1
|
Sabatke B, Rossi IV, Ramirez MI. Interaction vesicles as emerging mediators of host-pathogen molecular crosstalk and their implications for infection dynamics. FEBS Lett 2025. [PMID: 40313034 DOI: 10.1002/1873-3468.70055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 04/08/2025] [Accepted: 04/12/2025] [Indexed: 05/03/2025]
Abstract
Extracellular vesicles (EVs) are critical in cell communication, transfer of biomolecules, and host-pathogen interaction. A newly identified subset, "interaction vesicles" (iEVs), forms through host-pathogen contact, merging membrane elements from both. These iEVs may arise through multiple mechanisms, including direct cell-cell contact, membrane contact sites, uptake and repackaging of foreign EVs, and post-release fusion of EVs. These hybrid vesicles enable pathogens to modify host environments, aiding immune evasion and infection persistence. However, iEVs may also act in favor of the host, contributing to pathogen recognition and elimination. Advanced techniques, including proteomics and high-resolution microscopy, are beginning to clarify their composition and fusion. Yet, isolating these hybrid EVs remains challenging. Overcoming these barriers could enhance understanding of infection mechanisms and support diagnostic and therapeutic innovation.
Collapse
Affiliation(s)
- Bruna Sabatke
- Graduate Program in Microbiology, Pathology and Parasitology, Federal University of Paraná, Curitiba, Brazil
- EVAHPI - Extracellular Vesicles and Host-Parasite Interactions Research Group, Carlos Chagas Institute (Fiocruz-PR), Curitiba, Brazil
| | - Izadora Volpato Rossi
- Graduate Program in Microbiology, Pathology and Parasitology, Federal University of Paraná, Curitiba, Brazil
- EVAHPI - Extracellular Vesicles and Host-Parasite Interactions Research Group, Carlos Chagas Institute (Fiocruz-PR), Curitiba, Brazil
| | - Marcel I Ramirez
- Graduate Program in Microbiology, Pathology and Parasitology, Federal University of Paraná, Curitiba, Brazil
- EVAHPI - Extracellular Vesicles and Host-Parasite Interactions Research Group, Carlos Chagas Institute (Fiocruz-PR), Curitiba, Brazil
- Graduate Program in Cell and Molecular Biology, Federal University of Paraná, Curitiba, Brazil
| |
Collapse
|
2
|
Biquand E, Khau S, Papon N, Briard B. Extracellular vesicles: new bullets in the fungal armory. Trends Microbiol 2024; 32:1167-1169. [PMID: 38664155 DOI: 10.1016/j.tim.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 12/06/2024]
Abstract
Invasive fungal infections represent millions of deaths per year, but their pathophysiology remains insufficiently understood. Host-fungi interplay has been recently shown to include extracellular vesicles derived from fungi and host infected cells. In this forum article we discuss their emerging role in modulating the host immune response with particular emphasis on their regulatory involvement during Candida albicans infection.
Collapse
Affiliation(s)
- Elise Biquand
- Inserm, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France; Université de Tours, Tours, France
| | - Sandra Khau
- Inserm, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France; Université de Tours, Tours, France
| | - Nicolas Papon
- Université d'Angers, IRF, SFR ICAT, F-49000, Angers, France
| | - Benoit Briard
- Inserm, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France; Université de Tours, Tours, France.
| |
Collapse
|
3
|
Liu H, Ouyang Z, Li S. Advances of M1 macrophages-derived extracellular vesicles in tumor therapy. Biomed Pharmacother 2024; 181:117735. [PMID: 39644871 DOI: 10.1016/j.biopha.2024.117735] [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: 09/19/2024] [Revised: 11/28/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024] Open
Abstract
Extracellular vesicles derived from classically activated M1 macrophages (M1 EVs) have shown great potential in both tumor treatment and drug delivery. M1 EVs inherit specific biological messengers from their parent cells and possess the capability to activate immune cells residing in close or distant tumor tissues for antitumor therapy. Moreover, M1 EVs are commonly used as an attractive drug delivery system due to their tumor-targeting ability, biocompatibility, and non-toxic. They can effectively encapsulate various therapeutic cargoes through specific methods such as electroporation, co-incubation, sonication, extrusion, transfection, or click chemistry reaction. In this review, we provide a comprehensive summary of the advancements in M1 EVs for tumor therapy, discussing their application prospects and existing problems.
Collapse
Affiliation(s)
- Houli Liu
- School of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui Province 230012, China.
| | - Zhaorong Ouyang
- School of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui Province 230012, China
| | - Siyu Li
- School of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui Province 230012, China
| |
Collapse
|
4
|
Pushpamithran G, Blomgran R. Macrophage-derived extracellular vesicles from Ascaris lumbricoides antigen exposure enhance Mycobacterium tuberculosis growth control, reduce IL-1β, and contain miR-342-5p, miR-516b-5p, and miR-570-3p that regulate PI3K/AKT and MAPK signaling pathways. Front Immunol 2024; 15:1454881. [PMID: 39569198 PMCID: PMC11576181 DOI: 10.3389/fimmu.2024.1454881] [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: 06/25/2024] [Accepted: 10/07/2024] [Indexed: 11/22/2024] Open
Abstract
Background Helminth coinfection with tuberculosis (TB) can alter the phenotype and function of macrophages, which are the major host cells responsible for controlling Mycobacterium tuberculosis (Mtb). However, it is not known whether helminth infection stimulates the release of host-derived extracellular vesicles (EVs) to induce or maintain their regulatory network that suppresses TB immunity. We previously showed that pre-exposure of human monocyte-derived macrophages (hMDMs) with Ascaris lumbricoides protein antigens (ASC) results in reduced Mtb infection-driven proinflammation and gained bacterial control. This effect was entirely dependent on the presence of soluble components in the conditioned medium from helminth antigen-pre-exposed macrophages. Methods Our objective was to investigate the role of EVs released from helminth antigen-exposed hMDMs on Mtb-induced proinflammation and its effect on Mtb growth in hMDMs. Conditioned medium from 48-h pre-exposure with ASC or Schistosoma mansoni antigen (SM) was used to isolate EVs by ultracentrifugation. EVs were characterized by immunoblotting, flow cytometry, nanoparticle tracking assay, transmission electron microscopy, and a total of 377 microRNA (miRNA) from EVs screened by TaqMan array. Luciferase-expressing Mtb H37Rv was used to evaluate the impact of isolated EVs on Mtb growth control in hMDMs. Results EV characterization confirmed double-membraned EVs, with a mean size of 140 nm, expressing the classical exosome markers CD63, CD81, CD9, and flotillin-1. Specifically, EVs from the ASC conditioned medium increased the bacterial control in treatment-naïve hMDMs and attenuated Mtb-induced IL-1β at 5 days post-infection. Four miRNAs showed unique upregulation in response to ASC exposure in five donors. Pathway enrichment analysis showed that the MAPK and PI3K-AKT signaling pathways were regulated. Among the mRNA targets, relevant for regulating inflammatory responses and cellular stress pathways, CREB1 and MAPK13 were identified. In contrast, SM exposure showed significant regulation of the TGF-β signaling pathway with SMAD4 as a common target. Conclusion Overall, our findings suggest that miRNAs in EVs released from helminth-exposed macrophages regulate important signaling pathways that influence macrophage control of Mtb and reduce inflammation. Understanding these interactions between helminth-induced EVs, miRNAs, and macrophage responses may inform novel therapeutic strategies for TB management.
Collapse
Affiliation(s)
- Giggil Pushpamithran
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Robert Blomgran
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
5
|
Kulig K, Rapala-Kozik M, Karkowska-Kuleta J. Extracellular vesicle production: A bidirectional effect in the interplay between host and Candida fungi. CURRENT RESEARCH IN MICROBIAL SCIENCES 2024; 7:100255. [PMID: 39040088 PMCID: PMC11260599 DOI: 10.1016/j.crmicr.2024.100255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024] Open
Abstract
Candida fungi exploit various virulence strategies to invade the human host, while host cells employ diverse mechanisms to maintain homeostasis and respond to infection. Extracellular vesicles (EVs) are integral components of the multifaceted landscape of host-pathogen interactions, with their abundant production by all contributors involved in these complex and dynamic relations. Herein, we present the current state of knowledge regarding the host response by releasing EVs in reaction to Candida, as well as the influence of fungal EVs on the functionality of the confronted host cells. Fungal vesicles contribute to enhanced adhesion of pathogens to human cells as evidenced for C. auris, and may modulate the production of several cytokines, including IL-6, IL-8, IL-10, IL-12p40, TGF-β and TNF-α, thereby exerting pro-infective and pro-inflammatory effects, as described for C. albicans and other Candida species. Whereas the biosynthesis of EVs by host cells can dynamically modulate the proliferation and viability of fungal cells and affect the candidacidal functionality of other effector cells. The reciprocal influence of EVs from host cells and Candida pathogens is a key focus, explaining their significant role in cell signaling and interkingdom communication.
