1
|
dos Santos F, Vindel-Alfageme J, Ciordia S, Castro V, Orera I, Garaigorta U, Gastaminza P, Corrales F. Dynamic Cellular Proteome Remodeling during SARS-CoV-2 Infection. Identification of Plasma Protein Readouts. J Proteome Res 2025; 24:171-188. [PMID: 39593238 PMCID: PMC11705369 DOI: 10.1021/acs.jproteome.4c00566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 11/06/2024] [Accepted: 11/19/2024] [Indexed: 11/28/2024]
Abstract
The outbreak of COVID-19, led to an ongoing pandemic with devastating consequences for the global economy and human health. With the global spread of SARS-CoV-2, multidisciplinary initiatives were launched to explore new diagnostic, therapeutic, and vaccination strategies. From this perspective, proteomics could help to understand the mechanisms associated with SARS-CoV-2 infection and to identify new therapeutic options. A TMT-based quantitative proteomics and phosphoproteomics analysis was performed to study the proteome remodeling of human lung alveolar cells expressing human ACE2 (A549-ACE2) after infection with SARS-CoV-2. Detectability and the prognostic value of selected proteins was analyzed by targeted PRM. A total of 6802 proteins and 6428 phospho-sites were identified in A549-ACE2 cells after infection with SARS-CoV-2. The differential proteins here identified revealed that A549-ACE2 cells undergo a time-dependent regulation of essential processes, delineating the precise intervention of the cellular machinery by the viral proteins. From this mechanistic background and by applying machine learning modeling, 29 differential proteins were selected and detected in the serum of COVID-19 patients, 14 of which showed promising prognostic capacity. Targeting these proteins and the protein kinases responsible for the reported phosphorylation changes may provide efficient alternative strategies for the clinical management of COVID-19.
Collapse
Affiliation(s)
- Fátima
Milhano dos Santos
- Functional
Proteomics Laboratory, National Center for
Biotechnology (CNB-CSIC), Darwin 3, Madrid 28049, Spain
| | - Jorge Vindel-Alfageme
- Functional
Proteomics Laboratory, National Center for
Biotechnology (CNB-CSIC), Darwin 3, Madrid 28049, Spain
| | - Sergio Ciordia
- Functional
Proteomics Laboratory, National Center for
Biotechnology (CNB-CSIC), Darwin 3, Madrid 28049, Spain
| | - Victoria Castro
- Department
of Molecular and Cell Biology, National
Center for Biotechnology (CNB-CSIC), Darwin 3, Madrid 28049, Spain
| | - Irene Orera
- Proteomics
Research Core Facility, Instituto Aragonés
de Ciencias de la Salud (IACS), Zaragoza 50009, Spain
| | - Urtzi Garaigorta
- Department
of Molecular and Cell Biology, National
Center for Biotechnology (CNB-CSIC), Darwin 3, Madrid 28049, Spain
| | - Pablo Gastaminza
- Department
of Molecular and Cell Biology, National
Center for Biotechnology (CNB-CSIC), Darwin 3, Madrid 28049, Spain
| | - Fernando Corrales
- Functional
Proteomics Laboratory, National Center for
Biotechnology (CNB-CSIC), Darwin 3, Madrid 28049, Spain
| |
Collapse
|
2
|
Gebetsberger L, Malekshahi Z, Teutsch A, Tajti G, Fontaine F, Marella N, Mueller A, Prantl L, Stockinger H, Stoiber H, Ohradanova-Repic A. SARS-CoV-2 hijacks host CD55, CD59 and factor H to impair antibody-dependent complement-mediated lysis. Emerg Microbes Infect 2024; 13:2417868. [PMID: 39435487 PMCID: PMC11520101 DOI: 10.1080/22221751.2024.2417868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 09/18/2024] [Accepted: 10/13/2024] [Indexed: 10/23/2024]
Abstract
The complement system is a vital anti-microbial defence mechanism against circulating pathogens. Excessive complement activation can have deleterious outcomes for the host and is consequently tightly modulated by a set of membrane-associated and fluid-phase regulators of complement activation (RCAs). Here, we demonstrate that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) hijacks host cellular RCA members CD55 and CD59 and serum-derived Factor H (FH) to resist antibody-dependent complement-mediated lysis triggered by immunized human sera. Blockage of the biological functions of virion-associated CD55 and CD59 and competition of FH recruitment with functionally inactive recombinant FH-derived short consensus repeats SCR18-20 restore SARS-CoV-2 complement sensitivity in a synergistic manner. Moreover, complement-mediated virolysis is dependent on classical pathway activation and does not occur in the absence of virus-specific antibodies. Altogether, our findings present an intriguing immune escape mechanism that provides novel insights into the immunopathology observed in severe coronavirus disease 2019 (COVID-19).
