1
|
Mzyk A, Reyes-San-Martin C, Doğan Y, Woudstra W, Zhang Y, Yilmaz E, Bron R, de Haan-Visser W, Berg-Sorensen K, Schirhagl R. Quantum sensing to monitor changes in free radical generation by intracellular vesicles of polarized macrophages. Acta Biomater 2025:S1742-7061(25)00266-1. [PMID: 40222601 DOI: 10.1016/j.actbio.2025.04.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/21/2025] [Accepted: 04/10/2025] [Indexed: 04/15/2025]
Abstract
Macrophages are immune cells crucial in clearing our tissues from bacteria, viruses, dying cells, cell debris and other waste products. They also regulate inflammation by differentiating from non-activated (M0) cells into macrophages that initiate inflammation (pro-inflammatory macrophages, M1), or resolve inflammation (anti-inflammatory macrophages, M2). One of their key functions is to ingest pathogens within vesicles where they are degraded. The production of free radical (FR) plays an important role in this degradation process but also in macrophage differentiation and signaling. Here we used diamond-based quantum sensing to track free radical changes in vesicles with nanoscale resolution. We further followed the oxidative stress status, through free radical measurement during the macrophage activation process. We found that the three macrophage subtypes differed significantly in free radical generation in their vesicles. Additionally, we showed that the FR generation evolves over time in the different subtypes. We observed a 50 % increase in radical production in M0 after 24 h compared to the T1 values measured after 4 h of cell culture, a decrease in M1 and constant radical levels in M2 macrophages. STATEMENT OF SIGNIFICANCE: Here we use quantum sensing for the first time to investigate the role that free radicals play in immune cells when they differentiate to fulfill their functions in the immune system. We were able to measure free radical generation specifically in vesicles while the macrophages differentiated.
Collapse
Affiliation(s)
- Aldona Mzyk
- Groningen University, University Medical Center Groningen, Antonius Deusinglaan 1, 9713AV Groningen, the Netherlands; Technical University of Denmark, Ørsteds Plads, 349, 2800 Kgs, Lyngby, Denmark.
| | - Claudia Reyes-San-Martin
- Groningen University, University Medical Center Groningen, Antonius Deusinglaan 1, 9713AV Groningen, the Netherlands
| | - Yasemin Doğan
- Groningen University, University Medical Center Groningen, Antonius Deusinglaan 1, 9713AV Groningen, the Netherlands; Sabanci University, Orta Mahalle, Üniversite Caddesi, No: 27, Tuzla, 34956 Istanbul, Turkey
| | - Willem Woudstra
- Groningen University, University Medical Center Groningen, Antonius Deusinglaan 1, 9713AV Groningen, the Netherlands
| | - Yue Zhang
- Groningen University, University Medical Center Groningen, Antonius Deusinglaan 1, 9713AV Groningen, the Netherlands
| | - Ezgi Yilmaz
- Technical University of Denmark, Ørsteds Plads, 349, 2800 Kgs, Lyngby, Denmark
| | - Reinier Bron
- Groningen University, University Medical Center Groningen, Antonius Deusinglaan 1, 9713AV Groningen, the Netherlands
| | - Willy de Haan-Visser
- Groningen University, University Medical Center Groningen, Antonius Deusinglaan 1, 9713AV Groningen, the Netherlands
| | | | - Romana Schirhagl
- Groningen University, University Medical Center Groningen, Antonius Deusinglaan 1, 9713AV Groningen, the Netherlands.
| |
Collapse
|
2
|
Dai F, Liang W, Liu J, Guo M, Li C. Eeukaryotic-like Sppsk1 from Vibrio splendidus AJ01 mediates phagosome escape via inhibiting phagosome acidification and maturation. Cell Mol Life Sci 2025; 82:88. [PMID: 39985586 PMCID: PMC11846826 DOI: 10.1007/s00018-025-05610-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 12/16/2024] [Accepted: 01/30/2025] [Indexed: 02/24/2025]
Abstract
The intracellular pathogen has evolved sophisticated mechanisms to evade host immune defenses by secreting different virulence factors. In our previous study, the eukaryotic factor STPKLRR was identified from the intracellular pathogen Vibrio splendidus AJ01 and shown to facilitate promote AJ01 internalization by mediating actin-dependent coelomocytes phagocytosis. However, the molecular mechanisms underlying AJ01'escaped from the phagosome remained largely unclear. In this study, a novel eukaryotic-like factor was identified, containing both the Serine/Threonine/Tyrosine (STYKc) domain and protein phosphatase 2 C (PP2C) domain (denoted as Sppsk1), which was essential for AJ01 phagosome escape. Deletion of Sppsk1 significantly increased phagolysosome maturation and reduced the intracellular AJ01 levels compared to the wild AJ01. Mechanistic analysis showed that the STYKc domain of Sppsk1 directly phosphorylated phagosome H+ transport complex subunit ATP6V1C at Serine-356, resulting in the inhibition of phagosome acidification in coelomocytes and promoting AJ01 phagosome survival. Moreover, the PP2C domain of Sppsk1 dephosphorylated phosphatidylinositol-3-bisphosphate [PtdIns(3)P], converting it to PtdIns(3)P to phosphatidylinositol (PtdIns). Reduction of PtdIns(3)P on phagosomes hindered early endosome antigen 1 (EEA1) recruitment, thereby inhibiting phagosome maturation. These findings demonstrated that Sppsk1 in AJ01 could achieve phagosome escape by two strategies including inhibiting host coelomocytes' phagosome acidification and maturation, which advanced our knowledge of the general biology of pathogen-host interactions.
Collapse
Affiliation(s)
- Fa Dai
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Weikang Liang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Jiqing Liu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Ming Guo
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China.
| | - Chenghua Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China.
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, PR China.
| |
Collapse
|
3
|
Bamgbose TT, Schilke RM, Igiehon OO, Nkadi EH, Binwal M, Custis D, Bharrhan S, Schwarz B, Bohrnsen E, Bosio CM, Scott RS, Yurdagul Jr. A, Finck BN, Woolard MD. Lipin-1 restrains macrophage lipid synthesis to promote inflammation resolution. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:85-103. [PMID: 40073265 PMCID: PMC11844145 DOI: 10.1093/jimmun/vkae010] [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: 01/10/2024] [Accepted: 10/24/2024] [Indexed: 03/14/2025]
Abstract
Macrophages are critical to maintaining and restoring tissue homeostasis during inflammation. The lipid metabolic state of macrophages influences their function and polarization, which is crucial to the resolution of inflammation. The contribution of lipid synthesis to proinflammatory macrophage responses is well understood. However, how lipid synthesis regulates proresolving macrophage responses needs to be better understood. Lipin-1 is a phosphatidic acid phosphatase with a transcriptional coregulatory activity that regulates lipid metabolism. We previously demonstrated that lipin-1 supports proresolving macrophage responses, and here, myeloid-associated lipin-1 is required for inflammation resolution, yet how lipin-1-regulated cellular mechanisms promote macrophage proresolution responses is unknown. We demonstrated that the loss of lipin-1 in macrophages led to increased free fatty acid, neutral lipid, and ceramide content and increased phosphorylation of acetyl-CoA carboxylase. The inhibition of the first step of lipid synthesis, the transport of citrate from the mitochondria, reduced lipid content and restored efferocytosis and inflammation resolution in lipin-1mKO mice and macrophages. Our findings suggest macrophage-associated lipin-1 restrains lipid synthesis, promoting proresolving macrophage function in response to proresolving stimuli.
Collapse
Affiliation(s)
- Temitayo T Bamgbose
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Robert M Schilke
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Oluwakemi O Igiehon
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Ebubechukwu H Nkadi
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Monika Binwal
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - David Custis
- Research Core Facility, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Sushma Bharrhan
- Center for Applied Immunology and Pathological Processes, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Benjamin Schwarz
- Proteins and Chemistry Section, Research and Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT, United States
| | - Eric Bohrnsen
- Proteins and Chemistry Section, Research and Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT, United States
| | - Catharine M Bosio
- Immunity to Pulmonary Pathogens Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT, United States
| | - Rona S Scott
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
- Center for Applied Immunology and Pathological Processes, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Arif Yurdagul Jr.
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Brian N Finck
- Division of Nutritional Sciences and Obesity Medicine, Washington University School of Medicine in St. Louis, St Louis, MO, United States
| | - Matthew D Woolard
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
- Center for Applied Immunology and Pathological Processes, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| |
Collapse
|
4
|
Wang B, Cao X, Garcia-Mansfield K, Zhou J, Manousopoulou A, Pirrotte P, Wang Y, Wang LD, Feng M. Phosphoproteomic Profiling Reveals mTOR Signaling in Sustaining Macrophage Phagocytosis of Cancer Cells. Cancers (Basel) 2024; 16:4238. [PMID: 39766137 PMCID: PMC11674635 DOI: 10.3390/cancers16244238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Macrophage-mediated cancer cell phagocytosis has demonstrated considerable therapeutic potential. While the initiation of phagocytosis, facilitated by interactions between cancer cell surface signals and macrophage receptors, has been characterized, the mechanisms underlying its sustentation and attenuation post-initiation remain poorly understood. Methods: Through comprehensive phosphoproteomic profiling, we interrogated the temporal evolution of the phosphorylation profiles within macrophages during cancer cell phagocytosis. Results: Our findings reveal that activation of the mTOR pathway occurs following the initiation of phagocytosis and is crucial in sustaining phagocytosis of cancer cells. mTOR inhibition impaired the phagocytic capacity, but not affinity, of the macrophages toward the cancer cells by delaying phagosome maturation and impeding the transition between non-phagocytic and phagocytic states of macrophages. Conclusions: Our findings delineate the intricate landscape of macrophage phagocytosis and highlight the pivotal role of the mTOR pathway in mediating this process, offering valuable mechanistic insights for therapeutic interventions.
Collapse
Affiliation(s)
- Bixin Wang
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Xu Cao
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Krystine Garcia-Mansfield
- Cancer and Cell Biology Division, Translational Genomics Institute, Phoenix, AZ 85004, USA
- Integrated Mass Spectrometry Shared Resource, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Jingkai Zhou
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Antigoni Manousopoulou
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Patrick Pirrotte
- Cancer and Cell Biology Division, Translational Genomics Institute, Phoenix, AZ 85004, USA
- Integrated Mass Spectrometry Shared Resource, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Yingyu Wang
- Center for Informatics, City of Hope, Duarte, CA 91010, USA
| | - Leo D. Wang
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
- Department of Pediatrics, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Mingye Feng
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
5
|
Kovacs T, Cs. Szabo B, Kothalawala RC, Szekelyhidi V, Nagy P, Varga Z, Panyi G, Zakany F. Inhibition of the H V1 voltage-gated proton channel compromises the viability of human polarized macrophages in a polarization- and ceramide-dependent manner. Front Immunol 2024; 15:1487578. [PMID: 39742270 PMCID: PMC11685079 DOI: 10.3389/fimmu.2024.1487578] [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: 08/28/2024] [Accepted: 12/02/2024] [Indexed: 01/03/2025] Open
Abstract
The human voltage-gated proton channel (HV1) provides an efficient proton extrusion pathway from the cytoplasm contributing to the intracellular pH regulation and the oxidative burst. Although its pharmacological inhibition was previously shown to induce cell death in various cell types, no such effects have been examined in polarized macrophages albeit HV1 was suggested to play important roles in these cells. This study highlights that 5-chloro-2-guanidinobenzimidazole (ClGBI), the most widely applied HV1 inhibitor, reduces the viability of human THP-1-derived polarized macrophages at biologically relevant doses with M1 macrophages being the most, and M2 cells the least sensitive to this compound. ClGBI may exert this effect principally by blocking HV1 since the sensitivity of polarized macrophages correlates well with their HV1 expression levels; inhibitors of other macrophage ion channels that may be susceptible for off-target ClGBI effects cause no viability reductions; and Zn2+, another non-specific HV1 blocker, exerts similar effects. As a potential mechanism behind the ClGBI-induced cell death, we identify a complex pH dysregulation involving acidification of the cytoplasm and alkalinization of the lysosomes, which eventually result in membrane ceramide accumulation. Furthermore, ClGBI effects are alleviated by ARC39, a selective acid sphingomyelinase inhibitor supporting the unequivocal significance of ceramide accumulation in the process. Altogether, our results suggest that HV1 inhibition leads to cellular toxicity in polarized macrophages in a polarization-dependent manner, which occurs due to a pH dysregulation and concomitant ceramide overproduction mainly depending on the activity of acid sphingomyelinase. The reduced macrophage viability and plausible concomitant changes in homeostatic M1-M2 balance could contribute to both the therapeutic and potential side effects of HV1 inhibitors that show great promise in the treatment of neuroinflammation and malignant diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Florina Zakany
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
6
|
Maia Santos Urbancg Moncorvo F, Avendaño Leon OL, Curti C, Kabri Y, Redon S, Torres-Santos EC, Vanelle P. Enhancing Antileishmanial Activity of Amidoxime-Based Compounds Bearing a 4,5-Dihydrofuran Scaffold: In Vitro Screening Against Leishmania amazonensis. Molecules 2024; 29:5469. [PMID: 39598858 PMCID: PMC11597885 DOI: 10.3390/molecules29225469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024] Open
Abstract
Leishmaniasis, a protozoan disease affecting humans, exposes significant shortcomings in current treatments. In continuation to our previous findings on amidoxime-based antileishmanial compounds bearing a 4,5-dihydrofuran scaffold, twelve new amidoxime derivatives substituted at position 3 with an amide bearing a nitrogen heterocycle were synthesized. This series was designed to replace the sulfone and aryl group on a previously reported HIT. The synthesis of these compounds involved the following three-step pathway: manganese (III) acetate-based cyclization of a β-ketoester, followed by amidation with LiHMDS and a final reaction with hydroxylamine. Three of them, containing either bromine, chlorine, or methyl substitutions and featuring a pyridine moiety, showed an interesting toxicity-activity relationship in vitro. They exhibited IC50 values of 15.0 µM, 16.0 µM, and 17.0 µM against the promastigote form of the parasite and IC50 values of 0.5 µM, 0.6 µM, and 0.3 µM against the intracellular amastigote form, respectively. A selectivity index (SI) greater than 300 was established between the cytotoxic concentrations (in murine macrophages) and the effective concentrations (against the intracellular form of Leishmania amazonensis). This SI is at least seventy times higher than that observed for Pentamidine and twenty-five times higher than that observed for the reference HIT, as previously reported.