Collapse
Affiliation(s)
- Kamila Kulig
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| | - Maria Rapala-Kozik
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| | - Justyna Karkowska-Kuleta
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| |
Collapse
|
6
|
Lai WQ, Xia HF, Chen GH, Wang XL, Yang JG, Wu LZ, Zhao YF, Jia YL, Chen G. p-AKT/VPS4B regulates the small extracellular vesicle size in venous malformation endothelial cells. Oral Dis 2024; 30:1273-1285. [PMID: 37154262 DOI: 10.1111/odi.14608] [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: 10/29/2022] [Revised: 04/08/2023] [Accepted: 04/22/2023] [Indexed: 05/10/2023]
Abstract
OBJECTIVE Small extracellular vesicle (sEV)-mediated intercellular communication is increasingly the key for the understanding of venous malformations (VMs). This study aims to clarify the detailed changes of sEVs in VMs. SUBJECTS AND METHODS Fifteen VM patients without treatment history and twelve healthy donors were enrolled in the study. sEVs were isolated from both fresh lesions and cell supernatant, and were examined by western blotting, nanoparticle tracking analysis and transmission electron microscopy. Western blot analysis, immunohistochemistry and immunofluorescence were adopted to screening candidate regulator of sEV size. Specific inhibitors and siRNA were employed to validate the role of dysregulated p-AKT/vacuolar protein sorting-associated protein 4B (VPS4B) signaling on the size of sEVs in endothelial cells. RESULTS The size of sEVs derived from both VM lesion tissues and cell model was significantly increased. VPS4B, whose expression level was mostly significantly downregulated in VM endothelial cells, was responsible for the size change of sEVs. Targeting abnormal AKT activation corrected the size change of sEVs by recovering the expression level of VPS4B. CONCLUSION Downregulated VPS4B in endothelial cells, resulted from abnormally activated AKT signaling, contributed to the increased size of sEVs in VMs.
Collapse
Affiliation(s)
- Wen-Qiang Lai
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hou-Fu Xia
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Gao-Hong Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xiao-Le Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jie-Gang Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Lian-Zhi Wu
- Department of Obstetrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yi-Fang Zhao
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yu-Lin Jia
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
7
|
Lou K, Luo H, Jiang X, Feng S. Applications of emerging extracellular vesicles technologies in the treatment of inflammatory diseases. Front Immunol 2024; 15:1364401. [PMID: 38545101 PMCID: PMC10965547 DOI: 10.3389/fimmu.2024.1364401] [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: 01/02/2024] [Accepted: 03/04/2024] [Indexed: 04/17/2024] Open
Abstract
The emerging extracellular vesicles technologies is an advanced therapeutic approach showing promising potential for addressing inflammatory diseases. These techniques have been proven to have positive effects on immune modulation and anti-inflammatory responses. With these advancements, a comprehensive review and update on the role of extracellular vesicles in inflammatory diseases have become timely. This review aims to summarize the research progress of extracellular vesicle technologies such as plant-derived extracellular vesicles, milk-derived extracellular vesicles, mesenchymal stem cell-derived extracellular vesicles, macrophage-derived extracellular vesicles, etc., in the treatment of inflammatory diseases. It elucidates their potential significance in regulating inflammation, promoting tissue repair, and treating diseases. The goal is to provide insights for future research in this field, fostering the application and development of extracellular vesicle technology in the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Kecheng Lou
- Department of Urology, Lanxi People’s Hospital, Jinhua, Zhejiang, China
| | - Hui Luo
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Xinghua Jiang
- Department of Urology, Jingdezhen Second People’s Hospital, Jingdezhen, Jiangxi, China
| | - Shangzhi Feng
- Department of Urology, Jiujiang University Clinic College/Hospital, Jiujiang, Jiangxi, China
| |
Collapse
|
8
|
Santiago JV, Natu A, Ramelow CC, Rayaprolu S, Xiao H, Kumar V, Kumar P, Seyfried NT, Rangaraju S. Identification of State-Specific Proteomic and Transcriptomic Signatures of Microglia-Derived Extracellular Vesicles. Mol Cell Proteomics 2023; 22:100678. [PMID: 37952696 PMCID: PMC10755493 DOI: 10.1016/j.mcpro.2023.100678] [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: 08/07/2023] [Revised: 10/26/2023] [Accepted: 11/08/2023] [Indexed: 11/14/2023] Open
Abstract
Microglia are resident immune cells of the brain that play important roles in mediating inflammatory responses in several neurological diseases via direct and indirect mechanisms. One indirect mechanism may involve extracellular vesicle (EV) release, so that the molecular cargo transported by microglia-derived EVs can have functional effects by facilitating intercellular communication. The molecular composition of microglia-derived EVs, and how microglial activation states impact EV composition and EV-mediated effects in neuroinflammation, remain poorly understood. We hypothesize that microglia-derived EVs have unique molecular profiles that are determined by microglial activation state. Using size-exclusion chromatography to purify EVs from BV2 microglia, combined with proteomic (label-free quantitative mass spectrometry or LFQ-MS) and transcriptomic (mRNA and noncoding RNA seq) methods, we obtained comprehensive molecular profiles of microglia-derived EVs. LFQ-MS identified several classic EV proteins (tetraspanins, ESCRT machinery, and heat shock proteins), in addition to over 200 proteins not previously reported in the literature. Unique mRNA and microRNA signatures of microglia-derived EVs were also identified. After treating BV2 microglia with lipopolysaccharide (LPS), interleukin-10, or transforming growth factor beta, to mimic pro-inflammatory, anti-inflammatory, or homeostatic states, respectively, LFQ-MS and RNA seq revealed novel state-specific proteomic and transcriptomic signatures of microglia-derived EVs. Particularly, LPS treatment had the most profound impact on proteomic and transcriptomic compositions of microglia-derived EVs. Furthermore, we found that EVs derived from LPS-activated microglia were able to induce pro-inflammatory transcriptomic changes in resting responder microglia, confirming the ability of microglia-derived EVs to relay functionally relevant inflammatory signals. These comprehensive microglia-EV molecular datasets represent important resources for the neuroscience and omics communities and provide novel insights into the role of microglia-derived EVs in neuroinflammation.
Collapse
Affiliation(s)
- Juliet V Santiago
- Department of Neurology, Emory University, Atlanta, Georgia, USA; Center for Neurodegenerative Diseases, Emory University, Atlanta, Georgia, USA
| | - Aditya Natu
- Department of Neurology, Emory University, Atlanta, Georgia, USA; Center for Neurodegenerative Diseases, Emory University, Atlanta, Georgia, USA
| | - Christina C Ramelow
- Department of Neurology, Emory University, Atlanta, Georgia, USA; Center for Neurodegenerative Diseases, Emory University, Atlanta, Georgia, USA
| | - Sruti Rayaprolu
- Department of Neurology, Emory University, Atlanta, Georgia, USA; Center for Neurodegenerative Diseases, Emory University, Atlanta, Georgia, USA
| | - Hailian Xiao
- Department of Neurology, Emory University, Atlanta, Georgia, USA; Center for Neurodegenerative Diseases, Emory University, Atlanta, Georgia, USA
| | - Vishnu Kumar
- Department of Neurology, Emory University, Atlanta, Georgia, USA; Center for Neurodegenerative Diseases, Emory University, Atlanta, Georgia, USA
| | - Prateek Kumar
- Department of Neurology, Emory University, Atlanta, Georgia, USA; Center for Neurodegenerative Diseases, Emory University, Atlanta, Georgia, USA
| | - Nicholas T Seyfried
- Department of Neurology, Emory University, Atlanta, Georgia, USA; Center for Neurodegenerative Diseases, Emory University, Atlanta, Georgia, USA; Department of Biochemistry, Emory University, Atlanta, Georgia, USA
| | - Srikant Rangaraju
- Department of Neurology, Emory University, Atlanta, Georgia, USA; Center for Neurodegenerative Diseases, Emory University, Atlanta, Georgia, USA.
