12/2022] Open
Abstract
Macrophages have been associated with drug response and resistance in diverse settings, thus raising the possibility of using macrophage imaging as a companion diagnostic to inform personalized patient treatment strategies. Nanoparticle-based contrast agents are especially promising because they efficiently deliver fluorescent, magnetic, and/or radionuclide labels by leveraging the intrinsic capacity of macrophages to accumulate nanomaterials in their role as professional phagocytes. Unfortunately, current clinical imaging modalities are limited in their ability to quantify broad molecular programs that may explain (a) which particular cell subsets a given imaging agent is actually labeling, and (b) what mechanistic role those cells play in promoting drug response or resistance. Highly multiplexed single-cell approaches including single-cell RNA sequencing (scRNAseq) have emerged as resources to help answer these questions. In this review, we query recently published scRNAseq datasets to support companion macrophage imaging, with particular focus on using dextran-based nanoparticles to predict the action of anti-cancer nanotherapies and monoclonal antibodies.
Collapse
Affiliation(s)
- Sean Arlauckas
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114, USA
| | - Nuri Oh
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114, USA
| | - Ran Li
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114, USA.,Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114, USA.,Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02115, USA.,Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Miles A Miller
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114, USA.,Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
37
|
Lin XP, Mintern JD, Gleeson PA. Macropinocytosis in Different Cell Types: Similarities and Differences. MEMBRANES 2020; 10:membranes10080177. [PMID: 32756454 PMCID: PMC7463864 DOI: 10.3390/membranes10080177] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 07/29/2020] [Accepted: 07/30/2020] [Indexed: 12/14/2022]
Abstract
Macropinocytosis is a unique pathway of endocytosis characterised by the nonspecific internalisation of large amounts of extracellular fluid, solutes and membrane in large endocytic vesicles known as macropinosomes. Macropinocytosis is important in a range of physiological processes, including antigen presentation, nutrient sensing, recycling of plasma proteins, migration and signalling. It has become apparent in recent years from the study of specialised cells that there are multiple pathways of macropinocytosis utilised by different cell types, and some of these pathways are triggered by different stimuli. Understanding the physiological function of macropinocytosis requires knowledge of the regulation and fate of the macropinocytosis pathways in a range of cell types. Here, we compare the mechanisms of macropinocytosis in different primary and immortalised cells, identify the gaps in knowledge in the field and discuss the potential approaches to analyse the function of macropinocytosis in vivo.
Collapse
|
38
|
Patel VI, Booth JL, Dozmorov M, Brown BR, Metcalf JP. Anthrax Edema and Lethal Toxins Differentially Target Human Lung and Blood Phagocytes. Toxins (Basel) 2020; 12:toxins12070464. [PMID: 32698436 PMCID: PMC7405021 DOI: 10.3390/toxins12070464] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/14/2020] [Accepted: 07/15/2020] [Indexed: 12/26/2022] Open
Abstract
Bacillus anthracis, the causative agent of inhalation anthrax, is a serious concern as a bioterrorism weapon. The vegetative form produces two exotoxins: Lethal toxin (LT) and edema toxin (ET). We recently characterized and compared six human airway and alveolar-resident phagocyte (AARP) subsets at the transcriptional and functional levels. In this study, we examined the effects of LT and ET on these subsets and human leukocytes. AARPs and leukocytes do not express high levels of the toxin receptors, tumor endothelium marker-8 (TEM8) and capillary morphogenesis protein-2 (CMG2). Less than 20% expressed surface TEM8, while less than 15% expressed CMG2. All cell types bound or internalized protective antigen, the common component of the two toxins, in a dose-dependent manner. Most protective antigen was likely internalized via macropinocytosis. Cells were not sensitive to LT-induced apoptosis or necrosis at concentrations up to 1000 ng/mL. However, toxin exposure inhibited B. anthracis spore internalization. This inhibition was driven primarily by ET in AARPs and LT in leukocytes. These results support a model of inhalation anthrax in which spores germinate and produce toxins. ET inhibits pathogen phagocytosis by AARPs, allowing alveolar escape. In late-stage disease, LT inhibits phagocytosis by leukocytes, allowing bacterial replication in the bloodstream.