Collapse
Affiliation(s)
| | - Oscar Leonardo Avendaño Leon
- Aix Marseille Univ, CNRS, ICR UMR 7273, Equipe Pharmaco-Chimie Radicalaire, Faculté de Pharmacie, 27 Boulevard Jean Moulin, CS30064, CEDEX 05, 13385 Marseille, France; (O.L.A.L.); (C.C.); (Y.K.); (S.R.)
| | - Christophe Curti
- Aix Marseille Univ, CNRS, ICR UMR 7273, Equipe Pharmaco-Chimie Radicalaire, Faculté de Pharmacie, 27 Boulevard Jean Moulin, CS30064, CEDEX 05, 13385 Marseille, France; (O.L.A.L.); (C.C.); (Y.K.); (S.R.)
- Service Central de la Qualité et de l’Information Pharmaceutiques (SCQIP), Pharmacy Department, Assistance Publique—Hôpitaux de Marseille (AP-HM), 147 Bd. Baille, 13006 Marseille, France
| | - Youssef Kabri
- Aix Marseille Univ, CNRS, ICR UMR 7273, Equipe Pharmaco-Chimie Radicalaire, Faculté de Pharmacie, 27 Boulevard Jean Moulin, CS30064, CEDEX 05, 13385 Marseille, France; (O.L.A.L.); (C.C.); (Y.K.); (S.R.)
| | - Sébastien Redon
- Aix Marseille Univ, CNRS, ICR UMR 7273, Equipe Pharmaco-Chimie Radicalaire, Faculté de Pharmacie, 27 Boulevard Jean Moulin, CS30064, CEDEX 05, 13385 Marseille, France; (O.L.A.L.); (C.C.); (Y.K.); (S.R.)
| | - Eduardo Caio Torres-Santos
- Laboratório de Bioquímica de Tripanosomatídeos, Instituto Oswaldo Cruz—FIOCRUZ, Av. Brasil, 4365, Rio de Janeiro 21040-900, Brazil;
| | - Patrice Vanelle
- Aix Marseille Univ, CNRS, ICR UMR 7273, Equipe Pharmaco-Chimie Radicalaire, Faculté de Pharmacie, 27 Boulevard Jean Moulin, CS30064, CEDEX 05, 13385 Marseille, France; (O.L.A.L.); (C.C.); (Y.K.); (S.R.)
- Service Central de la Qualité et de l’Information Pharmaceutiques (SCQIP), Pharmacy Department, Assistance Publique—Hôpitaux de Marseille (AP-HM), 147 Bd. Baille, 13006 Marseille, France
| |
Collapse
|
7
|
Zangari M, Piccirilli F, Vaccari L, Radu C, Zacchi P, Bernareggi A, Leone S, Zabucchi G, Borelli V. Ferritin adsorption onto chrysotile asbestos fibers influences the protein secondary structure. Heliyon 2024; 10:e38966. [PMID: 39492902 PMCID: PMC11530830 DOI: 10.1016/j.heliyon.2024.e38966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 11/05/2024] Open
Abstract
Asbestos fiber exposure triggers chronic inflammation and cancer. Asbestos fibers can adsorb different types of proteins. The mechanism of this adsorption, not yet completely understood, has been studied in detail mainly with serum albumin and was shown to induce structural changes in the bound protein. The findings of these works regarded mainly the changes of the protein structure, independently of any relation with asbestos-related diseases. For the first time, we have focused our attention to the consequences of the interaction between asbestos fibers and ferritin, a protein involved in iron metabolism, which is strongly modified in asbestos-related diseases. Even if it is known that ferritin can be adsorbed by asbestos fibers, the results of this interaction for the ferritin secondary structure has not previously been studied. One consequence of asbestos-ferritin interaction, is the formation of the so-called ferruginous/asbestos bodies (ABs). In the AB-coating material, the secondary structure of ferritin is modified, and at present, it is unclear whether or not this modification is a direct consequence of the asbestos interaction. In the present study, chrysotile asbestos, more than other asbestos fiber types tested, was found to rapidly bind holo-ferritin, and the presence of iron seemed to play a key role in this process, since iron-free apo-ferritin was adsorbed at a lower level, and iron-saturated chrysotile lost its ferritin-adsorbing capacity. To directly study the details of ferritin adsorption on asbestos fibers, High Resolution Transmission Electron Microscopy (HR-TEM) was employed together with FTIR microspectroscopy and Infrared nanoscopy, which to the best of our knowledge, have not previously been used for this purpose. Chrysotile-bound apo-ferritin underwent a significant change in secondary structure, showing a shift from a prevalent α-helix to a β-sheet conformation. Conversely, the adsorbed holo-ferritin structure appeared to be only weakly modified. These findings add a new potential mechanism to the toxic activities of asbestos: the fibers can modify the structure, and very likely, the function of adsorbed proteins. This, in relation to ferritin, could be a key mechanism in cell iron homeostasis alteration, typically reported in asbestos-related diseases.
Collapse
Affiliation(s)
- Martina Zangari
- Department of Physics, University of Trieste, 34127, Trieste, Italy
- CERIC-ERIC, S.S. 14 - km 163,5, 34149, Basovizza, Trieste, Italy
| | - Federica Piccirilli
- Area Science Park, Padriciano 99, 34149, Trieste, Italy
- Elettra Sincrotrone Trieste, S.S. 14 - km 163,5, 34149, Basovizza, Trieste, Italy
| | - Lisa Vaccari
- Elettra Sincrotrone Trieste, S.S. 14 - km 163,5, 34149, Basovizza, Trieste, Italy
| | - Cristian Radu
- National Institute of Materials Physics, Atomistilor 405A, 077125, Magurele, jud. Ilfov, Romania
| | - Paola Zacchi
- Department of Life Science, University of Trieste, via via Valerio 28-28/1, 34127, Trieste, Italy
| | - Annalisa Bernareggi
- Department of Life Science, University of Trieste, via via Valerio 28-28/1, 34127, Trieste, Italy
| | - Sara Leone
- Fiber laboratory, Azienda Sanitaria Giiuliano Isontina (ASUGI), Trieste, Italy
| | - Giuliano Zabucchi
- Department of Life Science, University of Trieste, via via Valerio 28-28/1, 34127, Trieste, Italy
| | - Violetta Borelli
- Department of Life Science, University of Trieste, via via Valerio 28-28/1, 34127, Trieste, Italy
| |
Collapse
|
8
|
Xie XD, Dong SS, Liu RJ, Shi LL, Zhu T. Mechanism of Efferocytosis in Determining Ischaemic Stroke Resolution-Diving into Microglia/Macrophage Functions and Therapeutic Modality. Mol Neurobiol 2024; 61:7583-7602. [PMID: 38409642 DOI: 10.1007/s12035-024-04060-4] [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] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/17/2024] [Indexed: 02/28/2024]
Abstract
After ischaemic cerebral vascular injury, efferocytosis-a process known as the efficient clearance of apoptotic cells (ACs) by various phagocytes in both physiological and pathological states-is crucial for maintaining central nervous system (CNS) homeostasis and regaining prognosis. The mechanisms of efferocytosis in ischaemic stroke and its influence on preventing inflammation progression from secondary injury were still not fully understood, despite the fact that the fundamental process of efferocytosis has been described in a series of phases, including AC recognition, phagocyte engulfment, and subsequent degradation. The genetic reprogramming of macrophages and brain-resident microglia after an ischaemic stroke has been equated by some researchers to that of the peripheral blood and brain. Based on previous studies, some molecules, such as signal transducer and activator of transcription 6 (STAT6), peroxisome proliferator-activated receptor γ (PPARG), CD300A, and sigma non-opioid intracellular receptor 1 (SIGMAR1), were discovered to be largely associated with aspects of apoptotic cell elimination and accompanying neuroinflammation, such as inflammatory cytokine release, phenotype transformation, and suppressing of antigen presentation. Exacerbated stroke outcomes are brought on by defective efferocytosis and improper modulation of pertinent signalling pathways in blood-borne macrophages and brain microglia, which also results in subsequent tissue inflammatory damage. This review focuses on recent researches which contain a number of recently discovered mechanisms, such as studies on the relationship between benign efferocytosis and the regulation of inflammation in ischaemic stroke, the roles of some risk factors in disease progression, and current immune approaches that aim to promote efferocytosis to treat some autoimmune diseases. Understanding these pathways provides insight into novel pathophysiological processes and fresh characteristics, which can be used to build cerebral ischaemia targeting techniques.
Collapse
Affiliation(s)
- Xiao-Di Xie
- Department of Pathophysiology, School of Basic Medicine, Institute of Neuroregeneration & Neurorehabilitation, Qingdao University, No. 308 Ningxia Road, Qingdao, China
| | - Shan-Shan Dong
- Department of Pathophysiology, School of Basic Medicine, Institute of Neuroregeneration & Neurorehabilitation, Qingdao University, No. 308 Ningxia Road, Qingdao, China
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ru-Juan Liu
- Department of Pathophysiology, School of Basic Medicine, Institute of Neuroregeneration & Neurorehabilitation, Qingdao University, No. 308 Ningxia Road, Qingdao, China
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Liu-Liu Shi
- Department of Pathophysiology, School of Basic Medicine, Institute of Neuroregeneration & Neurorehabilitation, Qingdao University, No. 308 Ningxia Road, Qingdao, China
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ting Zhu
- Department of Pathophysiology, School of Basic Medicine, Institute of Neuroregeneration & Neurorehabilitation, Qingdao University, No. 308 Ningxia Road, Qingdao, China.
| |
Collapse
|
9
|
Allsup BL, Gharpure S, Bryson BD. Proximity labeling defines the phagosome lumen proteome of murine and primary human macrophages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.04.611277. [PMID: 39282337 PMCID: PMC11398489 DOI: 10.1101/2024.09.04.611277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
Proteomic analyses of the phagosome has significantly improved our understanding of the proteins which contribute to critical phagosome functions such as apoptotic cell clearance and microbial killing. However, previous methods of isolating phagosomes for proteomic analysis have relied on cell fractionation with some intrinsic limitations. Here, we present an alternative and modular proximity-labeling based strategy for mass spectrometry proteomic analysis of the phagosome lumen, termed PhagoID. We optimize proximity labeling in the phagosome and apply PhagoID to immortalized murine macrophages as well as primary human macrophages. Analysis of proteins detected by PhagoID in murine macrophages demonstrate that PhagoID corroborates previous proteomic studies, but also nominates novel proteins with unexpected residence at the phagosome for further study. A direct comparison between the proteins detected by PhagoID between mouse and human macrophages further reveals that human macrophage phagosomes have an increased abundance of proteins involved in the oxidative burst and antigen presentation. Our study develops and benchmarks a new approach to measure the protein composition of the phagosome and validates a subset of these findings, ultimately using PhagoID to grant further insight into the core constituent proteins and species differences at the phagosome lumen.
Collapse
Affiliation(s)
- Benjamin L Allsup
- Department of Biological Engineering, MIT, Cambridge, USA
- Ragon Institute of Mass General, Harvard, and MIT, Cambridge, USA
| | - Supriya Gharpure
- Ragon Institute of Mass General, Harvard, and MIT, Cambridge, USA
| | - Bryan D Bryson
- Department of Biological Engineering, MIT, Cambridge, USA
- Ragon Institute of Mass General, Harvard, and MIT, Cambridge, USA
| |
Collapse
|
10
|
Nadeem A, Lyons S, Kindopp A, Jamieson A, Roxbury D. Machine Learning-Assisted Near-Infrared Spectral Fingerprinting for Macrophage Phenotyping. ACS NANO 2024; 18:22874-22887. [PMID: 39148286 PMCID: PMC12020776 DOI: 10.1021/acsnano.4c03387] [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: 08/17/2024]
Abstract
Spectral fingerprinting has emerged as a powerful tool that is adept at identifying chemical compounds and deciphering complex interactions within cells and engineered nanomaterials. Using near-infrared (NIR) fluorescence spectral fingerprinting coupled with machine learning techniques, we uncover complex interactions between DNA-functionalized single-walled carbon nanotubes (DNA-SWCNTs) and live macrophage cells, enabling in situ phenotype discrimination. Utilizing Raman microscopy, we showcase statistically higher DNA-SWCNT uptake and a significantly lower defect ratio in M1 macrophages compared to M2 and naive phenotypes. NIR fluorescence data also indicate that distinctive intraendosomal environments of these cell types give rise to significant differences in many optical features, such as emission peak intensities, center wavelengths, and peak intensity ratios. Such features serve as distinctive markers for identifying different macrophage phenotypes. We further use a support vector machine (SVM) model trained on SWCNT fluorescence data to identify M1 and M2 macrophages, achieving an impressive accuracy of >95%. Finally, we observe that the stability of DNA-SWCNT complexes, influenced by DNA sequence length, is a crucial consideration for applications, such as cell phenotyping or mapping intraendosomal microenvironments using AI techniques. Our findings suggest that shorter DNA-sequences like GT6 give rise to more improved model accuracy (>87%) due to increased active interactions of SWCNTs with biomolecules in the endosomal microenvironment. Implications of this research extend to the development of nanomaterial-based platforms for cellular identification, holding promise for potential applications in real time monitoring of in vivo cellular differentiation.
Collapse
Affiliation(s)
- Aceer Nadeem
- Department of Chemical Engineering, University of Rhode Island, Kingston, RI 02881 USA
| | - Sarah Lyons
- Department of Chemical Engineering, University of Rhode Island, Kingston, RI 02881 USA
| | - Aidan Kindopp
- Department of Chemical Engineering, University of Rhode Island, Kingston, RI 02881 USA
| | - Amanda Jamieson
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912 USA
| | - Daniel Roxbury
- Department of Chemical Engineering, University of Rhode Island, Kingston, RI 02881 USA
| |
Collapse
|
11
|
De Rubis G, Chakraborty A, Paudel KR, Wang C, Kannaujiya V, Wich PR, Hansbro PM, Samuel CS, Oliver B, Dua K. Exploring the anti-inflammatory and anti-fibrotic activity of NFκB decoy oligodeoxynucleotide-loaded spermine-functionalized acetalated nanoparticles. Chem Biol Interact 2024; 396:111059. [PMID: 38761875 DOI: 10.1016/j.cbi.2024.111059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 05/12/2024] [Accepted: 05/15/2024] [Indexed: 05/20/2024]
Abstract
Chronic inflammation, oxidative stress, and airway remodelling represent the principal pathophysiological features of chronic respiratory disorders. Inflammation stimuli like lipopolysaccharide (LPS) activate macrophages and dendritic cells, with concomitant M1 polarization and release of pro-inflammatory cytokines. Chronic inflammation and oxidative stress lead to airway remodelling causing irreversible functional and structural alterations of the lungs. Airway remodelling is multifactorial, however, the hormone transforming growth factor-β (TGF-β) is one of the main contributors to fibrotic changes. The signalling pathways mediating inflammation and remodelling rely both on the transcription factor nuclear factor-κB (NFκB), underlying the potential of NFκB inhibition as a therapeutic strategy for chronic respiratory disorders. In this study, we encapsulated an NFκB-inhibiting decoy oligodeoxynucleotide (ODN) in spermine-functionalized acetalated dextran (SpAcDex) nanoparticles and tested the in vitro anti-inflammatory and anti-remodelling activity of this formulation. We show that NF-κB ODN nanoparticles counteract inflammation by reversing LPS-induced expression of the activation marker CD40 in myeloid cells and counteracts remodelling features by reversing the TGF-β-induced expression of collagen I and α-smooth muscle actin in human dermal fibroblast. In summary, our study highlights the great potential of inhibiting NFκB via decoy ODN as a therapeutic strategy tackling multiple pathophysiological features underlying chronic respiratory conditions.