| |
Collapse
|
9
|
Santiago JV, Natu A, Ramelow CC, Rayaprolu S, Xiao H, Kumar V, Seyfried NT, Rangaraju S. Identification of state-specific proteomic and transcriptomic signatures of microglia-derived extracellular vesicles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.28.551012. [PMID: 37546899 PMCID: PMC10402142 DOI: 10.1101/2023.07.28.551012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Microglia are resident immune cells of the brain that play important roles in mediating inflammatory responses in several neurological diseases via direct and indirect mechanisms. One indirect mechanism may involve extracellular vesicle (EV) release, so that the molecular cargo transported by microglia-derived EVs can have functional effects by facilitating intercellular communication. The molecular composition of microglia-derived EVs, and how microglial activation states impacts EV composition and EV-mediated effects in neuroinflammation, remain poorly understood. We hypothesize that microglia-derived EVs have unique molecular profiles that are determined by microglial activation state. Using size-exclusion chromatography to purify EVs from BV2 microglia, combined with proteomic (label-free quantitative mass spectrometry or LFQ-MS) and transcriptomic (mRNA and non-coding RNA seq) methods, we obtained comprehensive molecular profiles of microglia-derived EVs. LFQ-MS identified several classic EV proteins (tetraspanins, ESCRT machinery, and heat shock proteins), in addition to over 200 proteins not previously reported in the literature. Unique mRNA and microRNA signatures of microglia-derived EVs were also identified. After treating BV2 microglia with lipopolysaccharide (LPS), interleukin-10, or transforming growth factor beta, to mimic pro-inflammatory, anti-inflammatory, or homeostatic states, respectively, LFQ-MS and RNA seq revealed novel state-specific proteomic and transcriptomic signatures of microglia-derived EVs. Particularly, LPS treatment had the most profound impact on proteomic and transcriptomic compositions of microglia-derived EVs. Furthermore, we found that EVs derived from LPS-activated microglia were able to induce pro-inflammatory transcriptomic changes in resting responder microglia, confirming the ability of microglia-derived EVs to relay functionally-relevant inflammatory signals. These comprehensive microglia-EV molecular datasets represent important resources for the neuroscience and glial communities, and provide novel insights into the role of microglia-derived EVs in neuroinflammation.
Collapse
Affiliation(s)
- Juliet V. Santiago
- Department of Neurology, Emory University, 201 Dowman Drive Atlanta, Georgia, 30322, United States of America
- Center for Neurodegenerative Diseases, Emory University, Atlanta, GA 30322, USA
| | - Aditya Natu
- Department of Neurology, Emory University, 201 Dowman Drive Atlanta, Georgia, 30322, United States of America
- Center for Neurodegenerative Diseases, Emory University, Atlanta, GA 30322, USA
| | - Christina C. Ramelow
- Department of Neurology, Emory University, 201 Dowman Drive Atlanta, Georgia, 30322, United States of America
- Center for Neurodegenerative Diseases, Emory University, Atlanta, GA 30322, USA
| | - Sruti Rayaprolu
- Department of Neurology, Emory University, 201 Dowman Drive Atlanta, Georgia, 30322, United States of America
- Center for Neurodegenerative Diseases, Emory University, Atlanta, GA 30322, USA
| | - Hailian Xiao
- Department of Neurology, Emory University, 201 Dowman Drive Atlanta, Georgia, 30322, United States of America
- Center for Neurodegenerative Diseases, Emory University, Atlanta, GA 30322, USA
| | - Vishnu Kumar
- Department of Neurology, Emory University, 201 Dowman Drive Atlanta, Georgia, 30322, United States of America
- Center for Neurodegenerative Diseases, Emory University, Atlanta, GA 30322, USA
| | - Nicholas T. Seyfried
- Department of Neurology, Emory University, 201 Dowman Drive Atlanta, Georgia, 30322, United States of America
- Center for Neurodegenerative Diseases, Emory University, Atlanta, GA 30322, USA
- Department of Biochemistry, Emory University, Atlanta, GA 30322, USA
| | - Srikant Rangaraju
- Department of Neurology, Emory University, 201 Dowman Drive Atlanta, Georgia, 30322, United States of America
- Center for Neurodegenerative Diseases, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
10
|
Huang Q, Sun Y, Sun J, Peng L, Shang H, Wei D, Li C, Hu Z, Peng H. Proteomic Characterization of Peritoneal Extracellular Vesicles in a Mouse Model of Peritoneal Fibrosis. J Proteome Res 2023; 22:908-918. [PMID: 36648763 DOI: 10.1021/acs.jproteome.2c00713] [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] [Indexed: 01/18/2023]
Abstract
Peritoneal fibrosis progression is regarded as a significant cause of the loss of peritoneal function, markedly limiting the application of peritoneal dialysis (PD). However, the pathogenesis of peritoneal fibrosis remains to be elucidated. Tissue-derived extracellular vesicles (EVs) change their molecular cargos to adapt the environment alteration, mediating intercellular communications and play a significant role in organ fibrosis. Hence, we performed, for the first time, four-dimensional label-free quantitative liquid chromatography-tandem mass spectrometry proteomic analyses on EVs from normal peritoneal tissues and PD-induced fibrotic peritoneum in mice. We demonstrated the alterations of EV concentration and protein composition between normal control and PD groups. A total of 2339 proteins containing 967 differentially expressed proteins were identified. Notably, upregulated proteins in PD EVs were enriched in processes including response to wounding and leukocyte migration, which participated in the development of fibrosis. In addition, EV proteins of the PD group exhibited unique metabolic signature compared with those of the control group. The glycolysis-related proteins increased in PD EVs, while oxidative phosphorylation and fatty acid metabolism-related proteins decreased. We also evaluated the effect of cell-type specificity on EV proteins, suggesting that mesothelial cells mainly cause the alterations in the molecular composition of EVs. Our study provided a useful resource for further validation of the key regulator or therapeutic target of peritoneal fibrosis.
Collapse
Affiliation(s)
- Qiang Huang
- Nephrology Division, Department of Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Yuxiang Sun
- Nephrology Division, Department of Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Juan Sun
- Nephrology Division, Department of Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Long Peng
- Division of Cardiovascular Medicine, Department of Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Hongli Shang
- Nephrology Division, Department of Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Dandan Wei
- Nephrology Division, Department of Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Canming Li
- Nephrology Division, Department of Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Zhaoyong Hu
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Hui Peng
- Nephrology Division, Department of Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| |
Collapse
|
11
|
Wei Y, Wang Z, Liu Y, Liao B, Zong Y, Shi Y, Liao M, Wang J, Zhou X, Cheng L, Ren B. Extracellular vesicles of Candida albicans regulate its own growth through the l-arginine/nitric oxide pathway. Appl Microbiol Biotechnol 2022; 107:355-367. [PMCID: PMC9703431 DOI: 10.1007/s00253-022-12300-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/12/2022] [Accepted: 11/15/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Yu Wei
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| | - Zheng Wang
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| | - Yaqi Liu
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| | - Binyou Liao
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| | - Yawen Zong
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| | - Yangyang Shi
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| | - Min Liao
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| | - Jiannan Wang
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| | - Lei Cheng
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| | - Biao Ren
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| |
Collapse
|
12
|
Martínez-López R, Hernáez ML, Redondo E, Calvo G, Radau S, Pardo M, Gil C, Monteoliva L. Candida albicans Hyphal Extracellular Vesicles Are Different from Yeast Ones, Carrying an Active Proteasome Complex and Showing a Different Role in Host Immune Response. Microbiol Spectr 2022; 10:e0069822. [PMID: 35604172 PMCID: PMC9241596 DOI: 10.1128/spectrum.00698-22] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/13/2022] [Indexed: 12/20/2022] Open
Abstract
Candida albicans is the principal causative agent of lethal fungal infections, predominantly in immunocompromised hosts. Extracellular vesicles (EVs) have been described as crucial in the interaction of microorganisms with their host. Since the yeast-to-hypha transition is an important virulence trait with great impact in invasive candidiasis (IC), we have addressed the characterization of EVs secreted by hyphal cells (HEVs) from C. albicans, comparing them to yeast EVs (YEVs). YEVs comprised a larger population of bigger EVs with mainly cell wall proteins, while HEVs were smaller, in general, and had a much higher protein diversity. YEVs were able to rescue the sensitivity of a cell wall mutant against calcofluor white, presumably due to the larger amount of cell wall proteins they contained. On the other hand, HEVs also contained many cytoplasmic proteins related to protein metabolism and intracellular protein transport and the endosomal sorting complexes required for transport (ESCRT) pathway related to exosome biogenesis, pointing to an intracellular origin of HEVs. Interestingly, an active 20S proteasome complex was secreted exclusively in HEVs. Moreover, HEVs contained a greater number of virulence-related proteins. As for their immunogenic role, both types of EV presented immune reactivity with human sera from patients suffering invasive candidiasis; however, under our conditions, only HEVs showed a cytotoxic effect on human macrophages and could elicit the release of tumor necrosis factor alpha (TNF-α) by these macrophages. IMPORTANCE This first analysis of HEVs of C. albicans has shown clear differences between them and the YEVs of C. albicans, showing their relevance and possible use in the discovery of new diagnostic markers and treatment targets against C. albicans infections. The data obtained point to different mechanisms of biogenesis of YEVs and HEVs, as well as different involvements in cell biology and host interaction. YEVs played a more relevant role in cell wall maintenance, while HEVs were more closely related to virulence, as they had greater effects on human immune cells. Importantly, an active 20S proteosome complex was described as a fungal-EV cargo. A deeper study of its role and those of many other proteins exclusively detected in HEVs and involved in different relevant biological processes of this fungus could open up interesting new areas of research in the battle against C. albicans.