Collapse
Affiliation(s)
- Vineet I. Patel
- Department of Medicine, Pulmonary, Critical Care & Sleep Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (V.I.P.); (J.L.B.); (B.R.B.)
| | - J. Leland Booth
- Department of Medicine, Pulmonary, Critical Care & Sleep Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (V.I.P.); (J.L.B.); (B.R.B.)
| | - Mikhail Dozmorov
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Brent R. Brown
- Department of Medicine, Pulmonary, Critical Care & Sleep Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (V.I.P.); (J.L.B.); (B.R.B.)
| | - Jordan P. Metcalf
- Department of Medicine, Pulmonary, Critical Care & Sleep Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (V.I.P.); (J.L.B.); (B.R.B.)
- Department of Microbiology and Immunology, the University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA
- Correspondence:
| |
Collapse
|
39
|
Haider P, Kral-Pointner JB, Mayer J, Richter M, Kaun C, Brostjan C, Eilenberg W, Fischer MB, Speidl WS, Hengstenberg C, Huber K, Wojta J, Hohensinner P. Neutrophil Extracellular Trap Degradation by Differently Polarized Macrophage Subsets. Arterioscler Thromb Vasc Biol 2020; 40:2265-2278. [PMID: 32673525 PMCID: PMC7447175 DOI: 10.1161/atvbaha.120.314883] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Supplemental Digital Content is available in the text. Macrophages are immune cells, capable to remodel the extracellular matrix, which can harbor extracellular DNA incorporated into neutrophil extracellular traps (NETs). To study the breakdown of NETs we studied the capability of macrophage subsets to degrade these structures in vitro and in vivo in a murine thrombosis model. Furthermore, we analyzed human abdominal aortic aneurysm samples in support of our in vitro and in vivo results.
Collapse
Affiliation(s)
- Patrick Haider
- From the Division of Cardiology, Department of Medicine II (P. Haider, J.B.K.-P., J.M., M.R., C.K., W.S.S., C.H., J.W., P. Hohensinner), Medical University of Vienna, Austria
| | - Julia B Kral-Pointner
- From the Division of Cardiology, Department of Medicine II (P. Haider, J.B.K.-P., J.M., M.R., C.K., W.S.S., C.H., J.W., P. Hohensinner), Medical University of Vienna, Austria.,Ludwig Boltzmann Institute for Cardiovascular Research, Austria (J.B.K.-P., J.W.)
| | - Julia Mayer
- From the Division of Cardiology, Department of Medicine II (P. Haider, J.B.K.-P., J.M., M.R., C.K., W.S.S., C.H., J.W., P. Hohensinner), Medical University of Vienna, Austria
| | - Manuela Richter
- From the Division of Cardiology, Department of Medicine II (P. Haider, J.B.K.-P., J.M., M.R., C.K., W.S.S., C.H., J.W., P. Hohensinner), Medical University of Vienna, Austria
| | - Christoph Kaun
- From the Division of Cardiology, Department of Medicine II (P. Haider, J.B.K.-P., J.M., M.R., C.K., W.S.S., C.H., J.W., P. Hohensinner), Medical University of Vienna, Austria
| | - Christine Brostjan
- Division of Vascular Surgery and Surgical Research Laboratories, Department of Surgery (C.B., W.E.), Medical University of Vienna, Austria
| | - Wolf Eilenberg
- Division of Vascular Surgery and Surgical Research Laboratories, Department of Surgery (C.B., W.E.), Medical University of Vienna, Austria
| | - Michael B Fischer
- Department of Blood Group Serology and Transfusion Medicine (M.B.F.), Medical University of Vienna, Austria.,Department of Biomedical Research, Danube University Krems, Austria (M.B.F.)