Collapse
Affiliation(s)
- Laura Gebetsberger
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Zahra Malekshahi
- Medical University of Innsbruck, Institute of Virology, Innsbruck, Austria
| | - Aron Teutsch
- Medical University of Innsbruck, Institute of Virology, Innsbruck, Austria
| | - Gabor Tajti
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Frédéric Fontaine
- CeMM – Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Nara Marella
- CeMM – Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - André Mueller
- CeMM – Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Lena Prantl
- Medical University of Innsbruck, Institute of Virology, Innsbruck, Austria
| | - Hannes Stockinger
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Heribert Stoiber
- Medical University of Innsbruck, Institute of Virology, Innsbruck, Austria
| | - Anna Ohradanova-Repic
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| |
Collapse
|
3
|
Hulugalla K, Shofolawe-Bakare O, Toragall VB, Mohammad SA, Mayatt R, Hand K, Anderson J, Chism C, Misra SK, Shaikh T, Tanner EEL, Smith AE, Sharp JS, Fitzkee NC, Werfel T. Glycopolymeric Nanoparticles Enrich Less Immunogenic Protein Coronas, Reduce Mononuclear Phagocyte Clearance, and Improve Tumor Delivery Compared to PEGylated Nanoparticles. ACS NANO 2024; 18:30540-30560. [PMID: 39436672 DOI: 10.1021/acsnano.4c08922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
Nanoparticles (NPs) offer significant promise as drug delivery vehicles; however, their in vivo efficacy is often hindered by the formation of a protein corona (PC), which influences key physiological responses such as blood circulation time, biodistribution, cellular uptake, and intracellular localization. Understanding NP-PC interactions is crucial for optimizing NP design for biomedical applications. Traditional approaches have utilized hydrophilic polymer coatings like polyethylene glycol (PEG) to resist protein adsorption, but glycopolymer-coated nanoparticles have emerged as potential alternatives due to their biocompatibility and ability to reduce the adsorption of highly immunogenic proteins. In this study, we synthesized and characterized glycopolymer-based poly[2-(diisopropylamino)ethyl methacrylate-b-poly(methacrylamidoglucopyranose) (PDPA-b-PMAG) NPs as an alternative to PEGylated NPs. We characterized the polymers using a range of techniques to establish their molecular weight and chemical composition. PMAG and PEG-based NPs showed equivalent physicochemical properties with sizes of ∼100 nm, spherical morphology, and neutral surface charges. We next assessed the magnitude of protein adsorption on both NPs and catalogued the identity of the adsorbed proteins using mass spectrometry-based techniques. The PMAG NPs were found to adsorb fewer proteins in vitro as well as fewer immunogenic proteins such as Immunoglobulins and Complement proteins. Flow cytometry and confocal microscopy were employed to examine cellular uptake in RAW 264.7 macrophages and MDA-MB-231 tumor cells, where PMAG NPs showed higher uptake into tumor cells over macrophages. In vivo studies in BALB/c mice with orthotopic 4T1 breast cancer xenografts showed that PMAG NPs exhibited prolonged circulation times and enhanced tumor accumulation compared to PEGylated NPs. The biodistribution analysis also revealed greater selectivity for tumor tissue over the liver for PMAG NPs. These findings highlight the potential of glycopolymeric NPs to improve tumor targeting and reduce macrophage uptake compared to PEGylated NPs, offering significant advancements in cancer nanomedicine and immunotherapy.