Collapse
Affiliation(s)
- Gabriele De Rubis
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Amlan Chakraborty
- Division of Immunology, Immunity to Infection and Respiratory Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PL, UK; Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, VIC, 3800, Australia.
| | - Keshav Raj Paudel
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, 2007, Australia
| | - Chao Wang
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, VIC, 3800, Australia
| | - Vinod Kannaujiya
- School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia; Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Peter Richard Wich
- School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia; Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Philip Michael Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, 2007, Australia
| | - Chrishan S Samuel
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, VIC, 3800, Australia
| | - Brian Oliver
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Woolcock Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia.
| |
Collapse
|
12
|
Zhang Y, Béland LC, Roussel S, Bertrand N, Hébert SS, Vallières L. Optimization of a lipid nanoparticle-based protocol for RNA transfection into primary mononuclear phagocytes. J Leukoc Biol 2024; 115:1165-1176. [PMID: 38466819 DOI: 10.1093/jleuko/qiae059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 02/07/2024] [Accepted: 02/01/2024] [Indexed: 03/13/2024] Open
Abstract
The effective delivery of synthetic RNA into mononuclear phagocytes is a prerequisite for experimental research and therapeutic development. However, traditional methods are highly ineffective and toxic for these cells. Here, we aimed to optimize a transfection protocol for primary bone marrow-derived phagocytes, specifically dendritic cells and macrophages, using lipid nanoparticles generated by microfluidics. Our results show that a lipid mixture similar to that used in Moderna's COVID-19 messenger RNA vaccine outperforms the others tested. Improved messenger RNA transfection can be achieved by replacing uridine with methylpseudouridine but not methoxyuridine, which interferes with transfection. The addition of diphenyleneiodonium or apocynin can enhance transfection in a cell type-dependent manner without adverse effects, while apolipoprotein E provides no added value. These optimized transfection conditions can also be used for microRNA agonists and antagonists. In sum, this study offers a straightforward, highly efficient, reproducible, and nontoxic protocol to deliver RNA into different primary mononuclear phagocytes in culture.
Collapse
Affiliation(s)
- Yu Zhang
- Neuroscience Unit, University Hospital Center of Quebec-Laval University, 2705 Laurier Boulevard, Quebec City, Quebec G1V 4G2, Canada
| | - Louis-Charles Béland
- Neuroscience Unit, University Hospital Center of Quebec-Laval University, 2705 Laurier Boulevard, Quebec City, Quebec G1V 4G2, Canada
| | - Sabrina Roussel
- Endocrinology and Nephrology Unit, University Hospital Center of Quebec-Laval University, 2705 Laurier Boulevard, Quebec City, Quebec G1V 4G2, Canada
| | - Nicolas Bertrand
- Endocrinology and Nephrology Unit, University Hospital Center of Quebec-Laval University, 2705 Laurier Boulevard, Quebec City, Quebec G1V 4G2, Canada
| | - Sébastien S Hébert
- Neuroscience Unit, University Hospital Center of Quebec-Laval University, 2705 Laurier Boulevard, Quebec City, Quebec G1V 4G2, Canada
| | - Luc Vallières
- Neuroscience Unit, University Hospital Center of Quebec-Laval University, 2705 Laurier Boulevard, Quebec City, Quebec G1V 4G2, Canada
| |
Collapse
|
13
|
Sheng Y, Hu W, Chen S, Zhu X. Efferocytosis by macrophages in physiological and pathological conditions: regulatory pathways and molecular mechanisms. Front Immunol 2024; 15:1275203. [PMID: 38779685 PMCID: PMC11109379 DOI: 10.3389/fimmu.2024.1275203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 04/17/2024] [Indexed: 05/25/2024] Open
Abstract
Efferocytosis is defined as the highly effective phagocytic removal of apoptotic cells (ACs) by professional or non-professional phagocytes. Tissue-resident professional phagocytes ("efferocytes"), such as macrophages, have high phagocytic capacity and are crucial to resolve inflammation and aid in homeostasis. Recently, numerous exciting discoveries have revealed divergent (and even diametrically opposite) findings regarding metabolic immune reprogramming associated with efferocytosis by macrophages. In this review, we highlight the key metabolites involved in the three phases of efferocytosis and immune reprogramming of macrophages under physiological and pathological conditions. The next decade is expected to yield further breakthroughs in the regulatory pathways and molecular mechanisms connecting immunological outcomes to metabolic cues as well as avenues for "personalized" therapeutic intervention.
Collapse
Affiliation(s)
- Yan−Ran Sheng
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Wen−Ting Hu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Siman Chen
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Xiao−Yong Zhu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, China
| |
Collapse
|
14
|
Tang Y, Wu X, Li J, Li Y, Xu X, Li G, Zhang P, Qin C, Wu LJ, Tang Z, Tian DS. The Emerging Role of Microglial Hv1 as a Target for Immunomodulation in Myelin Repair. Aging Dis 2024; 15:1176-1203. [PMID: 38029392 PMCID: PMC11081154 DOI: 10.14336/ad.2023.1107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 11/07/2023] [Indexed: 12/01/2023] Open
Abstract
In the central nervous system (CNS), the myelin sheath ensures efficient interconnection between neurons and contributes to the regulation of the proper function of neuronal networks. The maintenance of myelin and the well-organized subtle process of myelin plasticity requires cooperation among myelin-forming cells, glial cells, and neural networks. The process of cooperation is fragile, and the balance is highly susceptible to disruption by microenvironment influences. Reactive microglia play a critical and complicated role in the demyelination and remyelination process. Recent studies have shown that the voltage-gated proton channel Hv1 is selectively expressed in microglia in CNS, which regulates intracellular pH and is involved in the production of reactive oxygen species, underlying multifaceted roles in maintaining microglia function. This paper begins by examining the molecular mechanisms of demyelination and emphasizes the crucial role of the microenvironment in demyelination. It focuses specifically on the role of Hv1 in myelin repair and its therapeutic potential in CNS demyelinating diseases.
Collapse
Affiliation(s)
- Yingxin Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xuan Wu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jiarui Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yuanwei Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xiaoxiao Xu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Gaigai Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Ping Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Chuan Qin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Zhouping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Dai-Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
15
|
Bates TA, Trank-Greene M, Nguyenla X, Anastas A, Gurmessa SK, Merutka IR, Dixon SD, Shumate A, Groncki AR, Parson MAH, Ingram JR, Barklis E, Burke JE, Shinde U, Ploegh HL, Tafesse FG. ESAT-6 undergoes self-association at phagosomal pH and an ESAT-6 specific nanobody restricts M. tuberculosis growth in macrophages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.16.553641. [PMID: 37645775 PMCID: PMC10462100 DOI: 10.1101/2023.08.16.553641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Mycobacterium tuberculosis (Mtb) is known to survive within macrophages by compromising the integrity of the phagosomal compartment in which it resides. This activity primarily relies on the ESX-1 secretion system, predominantly involving the protein duo ESAT-6 and CFP-10. CFP-10 likely acts as a chaperone, while ESAT-6 likely disrupts phagosomal membrane stability via a largely unknown mechanism. we employ a series of biochemical analyses, protein modeling techniques, and a novel ESAT-6-specific nanobody to gain insight into the ESAT-6's mode of action. First, we measure the binding kinetics of the tight 1:1 complex formed by ESAT-6 and CFP-10 at neutral pH. Subsequently, we demonstrate a rapid self-association of ESAT-6 into large complexes under acidic conditions, leading to the identification of a stable tetrameric ESAT-6 species. Using molecular dynamics simulations, we pinpoint the most probable interaction interface. Furthermore, we show that cytoplasmic expression of an anti-ESAT-6 nanobody blocks Mtb replication, thereby underlining the pivotal role of ESAT-6 in intracellular survival. Together, these data suggest that ESAT-6 acts by a pH dependent mechanism to establish two-way communication between the cytoplasm and the Mtb-containing phagosome.
Collapse
Affiliation(s)
- Timothy A Bates
- Department of Molecular Microbiology and Immunology, Oregon Health & Sciences University, Portland, Oregon, United States
| | - Mila Trank-Greene
- Department of Molecular Microbiology and Immunology, Oregon Health & Sciences University, Portland, Oregon, United States
| | - Xammy Nguyenla
- Department of Molecular Microbiology and Immunology, Oregon Health & Sciences University, Portland, Oregon, United States
| | - Aidan Anastas
- Department of Molecular Microbiology and Immunology, Oregon Health & Sciences University, Portland, Oregon, United States
| | - Sintayehu K Gurmessa
- Department of Molecular Microbiology and Immunology, Oregon Health & Sciences University, Portland, Oregon, United States
| | - Ilaria R Merutka
- Department of Molecular Microbiology and Immunology, Oregon Health & Sciences University, Portland, Oregon, United States
| | - Shandee D Dixon
- Department of Molecular Microbiology and Immunology, Oregon Health & Sciences University, Portland, Oregon, United States
| | - Anthony Shumate
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, United States
| | - Abigail R Groncki
- Department of Molecular Microbiology and Immunology, Oregon Health & Sciences University, Portland, Oregon, United States
| | - Matthew AH Parson
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, Canada
| | - Jessica R Ingram
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Eric Barklis
- Department of Molecular Microbiology and Immunology, Oregon Health & Sciences University, Portland, Oregon, United States
| | - John E Burke
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, Canada
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, Canada
| | - Ujwal Shinde
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, United States
| | - Hidde L Ploegh
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Fikadu G Tafesse
- Department of Molecular Microbiology and Immunology, Oregon Health & Sciences University, Portland, Oregon, United States
| |
Collapse
|
16
|
Nadeem A, Lyons S, Kindopp A, Jamieson A, Roxbury D. Machine Learning Assisted Spectral Fingerprinting for Immune Cell Phenotyping. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.05.583608. [PMID: 38496523 PMCID: PMC10942323 DOI: 10.1101/2024.03.05.583608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Spectral fingerprinting has emerged as a powerful tool, adept at identifying chemical compounds and deciphering complex interactions within cells and engineered nanomaterials. Using near-infrared (NIR) fluorescence spectral fingerprinting coupled with machine learning techniques, we uncover complex interactions between DNA-functionalized single-walled carbon nanotubes (DNA-SWCNTs) and live macrophage cells, enabling in situ phenotype discrimination. Through the use of Raman microscopy, we showcase statistically higher DNA-SWCNT uptake and a significantly lower defect ratio in M1 macrophages as compared to M2 and naïve phenotypes. NIR fluorescence data also indicate that distinctive intra-endosomal environments of these cell types give rise to significant differences in many optical features such as emission peak intensities, center wavelengths, and peak intensity ratios. Such features serve as distinctive markers for identifying different macrophage phenotypes. We further use a support vector machine (SVM) model trained on SWCNT fluorescence data to identify M1 and M2 macrophages, achieving an impressive accuracy of > 95%. Finally, we observe that the stability of DNA-SWCNT complexes, influenced by DNA sequence length, is a crucial consideration for applications such as cell phenotyping or mapping intra-endosomal microenvironments using AI techniques. Our findings suggest that shorter DNA-sequences like GT 6 give rise to more improved model accuracy (> 87%) due to increased active interactions of SWCNTs with biomolecules in the endosomal microenvironment. Implications of this research extend to the development of nanomaterial-based platforms for cellular identification, holding promise for potential applications in real time monitoring of in vivo cellular differentiation. TOC Graphic
Collapse
|
17
|
Gour N, Yong HM, Magesh A, Atakkatan A, Andrade F, Lajoie S, Dong X. A GPCR-neuropeptide axis dampens hyperactive neutrophils by promoting an alternative-like polarization during bacterial infection. Immunity 2024; 57:333-348.e6. [PMID: 38295799 PMCID: PMC10940224 DOI: 10.1016/j.immuni.2024.01.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 10/10/2023] [Accepted: 01/08/2024] [Indexed: 02/16/2024]
Abstract
The notion that neutrophils exist as a homogeneous population is being replaced with the knowledge that neutrophils adopt different functional states. Neutrophils can have a pro-inflammatory phenotype or an anti-inflammatory state, but how these states are regulated remains unclear. Here, we demonstrated that the neutrophil-expressed G-protein-coupled receptor (GPCR) Mrgpra1 is a negative regulator of neutrophil bactericidal functions. Mrgpra1-mediated signaling was driven by its ligand, neuropeptide FF (NPFF), which dictated the balance between pro- and anti-inflammatory programming. Specifically, the Mrgpra1-NPFF axis counter-regulated interferon (IFN) γ-mediated neutrophil polarization during acute lung infection by favoring an alternative-like polarization, suggesting that it may act to balance overzealous neutrophilic responses. Distinct, cross-regulated populations of neutrophils were the primary source of NPFF and IFNγ during infection. As a subset of neutrophils at steady state expressed NPFF, these findings could have broad implications in various infectious and inflammatory diseases. Therefore, a neutrophil-intrinsic pathway determines their cellular fate, function, and magnitude of infection.
Collapse
Affiliation(s)
- Naina Gour
- Solomon H. Snyder Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Hwan Mee Yong
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Aishwarya Magesh
- Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD, USA; Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Aishwarya Atakkatan
- Solomon H. Snyder Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD, USA; Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Felipe Andrade
- Division of Rheumatology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Stephane Lajoie
- Department of Otolaryngology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Xinzhong Dong
- Solomon H. Snyder Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
18
|
Lecoultre M, Chliate S, Espinoza FI, Tankov S, Dutoit V, Walker PR. Radio-chemotherapy of glioblastoma cells promotes phagocytosis by macrophages in vitro. Radiother Oncol 2024; 190:110049. [PMID: 38072365 DOI: 10.1016/j.radonc.2023.110049] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 11/03/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023]
Abstract
BACKGROUND AND PURPOSE Immunotherapy is actively explored in glioblastoma (GBM) to improve patient prognosis. Tumor-associated macrophages (TAMs) are abundant in GBM and harnessing their function for anti-tumor immunity is of interest. They are plastic cells that are influenced by the tumor microenvironment, by radio-chemotherapy and by their own phagocytic activity. Indeed, the engulfment of necrotic cells promotes pro-inflammatory (and anti-tumoral) functions while the engulfment of apoptotic cells promotes anti-inflammatory (and pro-tumoral) functions through efferocytosis. MATERIALS AND METHODS To model the effect of radio-chemotherapy on the GBM microenvironment, we exposed human macrophages to supernatant of treated GBM cells in vitro. Macrophages were derived from human monocytes and GBM cells from patient-resected tumors. GBM cells were exposed to therapeutically relevant doses of irradiation and chemotherapy. Apoptosis and phagocytic activity were assessed by flow cytometry. RESULTS The phagocytic activity of macrophages was increased, and it was correlated with the proportion of apoptotic GBM cells producing the supernatant. Whether uptake of apoptotic tumor cells could occur would depend upon the expression of efferocytosis-associated receptors. Indeed, we showed that efferocytosis-associated receptors, such as AXL, were upregulated. CONCLUSIONS AND PERSPECTIVES We showed that macrophage phagocytic activity increased when exposed to supernatant from GBM cells treated by radio-chemotherapy. However, as efferocytosis-associated receptors were up-regulated, this effect could be deleterious for the anti-GBM immune response. We speculate that by inducing GBM cell apoptosis in parallel to an increase in efferocytosis receptor expression, the impact of radio-chemotherapy on phagocytic activity could promote anti-inflammatory and pro-tumoral TAM functions.