| | - Walter S Speidl
- From the Division of Cardiology, Department of Medicine II (P. Haider, J.B.K.-P., J.M., M.R., C.K., W.S.S., C.H., J.W., P. Hohensinner), Medical University of Vienna, Austria
| | - Christian Hengstenberg
- From the Division of Cardiology, Department of Medicine II (P. Haider, J.B.K.-P., J.M., M.R., C.K., W.S.S., C.H., J.W., P. Hohensinner), Medical University of Vienna, Austria
| | - Kurt Huber
- Wilhelminenhospital, 3rd Department of Medicine, Cardiology and Intensive Care Medicine, Vienna, Austria (K.H.).,Sigmund Freud University, Medical Faculty, Vienna, Austria (K.H.)
| | - Johann Wojta
- From the Division of Cardiology, Department of Medicine II (P. Haider, J.B.K.-P., J.M., M.R., C.K., W.S.S., C.H., J.W., P. Hohensinner), Medical University of Vienna, Austria.,Ludwig Boltzmann Institute for Cardiovascular Research, Austria (J.B.K.-P., J.W.).,Medical University of Vienna, Core Facilities, Austria (J.W.)
| | - Philipp Hohensinner
- From the Division of Cardiology, Department of Medicine II (P. Haider, J.B.K.-P., J.M., M.R., C.K., W.S.S., C.H., J.W., P. Hohensinner), Medical University of Vienna, Austria
| |
Collapse
|
40
|
Hayashi Y, Takamiya M, Jensen PB, Ojea-Jiménez I, Claude H, Antony C, Kjaer-Sorensen K, Grabher C, Boesen T, Gilliland D, Oxvig C, Strähle U, Weiss C. Differential Nanoparticle Sequestration by Macrophages and Scavenger Endothelial Cells Visualized in Vivo in Real-Time and at Ultrastructural Resolution. ACS NANO 2020; 14:1665-1681. [PMID: 31922724 DOI: 10.1021/acsnano.9b07233] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Despite the common knowledge that the reticuloendothelial system is largely responsible for blood clearance of systemically administered nanoparticles, the sequestration mechanism remains a "black box". Using transgenic zebrafish embryos with cell type-specific fluorescent reporters and fluorescently labeled model nanoparticles (70 nm SiO2), we here demonstrate simultaneous three-color in vivo imaging of intravenously injected nanoparticles, macrophages, and scavenger endothelial cells (SECs). The trafficking processes were further revealed at ultrastructural resolution by transmission electron microscopy. We also find, using a correlative light-electron microscopy approach, that macrophages rapidly sequester nanoparticles via membrane adhesion and endocytosis (including macropinocytosis) within minutes after injection. In contrast, SECs trap single nanoparticles via scavenger receptor-mediated endocytosis, resulting in gradual sequestration with a time scale of hours. Inhibition of the scavenger receptors prevented SECs from accumulating nanoparticles but enhanced uptake in macrophages, indicating the competitive nature of nanoparticle clearance in vivo. To directly quantify the relative contributions of the two cell types to overall nanoparticle sequestration, the differential sequestration kinetics was studied within the first 30 min post-injection. This revealed a much higher and increasing relative contribution of SECs, as they by far outnumber macrophages in zebrafish embryos, suggesting the importance of the macrophage:SECs ratio in a given tissue. Further characterizing macrophages on their efficiency in nanoparticle clearance, we show that inflammatory stimuli diminish the uptake of nanoparticles per cell. Our study demonstrates the strength of transgenic zebrafish embryos for intravital real-time and ultrastructural imaging of nanomaterials that may provide mechanistic insights into nanoparticle clearance in rodent models and humans.