Collapse
Affiliation(s)
- Raquel Martínez-López
- Department of Microbiology and Parasitology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Madrid, Spain
- Ramon y Cajal Health Research Institute (IRYCIS), Madrid, Spain
| | | | - Esther Redondo
- Department of Microbiology and Parasitology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Madrid, Spain
| | - Guillermo Calvo
- Department of Microbiology and Parasitology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Madrid, Spain
| | - Sonja Radau
- Thermo Fisher Scientific GmbH, Dreieich, Germany
| | - Mercedes Pardo
- Functional Proteomics, The Institute of Cancer Research, London, United Kingdom
| | - Concha Gil
- Department of Microbiology and Parasitology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Madrid, Spain
- Ramon y Cajal Health Research Institute (IRYCIS), Madrid, Spain
- Proteomics Unit, Complutense University of Madrid, Madrid, Spain
| | - Lucía Monteoliva
- Department of Microbiology and Parasitology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Madrid, Spain
- Ramon y Cajal Health Research Institute (IRYCIS), Madrid, Spain
| |
Collapse
|
13
|
Proteomic Profiling and Functional Analysis of B Cell-Derived Exosomes upon Pneumocystis Infection. J Immunol Res 2022; 2022:5187166. [PMID: 35465354 PMCID: PMC9023222 DOI: 10.1155/2022/5187166] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/30/2022] [Indexed: 12/27/2022] Open
Abstract
Pneumocystis is a life-threatening fungal pathogen that frequently causes fatal pneumonia (PCP) in immunocompromised individuals. Recently, B cells have been reported to play a crucial role in the pathogenesis of PCP through producing antibodies and activating CD4+ T cell response. Exosomes are nanoscale small extracellular vesicles abundant with protein cargo and can mediate immune response during infectious disease. In this study, using tandem mass tag-based quantitative proteomics coupled with bioinformatic analysis, we attempted to characterize exosomes derived from B lymphocytes in response to PCP. Several proteins were verified by parallel reaction monitoring (PRM) analysis. Also, the effects of B cell exosomes on CD4+ T cell response and phagocytic function of macrophages were clarified. Briefly, 1701 proteins were identified from B cell exosomes, and the majority of them were reported in Vesiclepedia. A total of 51 differentially expressed proteins of B cell exosomes were found in response to PCP. They were mainly associated with immune response and transcription regulation. PRM analysis confirmed the significantly changed levels of histone H1.3, vimentin, and tyrosine-protein phosphatase nonreceptor type 6 (PTPN6). Moreover, a functional study revealed the proinflammatory profile of B cell exosomes on CD4+ T cell response in PCP. Taken together, our results suggest the involvement of exosomes derived from B cells in cell-to-cell communication, providing new information on the function of B cells in response to PCP.
Collapse
|
14
|
Ceballos-Garzon A, Monteoliva L, Gil C, Alvarez-Moreno C, Vega-Vela NE, Engelthaler DM, Bowers J, Le Pape P, Parra-Giraldo CM. Genotypic, proteomic, and phenotypic approaches to decipher the response to caspofungin and calcineurin inhibitors in clinical isolates of echinocandin-resistant Candida glabrata. J Antimicrob Chemother 2021; 77:585-597. [PMID: 34893830 PMCID: PMC8865013 DOI: 10.1093/jac/dkab454] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 11/09/2021] [Indexed: 01/20/2023] Open
Abstract
Background Echinocandin resistance represents a great concern, as these drugs are recommended as first-line therapy for invasive candidiasis. Echinocandin resistance is conferred by mutations in FKS genes. Nevertheless, pathways are crucial for enabling tolerance, evolution, and maintenance of resistance. Therefore, understanding the biological processes and proteins involved in the response to caspofungin may provide clues indicating new therapeutic targets. Objectives We determined the resistance mechanism and assessed the proteome response to caspofungin exposure. We then evaluated the phenotypic impact of calcineurin inhibition by FK506 and cephalosporine A (CsA) on caspofungin-resistant Candida glabrata isolates. Methods Twenty-five genes associated with caspofungin resistance were analysed by NGS, followed by studies of the quantitative proteomic response to caspofungin exposure. Then, susceptibility testing of caspofungin in presence of FK506 and CsA was performed. The effects of calcineurin inhibitor/caspofungin combinations on heat stress (40°C), oxidative stress (0.2 and 0.4 mM menadione) and on biofilm formation (polyurethane catheter) were analysed. Finally, a Galleria mellonella model using blastospores (1 × 109 cfu/mL) was developed to evaluate the impact of the combinations on larval survival. Results F659-del was found in the FKS2 gene of resistant strains. Proteomics data showed some up-regulated proteins are involved in cell-wall biosynthesis, response to stress and pathogenesis, some of them being members of calmodulin–calcineurin pathway. Therefore, the impact of calmodulin inhibition was explored. Calmodulin inhibition restored caspofungin susceptibility, decreased capacity to respond to stress conditions, and reduced biofilm formation and in vivo pathogenicity. Conclusions Our findings confirm that calmodulin-calcineurin-Crz1 could provide a relevant target in life-threatening invasive candidiasis.
Collapse
Affiliation(s)
- Andres Ceballos-Garzon
- Unidad de Proteómica y Micosis Humanas, Grupo de Enfermedades Infecciosas Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, D.C., Colombia
- Department of Parasitology and Medical Mycology, Faculty of Pharmacy, University of Nantes, Nantes Atlantique Universities, Nantes, France
| | - Lucia Monteoliva
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Concha Gil
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
- Unidad de Proteómica, Universidad Complutense de Madrid, Madrid, Spain
| | - Carlos Alvarez-Moreno
- Department of Internal Medicine, Faculty of Medicine, Universidad Nacional de Colombia, Bogotá, Colombia
- Clínica Universitaria Colombia, Clinica Colsanitas, Bogotá, Colombia
| | - Nelson E Vega-Vela
- Unidad de Proteómica y Micosis Humanas, Grupo de Enfermedades Infecciosas Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, D.C., Colombia
| | | | - Jolene Bowers
- Translational Genomics Research Institute, Flagstaff, AZ, USA
| | - Patrice Le Pape
- Department of Parasitology and Medical Mycology, Faculty of Pharmacy, University of Nantes, Nantes Atlantique Universities, Nantes, France
| | - Claudia M Parra-Giraldo
- Unidad de Proteómica y Micosis Humanas, Grupo de Enfermedades Infecciosas Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, D.C., Colombia
- Corresponding author. E-mail:
| |
Collapse
|
15
|
Arteaga-Blanco LA, Bou-Habib DC. The Role of Extracellular Vesicles from Human Macrophages on Host-Pathogen Interaction. Int J Mol Sci 2021; 22:ijms221910262. [PMID: 34638604 PMCID: PMC8508751 DOI: 10.3390/ijms221910262] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/20/2021] [Accepted: 06/21/2021] [Indexed: 12/17/2022] Open
Abstract
The nano-sized membrane enclosed extracellular vesicles (EVs) released by virtually all cell types play an essential role in intercellular communication via delivering bio-molecules, such as nucleic acids, proteins, lipids, and other molecules to recipient cells. By mediating an active and steady-state cell-to-cell communication, EVs contribute to regulating and preserving cellular homeostasis. On the other hand, EVs can also spread pathogen-derived molecules during infections, subverting the host immune responses during infections and thus worsening pathophysiological processes. In recent years, the biological functioning of EVs has become a widespread research field in basic and clinical branches of medical sciences due to their potential role in therapeutic applications for several diseases. This review aims to summarize the main recent findings regarding the implication of EVs shed by human macrophages (MΦ-EVs) and how they can modulate the host immune response to control or increase the damage caused by infectious agents. We will also present the methods used to describe MΦ-EVs, as well as the potential of these EVs as disease diagnostic tools for some human pathogens. We believe that an in-depth understanding of the host–pathogen interactions mediated by MΦ-EVs may trigger the development of innovative therapeutic strategies against infectious diseases.