Collapse
Affiliation(s)
- Kenneth Hulugalla
- Department of BioMolecular Sciences, University of Mississippi, University, Mississippi 38677, United States
| | - Oluwaseyi Shofolawe-Bakare
- Department of Chemical Engineering, University of Mississippi, University, Mississippi 38677, United States
| | - Veeresh B Toragall
- Department of Biomedical Engineering, University of Mississippi, University, Mississippi 38677, United States
| | - Sk Arif Mohammad
- Department of Biomedical Engineering, University of Mississippi, University, Mississippi 38677, United States
| | - Railey Mayatt
- Department of Chemistry, Mississippi State University, Starkville, Mississippi 39762, United States
| | - Kelsie Hand
- Department of Biomedical Engineering, University of Mississippi, University, Mississippi 38677, United States
| | - Joshua Anderson
- Department of Biomedical Engineering, University of Mississippi, University, Mississippi 38677, United States
| | - Claylee Chism
- Department of Chemistry and Biochemistry, University of Mississippi, University, Mississippi 38677, United States
| | - Sandeep K Misra
- Department of BioMolecular Sciences, University of Mississippi, University, Mississippi 38677, United States
| | - Tanveer Shaikh
- Department of Chemistry, Mississippi State University, Starkville, Mississippi 39762, United States
| | - Eden E L Tanner
- Department of Chemistry and Biochemistry, University of Mississippi, University, Mississippi 38677, United States
| | - Adam E Smith
- Department of Biomedical Engineering, University of Mississippi, University, Mississippi 38677, United States
- Department of Chemical Engineering, University of Mississippi, University, Mississippi 38677, United States
| | - Joshua S Sharp
- Department of BioMolecular Sciences, University of Mississippi, University, Mississippi 38677, United States
| | - Nicholas C Fitzkee
- Department of Chemistry, Mississippi State University, Starkville, Mississippi 39762, United States
| | - Thomas Werfel
- Department of BioMolecular Sciences, University of Mississippi, University, Mississippi 38677, United States
- Department of Biomedical Engineering, University of Mississippi, University, Mississippi 38677, United States
- Department of Chemical Engineering, University of Mississippi, University, Mississippi 38677, United States
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, Mississippi 39216, United States
| |
Collapse
|
4
|
Joharinia N, Bonneil É, Grandvaux N, Thibault P, Lippé R. Comprehensive proteomic analysis of HCoV-OC43 virions and virus-modulated extracellular vesicles. J Virol 2024; 98:e0085024. [PMID: 38953378 PMCID: PMC11265355 DOI: 10.1128/jvi.00850-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 06/13/2024] [Indexed: 07/04/2024] Open
Abstract
Viruses are obligate parasites that depend on the cellular machinery for their propagation. Several viruses also incorporate cellular proteins that facilitate viral spread. Defining these cellular proteins is critical to decipher viral life cycles and delineate novel therapeutic strategies. While numerous studies have explored the importance of host proteins in coronavirus spread, information about their presence in mature virions is limited. In this study, we developed a protocol to highly enrich mature HCoV-OC43 virions and characterize them by proteomics. Recognizing that cells release extracellular vesicles whose content is modulated by viruses, and given our ability to separate virions from these vesicles, we also analyzed their protein content in both uninfected and infected cells. We uncovered 69 unique cellular proteins associated with virions including 31 high-confidence hits. These proteins primarily regulate RNA metabolism, enzymatic activities, vesicular transport, cell adhesion, metabolite interconversion, and translation. We further discovered that the virus had a profound impact on exosome composition, incorporating 47 novel cellular proteins (11 high confidence) and excluding 92 others (61 high confidence) in virus-associated extracellular vesicles compared to uninfected cells. Moreover, a dsiRNA screen revealed that 11 of 18 select targets significantly impacted viral yields, including proteins found in virions or extracellular vesicles. Overall, this study provides new and important insights into the incorporation of numerous host proteins into HCoV-OC43 virions, their biological significance, and the ability of the virus to modulate extracellular vesicles. IMPORTANCE In recent years, coronaviruses have dominated global attention, making it crucial to develop methods to control them and prevent future pandemics. Besides viral proteins, host proteins play a significant role in viral propagation and offer potential therapeutic targets. Targeting host proteins is advantageous because they are less likely to mutate and develop resistance compared to viral proteins, a common issue with many antiviral treatments. In this study, we examined the protein content of the less virulent biosafety level 2 HCoV-OC43 virus as a stand-in for the more virulent SARS-CoV-2. Our findings reveal that several cellular proteins incorporated into the virion regulate viral spread. In addition, we report that the virus extensively modulates the content of extracellular vesicles, enhancing viral dissemination. This underscores the critical interplay between the virus, host proteins, and extracellular vesicles.