Collapse
Affiliation(s)
- Marc Lecoultre
- Faculty of Medicine, University of Geneva, Geneva, Switzerland; Immunobiology of Brain Tumours Laboratory, Center for Translational Research in Onco-Hematology, University of Geneva, Geneva Switzerland; Division of General Internal Medicine, Geneva University Hospital, Geneva, Switzerland
| | - Sylvie Chliate
- Faculty of Medicine, University of Geneva, Geneva, Switzerland; Immunobiology of Brain Tumours Laboratory, Center for Translational Research in Onco-Hematology, University of Geneva, Geneva Switzerland
| | - Felipe I Espinoza
- Faculty of Medicine, University of Geneva, Geneva, Switzerland; Immunobiology of Brain Tumours Laboratory, Center for Translational Research in Onco-Hematology, University of Geneva, Geneva Switzerland
| | - Stoyan Tankov
- Faculty of Medicine, University of Geneva, Geneva, Switzerland; Immunobiology of Brain Tumours Laboratory, Center for Translational Research in Onco-Hematology, University of Geneva, Geneva Switzerland
| | - Valérie Dutoit
- Faculty of Medicine, University of Geneva, Geneva, Switzerland; Immunobiology of Brain Tumours Laboratory, Center for Translational Research in Onco-Hematology, University of Geneva, Geneva Switzerland; Faculty of Medicine, Laboratory of Tumor Immunology and Center of Oncology, Geneva University Hospital, Geneva, Switzerland
| | - Paul R Walker
- Faculty of Medicine, University of Geneva, Geneva, Switzerland; Immunobiology of Brain Tumours Laboratory, Center for Translational Research in Onco-Hematology, University of Geneva, Geneva Switzerland.
| |
Collapse
|
19
|
Ni D, Zhou H, Wang P, Xu F, Li C. Visualizing Macrophage Phenotypes and Polarization in Diseases: From Biomarkers to Molecular Probes. PHENOMICS (CHAM, SWITZERLAND) 2023; 3:613-638. [PMID: 38223685 PMCID: PMC10781933 DOI: 10.1007/s43657-023-00129-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 08/06/2023] [Accepted: 08/10/2023] [Indexed: 01/16/2024]
Abstract
Macrophage is a kind of immune cell and performs multiple functions including pathogen phagocytosis, antigen presentation and tissue remodeling. To fulfill their functionally distinct roles, macrophages undergo polarization towards a spectrum of phenotypes, particularly the classically activated (M1) and alternatively activated (M2) subtypes. However, the binary M1/M2 phenotype fails to capture the complexity of macrophages subpopulations in vivo. Hence, it is crucial to employ spatiotemporal imaging techniques to visualize macrophage phenotypes and polarization, enabling the monitoring of disease progression and assessment of therapeutic responses to drug candidates. This review begins by discussing the origin, function and diversity of macrophage under physiological and pathological conditions. Subsequently, we summarize the identified macrophage phenotypes and their specific biomarkers. In addition, we present the imaging probes locating the lesions by visualizing macrophages with specific phenotype in vivo. Finally, we discuss the challenges and prospects associated with monitoring immune microenvironment and disease progression through imaging of macrophage phenotypes.
Collapse
Affiliation(s)
- Dan Ni
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, 201203 China
| | - Heqing Zhou
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Pengwei Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, 201203 China
| | - Fulin Xu
- Minhang Hospital, Fudan University, Shanghai, 201199 China
| | - Cong Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, 201203 China
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 201203 China
- Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Shanghai, 201203 China
| |
Collapse
|
20
|
Yu Y, Zhang Z, Yu Y. Timing of Phagosome Maturation Depends on Their Transport Switching from Actin to Microtubule Tracks. J Phys Chem B 2023; 127:9312-9322. [PMID: 37871280 PMCID: PMC10759163 DOI: 10.1021/acs.jpcb.3c05647] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Phagosomes, specialized membrane compartments responsible for digesting internalized pathogens, undergo sequential dynamic and biochemical changes as they mature from nascent phagosomes to degradative phagolysosomes. Maturation of phagosomes depends on their transport along actin filaments and microtubules. However, the specific quantitative relationship between the biochemical transformation and transport dynamics remains poorly characterized. The autonomous nature of phagosomes, moving and maturing at different rates, makes understanding this relationship challenging. Addressing this challenge, in this study we engineered particle sensors to image and quantify single phagosomes' maturation. We found that as phagosomes move from the actin cortex to microtubule tracks, the timing of their actin-to-microtubule transition governs the duration of the early phagosome stage before acquiring degradative capacities. Prolonged entrapment of phagosomes in the actin cortex extends the early phagosome stage by delaying the dissociation of early endosome markers and phagosome acidification. Conversely, a shortened transition from actin- to microtubule-based movements causes the opposite effect on phagosome maturation. These results suggest that the actin- and microtubule-based transport of phagosomes functions like a "clock" to coordinate the timing of biochemical events during phagosome maturation, which is crucial for effective pathogen degradation.
Collapse
Affiliation(s)
- Yanqi Yu
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405-7102, United States
| | - Zihan Zhang
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405-7102, United States
| | - Yan Yu
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405-7102, United States
| |
Collapse
|
21
|
Oates TCL, Moura PL, Cross S, Roberts K, Baum HE, Haydn‐Smith KL, Wilson MC, Heesom KJ, Severn CE, Toye AM. Defining the proteomic landscape of cultured macrophages and their polarization continuum. Immunol Cell Biol 2023; 101:947-963. [PMID: 37694300 PMCID: PMC10953363 DOI: 10.1111/imcb.12687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/28/2023] [Accepted: 08/18/2023] [Indexed: 09/12/2023]
Abstract
Macrophages have previously been characterized based on phenotypical and functional differences into suggested simplified subtypes of MØ, M1, M2a and M2c. These macrophage subtypes can be generated in a well-established primary monocyte culture model that produces cells expressing accepted subtype surface markers. To determine how these subtypes retain functional similarities and better understand their formation, we generated all four subtypes from the same donors. Comparative whole-cell proteomics confirmed that four distinct macrophage subtypes could be induced from the same donor material, with > 50% of 5435 identified proteins being significantly altered in abundance between subtypes. Functional assessment highlighted that these distinct protein expression profiles are primed to enable specific cell functions, indicating that this shifting proteome is predictive of meaningful changes in cell characteristics. Importantly, the 2552 proteins remained consistent in abundance across all macrophage subtypes examined, demonstrating maintenance of a stable core proteome that likely enables swift polarity changes. We next explored the cross-polarization capabilities of preactivated M1 macrophages treated with dexamethasone. Importantly, these treated cells undergo a partial repolarization toward the M2c surface markers but still retain the M1 functional phenotype. Our investigation of polarized macrophage subtypes therefore provides evidence of a sliding scale of macrophage functionality, with these data sets providing a valuable benchmark resource for further studies of macrophage polarity, with relevance for cell therapy development and drug discovery.
Collapse
Affiliation(s)
- Tiah CL Oates
- School of Biochemistry, Biomedical Sciences BuildingUniversity of BristolBristolUK
- National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Red Blood Cell ProductsUniversity of BristolBristolUK
| | - Pedro L Moura
- Center for Haematology and Regenerative Medicine, Department of Medicine (MedH)Karolinska InstitutetHuddingeSweden
| | | | - Kiren Roberts
- School of Biochemistry, Biomedical Sciences BuildingUniversity of BristolBristolUK
| | - Holly E Baum
- Max Planck Bristol Centre for Minimal Biology, School of ChemistryUniversity of BristolBristolUK
| | - Katy L Haydn‐Smith
- School of Biochemistry, Biomedical Sciences BuildingUniversity of BristolBristolUK
| | | | - Kate J Heesom
- Proteomics Facility, Biomedical Sciences BuildingUniversity of BristolBristolUK
| | - Charlotte E Severn
- School of Biochemistry, Biomedical Sciences BuildingUniversity of BristolBristolUK
- National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Red Blood Cell ProductsUniversity of BristolBristolUK
| | - Ashley M Toye
- School of Biochemistry, Biomedical Sciences BuildingUniversity of BristolBristolUK
- National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Red Blood Cell ProductsUniversity of BristolBristolUK
| |
Collapse
|
22
|
Bamgbose TT, Schilke RM, Igiehon OO, Nkadi EH, Custis D, Bharrhan S, Schwarz B, Bohrnsen E, Bosio CM, Scott RS, Yurdagul A, Finck BN, Woolard MD. Lipin-1 restrains macrophage lipid synthesis to promote inflammation resolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.23.563587. [PMID: 37961352 PMCID: PMC10634750 DOI: 10.1101/2023.10.23.563587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Macrophages are critical to maintaining and restoring tissue homeostasis during inflammation. The lipid metabolic state of macrophages influences their function, but a deeper understanding of how lipid metabolism is regulated in pro-resolving macrophage responses is needed. Lipin-1 is a phosphatidic acid phosphatase with a transcriptional coregulatory activity (TC) that regulates lipid metabolism. We previously demonstrated that lipin-1 supports pro-resolving macrophage responses, and here, myeloid-associated lipin-1 is required for inflammation resolution, yet how lipin-1-regulated cellular mechanisms promote macrophage pro-resolution responses is unknown. We demonstrated that the loss of lipin-1 in macrophages led to increased free fatty acid, neutral lipid, and ceramide content and increased phosphorylation of acetyl-CoA carboxylase. The inhibition of the first step of lipid synthesis and transport of citrate from the mitochondria in macrophages reduced lipid content and restored efferocytosis and inflammation resolution in lipin-1mKO macrophages and mice. Our findings suggest macrophage-associated lipin-1 restrains lipid synthesis, promoting pro-resolving macrophage function in response to pro-resolving stimuli.
Collapse
Affiliation(s)
- Temitayo T. Bamgbose
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Robert M. Schilke
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Oluwakemi O. Igiehon
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Ebubechukwu H. Nkadi
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - David Custis
- Research Core Facility, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Sushma Bharrhan
- Center for Applied Immunology and Pathological Processes (CAIPP), Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Benjamin Schwarz
- Proteins & Chemistry Section, Research and Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Disease, Hamilton, MT
| | - Eric Bohrnsen
- Proteins & Chemistry Section, Research and Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Disease, Hamilton, MT
| | - Catharine M. Bosio
- Immunity to Pulmonary Pathogens Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Disease, Hamilton, MT
| | - Rona S. Scott
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
- Center for Applied Immunology and Pathological Processes (CAIPP), Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Arif Yurdagul
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Brian N. Finck
- Division of Nutritional Sciences and Obesity Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Matthew D. Woolard
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
- Center for Applied Immunology and Pathological Processes (CAIPP), Louisiana State University Health Sciences Center, Shreveport, LA, United States
| |
Collapse
|
23
|
Cao C, Yin H, Yang B, Yue Q, Wu G, Gu M, Zhang Y, Fan Y, Dong X, Wang T, Wang C, Zhu X, Mao Y, Zhang X, Lei Z, Li C. Intra-Operative Definition of Glioma Infiltrative Margins by Visualizing Immunosuppressive Tumor-Associated Macrophages. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2304020. [PMID: 37544917 PMCID: PMC10558635 DOI: 10.1002/advs.202304020] [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: 06/19/2023] [Indexed: 08/08/2023]
Abstract
Accurate delineation of glioma infiltrative margins remains a challenge due to the low density of cancer cells in these regions. Here, a hierarchical imaging strategy to define glioma margins by locating the immunosuppressive tumor-associated macrophages (TAMs) is proposed. A pH ratiometric fluorescent probe CP2-M that targets immunosuppressive TAMs by binding to mannose receptor (CD206) is developed, and it subsequently senses the acidic phagosomal lumen, resulting in a remarkable fluorescence enhancement. With assistance of CP2-M, glioma xenografts in mouse models with a tumor-to-background ratio exceeding 3.0 for up to 6 h are successfully visualized. Furthermore, by intra-operatively mapping the pH distribution of exposed tissue after craniotomy, the glioma allograft in rat models is precisely excised. The overall survival of rat models significantly surpasses that achieved using clinically employed fluorescent probes. This work presents a novel strategy for locating glioma margins, thereby improving surgical outcomes for tumors with infiltrative characteristics.