Collapse
Affiliation(s)
- Luis A. Arteaga-Blanco
- Laboratory on Thymus Research, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro 21040-900, Brazil
- Correspondence: (L.A.A.-B.); or (D.C.B.-H.)
| | - Dumith Chequer Bou-Habib
- Laboratory on Thymus Research, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro 21040-900, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation, Rio de Janeiro 21040-900, Brazil
- Correspondence: (L.A.A.-B.); or (D.C.B.-H.)
| |
Collapse
|
16
|
Cryptococcus neoformans -Infected Macrophages Release Proinflammatory Extracellular Vesicles: Insight into Their Components by Multi-omics. mBio 2021; 12:mBio.00279-21. [PMID: 33785616 PMCID: PMC8092229 DOI: 10.1128/mbio.00279-21] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Cryptococcus neoformans causes cryptococcal meningitis, which is frequent in patients with HIV/AIDS, especially in less-developed countries. The incidence of cryptococcal meningitis is close to 1 million each year globally. Cryptococcus neoformans causes deadly mycosis in immunocompromised individuals. Macrophages are key cells fighting against microbes. Extracellular vesicles (EVs) are cell-to-cell communication mediators. The roles of EVs from infected host cells in the interaction with Cryptococcus remain uninvestigated. Here, EVs from viable C. neoformans-infected macrophages reduced fungal burdens but led to shorter survival of infected mice. In vitro, EVs induced naive macrophages to an inflammatory phenotype. Transcriptome analysis showed that EVs from viable C. neoformans-infected macrophages activated immune-related pathways, including p53 in naive human and murine macrophages. Conserved analysis demonstrated that basic cell biological processes, including cell cycle and division, were activated by infection-derived EVs from both murine and human infected macrophages. Combined proteomics, lipidomics, and metabolomics of EVs from infected macrophages showed regulation of pathways such as extracellular matrix (ECM) receptors and phosphatidylcholine. This form of intermacrophage communication could serve to prepare cells at more distant sites of infection to resist C. neoformans infection.
Collapse
|
17
|
Halder LD, Babych S, Palme DI, Mansouri-Ghahnavieh E, Ivanov L, Ashonibare V, Langenhorst D, Prusty B, Rambach G, Wich M, Trinks N, Blango MG, Kornitzer D, Terpitz U, Speth C, Jungnickel B, Beyersdorf N, Zipfel PF, Brakhage AA, Skerka C. Candida albicans Induces Cross-Kingdom miRNA Trafficking in Human Monocytes To Promote Fungal Growth. mBio 2021; 13:e0356321. [PMID: 35132877 PMCID: PMC8822622 DOI: 10.1128/mbio.03563-21] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 12/08/2021] [Indexed: 12/16/2022] Open
Abstract
In response to infections, human immune cells release extracellular vesicles (EVs) that carry a situationally adapted cocktail of proteins and nucleic acids, including microRNAs (miRNAs), to coordinate the immune response. In this study, we identified hsa-miR-21-5p and hsa-miR-24-3p as the most common miRNAs in exosomes released by human monocytes in response to the pathogenic fungus Candida albicans. Functional analysis of miRNAs revealed that hsa-miR-24-3p, but not hsa-miR-21-5p, acted across species and kingdoms, entering C. albicans and inducing fungal cell growth by inhibiting translation of the cyclin-dependent kinase inhibitor Sol1. Packaging of hsa-miR-24-3p into monocyte exosomes required binding of fungal soluble β-glucan to complement receptor 3 (CR3) and binding of mannan to Toll-like receptor 4 (TLR4), resulting in receptor colocalization. Together, our in vitro and in vivo findings reveal a novel cross-species evasion mechanism by which C. albicans exploits a human miRNA to promote fungal growth and survival in the host. IMPORTANCE Over the last decade, communication between immune cells by extracellular vesicle-associated miRNAs has emerged as an important regulator of the coordinated immune response. Therefore, a thorough understanding of the conversation occurring via miRNAs, especially during infection, may provide novel insights into both the host reaction to the microbe as well as the microbial response. This study provides evidence that the pathogenic fungus C. albicans communicates with human monocytes and induces the release of a human miRNA that promotes fungal growth. This mechanism represents an unexpected cross-species interaction and implies that an inhibition of specific miRNAs offers new possibilities for the treatment of human fungal infections.
Collapse
Affiliation(s)
- Luke D. Halder
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| | - Svitlana Babych
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| | - Diana I. Palme
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| | - Elham Mansouri-Ghahnavieh
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| | - Lia Ivanov
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| | - Victory Ashonibare
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| | - Daniela Langenhorst
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Bhupesh Prusty
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Günter Rambach
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Melissa Wich
- Department of Cell Biology, Institute of Biochemistry and Biophysics, Friedrich Schiller University, Jena, Germany
| | - Nora Trinks
- Department of Biotechnology and Biophysics, University of Würzburg, Würzburg, Germany
| | - Matthew G. Blango
- Junior Research Group RNA Biology of Fungal Infections, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| | - Daniel Kornitzer
- Department of Molecular Microbiology, B. Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Ulrich Terpitz
- Department of Biotechnology and Biophysics, University of Würzburg, Würzburg, Germany
| | - Cornelia Speth
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Berit Jungnickel
- Department of Cell Biology, Institute of Biochemistry and Biophysics, Friedrich Schiller University, Jena, Germany
| | - Niklas Beyersdorf
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Peter F. Zipfel
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
- Friedrich Schiller University, Jena, Germany
| | - Axel A. Brakhage
- Friedrich Schiller University, Jena, Germany
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| | - Christine Skerka
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| |
Collapse
|
18
|
Freitas MS, Pessoni AM, Coelho C, Bonato VLD, Rodrigues ML, Casadevall A, Almeida F. Interactions of Extracellular Vesicles from Pathogenic Fungi with Innate Leukocytes. Curr Top Microbiol Immunol 2021; 432:89-120. [DOI: 10.1007/978-3-030-83391-6_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
19
|
Garcia-Ceron D, Bleackley MR, Anderson MA. Fungal Extracellular Vesicles in Pathophysiology. Subcell Biochem 2021; 97:151-177. [PMID: 33779917 DOI: 10.1007/978-3-030-67171-6_7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Fungal pathogens are a concern in medicine and agriculture that has been exacerbated by the emergence of antifungal-resistant varieties that severely threaten human and animal health, as well as food security. This had led to the search for new and sustainable treatments for fungal diseases. Innovative solutions require a deeper understanding of the interactions between fungal pathogens and their hosts, and the key determinants of fungal virulence. Recently, a link has emerged between the release of extracellular vesicles (EVs) and fungal virulence that may contribute to finding new methods for fungal control. Fungal EVs carry pigments, carbohydrates, protein, nucleic acids and other macromolecules with similar functions as those found in EVs from other organisms, however certain fungal features, such as the fungal cell wall, impact EV release and cargo. Fungal EVs modulate immune responses in the host, have a role in cell-cell communication and transport molecules that function in virulence. Understanding the function of fungal EVs will expand our knowledge of host-pathogen interactions and may provide new and specific targets for antifungal drugs and agrichemicals.
Collapse
|
20
|
Wang Y, Zhao M, Liu S, Guo J, Lu Y, Cheng J, Liu J. Macrophage-derived extracellular vesicles: diverse mediators of pathology and therapeutics in multiple diseases. Cell Death Dis 2020; 11:924. [PMID: 33116121 PMCID: PMC7595091 DOI: 10.1038/s41419-020-03127-z] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/08/2020] [Accepted: 10/09/2020] [Indexed: 02/06/2023]
Abstract
Macrophages (Mφ) are primary innate immune cells that exhibit diverse functions in response to different pathogens or stimuli, and they are extensively involved in the pathology of various diseases. Extracellular vesicles (EVs) are small vesicles released by live cells. As vital messengers, macrophage-derived EVs (Mφ-EVs) can transfer multiple types of bioactive molecules from macrophages to recipient cells, modulating the biological function of recipient cells. In recent years, Mφ-EVs have emerged as vital mediators not only in the pathology of multiple diseases such as inflammatory diseases, fibrosis and cancers, but also as mediators of beneficial effects in immunoregulation, cancer therapy, infectious defense, and tissue repair. Although many investigations have been performed to explore the diverse functions of Mφ-EVs in disease pathology and intervention, few studies have comprehensively summarized their detailed biological roles as currently understood. In this review, we briefly introduced an overview of macrophage and EV biology, and primarily focusing on current findings and future perspectives with respect to the pathological and therapeutic effects of Mφ-EVs in various diseases.