Collapse
Affiliation(s)
- Yuya Hayashi
- Department of Molecular Biology and Genetics , Aarhus University , Gustav Wieds Vej 10 , 8000 Aarhus C , Denmark
- Institute of Toxicology and Genetics , Karlsruhe Institute of Technology (KIT) , Hermann-von-Helmholtz-Platz 1 , 76344 Eggenstein-Leopoldshafen , Germany
| | - Masanari Takamiya
- Institute of Toxicology and Genetics , Karlsruhe Institute of Technology (KIT) , Hermann-von-Helmholtz-Platz 1 , 76344 Eggenstein-Leopoldshafen , Germany
| | - Pia Bomholt Jensen
- iNANO Interdisciplinary Nanoscience Center , Aarhus University , Gustav Wieds Vej 14 , 8000 Aarhus C , Denmark
| | - Isaac Ojea-Jiménez
- Institute for Health and Consumer Protection , European Commission Joint Research Centre , Via E. Fermi 2749 , 21027 Ispra , Varese , Italy
| | - Hélicia Claude
- Institute of Toxicology and Genetics , Karlsruhe Institute of Technology (KIT) , Hermann-von-Helmholtz-Platz 1 , 76344 Eggenstein-Leopoldshafen , Germany
| | - Claude Antony
- Institute of Toxicology and Genetics , Karlsruhe Institute of Technology (KIT) , Hermann-von-Helmholtz-Platz 1 , 76344 Eggenstein-Leopoldshafen , Germany
| | - Kasper Kjaer-Sorensen
- Department of Molecular Biology and Genetics , Aarhus University , Gustav Wieds Vej 10 , 8000 Aarhus C , Denmark
| | - Clemens Grabher
- Institute of Toxicology and Genetics , Karlsruhe Institute of Technology (KIT) , Hermann-von-Helmholtz-Platz 1 , 76344 Eggenstein-Leopoldshafen , Germany
| | - Thomas Boesen
- Department of Molecular Biology and Genetics , Aarhus University , Gustav Wieds Vej 10 , 8000 Aarhus C , Denmark
- iNANO Interdisciplinary Nanoscience Center , Aarhus University , Gustav Wieds Vej 14 , 8000 Aarhus C , Denmark
| | - Douglas Gilliland
- Institute for Health and Consumer Protection , European Commission Joint Research Centre , Via E. Fermi 2749 , 21027 Ispra , Varese , Italy
| | - Claus Oxvig
- Department of Molecular Biology and Genetics , Aarhus University , Gustav Wieds Vej 10 , 8000 Aarhus C , Denmark
| | - Uwe Strähle
- Institute of Toxicology and Genetics , Karlsruhe Institute of Technology (KIT) , Hermann-von-Helmholtz-Platz 1 , 76344 Eggenstein-Leopoldshafen , Germany
| | - Carsten Weiss
- Institute of Toxicology and Genetics , Karlsruhe Institute of Technology (KIT) , Hermann-von-Helmholtz-Platz 1 , 76344 Eggenstein-Leopoldshafen , Germany
| |
Collapse
|
41
|
Doodnauth SA, Grinstein S, Maxson ME. Constitutive and stimulated macropinocytosis in macrophages: roles in immunity and in the pathogenesis of atherosclerosis. Philos Trans R Soc Lond B Biol Sci 2020; 374:20180147. [PMID: 30967001 DOI: 10.1098/rstb.2018.0147] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Macrophages respond to several stimuli by forming florid membrane ruffles that lead to fluid uptake by macropinocytosis. This type of induced macropinocytosis, executed by a variety of non-malignant and malignant cells, is initiated by transmembrane receptors and is involved in nutrient acquisition and mTOR signalling. However, macrophages also perform a unique type of constitutive ruffling and macropinocytosis that is dependent on the presence of extracellular calcium. Calcium-sensing receptors are responsible for this activity. This distinct form of macropinocytosis enables macrophages to continuously sample their microenvironment for antigenic molecules and for pathogen- and danger-associated molecular patterns, as part of their immune surveillance functions. Interestingly, even within the monocyte lineage, there are differences in macropinocytic ability that reflect the polarized functional roles of distinct macrophage subsets. This review discusses the shared and distinct features of both induced and constitutive macropinocytosis displayed by the macrophage lineage and their roles in physiology, immunity and pathophysiology. In particular, we analyse the role of macropinocytosis in the uptake of modified low-density lipoprotein (LDL) and its contribution to foam cell and atherosclerotic plaque formation. We propose a combined role of scavenger receptors and constitutive macropinocytosis in oxidized LDL uptake, a process we have termed 'receptor-assisted macropinocytosis'. This article is part of the Theo Murphy meeting issue 'Macropinocytosis'.