Collapse
Affiliation(s)
- Chong Cao
- Key Laboratory of Smart Drug Delivery Ministry of EducationInnovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of EducationSchool of PharmacyDepartment of Neurosurgery, Huashan HospitalFudan UniversityShanghai201203China
| | - Hang Yin
- Key Laboratory of Smart Drug Delivery Ministry of EducationInnovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of EducationSchool of PharmacyDepartment of Neurosurgery, Huashan HospitalFudan UniversityShanghai201203China
| | - Biao Yang
- Key Laboratory of Smart Drug Delivery Ministry of EducationInnovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of EducationSchool of PharmacyDepartment of Neurosurgery, Huashan HospitalFudan UniversityShanghai201203China
| | - Qi Yue
- Key Laboratory of Smart Drug Delivery Ministry of EducationInnovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of EducationSchool of PharmacyDepartment of Neurosurgery, Huashan HospitalFudan UniversityShanghai201203China
| | - Guoqing Wu
- School of Information Science and TechnologyFudan UniversityShanghai200438China
| | - Meng Gu
- Key Laboratory of Smart Drug Delivery Ministry of EducationInnovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of EducationSchool of PharmacyDepartment of Neurosurgery, Huashan HospitalFudan UniversityShanghai201203China
| | - Yuwen Zhang
- Institute of Science and Technology for Brain‐Inspired IntelligenceMOE Key Laboratory of Computational Neuroscience and Brain‐Inspired IntelligenceMOE Frontiers Center for Brain ScienceFudan University220 Handan RoadShanghai200433China
| | - Yang Fan
- Key Laboratory of Smart Drug Delivery Ministry of EducationInnovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of EducationSchool of PharmacyDepartment of Neurosurgery, Huashan HospitalFudan UniversityShanghai201203China
| | - Xiaoyan Dong
- Key Laboratory of Smart Drug Delivery Ministry of EducationInnovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of EducationSchool of PharmacyDepartment of Neurosurgery, Huashan HospitalFudan UniversityShanghai201203China
| | - Ting Wang
- Key Laboratory of Smart Drug Delivery Ministry of EducationInnovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of EducationSchool of PharmacyDepartment of Neurosurgery, Huashan HospitalFudan UniversityShanghai201203China
| | - Cong Wang
- Key Laboratory of Smart Drug Delivery Ministry of EducationInnovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of EducationSchool of PharmacyDepartment of Neurosurgery, Huashan HospitalFudan UniversityShanghai201203China
| | - Xiao Zhu
- Key Laboratory of Smart Drug Delivery Ministry of EducationInnovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of EducationSchool of PharmacyDepartment of Neurosurgery, Huashan HospitalFudan UniversityShanghai201203China
| | - Ying Mao
- Key Laboratory of Smart Drug Delivery Ministry of EducationInnovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of EducationSchool of PharmacyDepartment of Neurosurgery, Huashan HospitalFudan UniversityShanghai201203China
| | - Xiao‐Yong Zhang
- Institute of Science and Technology for Brain‐Inspired IntelligenceMOE Key Laboratory of Computational Neuroscience and Brain‐Inspired IntelligenceMOE Frontiers Center for Brain ScienceFudan University220 Handan RoadShanghai200433China
| | - Zuhai Lei
- Key Laboratory of Smart Drug Delivery Ministry of EducationInnovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of EducationSchool of PharmacyDepartment of Neurosurgery, Huashan HospitalFudan UniversityShanghai201203China
| | - Cong Li
- Key Laboratory of Smart Drug Delivery Ministry of EducationInnovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of EducationSchool of PharmacyDepartment of Neurosurgery, Huashan HospitalFudan UniversityShanghai201203China
- State Key Laboratory of Medical NeurobiologyZhongshan HospitalFudan UniversityShanghai200032China
| |
Collapse
|
24
|
Mylvaganam S, Freeman SA. The resolution of phagosomes. Immunol Rev 2023; 319:45-64. [PMID: 37551912 DOI: 10.1111/imr.13260] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/18/2023] [Indexed: 08/09/2023]
Abstract
Phagocytosis is a fundamental immunobiological process responsible for the removal of harmful particulates. While the number of phagocytic events achieved by a single phagocyte can be remarkable, exceeding hundreds per day, the same phagocytic cells are relatively long-lived. It should therefore be obvious that phagocytic meals must be resolved in order to maintain the responsiveness of the phagocyte and to avoid storage defects. In this article, we discuss the mechanisms involved in the resolution process, including solute transport pathways and membrane traffic. We describe how products liberated in phagolysosomes support phagocyte metabolism and the immune response. We also speculate on mechanisms involved in the redistribution of phagosomal metabolites back to circulation. Finally, we highlight the pathologies owed to impaired phagosome resolution, which range from storage disorders to neurodegenerative diseases.
Collapse
Affiliation(s)
- Sivakami Mylvaganam
- Program in Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Spencer A Freeman
- Program in Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
25
|
Duan W, Wang C, Jiang Y, Sui A, Li Z, Wang L, Lei Z, Aime S, Yu J, Li C. A Ratiometric SERS Probe for Imaging the Macrophage Phenotypes in Live Mice with Epilepsy and Brain Tumor. Adv Healthc Mater 2023; 12:e2301000. [PMID: 37580893 DOI: 10.1002/adhm.202301000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/17/2023] [Indexed: 08/16/2023]
Abstract
Macrophage performs multiple functions such as pathogen phagocytosis, antigen presentation, and tissue remodeling by polarizing toward a spectrum of phenotypes. Dynamic imaging of macrophage phenotypes is critical for evaluating disease progression and the therapeutic response of drug candidates. However, current technologies cannot identify macrophage phenotypes in vivo. Herein, a surface-enhanced Raman scattering nanoprobe, AH1, which enables the accurate determination of physiological pH with high sensitivity and tissue penetration depth through ratiometric Raman signals is developed. Due to the phenotype-dependent metabolic reprogramming, AH1 can effectively identify macrophage subpopulations by measuring the acidity levels in phagosomes. After intravenous administration, AH1 not only visualizes the spatial distribution of macrophage phenotypes in brain tumors and epileptic regions of mouse models, but also reveals the repolarization of macrophages in brain lesions after drug intervention. This work provides a new tool for dynamically monitoring the disease-associated immune microenvironment and evaluating the efficacy of immune-therapeutics in vivo.
Collapse
Affiliation(s)
- Wenjia Duan
- Key Laboratory of Smart Drug Delivery, Ministry of Education; Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
- Department of Forensic Toxicology, Academy of Forensic Science, Ministry of Justice, Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Science Platform, Shanghai, 200063, China
| | - Cong Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education; Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
- State Key Laboratory of Medical Neurobiology, Zhongshan Hospital, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 201203, China
| | - Yiqing Jiang
- Key Laboratory of Smart Drug Delivery, Ministry of Education; Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - An Sui
- School of Information Science and Technology, Fudan University, Shanghai, 200433, China
| | - Zhi Li
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Lu Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education; Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Zuhai Lei
- Key Laboratory of Smart Drug Delivery, Ministry of Education; Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Silvio Aime
- Department of Molecular Biotechnologies, Health Sciences Molecular Imaging Center, University of Torino, Torino, 10126, Italy
| | - Jinhua Yu
- School of Information Science and Technology, Fudan University, Shanghai, 200433, China
| | - Cong Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education; Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
- State Key Laboratory of Medical Neurobiology, Zhongshan Hospital, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 201203, China
| |
Collapse
|
26
|
Méndez-Alejandre A, Raymond BBA, Trost M, Marín-Rubio JL. Bi-functional particles for real-time phagosome acidification and proteolysis multiplex assay in macrophages. Front Immunol 2023; 14:1204223. [PMID: 37638042 PMCID: PMC10456865 DOI: 10.3389/fimmu.2023.1204223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/17/2023] [Indexed: 08/29/2023] Open
Abstract
Phagosome acidification and proteolysis are essential processes in the immune response to contain and eliminate pathogens. In recent years, there has been an increased desire for a rapid and accurate method of assessing these processes in real-time. Here, we outline the development of a multiplexed assay that allows simultaneous monitoring of phagosome acidification and proteolysis in the same sample using silica beads conjugated to pHrodo and DQ BSA. We describe in detail how to prepare the bi-functional particles and show proof of concept using differentially activated macrophages. This multiplexed spectrophotometric assay allows rapid and accurate assessment of phagosome acidification and proteolysis in real-time and could provide valuable information for understanding the immune response to pathogen invasion.
Collapse
Affiliation(s)
- Alba Méndez-Alejandre
- Laboratory for Biological Mass Spectrometry, Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, United Kingdom
- Biology Department, Autonomous University of Madrid, Madrid, Spain
| | | | - Matthias Trost
- Laboratory for Biological Mass Spectrometry, Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - José Luis Marín-Rubio
- Laboratory for Biological Mass Spectrometry, Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| |
Collapse
|
27
|
Wu X, Singla S, Liu JJ, Hong L. The role of macrophage ion channels in the progression of atherosclerosis. Front Immunol 2023; 14:1225178. [PMID: 37588590 PMCID: PMC10425548 DOI: 10.3389/fimmu.2023.1225178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/10/2023] [Indexed: 08/18/2023] Open
Abstract
Atherosclerosis is a complex inflammatory disease that affects the arteries and can lead to severe complications such as heart attack and stroke. Macrophages, a type of immune cell, play a crucial role in atherosclerosis initiation and progression. Emerging studies revealed that ion channels regulate macrophage activation, polarization, phagocytosis, and cytokine secretion. Moreover, macrophage ion channel dysfunction is implicated in macrophage-derived foam cell formation and atherogenesis. In this context, exploring the regulatory role of ion channels in macrophage function and their impacts on the progression of atherosclerosis emerges as a promising avenue for research. Studies in the field will provide insights into novel therapeutic targets for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Xin Wu
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Sidhant Singla
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Jianhua J. Liu
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, United States
| | - Liang Hong
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
28
|
Zhang K, Sowers ML, Cherryhomes EI, Singh VK, Mishra A, Restrepo BI, Khan A, Jagannath C. Sirtuin-dependent metabolic and epigenetic regulation of macrophages during tuberculosis. Front Immunol 2023; 14:1121495. [PMID: 36993975 PMCID: PMC10040548 DOI: 10.3389/fimmu.2023.1121495] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 02/01/2023] [Indexed: 03/14/2023] Open
Abstract
Macrophages are the preeminent phagocytic cells which control multiple infections. Tuberculosis a leading cause of death in mankind and the causative organism Mycobacterium tuberculosis (MTB) infects and persists in macrophages. Macrophages use reactive oxygen and nitrogen species (ROS/RNS) and autophagy to kill and degrade microbes including MTB. Glucose metabolism regulates the macrophage-mediated antimicrobial mechanisms. Whereas glucose is essential for the growth of cells in immune cells, glucose metabolism and its downsteam metabolic pathways generate key mediators which are essential co-substrates for post-translational modifications of histone proteins, which in turn, epigenetically regulate gene expression. Herein, we describe the role of sirtuins which are NAD+-dependent histone histone/protein deacetylases during the epigenetic regulation of autophagy, the production of ROS/RNS, acetyl-CoA, NAD+, and S-adenosine methionine (SAM), and illustrate the cross-talk between immunometabolism and epigenetics on macrophage activation. We highlight sirtuins as emerging therapeutic targets for modifying immunometabolism to alter macrophage phenotype and antimicrobial function.
Collapse
Affiliation(s)
- Kangling Zhang
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Mark L. Sowers
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Ellie I. Cherryhomes
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Vipul K. Singh
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, United States
| | - Abhishek Mishra
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, United States
| | - Blanca I. Restrepo
- University of Texas Health Houston, School of Public Health, Brownsville, TX, United States
| | - Arshad Khan
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, United States
| | - Chinnaswamy Jagannath
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, United States
| |
Collapse
|
29
|
Hickman E, Smyth T, Cobos-Uribe C, Immormino R, Rebuli ME, Moran T, Alexis NE, Jaspers I. Expanded characterization of in vitro polarized M0, M1, and M2 human monocyte-derived macrophages: Bioenergetic and secreted mediator profiles. PLoS One 2023; 18:e0279037. [PMID: 36862675 PMCID: PMC9980743 DOI: 10.1371/journal.pone.0279037] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 11/28/2022] [Indexed: 03/03/2023] Open
Abstract
Respiratory macrophage subpopulations exhibit unique phenotypes depending on their location within the respiratory tract, posing a challenge to in vitro macrophage model systems. Soluble mediator secretion, surface marker expression, gene signatures, and phagocytosis are among the characteristics that are typically independently measured to phenotype these cells. Bioenergetics is emerging as a key central regulator of macrophage function and phenotype but is often not included in the characterization of human monocyte-derived macrophage (hMDM) models. The objective of this study was to expand the phenotype characterization of naïve hMDMs, and their M1 and M2 subsets by measuring cellular bioenergetic outcomes and including an expanded cytokine profile. Known markers of M0, M1 and M2 phenotypes were also measured and integrated into the phenotype characterization. Peripheral blood monocytes from healthy volunteers were differentiated into hMDM and polarized with either IFN-γ + LPS (M1) or IL-4 (M2). As expected, our M0, M1, and M2 hMDMs exhibited cell surface marker, phagocytosis, and gene expression profiles indicative of their different phenotypes. M2 hMDMs however were uniquely characterized and different from M1 hMDMs by being preferentially dependent on oxidativte phosphorylation for their ATP generation and by secreting a distinct cluster of soluble mediators (MCP4, MDC, and TARC). In contrast, M1 hMDMs secreted prototypic pro-inflammatory cytokines (MCP1, eotaxin, eotaxin-3, IL12p70, IL-1α, IL15, TNF-β, IL-6, TNF-α, IL12p40, IL-13, and IL-2), but demonstrated a relatively constitutively heightened bioenergetic state, and relied on glycolysis for ATP generation. These data are similar to the bioenergetic profiles we previously observed in vivo in sputum (M1) and BAL (M2)-derived macrophages in healthy volunteers, supporting the notion that polarized hMDMs can provide an acceptable in vitro model to study specific human respiratory macrophage subtypes.
Collapse
Affiliation(s)
- Elise Hickman
- Center for Environmental Medicine, Asthma, and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Curriculum in Toxicology & Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Timothy Smyth
- Center for Environmental Medicine, Asthma, and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Curriculum in Toxicology & Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Catalina Cobos-Uribe
- Center for Environmental Medicine, Asthma, and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Curriculum in Toxicology & Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Robert Immormino
- Center for Environmental Medicine, Asthma, and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Meghan E. Rebuli
- Center for Environmental Medicine, Asthma, and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Curriculum in Toxicology & Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Timothy Moran
- Center for Environmental Medicine, Asthma, and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Curriculum in Toxicology & Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Neil E. Alexis
- Center for Environmental Medicine, Asthma, and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Curriculum in Toxicology & Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Ilona Jaspers
- Center for Environmental Medicine, Asthma, and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Curriculum in Toxicology & Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
30
|
Baruah M, Kwon HY, Cho H, Chang YT, Samanta A. A Photoinduced Electron Transfer-Based Hypochlorite-Specific Fluorescent Probe for Selective Imaging of Proinflammatory M1 in a Rheumatoid Arthritis Model. Anal Chem 2023; 95:4147-4154. [PMID: 36800528 DOI: 10.1021/acs.analchem.2c05218] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
The differentiation of the distinct phenotypes of macrophages is essential for monitoring the stage of inflammatory diseases for accurate diagnosis and treatment. Recent studies revealed that the level of hypochlorite (OCl-) varies from activated M1 macrophages (killing pathogens) to M2 (resolution of inflammation) during inflammation. Thus, we developed a simple and efficient fluorescent probe for discriminating M1 from M0 and M2. Herein, fluorescent-based imaging is applied as an alternative to immunohistochemistry, which is challenging due to the tedious process and high cost. We developed a hypochlorite-specific probe PMS-T to differentiate M1 and M2, employing a metabolism-oriented live-cell distinction. This probe enables the detection of inflammatory rheumatoid arthritis in an ex vivo mouse model. Thus, it can be a potential chemical tool for monitoring inflammatory diseases, including rheumatoid arthritis, that may overcome the existing barriers of immunohistochemistry.