Collapse
Affiliation(s)
- Negar Joharinia
- Azrieli Research center of the CHU Sainte-Justine, Montreal, Quebec, Canada
- Department of Microbiology, Infectiology and Immunology, University of Montreal, Montreal, Quebec, Canada
| | - Éric Bonneil
- IRIC, University of Montreal, Montreal, Quebec, Canada
| | - Nathalie Grandvaux
- Research center of the CHUM (CRCHUM), Montreal, Quebec, Canada
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Pierre Thibault
- IRIC, University of Montreal, Montreal, Quebec, Canada
- Department of Chemistry, University of Montreal, Montreal, Quebec, Canada
| | - Roger Lippé
- Azrieli Research center of the CHU Sainte-Justine, Montreal, Quebec, Canada
- Department of Pathology and Cell biology, University of Montreal, Montreal, Quebec, Canada
| |
Collapse
|
5
|
Sołkiewicz K, Kokot I, Kacperczyk M, Dymicka-Piekarska V, Dorf J, Kratz EM. Serum Clusterin Concentration and Its Glycosylation Changes as Potential New Diagnostic Markers of SARS-CoV-2 Infection and Recovery Process. Int J Mol Sci 2024; 25:4198. [PMID: 38673784 PMCID: PMC11049940 DOI: 10.3390/ijms25084198] [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: 03/05/2024] [Revised: 04/05/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
COVID-19 is an infectious disease caused by the SARS-CoV-2 virus. Glycoprotein clusterin (CLU) has many functions such as phagocyte recruitment, complement system inhibition, apoptosis inhibition, hormone and lipid transport, as well as in the immune response. The study aimed to assess the changes in CLU concentrations and the profile and degree of CLU glycosylation between patients with severe COVID-19, convalescents, and healthy subjects (control). The profile and degree of serum CLU N-glycosylation were analyzed using lectin-ELISA with specific lectins. CLU concentrations were significantly lower and relative reactivities of CLU glycans with SNA (Sambucus nigra agglutinin) were significantly higher in severe COVID-19 patients in comparison to convalescents and the control group. The relative reactivities of CLU glycans with MAA (Maackia amurensis agglutinin), together with relative reactivity with LCA (Lens culinaris agglutinin), were also significantly higher in patients with severe COVID-19 than in convalescents and the control group, but they also significantly differed between convalescents and control. The development of acute inflammation in the course of severe COVID-19 is associated with a decrease in CLU concentration, accompanied by an increase in the expression of α2,3-linked sialic acid, and core fucose. Both of these parameters can be included as useful glycomarkers differentiating patients with severe COVID-19 from convalescents and the control group, as well as convalescents and healthy subjects.
Collapse
Affiliation(s)
- Katarzyna Sołkiewicz
- Department of Laboratory Diagnostics, Division of Laboratory Diagnostics, Faculty of Pharmacy, Wroclaw Medical University, Borowska Street 211a, 50-556 Wroclaw, Poland; (I.K.); (M.K.)
| | - Izabela Kokot
- Department of Laboratory Diagnostics, Division of Laboratory Diagnostics, Faculty of Pharmacy, Wroclaw Medical University, Borowska Street 211a, 50-556 Wroclaw, Poland; (I.K.); (M.K.)
| | - Monika Kacperczyk
- Department of Laboratory Diagnostics, Division of Laboratory Diagnostics, Faculty of Pharmacy, Wroclaw Medical University, Borowska Street 211a, 50-556 Wroclaw, Poland; (I.K.); (M.K.)
| | - Violetta Dymicka-Piekarska
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, Waszyngtona 15A St., 15-269 Bialystok, Poland; (V.D.-P.); (J.D.)
| | - Justyna Dorf
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, Waszyngtona 15A St., 15-269 Bialystok, Poland; (V.D.-P.); (J.D.)
| | - Ewa Maria Kratz
- Department of Laboratory Diagnostics, Division of Laboratory Diagnostics, Faculty of Pharmacy, Wroclaw Medical University, Borowska Street 211a, 50-556 Wroclaw, Poland; (I.K.); (M.K.)
| |
Collapse
|
6
|
Dhaliwal M, Muthu V, Sharma A, Raj K, Rudramurthy SM, Agarwal R, Kaur H, Rawat A, Singh S, Chakrabarti A. Immune and metabolic perturbations in COVID-19-associated pulmonary mucormycosis: A transcriptome analysis of innate immune cells. Mycoses 2024; 67:e13679. [PMID: 38214399 DOI: 10.1111/myc.13679] [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: 10/18/2023] [Accepted: 11/23/2023] [Indexed: 01/13/2024]
Abstract
BACKGROUND AND OBJECTIVES The mechanisms underlying COVID-19-associated pulmonary mucormycosis (CAPM) remain unclear. We use a transcriptomic analysis of the innate immune cells to investigate the host immune and metabolic response pathways in patients with CAPM. PATIENTS AND METHODS We enrolled subjects with CAPM (n = 5), pulmonary mucormycosis (PM) without COVID-19 (n = 5), COVID-19 (without mucormycosis, n = 5), healthy controls (n = 5) without comorbid illness and negative for SARS-CoV-2. Peripheral blood samples from cases were collected before initiating antifungal therapy, and neutrophils and monocytes were isolated. RNA sequencing was performed using Illumina HiSeqX from monocytes and neutrophils. Raw reads were aligned with HISAT-2 pipeline and DESeq2 was used for differential gene expression. Gene ontology (GO) and metabolic pathway analysis were performed using Shiny GO application and R packages (ggplot2, Pathview). RESULTS The derangement of core immune and metabolic responses in CAPM patients was noted. Pattern recognition receptors, dectin-2, MCL, FcRγ receptors and CLEC-2, were upregulated, but signalling pathways such as JAK-STAT, IL-17 and CARD-9 were downregulated; mTOR and MAP-kinase signalling were elevated in monocytes from CAPM patients. The complement receptors, NETosis, and pro-inflammatory responses, such as S100A8/A9, lipocalin and MMP9, were elevated. The major metabolic pathways of glucose metabolism-glycolysis/gluconeogenesis, pentose phosphate pathway, HIF signalling and iron metabolism-ferroptosis were also upregulated in CAPM. CONCLUSIONS We identified significant alterations in the metabolic pathways possibly leading to cellular iron overload and a hyperglycaemic state. Immune responses revealed altered recognition, signalling, effector functions and a pro-inflammatory state in monocytes and neutrophils from CAPM patients.