Collapse
Affiliation(s)
- Sasha A Doodnauth
- 1 Princess Margaret Cancer Center, University Health Network , Toronto, ON , Canada M5G 1L7.,2 Department of Medical Biophysics, University of Toronto , Toronto, ON , Canada M5G 1L7
| | - Sergio Grinstein
- 3 Program in Cell Biology, Hospital for Sick Children , 686 Bay Street, Toronto, ON , Canada M5G 0A4.,4 Department of Biochemistry, University of Toronto , 1 King's Circle, Toronto, ON , Canada M5S 1A8.,5 Keenan Research Centre of the Li Ka Shing Knowledge Institute , St. Michael's Hospital, 290 Victoria Street, Toronto, ON , Canada M5C 1N8
| | - Michelle E Maxson
- 3 Program in Cell Biology, Hospital for Sick Children , 686 Bay Street, Toronto, ON , Canada M5G 0A4
| |
Collapse
|
42
|
Szwed M, Torgersen ML, Kumari RV, Yadava SK, Pust S, Iversen TG, Skotland T, Giri J, Sandvig K. Biological response and cytotoxicity induced by lipid nanocapsules. J Nanobiotechnology 2020; 18:5. [PMID: 31907052 PMCID: PMC6943936 DOI: 10.1186/s12951-019-0567-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 12/22/2019] [Indexed: 12/13/2022] Open
Abstract
Background Lipid nanocapsules (LNCs) are promising vehicles for drug delivery. However, since not much was known about cellular toxicity of these nanoparticles in themselves, we have here investigated the mechanisms involved in LNC-induced intoxication of the three breast cancer cell lines MCF-7, MDA-MD-231 and MDA-MB-468. The LNCs used were made of Labrafac™ Lipophile WL1349, Lipoid® S75 and Solutol® HS15. Results High resolution SIM microscopy showed that the DiD-labeled LNCs ended up in lysosomes close to the membrane. Empty LNCs, i.e. without encapsulated drug, induced not only increased lysosomal pH, but also acidification of the cytosol and a rapid inhibition of protein synthesis. The cytotoxicity of the LNCs were measured for up to 72 h of incubation using the MTT assay and ATP measurements in all three cell lines, and revealed that MDA-MB-468 was the most sensitive cell line and MCF-7 the least sensitive cell line to these LNCs. The LNCs induced generation of reactive free oxygen species and lipid peroxidation. Experiments with knock-down of kinases in the near-haploid cell line HAP1 indicated that the kinase HRI is essential for the observed phosphorylation of eIF2α. Nrf2 and ATF4 seem to play a protective role against the LNCs in MDA-MB-231 cells, as knock-down of these factors sensitizes the cells to the LNCs. This is in contrast to MCF-7 cells where the knock-down of these factors had a minor effect on the toxicity of the LNCs. Inhibitors of ferroptosis provided a large protection against LNC toxicity in MDA-MB-231 cells, but not in MCF-7 cells. Conclusions High doses of LNCs showed a different degree of toxicity on the three cell lines studied, i.e. MCF-7, MDA-MD-231 and MDA-MB-468 and affected signaling factors and the cell fate differently in these cell lines.
Collapse
Affiliation(s)
- Marzena Szwed
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway
| | - Maria Lyngaas Torgersen
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway
| | - Remya Valsala Kumari
- Department of Biomedical Engineering, Indian Institute of Technology, Hyderabad, India
| | - Sunil Kumar Yadava
- Department of Biomedical Engineering, Indian Institute of Technology, Hyderabad, India
| | - Sascha Pust
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway
| | - Tore Geir Iversen
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway
| | - Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway
| | - Jyotsnendu Giri
- Department of Biomedical Engineering, Indian Institute of Technology, Hyderabad, India.