Collapse
Affiliation(s)
- Mousumi Baruah
- Molecular Sensors and Therapeutics (MST) Research Laboratory, Department of Chemistry, School of Natural Sciences, Shiv Nadar University, Delhi NCR, NH 91, Tehsil Dadri, Greater Noida, Uttar Pradesh 201314, India
| | - Haw-Young Kwon
- Center for Self-Assembly and Complexity, Institute for Basic Science (IBS), Pohang 37673, Republic of Korea
| | - Heewon Cho
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Young-Tae Chang
- Center for Self-Assembly and Complexity, Institute for Basic Science (IBS), Pohang 37673, Republic of Korea.,Department of Chemistry, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Animesh Samanta
- Molecular Sensors and Therapeutics (MST) Research Laboratory, Department of Chemistry, School of Natural Sciences, Shiv Nadar University, Delhi NCR, NH 91, Tehsil Dadri, Greater Noida, Uttar Pradesh 201314, India
| |
Collapse
|
31
|
Kendall RL, Ray JL, Hamilton RF, Holian A. Self-replicating murine ex vivo cultured alveolar macrophages as a model for toxicological studies of particle-induced inflammation. Toxicol Appl Pharmacol 2023; 461:116400. [PMID: 36702314 PMCID: PMC10022441 DOI: 10.1016/j.taap.2023.116400] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 01/25/2023]
Abstract
Alveolar macrophages (AM) are integral to maintaining homeostasis within the lungs following exposure to inhaled particles. However, due to the high animal number requirements for in vitro research with primary AM, there remains a need for validated cell models that replicate alveolar macrophages in form and function to better understand the mechanisms that contribute to particle-induced inflammation and disease. A novel, easily adaptable, culture model that facilitates the continued expansion of murine alveolar macrophages for several months, termed murine ex vivo cultured AM (mexAM) has been recently described. Therefore, the present work evaluated the use of mexAMs as a suitable model for primary AM interactions with nano- and micro-sized particles. mexAM displayed a comparable profile of functional phenotype gene expression as primary AM and similar particle uptake capabilities. The NLRP3 inflammasome-driven IL-1β inflammatory response to crystalline silica and various nanoparticles was also assessed, as well as the effects of cationic amphiphilic drugs to block particle-induced inflammation. For all endpoints, mexAM showed a comparable response to primary AM. Altogether, the present work supports the use of mexAM as a validated replacement for primary AM cultures thereby reducing animal numbers and serving as an effective model for mechanistic investigation of inflammatory pathways in particle-induced respiratory disease.
Collapse
Affiliation(s)
- Rebekah L Kendall
- Center for Environmental Health Sciences, University of Montana, Missoula, MT, United States of America.
| | - Jessica L Ray
- Center for Environmental Health Sciences, University of Montana, Missoula, MT, United States of America
| | - Raymond F Hamilton
- Center for Environmental Health Sciences, University of Montana, Missoula, MT, United States of America
| | - Andrij Holian
- Center for Environmental Health Sciences, University of Montana, Missoula, MT, United States of America
| |
Collapse
|
32
|
Roy S, Sharma A, Ghosh S. Mechanistic crosstalk of extracellular calcium-mediated regulation of maturation and plasticity in human monocytes. Biochem Biophys Res Commun 2023; 643:39-47. [PMID: 36586157 DOI: 10.1016/j.bbrc.2022.12.054] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/19/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Innate immune cells play a pivotal role in controlling tissue repair and rejection after biomaterial implantation. Calcium supplementation regulates cellular responses and alter the pathophysiology of various diseases. A series of macrophage activations through differential plasticity has been observed after cell-to-material interactions. We investigated the role of calcium supplementation in controlling macrophage phenotypes in pro-inflammatory and pre-reparative states. Oxidative defence and mitochondria involvement in cellular plasticity and the sequential M0 to M1 and M1 to M2 transitions were observed after calcium supplementation. This study describes the molecular mechanism of reactive oxygen species and drives the interconnected cellular plasticity of macrophages in the presence of calcium. Gene expression, and immunostaining, revealed a relationship between MHC class II maturation and cellular plasticity. This study elucidated the role of controlled calcium supplementation under various conditions. These findings underscore the molecular mechanism of calcium-mediated immune induction and its favourable use in different calcium-containing biomaterials., essential for tissue regeneration.
Collapse
Affiliation(s)
- Subhadeep Roy
- Regenerative Engineering Laboratory, Department of Textile and Fiber Engineering, Indian Institute of Technology, New Delhi, 110016, India
| | - Aarushi Sharma
- Regenerative Engineering Laboratory, Department of Textile and Fiber Engineering, Indian Institute of Technology, New Delhi, 110016, India
| | - Sourabh Ghosh
- Regenerative Engineering Laboratory, Department of Textile and Fiber Engineering, Indian Institute of Technology, New Delhi, 110016, India.
| |
Collapse
|
33
|
Lail SS, Balce DR, Canton J, Yates RM. Approaches to Measuring Reductive and Oxidative Events in Phagosomes. Methods Mol Biol 2023; 2692:139-152. [PMID: 37365466 DOI: 10.1007/978-1-0716-3338-0_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
The phagosome is a redox-active organelle. Numerous reductive and oxidative systems play both direct and indirect roles in phagosomal function. With the advent of newer methodologies to study these redox events in live cells, the details of how redox conditions change within the maturing phagosome, how they are regulated, and how they influence other phagosomal functions can be investigated. In this chapter, we detail phagosome-specific, fluorescence-based assays that measure disulfide reduction and the production of reactive oxygen species in live phagocytes such as macrophages and dendritic cells, in real time.
Collapse
Affiliation(s)
- Shranjit S Lail
- Department of Medical Science, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Dale R Balce
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Johnathan Canton
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute of Chronic Disease, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Robin M Yates
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada.
- Snyder Institute of Chronic Disease, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
34
|
Pacifici N, Cruz-Acuña M, Diener A, Tu A, Senthil N, Han H, Lewis JS. Vomocytosis of Cryptococcus neoformans cells from murine, bone marrow-derived dendritic cells. PLoS One 2023; 18:e0280692. [PMID: 36928392 PMCID: PMC10019626 DOI: 10.1371/journal.pone.0280692] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 01/05/2023] [Indexed: 03/18/2023] Open
Abstract
Cryptococcus neoformans (CN) cells survive within the acidic phagolysosome of macrophages (MΦ) for extended times, then escape without impacting the viability of the host cell via a phenomenon that has been coined 'vomocytosis'. Through this mechanism, CN disseminate throughout the body, sometimes resulting in a potentially fatal condition-Cryptococcal Meningitis (CM). Justifiably, vomocytosis studies have focused primarily on MΦ, as alveolar MΦ within the lung act as first responders that ultimately expel this fungal pathogen. Herein, we hypothesize that dendritic cells (DCs), an innate immune cell with attributes that include phagocytosis and antigen presentation, can also act as 'vomocytes'. Presciently, this report shows that vomocytosis of CN indeed occurs from murine, bone marrow-derived DCs. Primarily through time-lapse microscopy imaging, we show that rates of vomocytosis events from DCs are comparable to those seen from MΦ and further, are independent of the presence of the CN capsule and infection ratios. Moreover, the phagosome-altering drug bafilomycin A inhibits this phenomenon from DCs. Although DC immunophenotype does not affect the total number of vomocytic events, we observed differences in the numbers of CN per phagosome and expulsion times. Interestingly, these observations were similar in murine, bone marrow-derived MΦ. This work not only demonstrates the vomocytic ability of DCs, but also investigates the complexity of vomocytosis regulation in this cell type and MΦ under multiple modulatory conditions. Understanding the vomocytic behavior of different phagocytes and their phenotypic subtypes is needed to help elucidate the full picture of the dynamic interplay between CN and the immune system. Critically, deeper insight into vomocytosis could reveal novel approaches to treat CM, as well as other immune-related conditions.
Collapse
Affiliation(s)
- Noah Pacifici
- Department of Biomedical Engineering, University of California—Davis, Davis, CA, United States of America
| | - Melissa Cruz-Acuña
- Department of Biomedical Engineering, University of California—Davis, Davis, CA, United States of America
| | - Agustina Diener
- Department of Biomedical Engineering, University of California—Davis, Davis, CA, United States of America
| | - Allen Tu
- Department of Biomedical Engineering, University of California—Davis, Davis, CA, United States of America
| | - Neeraj Senthil
- Department of Biomedical Engineering, University of California—Davis, Davis, CA, United States of America
| | - Hyunsoo Han
- Department of Biomedical Engineering, University of California—Davis, Davis, CA, United States of America
| | - Jamal S. Lewis
- Department of Biomedical Engineering, University of California—Davis, Davis, CA, United States of America
- J. Crayton Pruitt Family Department of Biomedical Engineering, Gainesville, FL, United States of America
- * E-mail:
| |
Collapse
|
35
|
Gonzales GA, Canton J. Measuring Phagosomal pH by Fluorescence Microscopy. Methods Mol Biol 2023; 2692:153-169. [PMID: 37365467 DOI: 10.1007/978-1-0716-3338-0_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
Dual-wavelength and dual-fluorophore ratiometric imaging has become a powerful tool for the study of pH in intracellular compartments. It allows for the dynamic imaging of live cells while accounting for changes in the focal plane, differential loading of the fluorescent probe, and photobleaching caused by repeated image acquisitions. Ratiometric microscopic imaging has the added advantage over whole-population methods of being able to resolve individual cells and even individual organelles. In this chapter, we provide a detailed discussion of the basic principles of ratiometric imaging and its application to the measurement of phagosomal pH, including probe selection, the necessary instrumentation, and calibration methods.
Collapse
Affiliation(s)
- Gerone A Gonzales
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Johnathan Canton
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada.
- Calvin, Joan and Phoebe Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
36
|
Torres A, Collin-Faure V, Fenel D, Sergent JA, Rabilloud T. About the Transient Effects of Synthetic Amorphous Silica: An In Vitro Study on Macrophages. Int J Mol Sci 2022; 24:ijms24010220. [PMID: 36613664 PMCID: PMC9820141 DOI: 10.3390/ijms24010220] [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: 09/27/2022] [Revised: 12/17/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022] Open
Abstract
Silica (either crystalline or amorphous) is widely used for different applications and its toxicological assessment depends on its characteristics and intended use. As sustained inflammation induced by crystalline silica is at the root of silicosis, investigating the inflammatory effects induced by amorphous silicas and their persistence is needed. For the development of new grades of synthetic amorphous silicas, it is also desirable to be able to understand better the factors underlying potential adverse effects. Therefore, we used an optimized in vitro macrophage system to investigate the effects of amorphous silicas, and their persistence. By using different amorphous silicas, we demonstrated that the main driver for the adverse effects is a low size of the overall particle/agglomerate; the second driver being a low size of the primary particle. We also demonstrated that the effects were transient. By using silicon dosage in cells, we showed that the transient effects are coupled with a decrease of intracellular silicon levels over time after exposure. To further investigate this phenomenon, a mild enzymatic cell lysis allowed us to show that amorphous silicas are degraded in macrophages over time, explaining the decrease in silicon content and thus the transiency of the effects of amorphous silicas on macrophages.
Collapse
Affiliation(s)
- Anaëlle Torres
- Solvay/GBU Silica, 69003 Lyon, France
- Chemistry and Biology of Metals, University Grenoble Alpes, CNRS UMR5249, CEA, IRIG-LCBM, 38054 Grenoble, France
- Correspondence: (A.T.); (T.R.)
| | - Véronique Collin-Faure
- Chemistry and Biology of Metals, University Grenoble Alpes, CNRS UMR5249, CEA, IRIG-LCBM, 38054 Grenoble, France
| | - Daphna Fenel
- Institut de Biologie Structurale, Université Grenoble Alpes, CEA, CNRS, 38000 Grenoble, France
- Integrated Structural Biology Grenoble (ISBG) CNRS, CEA, Université Grenoble Alpes, EMBL, 38000 Grenoble, France
| | | | - Thierry Rabilloud
- Chemistry and Biology of Metals, University Grenoble Alpes, CNRS UMR5249, CEA, IRIG-LCBM, 38054 Grenoble, France
- Correspondence: (A.T.); (T.R.)
| |
Collapse
|
37
|
Miyazaki H, Kinoshita M, Nakashima H, Nakamura S, Saitoh D. Pioglitazone Modifies Kupffer Cell Function and Protects against Escherichia coli-Induced Bacteremia in Burned Mice. Int J Mol Sci 2022; 23:12746. [PMID: 36361535 PMCID: PMC9657905 DOI: 10.3390/ijms232112746] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/12/2022] [Accepted: 10/20/2022] [Indexed: 01/24/2023] Open
Abstract
Infectious complications and subsequent sepsis in severely burned patients lead to high morbidity and mortality in response to uncontrolled innate immune responses mediated by macrophages. Peroxisome proliferator-activated receptor gamma (PPARγ) has anti-inflammatory activity and acts as a master regulator of macrophage polarization. In this study, we investigated whether the administration of a PPARγ agonist could modulate the Kupffer cell phenotype and thereby ameliorate the dysregulated innate response during post-burn bacterial infection. C57BL/6 mice were subjected to severe burns and randomized to receive either the PPARγ agonist, pioglitazone, or the vehicle control five days after injury, followed by the subsequent analysis of hepatic macrophages. Survival from the bacterial infection was monitored for seven days. Pioglitazone protected burned mice against bacterial infection. A single treatment with pioglitazone significantly enhanced phagocytosis, phagosome acidification, bacterial clearance, and reduction in inflammatory mediators in Kupffer cells. In conclusion, PPARγ activation by pioglitazone prevents clinical deterioration due to post-burn bacterial infection and improves survival. Our findings suggest that pioglitazone may be an effective therapeutic candidate for post-burn infectious complications.
Collapse
Affiliation(s)
- Hiromi Miyazaki
- Division of Biomedical Engineering, National Defense Medical College Research Institute, Saitama 359-8513, Japan
| | - Manabu Kinoshita
- Department of Immunology and Microbiology, National Defense Medical College, Saitama 359-8513, Japan
| | - Hiroyuki Nakashima
- Department of Immunology and Microbiology, National Defense Medical College, Saitama 359-8513, Japan
| | - Shingo Nakamura
- Division of Biomedical Engineering, National Defense Medical College Research Institute, Saitama 359-8513, Japan
| | - Daizoh Saitoh
- Division of Traumatology, National Defense Medical College Research Institute, Saitama 359-8513, Japan
| |
Collapse
|
38
|
Staphylococcus lugdunensis Uses the Agr Regulatory System to Resist Killing by Host Innate Immune Effectors. Infect Immun 2022; 90:e0009922. [PMID: 36069592 PMCID: PMC9584346 DOI: 10.1128/iai.00099-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Coagulase-negative staphylococci (CoNS) are frequently commensal bacteria that rarely cause disease in mammals. Staphylococcus lugdunensis is an exceptional CoNS that causes disease in humans similar to virulent Staphylococcus aureus, but the factors that enhance the virulence of this bacterium remain ill defined. Here, we used random transposon insertion mutagenesis to identify the agr quorum sensing system as a regulator of hemolysins in S. lugdunensis. Using RNA sequencing (RNA-seq), we revealed that agr regulates dozens of genes, including hemolytic S. lugdunensis synergistic hemolysins (SLUSH) peptides and the protease lugdulysin. A murine bacteremia model was used to show that mice infected systemically with wild-type S. lugdunensis do not show overt signs of disease despite there being high numbers of bacteria in the livers and kidneys of mice. Moreover, proliferation of the agr mutant in these organs was no different from that of the wild-type strain, leaving the role of the SLUSH peptides and the metalloprotease lugdulysin in pathogenesis still unclear. Nonetheless, the tropism of S. lugdunensis for humans led us to investigate the role of virulence factors in other ways. We show that agr-regulated effectors, but not SLUSH or lugdulysin alone, are important for S. lugdunensis survival in whole human blood. Moreover, we demonstrate that Agr contributes to survival of S. lugdunensis during encounters with murine and primary human macrophages. These findings demonstrate that, in S. lugdunensis, Agr regulates expression of virulence factors and is required for resistance to host innate antimicrobial defenses. This study therefore provides insight into strategies that this Staphylococcus species uses to cause disease.