Collapse
Affiliation(s)
- Manpreet Dhaliwal
- Department of Paediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Valliappan Muthu
- Department of Pulmonary Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Arunima Sharma
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Khem Raj
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Shivaprakash M Rudramurthy
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ritesh Agarwal
- Department of Pulmonary Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Harsimran Kaur
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Amit Rawat
- Department of Paediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Surjit Singh
- Department of Paediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Arunaloke Chakrabarti
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
7
|
Cheng H, Wang S, Huang A, Ma J, Gao D, Li M, Chen H, Guo K. HSF1 is involved in immunotherapeutic response through regulating APOJ/STAT3-mediated PD-L1 expression in hepatocellular carcinoma. Cancer Biol Ther 2023; 24:1-9. [PMID: 36482717 PMCID: PMC9746510 DOI: 10.1080/15384047.2022.2156242] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 11/17/2022] [Accepted: 11/23/2022] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular cancer (HCC) is a serious illness with high prevalence and mortality throughout the whole world. For advanced HCC, immunotherapy is somewhat impactful and encouraging. Nevertheless, a substantial proportion of patients with advanced HCC are still unable to achieve a durable response, owing to heterogeneity from clonal variability and differential expression of the PD-1/PD-L1 axis. Recently, heat shock factor 1 (HSF1) is recognized as an important component of tumor immunotherapeutic response as well as related to PD-L1 expression in cancer. However, the mechanism of HSF1 regulating PD-L1 in cancer, especially in HCC, is still not fully clear. In this study, we observed the significantly positive correlation between HSF1 expression and PD-L1 expression in HCC samples; meanwhile combination expressions of HSF1 and PD-L1 served as the signature for predicting prognosis of patients with HCC. Mechanistically, HSF1 upregulated PD-L1 expression by inducing APOJ expression and activating STAT3 signaling pathway in HCC. In addition, we explored further the potential values of targeting the HSF1-APOJ-STAT3 axis against CD8+ T cells-mediated cancer cells cytotoxicity. These findings unveiled the important involvement of HSF1 in regulating PD-L1 expression in HCC as well as provided a novel invention component for improving the clinical response rate and efficacy of PD-1/PD-L1 blockade.
Collapse
Affiliation(s)
- Hongxia Cheng
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion Ministry of Education, Shanghai, People’s Republic of China
| | - Sikai Wang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion Ministry of Education, Shanghai, People’s Republic of China
| | - Aidan Huang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion Ministry of Education, Shanghai, People’s Republic of China
- Guangxi Zhuang Autonomous Region Women and Children Care Hospital, Guangxi, China
| | - Jing Ma
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion Ministry of Education, Shanghai, People’s Republic of China
| | - Dongmei Gao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion Ministry of Education, Shanghai, People’s Republic of China
| | - Miaomiao Li
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion Ministry of Education, Shanghai, People’s Republic of China
| | - Huaping Chen
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University Nanning, Guangxi, China
| | - Kun Guo
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion Ministry of Education, Shanghai, People’s Republic of China
| |
Collapse
|
8
|
Zhang PH, Wu DB, Liu J, Wen JT, Chen ES, Xiao CH. Proteomics analysis of lung tissue reveals protein makers for the lung injury of adjuvant arthritis rats. Mol Med Rep 2023; 28:163. [PMID: 37449522 PMCID: PMC10407615 DOI: 10.3892/mmr.2023.13051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 06/15/2023] [Indexed: 07/18/2023] Open
Abstract