Collapse
Affiliation(s)
- Yizhuo Wang
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Meng Zhao
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Shuyun Liu
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jun Guo
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yanrong Lu
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| | - Jingqiu Cheng
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| | - Jingping Liu
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
21
|
Rizzo J, Rodrigues ML, Janbon G. Extracellular Vesicles in Fungi: Past, Present, and Future Perspectives. Front Cell Infect Microbiol 2020; 10:346. [PMID: 32760680 PMCID: PMC7373726 DOI: 10.3389/fcimb.2020.00346] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 06/05/2020] [Indexed: 12/21/2022] Open
Abstract
Extracellular vesicles (EVs) have garnered much interest in the cell biology and biomedical research fields. Many studies have reported the existence of EVs in all types of living cells, including in fifteen different fungal genera. EVs play diverse biological roles, from the regulation of physiological events and response to specific environmental conditions to the mediation of highly complex interkingdom communications. This review will provide a historical perspective on EVs produced by fungi and an overview of the recent discoveries in the field. We will also review the current knowledge about EV biogenesis and cargo, their role in cell-to-cell interactions, and methods of EV analysis. Finally, we will discuss the perspectives of EVs as vehicles for the delivery of biologically active molecules.
Collapse
Affiliation(s)
- Juliana Rizzo
- Unité Biologie des ARN des Pathogènes Fongiques, Département de Mycologie, Institut Pasteur, Paris, France
| | - Marcio L. Rodrigues
- Instituto Carlos Chagas, Fundação Oswaldo Cruz, Curitiba, Brazil
- Instituto de Microbiologia Paulo de Góes, Universidade Federal Do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Guilhem Janbon
- Unité Biologie des ARN des Pathogènes Fongiques, Département de Mycologie, Institut Pasteur, Paris, France
| |
Collapse
|
22
|
Halder LD, Jo EAH, Hasan MZ, Ferreira-Gomes M, Krüger T, Westermann M, Palme DI, Rambach G, Beyersdorf N, Speth C, Jacobsen ID, Kniemeyer O, Jungnickel B, Zipfel PF, Skerka C. Immune modulation by complement receptor 3-dependent human monocyte TGF-β1-transporting vesicles. Nat Commun 2020; 11:2331. [PMID: 32393780 PMCID: PMC7214408 DOI: 10.1038/s41467-020-16241-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 04/22/2020] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles have an important function in cellular communication. Here, we show that human and mouse monocytes release TGF-β1-transporting vesicles in response to the pathogenic fungus Candida albicans. Soluble β-glucan from C. albicans binds to complement receptor 3 (CR3, also known as CD11b/CD18) on monocytes and induces the release of TGF-β1-transporting vesicles. CR3-dependence is demonstrated using CR3-deficient (CD11b knockout) monocytes generated by CRISPR-CAS9 genome editing and isolated from CR3-deficient (CD11b knockout) mice. These vesicles reduce the pro-inflammatory response in human M1-macrophages as well as in whole blood. Binding of the vesicle-transported TGF-β1 to the TGF-β receptor inhibits IL1B transcription via the SMAD7 pathway in whole blood and induces TGFB1 transcription in endothelial cells, which is resolved upon TGF-β1 inhibition. Notably, human complement-opsonized apoptotic bodies induce production of similar TGF-β1-transporting vesicles in monocytes, suggesting that the early immune response might be suppressed through this CR3-dependent anti-inflammatory vesicle pathway.
Collapse
Affiliation(s)
- Luke D Halder
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, 07745, Jena, Germany
| | - Emeraldo A H Jo
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, 07745, Jena, Germany
| | - Mohammad Z Hasan
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, 07745, Jena, Germany
| | - Marta Ferreira-Gomes
- Department of Cell Biology, Institute of Biochemistry and Biophysics, Friedrich Schiller University, 07745, Jena, Germany
| | - Thomas Krüger
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology, 07745, Jena, Germany
| | - Martin Westermann
- Electron Microscopy Center, University Hospital Jena, 07743, Jena, Germany
| | - Diana I Palme
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, 07745, Jena, Germany
| | - Günter Rambach
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, A-6020, Innsbruck, Austria
| | - Niklas Beyersdorf
- Institute for Virology and Immunobiology, University of Würzburg, 97070, Würzburg, Germany
| | - Cornelia Speth
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, A-6020, Innsbruck, Austria
| | - Ilse D Jacobsen
- Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology, 07745, Jena, Germany.,Friedrich Schiller University, 07743, Jena, Germany
| | - Olaf Kniemeyer
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology, 07745, Jena, Germany
| | - Berit Jungnickel
- Department of Cell Biology, Institute of Biochemistry and Biophysics, Friedrich Schiller University, 07745, Jena, Germany
| | - Peter F Zipfel
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, 07745, Jena, Germany.,Friedrich Schiller University, 07743, Jena, Germany
| | - Christine Skerka
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, 07745, Jena, Germany.
| |
Collapse
|
23
|
Pellon A, Sadeghi Nasab SD, Moyes DL. New Insights in Candida albicans Innate Immunity at the Mucosa: Toxins, Epithelium, Metabolism, and Beyond. Front Cell Infect Microbiol 2020; 10:81. [PMID: 32195196 PMCID: PMC7062647 DOI: 10.3389/fcimb.2020.00081] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/18/2020] [Indexed: 12/16/2022] Open
Abstract
The mucosal surfaces of the human body are challenged by millions of microbes on a daily basis. Co-evolution with these microbes has led to the development of plastic mechanisms in both host and microorganisms that regulate the balance between preserving beneficial microbes and clearing pathogens. Candida albicans is a fungal pathobiont present in most healthy individuals that, under certain circumstances, can become pathogenic and cause everything from mild mucosal infections to life-threatening systemic diseases. As an essential part of the innate immunity in mucosae, epithelial cells elaborate complex immune responses that discriminate between commensal and pathogenic microbes, including C. albicans. Recently, several significant advances have been made identifying new pieces in the puzzle of host-microbe interactions. This review will summarize these advances in the context of our current knowledge of anti-Candida mucosal immunity, and their impact on epithelial immune responses to this fungal pathogen.
Collapse
Affiliation(s)
- Aize Pellon
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Shervin Dokht Sadeghi Nasab
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - David L Moyes
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| |
Collapse
|
24
|
González Plaza JJ. Small RNAs in cell-to-cell communications during bacterial infection. FEMS Microbiol Lett 2019; 365:4830097. [PMID: 29390095 DOI: 10.1093/femsle/fny024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 01/25/2018] [Indexed: 01/18/2023] Open
Abstract
Intercellular communication is a widespread phenomenon in all domains of life. Bacteria have developed many ways of communicating with one another and with other species, either prokaryotic or eukaryotic. RNA has been a key molecule since the beginning of life on Earth, and is one of the carriers of information. Given the current antibiotic crisis, understanding the way in which pathogens communicate can lead towards improved ways to control infections when antimicrobial therapy is not possible. Different subspecies of RNA, non-coding, and of small size, designated here as ncRNAs, have been in recent years the subject of a great research effort, and results have contributed to a growing field of knowledge. This review focuses on four different aspects of ncRNA involvement in cell-to-cell communications during bacterial infections: pathogen recognition by the host, alteration of host microRNA profiles, production of domestic and secreted forms of ncRNAs and subversion of the host responses. The current review article focuses on the most recent discoveries in the field and gives an integrative idea based on the discussed studies.