Collapse
|
39
|
Theranostic Properties of Crystalline Aluminum Phthalocyanine Nanoparticles as a Photosensitizer. Pharmaceutics 2022; 14:pharmaceutics14102122. [PMID: 36297557 PMCID: PMC9611939 DOI: 10.3390/pharmaceutics14102122] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 09/28/2022] [Accepted: 10/05/2022] [Indexed: 12/03/2022] Open
Abstract
The study of phthalocyanines, known photosensitizers, for biomedical applications has been of high research interest for several decades. Of specific interest, nanophotosensitizers are crystalline aluminum phthalocyanine nanoparticles (AlPc NPs). In crystalline form, they are water-insoluble and atoxic, but upon contact with tumors, immune cells, or pathogenic microflora, they change their spectroscopic properties (acquire the ability to fluoresce and become phototoxic), which makes them upcoming agents for selective phototheranostics. Aqueous colloids of crystalline AlPc NPs with a hydrodynamic size of 104 ± 54 nm were obtained using ultrasonic dispersal and centrifugation. Intracellular accumulation and localization of AlPc were studied on HeLa and THP-1 cell cultures and macrophages (M0, M1, M2) by fluorescence microscopy. Crystallinity was assessed by XRD spectroscopy. Time-resolved spectroscopy was used to obtain characteristic fluorescence kinetics of AlPc NPs upon interaction with cell cultures. The photodynamic efficiency and fluorescence quantum yield of AlPc NPs in HeLa and THP-1 cells were evaluated. After entering the cells, AlPc NPs localized in lysosomes and fluorescence corresponding to individual AlPc molecules were observed, as well as destruction of lysosomes and a rapid decrease in fluorescence intensity during photodynamic action. The photodynamic efficiency of AlPc NPs in THP-1 cells was almost 1.8-fold that of the molecular form of AlPc (Photosens). A new mechanism for the occurrence of fluorescence and phototoxicity of AlPc NPs in interaction with cells is proposed.
Collapse
|
40
|
Yu Y, Zhang Z, Walpole GFW, Yu Y. Kinetics of phagosome maturation is coupled to their intracellular motility. Commun Biol 2022; 5:1014. [PMID: 36163370 PMCID: PMC9512794 DOI: 10.1038/s42003-022-03988-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 09/12/2022] [Indexed: 11/09/2022] Open
Abstract
Immune cells degrade internalized pathogens in phagosomes through sequential biochemical changes. The degradation must be fast enough for effective infection control. The presumption is that each phagosome degrades cargos autonomously with a distinct but stochastic kinetic rate. However, here we show that the degradation kinetics of individual phagosomes is not stochastic but coupled to their intracellular motility. By engineering RotSensors that are optically anisotropic, magnetic responsive, and fluorogenic in response to degradation activities in phagosomes, we monitored cargo degradation kinetics in single phagosomes simultaneously with their translational and rotational dynamics. We show that phagosomes that move faster centripetally are more likely to encounter and fuse with lysosomes, thereby acidifying faster and degrading cargos more efficiently. The degradation rates increase nearly linearly with the translational and rotational velocities of phagosomes. Our results indicate that the centripetal motion of phagosomes functions as a clock for controlling the progression of cargo degradation.
Collapse
Affiliation(s)
- Yanqi Yu
- Department of Chemistry, Indiana University, Bloomington, IN, 47405-7102, USA
| | - Zihan Zhang
- Department of Chemistry, Indiana University, Bloomington, IN, 47405-7102, USA
| | - Glenn F W Walpole
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Yan Yu
- Department of Chemistry, Indiana University, Bloomington, IN, 47405-7102, USA.
| |
Collapse
|
41
|
Santiago-Burgos EJ, Stuckey PV, Santiago-Tirado FH. Real-time visualization of phagosomal pH manipulation by Cryptococcus neoformans in an immune signal-dependent way. Front Cell Infect Microbiol 2022; 12:967486. [PMID: 36211949 PMCID: PMC9538179 DOI: 10.3389/fcimb.2022.967486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 09/08/2022] [Indexed: 11/21/2022] Open
Abstract
Understanding of how intracellular pathogens survive in their host cells is important to improve management of their diseases. This has been fruitful for intracellular bacteria, but it is an understudied area in fungal pathogens. Here we start elucidating and characterizing the strategies used by one of the commonest fungal pathogens, Cryptococcus neoformans, to survive intracellularly. The ability of the fungus to survive inside host cells is one of the main drivers of disease progression, yet it is unclear whether C. neoformans resides in a fully acidified, partially acidic, or neutral phagosome. Using a dye that only fluoresce under acidic conditions to stain C. neoformans, a hypha-defective Candida albicans mutant, and the nonpathogenic Saccharomyces cerevisiae, we characterized the fungal behaviors in infected macrophages by live microscopy. The main behavior in the C. albicans mutant strain and S. cerevisiae-phagosomes was rapid acidification after internalization, which remained for the duration of the imaging. In contrast, a significant number of C. neoformans-phagosomes exhibited alternative behaviors distinct from the normal phagosomal maturation: some phagosomes acidified with subsequent loss of acidification, and other phagosomes never acidified. Moreover, the frequency of these behaviors was affected by the immune status of the host cell. We applied the same technique to a flow cytometry analysis and found that a substantial percentage of C. neoformans-phagosomes showed impaired acidification, whereas almost 100% of the S. cerevisiae-phagosomes acidify. Lastly, using a membrane-damage reporter, we show phagosome permeabilization correlates with acidification alterations, but it is not the only strategy that C. neoformans uses to manipulate phagosomal acidification. The different behaviors described here provide an explanation to the confounding literature regarding cryptococcal-phagosome acidification and the methods can be applied to study other intracellular fungal pathogens.
Collapse
Affiliation(s)
| | - Peter V. Stuckey
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States
| | - Felipe H. Santiago-Tirado
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States
- Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN, United States
- Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, IN, United States
- *Correspondence: Felipe H. Santiago-Tirado,
| |
Collapse
|
42
|
Wojnilowicz M, Laznickova P, Ju Y, Ang CS, Tidu F, Bendickova K, Forte G, Plebanski M, Caruso F, Cavalieri F, Fric J. Influence of protein corona on the interaction of glycogen-siRNA constructs with ex vivo human blood immune cells. BIOMATERIALS ADVANCES 2022; 140:213083. [PMID: 36027666 DOI: 10.1016/j.bioadv.2022.213083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 06/28/2022] [Accepted: 08/11/2022] [Indexed: 06/15/2023]
Abstract
Glycogen-nucleic acid constructs i.e., glycoplexes are emerging promising platforms for the alteration of gene expression and transcription. Understanding the interaction of glycoplexes with human blood components, such as serum proteins and peripheral blood mononuclear cells (PBMCs), is important to overcome immune cell activation and control biodistribution upon administration of the glycoplexes in vivo. Herein, we investigated the interactions of polyethylene glycol (PEG)ylated and non-PEGylated glycoplexes carrying siRNA molecules with PBMCs isolated from the blood of healthy donors. We found that both types of glycoplexes were non-toxic and were primarily phagocytosed by monocytes without triggering a pro-inflammatory interleukin 6 cytokine production. Furthermore, we investigated the role of the protein corona on controlling the internalization efficiency in immune cells - we found that the adsorption of serum proteins, in particular haptoglobin, alpha-1-antitrypsin and apolipoprotein A-II, onto the non-PEGylated glycoplexes, significantly reduced the uptake of the glycoplexes by PBMCs. Moreover, the non-PEGylated glycoplexes were efficient in the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) knockdown in monocytic THP-1 cell line. This study provides an insight into the rational design of glycogen-based nanocarriers for the safe delivery of siRNA without eliciting unwanted immune cell activation and efficient siRNA activity upon its delivery.
Collapse
Affiliation(s)
- Marcin Wojnilowicz
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Petra Laznickova
- Center for Translational Medicine, International Clinical Research Center (ICRC), St Anne's University Hospital Brno, Pekarska 53, 656 91 Brno, Czech Republic; Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00 Brno-Bohunice, Czech Republic
| | - Yi Ju
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia; School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria 3083, Australia
| | - Ching-Seng Ang
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Federico Tidu
- Center for Translational Medicine, International Clinical Research Center (ICRC), St Anne's University Hospital Brno, Pekarska 53, 656 91 Brno, Czech Republic; Division of Cancer Biology, The Institute of Cancer Research: London, 123 Old Brompton Road, London SW73RP, United Kingdom
| | - Kamila Bendickova
- Center for Translational Medicine, International Clinical Research Center (ICRC), St Anne's University Hospital Brno, Pekarska 53, 656 91 Brno, Czech Republic
| | - Giancarlo Forte
- Center for Translational Medicine, International Clinical Research Center (ICRC), St Anne's University Hospital Brno, Pekarska 53, 656 91 Brno, Czech Republic
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria 3083, Australia
| | - Frank Caruso
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia.
| | - Francesca Cavalieri
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia; School of Science, RMIT University, Victoria 3000, Australia; Dipartimento di Scienze e Tecnologie Chimiche, Universita' degli Studi di Roma "Tor Vergata", Via della Ricerca Scientifica 1, 00133 Rome, Italy.
| | - Jan Fric
- Center for Translational Medicine, International Clinical Research Center (ICRC), St Anne's University Hospital Brno, Pekarska 53, 656 91 Brno, Czech Republic; Institute of Hematology and Blood Transfusion, U Nemocnice 2094, 128 20 Prague 2, Czech Republic.
| |
Collapse
|
43
|
Koda K, Keller S, Kojima R, Kamiya M, Urano Y. Measuring the pH of Acidic Vesicles in Live Cells with an Optimized Fluorescence Lifetime Imaging Probe. Anal Chem 2022; 94:11264-11271. [PMID: 35913787 DOI: 10.1021/acs.analchem.2c01840] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Acidification of intracellular vesicles, such as endosomes and lysosomes, is a key pathway for regulating the function of internal proteins. Most conventional methods of measuring pH are not satisfactory for quantifying the pH inside these vesicles. Here, we investigated the molecular requirements for a fluorescence probe to measure the intravesicular acidic pH in living cells by means of fluorescence lifetime imaging microscopy (FLIM). The developed probe, m-DiMeNAF488, exhibits a pH-dependent equilibrium between highly fluorescent and moderately fluorescent forms, which has distinct and detectable fluorescence lifetimes of 4.36 and 0.58 ns, respectively. The pKa(τ) value of m-DiMeNAF488 was determined to be 4.58, which would be favorable for evaluating the pH in the acidic vesicles. We were able to monitor the pH changes in phagosomes during phagocytosis by means of FLIM using m-DiMeNAF488. This probe is expected to be a useful tool for investigating acidic pH-regulated biological phenomena.
Collapse
Affiliation(s)
| | | | - Ryosuke Kojima
- PRESTO, Japan Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
| | - Mako Kamiya
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259, Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Yasuteru Urano
- AMED-CREST, Japan Agency for Medical Research and Development, 1-7-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| |
Collapse
|
44
|
Roy Chowdhury A, Sah S, Varshney U, Chakravortty D. Salmonella Typhimurium outer membrane protein A (OmpA) renders protection from nitrosative stress of macrophages by maintaining the stability of bacterial outer membrane. PLoS Pathog 2022; 18:e1010708. [PMID: 35969640 PMCID: PMC9410544 DOI: 10.1371/journal.ppat.1010708] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 08/25/2022] [Accepted: 06/27/2022] [Indexed: 11/18/2022] Open
Abstract
Bacterial porins are highly conserved outer membrane proteins used in the selective transport of charged molecules across the membrane. In addition to their significant contributions to the pathogenesis of Gram-negative bacteria, their role(s) in salmonellosis remains elusive. In this study, we investigated the role of outer membrane protein A (OmpA), one of the major outer membrane porins of Salmonella, in the pathogenesis of Salmonella Typhimurium (STM). Our study revealed that OmpA plays an important role in the intracellular virulence of Salmonella. An ompA deficient strain of Salmonella (STM ΔompA) showed compromised proliferation in macrophages. We found that the SPI-2 encoded virulence factors such as sifA and ssaV are downregulated in STM ΔompA. The poor colocalization of STM ΔompA with LAMP-1 showed that disruption of SCV facilitated its release into the cytosol of macrophages, where it was assaulted by reactive nitrogen intermediates (RNI). The enhanced recruitment of nitrotyrosine on the cytosolic population of STM ΔompAΔsifA and ΔompAΔssaV compared to STM ΔsifA and ΔssaV showed an additional role of OmpA in protecting the bacteria from host nitrosative stress. Further, we showed that the generation of greater redox burst could be responsible for enhanced sensitivity of STM ΔompA to the nitrosative stress. The expression of several other outer membrane porins such as ompC, ompD, and ompF was upregulated in STM ΔompA. We found that in the absence of ompA, the enhanced expression of ompF increased the outer membrane porosity of Salmonella and made it susceptible to in vitro and in vivo nitrosative stress. Our study illustrates a novel mechanism for the strategic utilization of OmpA by Salmonella to protect itself from the nitrosative stress of macrophages.