Lung injury is one of the common extra‑articular lesions in rheumatoid arthritis (RA). Due to its insidious onset and no obvious clinical symptoms, it can be easily dismissed in the early stage of diagnosis, which is one of the reasons that leads to a decline of the quality of life and subsequent death of patients with RA. However, its pathogenesis is still unclear and there is a lack of effective therapeutic targets. In the present study, tandem mass tag‑labeled proteomics was used to research the lung tissue proteins in RA model (adjuvant arthritis, AA) rats that had secondary lung injury. The aim of the present study was to identify the differentially expressed proteins related to RA‑lung injury, determine their potential role in the pathogenesis of RA‑lung injury and provide potential targets for clinical treatment. Lung tissue samples were collected from AA‑lung injury and normal rats. The differentially expressed proteins (DEPs) were identified by tandem mass spectrometry. Bioinformatic analysis was used to assess the biological processes and signaling pathways associated with these DEPs. A total of 310 DEPs were found, of which 244 were upregulated and 66 were downregulated. KEGG anlysis showed that 'fatty acid degradation', 'fatty acid metabolism', 'fatty acid elongation', 'complement and coagulation cascades', 'peroxisome proliferator‑activated receptor signaling pathway' and 'hypoxia‑inducible factor signaling pathway' were significantly upregulated in the lung tissues of AA‑lung injury. Immunofluorescence staining confirmed the increased expression of clusterin, serine protease inhibitors and complement 1qc in lung tissue of rats with AA lung injury. In the present study, the results revealed the significance of certain DEPs (for example, C9, C1qc and Clu) in the occurrence and development of RA‑lung injury and provided support through experiments to identify potential biomarkers for the early diagnosis and prevention of RA‑lung injury.
Collapse
Affiliation(s)
- Ping-Heng Zhang
- Rheumatology and Immunology Department, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510315, P.R. China
- Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Dan-Bin Wu
- Department of Traditional Chinese Medicine, School of Medicine, First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Jian Liu
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui 230038, P.R. China
| | - Jian-Ting Wen
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui 230038, P.R. China
| | - En-Sheng Chen
- Rheumatology and Immunology Department, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510315, P.R. China
| | - Chang-Hong Xiao
- Rheumatology and Immunology Department, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510315, P.R. China
| |
Collapse
|
9
|
Fox CR, Kedarinath K, Neal CJ, Sheiber J, Kolanthai E, Kumar U, Drake C, Seal S, Parks GD. Broad-Spectrum, Potent, and Durable Ceria Nanoparticles Inactivate RNA Virus Infectivity by Targeting Virion Surfaces and Disrupting Virus-Receptor Interactions. Molecules 2023; 28:5190. [PMID: 37446852 DOI: 10.3390/molecules28135190] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 06/29/2023] [Accepted: 07/01/2023] [Indexed: 07/15/2023] Open
Abstract
There is intense interest in developing long-lasting, potent, and broad-spectrum antiviral disinfectants. Ceria nanoparticles (CNPs) can undergo surface redox reactions (Ce3+ ↔ Ce4+) to generate ROS without requiring an external driving force. Here, we tested the mechanism behind our prior finding of potent inactivation of enveloped and non-enveloped RNA viruses by silver-modified CNPs, AgCNP1 and AgCNP2. Treatment of human respiratory viruses, coronavirus OC43 and parainfluenza virus type 5 (PIV5) with AgCNP1 and 2, respectively, prevented virus interactions with host cell receptors and resulted in virion aggregation. Rhinovirus 14 (RV14) mutants were selected to be resistant to inactivation by AgCNP2. Sequence analysis of the resistant virus genomes predicted two amino acid changes in surface-located residues D91V and F177L within capsid protein VP1. Consistent with the regenerative properties of CNPs, surface-applied AgCNP1 and 2 inactivated a wide range of structurally diverse viruses, including enveloped (OC43, SARS-CoV-2, and PIV5) and non-enveloped RNA viruses (RV14 and feline calicivirus; FCV). Remarkably, a single application of AgCNP1 and 2 potently inactivated up to four sequential rounds of virus challenge. Our results show broad-spectrum and long-lasting anti-viral activity of AgCNP nanoparticles, due to targeting of viral surface proteins to disrupt interactions with cellular receptors.