Collapse
Affiliation(s)
- Juan José González Plaza
- Research Department, University Hospital for Infectious Diseases "Dr. Fran Mihaljevic", Zagreb, Croatia
| |
Collapse
|
25
|
Pieters BCH, Cappariello A, van den Bosch MHJ, van Lent PLEM, Teti A, van de Loo FAJ. Macrophage-Derived Extracellular Vesicles as Carriers of Alarmins and Their Potential Involvement in Bone Homeostasis. Front Immunol 2019; 10:1901. [PMID: 31440259 PMCID: PMC6694442 DOI: 10.3389/fimmu.2019.01901] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 07/26/2019] [Indexed: 02/04/2023] Open
Abstract
Extracellular vesicles are a heterogeneous group of cell-derived membranous structures, which facilitate intercellular communication. Recent studies have highlighted the importance of extracellular vesicles in bone homeostasis, as mediators of crosstalk between different bone-resident cells. Osteoblasts and osteoclasts are capable of releasing various types of extracellular vesicles that promote both osteogenesis, as well as, osteoclastogenesis, maintaining bone homeostasis. However, the contribution of immune cell-derived extracellular vesicles in bone homeostasis remains largely unknown. Recent proteomic studies showed that alarmins are abundantly present in/on macrophage-derived EVs. In this review we will describe these alarmins in the context of bone matrix regulation and discuss the potential contribution macrophage-derived EVs may have in this process.
Collapse
Affiliation(s)
- Bartijn C H Pieters
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Alfredo Cappariello
- Research Laboratories - Department of Oncohematology IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | | | - Peter L E M van Lent
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Anna Teti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Fons A J van de Loo
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
26
|
Molinier-Frenkel V, Prévost-Blondel A, Castellano F. The IL4I1 Enzyme: A New Player in the Immunosuppressive Tumor Microenvironment. Cells 2019; 8:E757. [PMID: 31330829 PMCID: PMC6678094 DOI: 10.3390/cells8070757] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 07/18/2019] [Accepted: 07/19/2019] [Indexed: 12/14/2022] Open
Abstract
The high metabolic needs of T lymphocytes in response to activation make them particularly vulnerable to modifications of their biochemical milieu. Immunosuppressive enzymes produced in the tumor microenvironment modify nutrient availability by catabolizing essential or semi-essential amino acids and producing toxic catabolites, thus participating in the local sabotage of the antitumor immune response. L-amino-acid oxidases are FAD-bound enzymes found throughout evolution, from bacteria to mammals, and are often endowed with anti-infectious properties. IL4I1 is a secreted L-phenylalanine oxidase mainly produced by inflammatory antigen-presenting cells-in particular, macrophages present in T helper type 1 granulomas and in various types of tumors. In the last decade, it has been shown that IL4I1 is involved in the fine control of B- and T-cell adaptive immune responses. Preclinical models have revealed its role in cancer immune evasion. Recent clinical data highlight IL4I1 as a new potential prognostic marker in human melanoma. As a secreted enzyme, IL4I1 may represent an easily targetable molecule for cancer immunotherapy.
Collapse
Affiliation(s)
- Valérie Molinier-Frenkel
- INSERM, U955, Team 09, 94010 Créteil, France.
- Faculty of Medicine, University Paris Est, 94010 Créteil, France.
- AP-HP, H. Mondor - A. Chenevier Hospital, Biological Immunology Service, 94010 Créteil, France.
| | - Armelle Prévost-Blondel
- INSERM, U1016, Institute Cochin, 75014 Paris, France
- CNRS, UMR8104, 75014 Paris, France
- University Paris Descartes, Sorbonne Paris Cité, 75014 Paris, France
| | - Flavia Castellano
- INSERM, U955, Team 09, 94010 Créteil, France.
- Faculty of Medicine, University Paris Est, 94010 Créteil, France.
| |
Collapse
|
27
|
Mamrot J, Balachandran S, Steele EJ, Lindley RA. Molecular model linking Th2 polarized M2 tumour-associated macrophages with deaminase-mediated cancer progression mutation signatures. Scand J Immunol 2019; 89:e12760. [PMID: 30802996 PMCID: PMC6850162 DOI: 10.1111/sji.12760] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 02/19/2019] [Indexed: 12/11/2022]
Abstract
A new and diverse range of somatic mutation signatures are observed in late-stage cancers, but the underlying reasons are not fully understood. We advance a "combinatorial association model" for deaminase binding domain (DBD) diversification to explain the generation of previously observed cancer-progression associated mutation signatures. We also propose that changes in the polarization of tumour-associated macrophages (TAMs) are accompanied by the expression of deaminases with a new and diverse range of DBDs, and thus accounting for the generation of new somatic mutation signatures. The mechanism proposed is molecularly reminiscent of combinatorial association of heavy (H) and light (L) protein chains following V(D)J recombination of immunoglobulin molecules (and similarly for protein chains in heterodimers α/β and γ/δ of V(D)Js of T Cell Receptors) required for pathogen antigen recognition by B cells and T cells, respectively. We also discuss whether extracellular vesicles (EVs) emanating from tumour enhancing M2-polarized macrophages represent a likely source of the de novo deaminase DBDs. We conclude that M2-polarized macrophages extruding EVs loaded with deaminase proteins or deaminase-specific transcription/translation regulatory factors and like information may directly trigger deaminase diversification within cancer cells, and thus account for the many new somatic mutation signatures that are indicative of cancer progression. This hypothesis now has a plausible evidentiary base, and it is worth direct testing in future investigations. A long-term objective would be to identify molecular biomarkers predicting cancer progression (or metastatic disease) and to support the development of new drug targets before metastatic pathways are activated.
Collapse
Affiliation(s)
| | - Siddharth Balachandran
- Blood Cell Development and Function ProgramFox Chase Cancer CenterPhiladelphiaPennsylvania
| | - Edward J. Steele
- CYO’Connor ERADE Village FoundationPerthWestern AustraliaAustralia
- Melville Analytics Pty LtdMelbourneVictoriaAustralia
| | - Robyn A. Lindley
- GMDxCo Pty LtdMelbourneVictoriaAustralia
- Faculty of Medicine, Dentistry & Health Sciences, Department of Clinical PathologyUniversity of MelbourneMelbourneVictoriaAustralia
| |
Collapse
|
28
|
Vaz C, Reales-Calderon JA, Pitarch A, Vellosillo P, Trevisan M, Hernáez ML, Monteoliva L, Gil C. Enrichment of ATP Binding Proteins Unveils Proteomic Alterations in Human Macrophage Cell Death, Inflammatory Response, and Protein Synthesis after Interaction with Candida albicans. J Proteome Res 2019; 18:2139-2159. [PMID: 30985132 DOI: 10.1021/acs.jproteome.9b00032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Macrophages are involved in the primary human response to Candida albicans. After pathogen recognition, signaling pathways are activated, leading to the production of cytokines, chemokines, and antimicrobial peptides. ATP binding proteins are crucial for this regulation. Here, a quantitative proteomic and phosphoproteomic approach was carried out for the study of human macrophage ATP-binding proteins after interaction with C. albicans. From a total of 547 nonredundant quantified proteins, 137 were ATP binding proteins and 59 were detected as differentially abundant. From the differentially abundant ATP-binding proteins, 6 were kinases (MAP2K2, SYK, STK3, MAP3K2, NDKA, and SRPK1), most of them involved in signaling pathways. Furthermore, 85 phosphopeptides were quantified. Macrophage proteomic alterations including an increase of protein synthesis with a consistent decrease in proteolysis were observed. Besides, macrophages showed changes in proteins of endosomal trafficking together with mitochondrial proteins, including some involved in the response to oxidative stress. Regarding cell death mechanisms, an increase of antiapoptotic over pro-apoptotic signals is suggested. Furthermore, a high pro-inflammatory response was detected, together with no upregulation of key mi-RNAs involved in the negative feedback of this response. These findings illustrate a strategy to deepen the knowledge of the complex interactions between the host and the clinically important pathogen C. albicans.