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway. .,Department of Biosciences, University of Oslo, Oslo, Norway.
| |
Collapse
|
43
|
Salloum G, Jakubik CT, Erami Z, Heitz SD, Bresnick AR, Backer JM. PI3Kβ is selectively required for growth factor-stimulated macropinocytosis. J Cell Sci 2019; 132:jcs.231639. [PMID: 31409694 DOI: 10.1242/jcs.231639] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 07/16/2019] [Indexed: 12/16/2022] Open
Abstract
Macropinocytosis is an actin-dependent but clathrin-independent endocytic process by which cells nonselectively take up large aliquots of extracellular material. Macropinocytosis is used for immune surveillance by dendritic cells, as a route of infection by viruses and protozoa, and as a nutrient uptake pathway in tumor cells. In this study, we explore the role of class I phosphoinositide 3-kinases (PI3Ks) during ligand-stimulated macropinocytosis. We find that macropinocytosis in response to receptor tyrosine kinase activation is strikingly dependent on a single class I PI3K isoform, namely PI3Kβ (containing the p110β catalytic subunit encoded by PIK3CB). Loss of PI3Kβ expression or activity blocks macropinocytosis at early steps, before the formation of circular dorsal ruffles, but also plays a role in later steps, downstream from Rac1 activation. PI3Kβ is also required for the elevated levels of constitutive macropinocytosis found in tumor cells that are defective for the PTEN tumor suppressor. Our data shed new light on PI3K signaling during macropinocytosis, and suggest new therapeutic uses for pharmacological inhibitors of PI3Kβ.
Collapse
Affiliation(s)
- Gilbert Salloum
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Charles T Jakubik
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Zahra Erami
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Samantha D Heitz
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Anne R Bresnick
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jonathan M Backer
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA .,Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
44
|
Williams TD, Paschke PI, Kay RR. Function of small GTPases in Dictyostelium macropinocytosis. Philos Trans R Soc Lond B Biol Sci 2019; 374:20180150. [PMID: 30967009 PMCID: PMC6304742 DOI: 10.1098/rstb.2018.0150] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2018] [Indexed: 12/17/2022] Open
Abstract
Macropinocytosis-the large-scale, non-specific uptake of fluid by cells-is used by Dictyostelium discoideum amoebae to obtain nutrients. These cells form circular ruffles around regions of membrane defined by a patch of phosphatidylinositol (3,4,5)-trisphosphate (PIP3) and the activated forms of the small G-proteins Ras and Rac. When this ruffle closes, a vesicle of the medium is delivered to the cell interior for further processing. It is accepted that PIP3 is required for efficient macropinocytosis. Here, we assess the roles of Ras and Rac in Dictyostelium macropinocytosis. Gain-of-function experiments show that macropinocytosis is stimulated by persistent Ras activation and genetic analysis suggests that RasG and RasS are the key Ras proteins involved. Among the activating guanine exchange factors (GEFs), GefF is implicated in macropinocytosis by an insertional mutant. The individual roles of Rho family proteins are little understood but activation of at least some may be independent of PIP3. This article is part of the Theo Murphy meeting issue 'Macropinocytosis'.
Collapse
Affiliation(s)
| | | | - Robert R. Kay
- MRC-Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| |
Collapse
|
45
|
Fusser M, Øverbye A, Pandya AD, Mørch Ý, Borgos SE, Kildal W, Snipstad S, Sulheim E, Fleten KG, Askautrud HA, Engebraaten O, Flatmark K, Iversen TG, Sandvig K, Skotland T, Mælandsmo GM. Cabazitaxel-loaded Poly(2-ethylbutyl cyanoacrylate) nanoparticles improve treatment efficacy in a patient derived breast cancer xenograft. J Control Release 2018; 293:183-192. [PMID: 30529259 DOI: 10.1016/j.jconrel.2018.11.029] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 11/28/2018] [Accepted: 11/30/2018] [Indexed: 01/07/2023]
Abstract