Collapse
Affiliation(s)
- Candace R Fox
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Kritika Kedarinath
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Craig J Neal
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32816, USA
| | - Jeremy Sheiber
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Elayaraja Kolanthai
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32816, USA
| | - Udit Kumar
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32816, USA
| | | | - Sudipta Seal
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32816, USA
- Nano Science Technology Center, University of Central Florida, Orlando, FL 32816, USA
- Biionix Cluster, College of Medicine, University of Central Florida, Orlando, FL 32816, USA
| | - Griffith D Parks
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| |
Collapse
|
10
|
Wilson MR, Satapathy S, Vendruscolo M. Extracellular protein homeostasis in neurodegenerative diseases. Nat Rev Neurol 2023; 19:235-245. [PMID: 36828943 DOI: 10.1038/s41582-023-00786-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/02/2023] [Indexed: 02/26/2023]
Abstract
The protein homeostasis (proteostasis) system encompasses the cellular processes that regulate protein synthesis, folding, concentration, trafficking and degradation. In the case of intracellular proteostasis, the identity and nature of these processes have been extensively studied and are relatively well known. By contrast, the mechanisms of extracellular proteostasis are yet to be fully elucidated, although evidence is accumulating that their age-related progressive impairment might contribute to neuronal death in neurodegenerative diseases. Constitutively secreted extracellular chaperones are emerging as key players in processes that operate to protect neurons and other brain cells by neutralizing the toxicity of extracellular protein aggregates and promoting their safe clearance and disposal. Growing evidence indicates that these extracellular chaperones exert multiple effects to promote cell viability and protect neurons against pathologies arising from the misfolding and aggregation of proteins in the synaptic space and interstitial fluid. In this Review, we outline the current knowledge of the mechanisms of extracellular proteostasis linked to neurodegenerative diseases, and we examine the latest understanding of key molecules and processes that protect the brain from the pathological consequences of extracellular protein aggregation and proteotoxicity. Finally, we contemplate possible therapeutic opportunities for neurodegenerative diseases on the basis of this emerging knowledge.
Collapse
Affiliation(s)
- Mark R Wilson
- School of Chemistry and Molecular Bioscience, Molecular Horizons Research Institute, University of Wollongong, Wollongong, New South Wales, Australia.
| | - Sandeep Satapathy
- Blavatnik Institute of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| |
Collapse
|
11
|
Pagani L, Chinello C, Risca G, Capitoli G, Criscuolo L, Lombardi A, Ungaro R, Mangioni D, Piga I, Muscatello A, Blasi F, Favalli A, Martinovic M, Gori A, Bandera A, Grifantini R, Magni F. Plasma Proteomic Variables Related to COVID-19 Severity: An Untargeted nLC-MS/MS Investigation. Int J Mol Sci 2023; 24:ijms24043570. [PMID: 36834989 PMCID: PMC9962231 DOI: 10.3390/ijms24043570] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/26/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) infection leads to a wide range of clinical manifestations and determines the need for personalized and precision medicine. To better understand the biological determinants of this heterogeneity, we explored the plasma proteome of 43 COVID-19 patients with different outcomes by an untargeted liquid chromatography-mass spectrometry approach. The comparison between asymptomatic or pauci-symptomatic subjects (MILDs), and hospitalised patients in need of oxygen support therapy (SEVEREs) highlighted 29 proteins emerged as differentially expressed: 12 overexpressed in MILDs and 17 in SEVEREs. Moreover, a supervised analysis based on a decision-tree recognised three proteins (Fetuin-A, Ig lambda-2chain-C-region, Vitronectin) that are able to robustly discriminate between the two classes independently from the infection stage. In silico functional annotation of the 29 deregulated proteins pinpointed several functions possibly related to the severity; no pathway was associated exclusively to MILDs, while several only to SEVEREs, and some associated to both MILDs and SEVEREs; SARS-CoV-2 signalling pathway was significantly enriched by proteins up-expressed in SEVEREs (SAA1/2, CRP, HP, LRG1) and in MILDs (GSN, HRG). In conclusion, our analysis could provide key information for 'proteomically' defining possible upstream mechanisms and mediators triggering or limiting the domino effect of the immune-related response and characterizing severe exacerbations.