Collapse
Affiliation(s)
- Catarina Vaz
- Departamento de Microbiologı́a y Parasitología, Facultad de Farmacia , Universidad Complutense de Madrid , 28040 Madrid , Spain.,Instituto Ramón y Cajal de Investigación Sanitaria IRYCIS , 28034 Madrid , Spain
| | - Jose Antonio Reales-Calderon
- Departamento de Microbiologı́a y Parasitología, Facultad de Farmacia , Universidad Complutense de Madrid , 28040 Madrid , Spain.,Instituto Ramón y Cajal de Investigación Sanitaria IRYCIS , 28034 Madrid , Spain
| | - Aida Pitarch
- Departamento de Microbiologı́a y Parasitología, Facultad de Farmacia , Universidad Complutense de Madrid , 28040 Madrid , Spain.,Instituto Ramón y Cajal de Investigación Sanitaria IRYCIS , 28034 Madrid , Spain
| | - Perceval Vellosillo
- Departamento de Microbiologı́a y Parasitología, Facultad de Farmacia , Universidad Complutense de Madrid , 28040 Madrid , Spain
| | - Marco Trevisan
- Laboratorio de Proteómica Cardiovascular , Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) , 28029 Madrid , Spain
| | - María Luisa Hernáez
- Unidad de Proteómica , Universidad Complutense de Madrid , 28040 Madrid , Spain
| | - Lucía Monteoliva
- Departamento de Microbiologı́a y Parasitología, Facultad de Farmacia , Universidad Complutense de Madrid , 28040 Madrid , Spain.,Instituto Ramón y Cajal de Investigación Sanitaria IRYCIS , 28034 Madrid , Spain
| | - Concha Gil
- Departamento de Microbiologı́a y Parasitología, Facultad de Farmacia , Universidad Complutense de Madrid , 28040 Madrid , Spain.,Instituto Ramón y Cajal de Investigación Sanitaria IRYCIS , 28034 Madrid , Spain.,Unidad de Proteómica , Universidad Complutense de Madrid , 28040 Madrid , Spain
| |
Collapse
|
29
|
Wang P, Wang H, Huang Q, Peng C, Yao L, Chen H, Qiu Z, Wu Y, Wang L, Chen W. Exosomes from M1-Polarized Macrophages Enhance Paclitaxel Antitumor Activity by Activating Macrophages-Mediated Inflammation. Theranostics 2019; 9:1714-1727. [PMID: 31037133 PMCID: PMC6485189 DOI: 10.7150/thno.30716] [Citation(s) in RCA: 318] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 01/07/2019] [Indexed: 12/19/2022] Open
Abstract
Objective: Exosomes (Exos) are membrane-encased vesicles derived by nearly all cell types for intercellular communication and regulation. They also received attention for their use as natural therapeutic platforms and drug delivery system. Classically activated M1 macrophages suppress tumor growth by releasing pro-inflammatory factors. This study investigated the suitability of M1-exosomes (M1-Exos) as drug carrier and their effect on the NF-κB signal pathway and further detected whether macrophages repolarization can potentiate the antitumor activities of chemotherapeutics. Methods: M1-Exos were isolated from M1-macrophages by ultracentrifugation and characterized by transmission electron, nanoparticle tracking analysis, dynamic light scattering and western blot. Then M1-Exos were used as Paclitaxel (PTX) carriers to prepare a nano-formulation (PTX- M1-Exos). A relatively simple slight sonication method was used to prepare the drug delivery system (PTX-M1-Exos). The cytotoxicity of PTX-M1-Exos on cancer cells was detected by MTT and flow cytometry in vitro. 4T1 tumor bearing mice were used to perform the therapeutic effect of PTX-M1-Exos in vivo. Results: The expression of caspase-3 in breast cancer cells was increased when co-incubated with macrophages in the presence of M1-Exos in vitro. The production of pro-inflammatory cytokines was increased after exposure of macrophages in M1-Exos. M1-Exos provided a pro-inflammatory environment which enhanced the anti-tumor activity via caspase-3 mediated pathway. The treatment of M1-Exos to the tumor bearing mice exhibit anti-tumor effects in vivo. Meanwhile, the treatment of PTX-M1-Exos demonstrated higher anti-tumor effects than the M1-Exos or PTX group. Conclusion: The results in our study indicate that the M1-Exos act as the carrier to deliver PTX into the tumor tissues, and also enhance the anti-tumor effects of chemotherapeutics in tumor bearing mice.
Collapse
Affiliation(s)
- Piaopiao Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
- Institute of Drug Metabolism, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Huihui Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
- Institute of Drug Metabolism, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Qianqian Huang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
- Institute of Drug Metabolism, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Can Peng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
- Institute of Drug Metabolism, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui, 230012, China
- Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, Anhui, 230012, China
- Synergetic Innovation Center of Anhui Authentic Chinese Medicine Quality Improvement, Hefei, Anhui, 230012, China
| | - Liang Yao
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
- Institute of Drug Metabolism, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Hong Chen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
- Institute of Drug Metabolism, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Zhen Qiu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
- Institute of Drug Metabolism, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Yifan Wu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
- Institute of Drug Metabolism, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Lei Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui, 230012, China
- Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Weidong Chen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
- Institute of Drug Metabolism, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui, 230012, China
- Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, Anhui, 230012, China
- Synergetic Innovation Center of Anhui Authentic Chinese Medicine Quality Improvement, Hefei, Anhui, 230012, China
| |
Collapse
|
30
|
Yang Y, Boza-Serrano A, Dunning CJR, Clausen BH, Lambertsen KL, Deierborg T. Inflammation leads to distinct populations of extracellular vesicles from microglia. J Neuroinflammation 2018; 15:168. [PMID: 29807527 PMCID: PMC5972400 DOI: 10.1186/s12974-018-1204-7] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 05/15/2018] [Indexed: 11/10/2022] Open
Abstract
Background Activated microglia play an essential role in inflammatory responses elicited in the central nervous system (CNS). Microglia-derived extracellular vesicles (EVs) are suggested to be involved in propagation of inflammatory signals and in the modulation of cell-to-cell communication. However, there is a lack of knowledge on the regulation of EVs and how this in turn facilitates the communication between cells in the brain. Here, we characterized microglial EVs under inflammatory conditions and investigated the effects of inflammation on the EV size, quantity, and protein content. Methods We have utilized western blot, nanoparticle tracking analysis (NTA), and mass spectrometry to characterize EVs and examine the alterations of secreted EVs from a microglial cell line (BV2) following lipopolysaccharide (LPS) and tumor necrosis factor (TNF) inhibitor (etanercept) treatments, or either alone. The inflammatory responses were measured with multiplex cytokine ELISA and western blot. We also subjected TNF knockout mice to experimental stroke (permanent middle cerebral artery occlusion) and validated the effect of TNF inhibition on EV release. Results Our analysis of EVs originating from activated BV2 microglia revealed a significant increase in the intravesicular levels of TNF and interleukin (IL)-6. We also observed that the number of EVs released was reduced both in vitro and in vivo when inflammation was inhibited via the TNF pathway. Finally, via mass spectrometry, we identified 49 unique proteins in EVs released from LPS-activated microglia compared to control EVs (58 proteins in EVs released from LPS-activated microglia and 37 from control EVs). According to Gene Ontology (GO) analysis, we found a large increase of proteins related to translation and transcription in EVs from LPS. Importantly, we showed a distinct profile of proteins found in EVs released from LPS treated cells compared to control. Conclusions We demonstrate altered EV production in BV2 microglial cells and altered cytokine levels and protein composition carried by EVs in response to LPS challenge. Our findings provide new insights into the potential roles of EVs that could be related to the pathogenesis in neuroinflammatory diseases. Electronic supplementary material The online version of this article (10.1186/s12974-018-1204-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yiyi Yang
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden.
| | - Antonio Boza-Serrano
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | | | - Bettina Hjelm Clausen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,BRIGDE-Brain Research-Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Kate Lykke Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,BRIGDE-Brain Research-Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Neurology, Odense University Hospital, Odense, Denmark
| | - Tomas Deierborg
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden.
| |
Collapse
|
31
|
Monocarboxylate transporter 1 (MCT1), a tool to stratify acute myeloid leukemia (AML) patients and a vehicle to kill cancer cells. Oncotarget 2017; 8:82803-82823. [PMID: 29137304 PMCID: PMC5669930 DOI: 10.18632/oncotarget.20294] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 06/27/2017] [Indexed: 12/12/2022] Open
Abstract
Dysregulation of glucose/lactate dynamics plays a role in cancer progression, and MCTs are key elements in metabolic remodeling. VEGF is a relevant growth factor in the maintenance of bone marrow microenvironment and it is also important in hematological diseases. Our aim was to investigate the role of VEGF in the metabolic adaptation of Acute myeloid leukemia (AML) cells by evaluating the metabolic profiles and cell features according to the AML lineage and testing lactate as a metabolic coin. Our in vitro results showed that AML promyelocytic (HL60) and monocytic (THP1) (but not erythroid- HEL) lineages are well adapted to VEGF and lactate rich environment. Their metabolic adaptation relies on high rates of glycolysis to generate intermediates for PPP to support cell proliferation, and on the consumption of glycolysis-generated lactate to supply biomass and energy production. VEGF orchestrates this metabolic network by regulating MCT1 expression. Bromopyruvic acid (BPA) was proven to be an effective cytotoxic in AML, possibly transported by MCT1. Our study reinforces that targeting metabolism can be a good strategy to fight cancer. MCT1 expression at the time of diagnosis can assist on the identification of AML patients that will benefit from BPA therapy. Additionally, MCT1 can be used in targeted delivery of conventional cytotoxic drugs.
Collapse
|