The effect of poly(2-ethyl-butyl cyanoacrylate) nanoparticles containing the cytotoxic drug cabazitaxel was studied in three breast cancer cell lines and one basal-like patient-derived xenograft model grown in the mammary fat pad of immunodeficient mice. Nanoparticle-encapsulated cabazitaxel had a much better efficacy than similar concentrations of free drug in the basal-like patient-derived xenograft and resulted in complete remission of 6 out of 8 tumors, whereas free drug gave complete remission only with 2 out of 9 tumors. To investigate the different efficacies obtained with nanoparticle-encapsulated versus free cabazitaxel, mass spectrometry quantification of cabazitaxel was performed in mice plasma and selected tissue samples. Nanoparticle-encapsulated drug had a longer circulation time in blood. There was approximately a three times higher drug concentration in tumor tissue 24 h after injection, and two times higher 96 h after injection of nanoparticles with drug compared to the free drug. The tissue biodistribution obtained after 24 h using mass spectrometry analyses correlates well with biodistribution data obtained using IVIS® Spectrum in vivo imaging of nanoparticles labeled with the fluorescent substance NR668, indicating that these data also are representative for the nanoparticle distribution. Furthermore, immunohistochemistry was used to estimate infiltration of macrophages into the tumor tissue following injection of nanoparticle-encapsulated and free cabazitaxel. The higher infiltration of anti-tumorigenic versus pro-tumorigenic macrophages in tumors treated with the nanoparticles might also contribute to the improved effect obtained with the nanoparticle-encapsulated drug. Tumor infiltration of pro-tumorigenic macrophages was four times lower when using nanoparticles containing cabazitaxel than when using particles without drug, and we speculate that the very good therapeutic efficacy obtained with our cabazitaxel-containing particles may be due to their ability to reduce the level of pro-tumorigenic macrophages in the tumor. In summary, encapsulation of cabazitaxel in poly(2-ethyl-butyl cyanoacrylate) nanoparticles seems promising for treatment of breast cancer.
Collapse
Affiliation(s)
- Markus Fusser
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Anders Øverbye
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Abhilash D Pandya
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Ýrr Mørch
- Department of Biotechnology and Nanomedicine, SINTEF AS, Trondheim, Norway
| | - Sven Even Borgos
- Department of Biotechnology and Nanomedicine, SINTEF AS, Trondheim, Norway
| | - Wanja Kildal
- Institute for Cancer Genetics and Informatics, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Sofie Snipstad
- Department of Biotechnology and Nanomedicine, SINTEF AS, Trondheim, Norway; Department of Physics, The Norwegian University of Science and Technology, Trondheim, Norway
| | - Einar Sulheim
- Department of Biotechnology and Nanomedicine, SINTEF AS, Trondheim, Norway; Department of Physics, The Norwegian University of Science and Technology, Trondheim, Norway
| | - Karianne Giller Fleten
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Hanne Arenberg Askautrud
- Institute for Cancer Genetics and Informatics, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Olav Engebraaten
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway; Institute for Clinical Medicine, The Medical Faculty, University of Oslo, Oslo, Norway
| | - Kjersti Flatmark
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway; Institute for Clinical Medicine, The Medical Faculty, University of Oslo, Oslo, Norway
| | - Tore Geir Iversen
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway; Department of Biosciences, University of Oslo, Oslo, Norway
| | - Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.
| | - Gunhild M Mælandsmo
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway; Department of Pharmacy, University of Tromsø, Tromsø, Norway
| |
Collapse
|
46
|
Canton J. Macropinocytosis: New Insights Into Its Underappreciated Role in Innate Immune Cell Surveillance. Front Immunol 2018; 9:2286. [PMID: 30333835 PMCID: PMC6176211 DOI: 10.3389/fimmu.2018.02286] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 09/14/2018] [Indexed: 12/25/2022] Open
Abstract
Macropinocytosis has received increasing attention in recent years for its various roles in nutrient acquisition, immune surveillance, and virus and cancer pathologies. In most cases macropinocytosis is initiated by the sudden increase in an external stimulus such as a growth factor. This "induced" form of macropinocytosis has been the subject of much of the work addressing its mechanism and function over the years. An alternative, "constitutive" form of macropinocytosis restricted to primary innate immune cells also exists, although its mechanism has remained severely understudied. This mini-review focuses on the very recent advances that have shed new light on the initiation, formation and functional relevance of constitutive macropinocytosis in primary innate immune cells. An emphasis is placed on how this new understanding of constitutive macropinocytosis is helping to define the sentinel function of innate immune cells including polarized macrophages and dendritic cells.
Collapse
Affiliation(s)
- Johnathan Canton
- Immunobiology Laboratory, The Francis Crick Institute, London, United Kingdom
| |
Collapse
|