Collapse
Affiliation(s)
- Lisa Pagani
- Proteomics and Metabolomics Unit, School of Medicine and Surgery, University of Milano-Bicocca, 20854 Vedano al Lambro, Italy
| | - Clizia Chinello
- Proteomics and Metabolomics Unit, School of Medicine and Surgery, University of Milano-Bicocca, 20854 Vedano al Lambro, Italy
- Correspondence: ; Tel.:+39-333-5905725
| | - Giulia Risca
- Bicocca Bioinformatics Biostatistics and Bioimaging Centre—B4, School of Medicine and Surgery, University of Milano-Bicocca, 20854 Vedano al Lambro, Italy
| | - Giulia Capitoli
- Bicocca Bioinformatics Biostatistics and Bioimaging Centre—B4, School of Medicine and Surgery, University of Milano-Bicocca, 20854 Vedano al Lambro, Italy
| | - Lucrezia Criscuolo
- Proteomics and Metabolomics Unit, School of Medicine and Surgery, University of Milano-Bicocca, 20854 Vedano al Lambro, Italy
| | - Andrea Lombardi
- Department of Pathophysiology and Transplantation, University of Milano, 20122 Milano, Italy
- Infectious Diseases Unit, IRCCS Ca’ Granda Ospedale Maggiore Policlinico Foundation, 20122 Milano, Italy
| | - Riccardo Ungaro
- Infectious Diseases Unit, IRCCS Ca’ Granda Ospedale Maggiore Policlinico Foundation, 20122 Milano, Italy
| | - Davide Mangioni
- Department of Pathophysiology and Transplantation, University of Milano, 20122 Milano, Italy
- Infectious Diseases Unit, IRCCS Ca’ Granda Ospedale Maggiore Policlinico Foundation, 20122 Milano, Italy
| | - Isabella Piga
- Proteomics and Metabolomics Unit, School of Medicine and Surgery, University of Milano-Bicocca, 20854 Vedano al Lambro, Italy
| | - Antonio Muscatello
- Infectious Diseases Unit, IRCCS Ca’ Granda Ospedale Maggiore Policlinico Foundation, 20122 Milano, Italy
| | - Francesco Blasi
- Department of Pathophysiology and Transplantation, University of Milano, 20122 Milano, Italy
- Respiratory Unit and Cystic Fibrosis Adult Center, Internal Medicine Department, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy
| | - Andrea Favalli
- Istituto Nazionale di Genetica Molecolare (INGM), 20122 Milano, Italy
| | | | - Andrea Gori
- Department of Pathophysiology and Transplantation, University of Milano, 20122 Milano, Italy
- Infectious Diseases Unit, IRCCS Ca’ Granda Ospedale Maggiore Policlinico Foundation, 20122 Milano, Italy
| | - Alessandra Bandera
- Department of Pathophysiology and Transplantation, University of Milano, 20122 Milano, Italy
- Infectious Diseases Unit, IRCCS Ca’ Granda Ospedale Maggiore Policlinico Foundation, 20122 Milano, Italy
| | - Renata Grifantini
- Istituto Nazionale di Genetica Molecolare (INGM), 20122 Milano, Italy
| | - Fulvio Magni
- Proteomics and Metabolomics Unit, School of Medicine and Surgery, University of Milano-Bicocca, 20854 Vedano al Lambro, Italy
| |
Collapse
|
12
|
Varudkar N, Shiffer EM, Oyer JL, Copik A, Parks GD. Delivery of a novel membrane-anchored Fc chimera enhances NK cell-mediated killing of tumor cells and persistently virus-infected cells. PLoS One 2023; 18:e0285532. [PMID: 37146009 PMCID: PMC10162523 DOI: 10.1371/journal.pone.0285532] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 04/25/2023] [Indexed: 05/07/2023] Open
Abstract
Antibody-dependent cellular cytotoxicity (ADCC) is one of the most powerful mechanisms for Natural Killer (NK) cells to kill cancer cells or virus-infected cells. A novel chimeric protein (NA-Fc) was created, which when expressed in cells, positions an IgG Fc domain on the plasma membrane, mimicking the orientation of IgG bound to the cell surface. This NA-Fc chimera was tested with PM21-NK cells, produced through a previously developed particle-based method which yields superior NK cells for immunotherapeutic applications. Real time viability assays revealed higher PM21-NK killing of both ovarian and lung cancer cells expressing NA-Fc, which correlated with increased release of TNF-α and IFN-γ cytokines from NK cells and was dependent on CD16-Fc interactions. Lentivirus delivery of NA-Fc to target cells increased the rate of PM21-NK cell killing of A549 and H1299 lung, SKOV3 ovarian and A375 melanoma cancer cells. This NA-Fc-directed killing was extended to virus infected cells, where delivery of NA-Fc to lung cells that were persistently infected with Parainfluenza virus resulted in increased killing by PM21-NK cells. In contrast to its effect on PM21-NK cells, the NA-Fc molecule did not enhance complement mediated lysis of lung cancer cells. Our study lays the foundation for application of the novel NA-Fc chimera that could be delivered specifically to tumors during oncolytic virotherapy to mark target cells for ADCC by co-treatment with adoptive NK cells. This strategy would potentially eliminate the need to search for unique cancer specific antigens for development of new antibody therapeutics.
Collapse
Affiliation(s)
- Namita Varudkar
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States of America
| | - Elisabeth M Shiffer
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States of America
| | - Jeremiah L Oyer
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States of America
| | - Alicja Copik
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States of America
| | - Griffith D Parks
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States of America
| |
Collapse
|