Collapse
Affiliation(s)
- Atish Roy Chowdhury
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| | - Shivjee Sah
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| | - Umesh Varshney
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| | - Dipshikha Chakravortty
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| |
Collapse
|
45
|
Titania nanospikes activate macrophage phagocytosis by ligand-independent contact stimulation. Sci Rep 2022; 12:12250. [PMID: 35851278 PMCID: PMC9293906 DOI: 10.1038/s41598-022-16214-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 07/06/2022] [Indexed: 11/21/2022] Open
Abstract
Macrophage phagocytosis is an important research target to combat various inflammatory or autoimmune diseases; however, the phenomenon has never been controlled by artificial means. Titania nanospikes created by alkaline etching treatment can tune macrophage polarization toward a M1-like type and might regulate macrophage phagocytosis. This in vitro study aimed to determine whether the two-dimensional titania nanosurfaces created by alkaline etching treatment activated the macrophage phagocytosis by nanospike-mediated contact stimulation. On two-dimensional pure titanium sheets, alkaline etching treatments with different protocols created superhydrophilic nanosurfaces with hydroxyl function groups and moderate or dense nanospikes. Both types of titania nanosurfaces promoted the phagocytic activity of the mouse macrophage-like cell line, J774A.1, through upregulation of M1 polarization markers and phagocytosis-related receptors, such as toll-like receptors (TLR2 and 4). In contrast, the hydrophobic smooth or micro-roughened titanium surfaces did not activate macrophage phagocytosis or the expression of related receptors. These phenomena remained unchanged even under the antibody blockade of macrophage TLR2 but were either suppressed or augmented for each surface excited by ultraviolet irradiation. Titania nanospikes induced paxillin expression and provided physical stimuli to macrophages, the extent of which was positively correlated with TLR expression levels. Ligand stimulation with lipopolysaccharide did not upregulate macrophage TLR expression but further enhanced M1 marker expression by titania nanosurfaces. These results showed that the two-dimensional titania nanosurfaces activated macrophage phagocytosis by enhancing expression of phagocytosis-related receptors through nanospike-mediated contact stimulation, in assistance with physical surface properties, in a ligand-independent manner.
Collapse
|
46
|
Dragotakes Q, Jacobs E, Ramirez LS, Yoon OI, Perez-Stable C, Eden H, Pagnotta J, Vij R, Bergman A, D’Alessio F, Casadevall A. Bet-hedging antimicrobial strategies in macrophage phagosome acidification drive the dynamics of Cryptococcus neoformans intracellular escape mechanisms. PLoS Pathog 2022; 18:e1010697. [PMID: 35816543 PMCID: PMC9302974 DOI: 10.1371/journal.ppat.1010697] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/21/2022] [Accepted: 06/23/2022] [Indexed: 11/19/2022] Open
Abstract
The fungus Cryptococcus neoformans is a major human pathogen with a remarkable intracellular survival strategy that includes exiting macrophages through non-lytic exocytosis (Vomocytosis) and transferring between macrophages (Dragotcytosis) by a mechanism that involves sequential events of non-lytic exocytosis and phagocytosis. Vomocytosis and Dragotcytosis are fungal driven processes, but their triggers are not understood. We hypothesized that the dynamics of Dragotcytosis could inherit the stochasticity of phagolysosome acidification and that Dragotcytosis was triggered by fungal cell stress. Consistent with this view, fungal cells involved in Dragotcytosis reside in phagolysosomes characterized by low pH and/or high oxidative stress. Using fluorescent microscopy, qPCR, live cell video microscopy, and fungal growth assays we found that the that mitigating pH or oxidative stress reduced Dragotcytosis frequency, whereas ROS susceptible mutants of C. neoformans underwent Dragotcytosis more frequently. Dragotcytosis initiation was linked to phagolysosomal pH, oxidative stresses, and macrophage polarization state. Dragotcytosis manifested stochastic dynamics thus paralleling the dynamics of phagosomal acidification, which correlated with the inhospitality of phagolysosomes in differently polarized macrophages. Hence, randomness in phagosomal acidification randomly created a population of inhospitable phagosomes where fungal cell stress triggered stochastic C. neoformans non-lytic exocytosis dynamics to escape a non-permissive intracellular macrophage environment.
Collapse
Affiliation(s)
- Quigly Dragotakes
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Ella Jacobs
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Lia Sanchez Ramirez
- Department of Molecular and Cell Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Olivia Insun Yoon
- Department of Molecular and Cell Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Caitlin Perez-Stable
- Department of Molecular and Cell Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Hope Eden
- Department of Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Jenlu Pagnotta
- Department of Molecular and Cell Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Raghav Vij
- Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| | - Aviv Bergman
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, New York City, New York, United States of America
- Santa Fe Institute, Santa Fe, New Mexico, United States of America
| | - Franco D’Alessio
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| |
Collapse
|
47
|
Faria-Gonçalves P, Oliveira AS, Gaspar C, Rodrigues L, Palmeira-de-Oliveira R, Martinez-de-Oliveira J, Gonçalves T, Palmeira-de-Oliveira A, Rolo J. Vulvovaginal Candida albicans Clinical Isolates’ Resistance to Phagocytosis In-Vitro. Life (Basel) 2022; 12:life12060838. [PMID: 35743869 PMCID: PMC9225182 DOI: 10.3390/life12060838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 11/16/2022] Open
Abstract
Previous studies have revealed that Candida albicans isolates involved in chronic vulvovaginal candidosis (cVVC) phenotypically express less virulent traits than clinical isolates involved in sporadic infections. In this study, we aimed to further explore this finding by studying the behaviour of those same clinical isolates in in-vitro models of infection. Eighteen clinical Candida albicans isolates were collected from women suffering sporadic (eight isolates) or chronic infections (ten isolates). Adhesion to HeLa cells (human cervical cancer epithelial cell line) and resistance to phagocytosis by RAW 264.7 cells (murine macrophages cell line) were tested in-vitro. In addition, phenotypic expression of virulence factors related with either adhesion or resistance to phagocytosis was tested in-vitro. Results indicated that yeast isolates involved in sporadic infection adhered in a higher proportion of HeLa cells than those of chronic infections, which was related with their ability to produce biofilm (p < 0.05). The ability to evade phagocytosis was related to an elevated production of proteases (p < 0.05) by chronic isolates, while sporadic isolates’ resistance to phagocytosis was related to a higher hydrophobicity of cell walls (p < 0.05). We conclude that the evasion of macrophage-mediated phagocytosis related to the production of proteases might be an important factor involved in the recurrence of vulvovaginal candidosis infection.
Collapse
Affiliation(s)
- Paula Faria-Gonçalves
- CICS-UBI—Health Sciences Research Center, University of Beira Interior, 6200-506 Covilhã, Portugal; (P.F.-G.); (A.S.O.); (C.G.); (R.P.-d.-O.); (J.M.-d.-O.); (A.P.-d.-O.)
- FCS-UBI—Faculty of Health Sciences, University of Beira Interior, 6200-506 Covilhã, Portugal
- FMUMN—Faculty of Medicine, University Mandume Ya Ndemufayo, Lubango 3FJP+27X, Angola
| | - Ana Sofia Oliveira
- CICS-UBI—Health Sciences Research Center, University of Beira Interior, 6200-506 Covilhã, Portugal; (P.F.-G.); (A.S.O.); (C.G.); (R.P.-d.-O.); (J.M.-d.-O.); (A.P.-d.-O.)
- FCS-UBI—Faculty of Health Sciences, University of Beira Interior, 6200-506 Covilhã, Portugal
| | - Carlos Gaspar
- CICS-UBI—Health Sciences Research Center, University of Beira Interior, 6200-506 Covilhã, Portugal; (P.F.-G.); (A.S.O.); (C.G.); (R.P.-d.-O.); (J.M.-d.-O.); (A.P.-d.-O.)
- FCS-UBI—Faculty of Health Sciences, University of Beira Interior, 6200-506 Covilhã, Portugal
- Labfit-HPRD—Health Products Research and Development Lda, 6200-284 Covilhã, Portugal
| | - Lisa Rodrigues
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (L.R.); (T.G.)
- FMUC—Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Rita Palmeira-de-Oliveira
- CICS-UBI—Health Sciences Research Center, University of Beira Interior, 6200-506 Covilhã, Portugal; (P.F.-G.); (A.S.O.); (C.G.); (R.P.-d.-O.); (J.M.-d.-O.); (A.P.-d.-O.)
- FCS-UBI—Faculty of Health Sciences, University of Beira Interior, 6200-506 Covilhã, Portugal
- Labfit-HPRD—Health Products Research and Development Lda, 6200-284 Covilhã, Portugal
| | - José Martinez-de-Oliveira
- CICS-UBI—Health Sciences Research Center, University of Beira Interior, 6200-506 Covilhã, Portugal; (P.F.-G.); (A.S.O.); (C.G.); (R.P.-d.-O.); (J.M.-d.-O.); (A.P.-d.-O.)
| | - Teresa Gonçalves
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (L.R.); (T.G.)
- FMUC—Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Ana Palmeira-de-Oliveira
- CICS-UBI—Health Sciences Research Center, University of Beira Interior, 6200-506 Covilhã, Portugal; (P.F.-G.); (A.S.O.); (C.G.); (R.P.-d.-O.); (J.M.-d.-O.); (A.P.-d.-O.)
- FCS-UBI—Faculty of Health Sciences, University of Beira Interior, 6200-506 Covilhã, Portugal
- Labfit-HPRD—Health Products Research and Development Lda, 6200-284 Covilhã, Portugal
| | - Joana Rolo
- CICS-UBI—Health Sciences Research Center, University of Beira Interior, 6200-506 Covilhã, Portugal; (P.F.-G.); (A.S.O.); (C.G.); (R.P.-d.-O.); (J.M.-d.-O.); (A.P.-d.-O.)
- Correspondence:
| |
Collapse
|
48
|
Macrophages disseminate pathogen associated molecular patterns through the direct extracellular release of the soluble content of their phagolysosomes. Nat Commun 2022; 13:3072. [PMID: 35654768 PMCID: PMC9163141 DOI: 10.1038/s41467-022-30654-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 05/04/2022] [Indexed: 12/12/2022] Open
Abstract
Recognition of pathogen-or-damage-associated molecular patterns is critical to inflammation. However, most pathogen-or-damage-associated molecular patterns exist within intact microbes/cells and are typically part of non-diffusible, stable macromolecules that are not optimally immunostimulatory or available for immune detection. Partial digestion of microbes/cells following phagocytosis potentially generates new diffusible pathogen-or-damage-associated molecular patterns, however, our current understanding of phagosomal biology would have these molecules sequestered and destroyed within phagolysosomes. Here, we show the controlled release of partially-digested, soluble material from phagolysosomes of macrophages through transient, iterative fusion-fission events between mature phagolysosomes and the plasma membrane, a process we term eructophagy. Eructophagy is most active in proinflammatory macrophages and further induced by toll like receptor engagement. Eructophagy is mediated by genes encoding proteins required for autophagy and can activate vicinal cells by release of phagolysosomally-processed, partially-digested pathogen associated molecular patterns. We propose that eructophagy allows macrophages to amplify local inflammation through the processing and dissemination of pathogen-or-damage-associated molecular patterns. The detection of conserved motifs by pattern recognition receptors is a crucial component of the innate detection of pathogens and danger signals via conserved pattern recognition receptors. Here the authors define a pathway that transfers partially digested material from the phagolysosomal pathway of macrophages to release at the plasma membrane which is associated with enhanced inflammatory potential, by a process they introduce as eructophagy.
Collapse
|
49
|
Silberberg E, Filep JG, Ariel A. Weathering the Storm: Harnessing the Resolution of Inflammation to Limit COVID-19 Pathogenesis. Front Immunol 2022; 13:863449. [PMID: 35615359 PMCID: PMC9124752 DOI: 10.3389/fimmu.2022.863449] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/22/2022] [Indexed: 12/13/2022] Open
Abstract
The resolution of inflammation is a temporally and spatially coordinated process that in its innate manifestations, primarily involves neutrophils and macrophages. The shutdown of infection or injury-induced acute inflammation requires termination of neutrophil accumulation within the affected sites, neutrophil demise, and clearance by phagocytes (efferocytosis), such as tissue-resident and monocyte-derived macrophages. This must be followed by macrophage reprogramming from the inflammatory to reparative and consequently resolution-promoting phenotypes and the production of resolution-promoting lipid and protein mediators that limit responses in various cell types and promote tissue repair and return to homeostatic architecture and function. Recent studies suggest that these events, and macrophage reprogramming to pro-resolving phenotypes in particular, are not only important in the acute setting, but might be paramount in limiting chronic inflammation, autoimmunity, and various uncontrolled cytokine-driven pathologies. The SARS-CoV-2 (COVID-19) pandemic has caused a worldwide health and economic crisis. Severe COVID-19 cases that lead to high morbidity are tightly associated with an exuberant cytokine storm that seems to trigger shock-like pathologies, leading to vascular and multiorgan failures. In other cases, the cytokine storm can lead to diffuse alveolar damage that results in acute respiratory distress syndrome (ARDS) and lung failure. Here, we address recent advances on effectors in the resolution of inflammation and discuss how pro-resolution mechanisms with particular emphasis on macrophage reprogramming, might be harnessed to limit the universal COVID-19 health threat.
Collapse
Affiliation(s)
- Esther Silberberg
- Department of Biology and Human Biology, University of Haifa, Haifa, Israel
| | - János G. Filep
- Department of Pathology and Cell Biology, University of Montreal, Montreal, QC, Canada
- Research Center, Maisonneuve-Rosemont Hospital, Montreal, QC, Canada
- *Correspondence: Amiram Ariel, ; János G. Filep,
| | - Amiram Ariel
- Department of Biology and Human Biology, University of Haifa, Haifa, Israel
- *Correspondence: Amiram Ariel, ; János G. Filep,
| |
Collapse
|
50
|
Arnhold J, Malle E. Halogenation Activity of Mammalian Heme Peroxidases. Antioxidants (Basel) 2022; 11:antiox11050890. [PMID: 35624754 PMCID: PMC9138014 DOI: 10.3390/antiox11050890] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 12/10/2022] Open
Abstract
Mammalian heme peroxidases are fascinating due to their unique peculiarity of oxidizing (pseudo)halides under physiologically relevant conditions. These proteins are able either to incorporate oxidized halides into substrates adjacent to the active site or to generate different oxidized (pseudo)halogenated species, which can take part in multiple (pseudo)halogenation and oxidation reactions with cell and tissue constituents. The present article reviews basic biochemical and redox mechanisms of (pseudo)halogenation activity as well as the physiological role of heme peroxidases. Thyroid peroxidase and peroxidasin are key enzymes for thyroid hormone synthesis and the formation of functional cross-links in collagen IV during basement membrane formation. Special attention is directed to the properties, enzymatic mechanisms, and resulting (pseudo)halogenated products of the immunologically relevant proteins such as myeloperoxidase, eosinophil peroxidase, and lactoperoxidase. The potential role of the (pseudo)halogenated products (hypochlorous acid, hypobromous acid, hypothiocyanite, and cyanate) of these three heme peroxidases is further discussed.
Collapse
Affiliation(s)
- Jürgen Arnhold
- Medical Faculty, Institute of Medical Physics and Biophysics, Leipzig University, 04107 Leipzig, Germany
- Correspondence: (J.A.); or (E.M.)
| | - Ernst Malle
- Gottfried Schatz Research Center, Division of Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
- Correspondence: (J.A.); or (E.M.)
| |
Collapse
|