1
|
Rydland A, Heinicke F, Nyman TA, Trøseid AMS, Flåm ST, Stensland M, Gehin J, Eikeland J, Øvstebø R, Mjaavatten MD, Lie BA. Newly-diagnosed rheumatoid arthritis patients have elevated levels of plasma extracellular vesicles with protein cargo altered towards inflammatory processes. Sci Rep 2025; 15:11632. [PMID: 40185859 PMCID: PMC11971360 DOI: 10.1038/s41598-025-96325-8] [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: 12/16/2024] [Accepted: 03/27/2025] [Indexed: 04/07/2025] Open
Abstract
Extracellular vesicles (EVs) are implicated in rheumatoid arthritis (RA) but have mainly been assessed in RA patients taking disease modifying anti-rheumatic drugs. EVs are nanoparticles important in cell-cell communication and their molecular cargo are biomarker candidates. We characterized the protein profiles of EVs from blood plasma from newly diagnosed, treatment naïve RA patients (N = 32) and compared them to healthy controls (N = 20), by size exclusion chromatography-based EV enrichment coupled with high-resolution quantitative proteomics. The RA patients had higher EV concentration and larger EVs than controls. A total of 682 EV proteins were reliably quantified, and the overall profiles were distinctly different between patients and controls. Specifically, 26 proteins were significantly upregulated and 31 downregulated in RA patients, with several proteins acting in inflammatory networks and with immunologically important upstream regulators. The RA associated EVs appear, based on the tissue expression of their cargo proteins, to originate mainly from hepatocytes or immune cells, like neutrophils. Interestingly, the strongest RA associated EV proteins were inflammatory molecules, like SAA1 and S100A9, already suggested as biomarkers in RA. Furthermore, the RA associated EV proteins were generally not correlated with total serum protein levels, stressing the importance of EV transport of inflammatory proteins in RA pathogenesis.
Collapse
Affiliation(s)
- Anne Rydland
- Department of Medical Genetics, University of Oslo and Oslo University Hospital, Postboks 4956 Nydalen, OUS HF Ullevål sykehus, 0424, Oslo, Norway.
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway.
| | - Fatima Heinicke
- Department of Medical Genetics, University of Oslo and Oslo University Hospital, Postboks 4956 Nydalen, OUS HF Ullevål sykehus, 0424, Oslo, Norway
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
- Department of Biostatistics, Institute of Basic Medical Sciences, Oslo Centre for Biostatistics and Epidemiology, University of Oslo, Oslo, Norway
| | - Tuula A Nyman
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Anne-Marie Siebke Trøseid
- The Blood Cell Research Group, Department of Medical Biochemistry, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Siri T Flåm
- Department of Medical Genetics, University of Oslo and Oslo University Hospital, Postboks 4956 Nydalen, OUS HF Ullevål sykehus, 0424, Oslo, Norway
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Maria Stensland
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Johanna Gehin
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Joakim Eikeland
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Reidun Øvstebø
- The Blood Cell Research Group, Department of Medical Biochemistry, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Maria Dahl Mjaavatten
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
- Division of Rheumatology, Diakonhjemmet Hospital, Oslo, Norway
| | - Benedicte A Lie
- Department of Medical Genetics, University of Oslo and Oslo University Hospital, Postboks 4956 Nydalen, OUS HF Ullevål sykehus, 0424, Oslo, Norway.
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway.
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
2
|
Trezza A, Roncaglia B, Visibelli A, Barletta R, Peruzzi L, Marzocchi B, Braconi D, Spiga O, Santucci A. Integrated Clinomics and Molecular Dynamics Simulation Approaches Reveal the SAA1.1 Allele as a Biomarker in Alkaptonuria Disease Severity. Biomolecules 2025; 15:194. [PMID: 40001497 PMCID: PMC11853296 DOI: 10.3390/biom15020194] [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: 12/05/2024] [Revised: 01/16/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Alkaptonuria (AKU) is a rare metabolic disorder characterized by the accumulation of homogentisic acid (HGA), leading to progressive ochronosis and joint degeneration. While much is known about HGA's role in tissue damage, the molecular mechanisms underlying acute inflammation in AKU remain poorly understood. Serum amyloid A (SAA) proteins are key mediators of the inflammatory response, yet their potential as biomarkers for inflammation in AKU has not been explored. This study investigated the role of the SAA1.1 allele as a biomarker for the severity of acute inflammation in AKU. Data from the ApreciseKUre Precision Medicine Ecosystem were analyzed to assess the relationship between SAA1 allelic variants and inflammatory markers. Molecular dynamics simulations compared the structural dynamics of SAA1.1 and SAA1.2 isoforms, with standard modeling and analysis pipelines employed. Using a clinomics approach, we showed that AKU patients expressing the SAA1.1 allele have significantly higher acute inflammation-related markers. Extensive molecular dynamics simulations revealed that the SAA1.1 isoform lent high structural instability of the C-terminal domain, accelerating the formation of amyloid fibrils and exacerbating the inflammatory condition. These findings would identify the SAA1.1 allele as a novel genetic biomarker for the progression of secondary amyloidosis in AKU and its severity. Furthermore, new molecular insights into the inflammatory mechanisms of AKU were provided, suggesting potential therapeutic approaches aimed at stabilizing SAA1.1 protein and preventing amyloid fibril formation, with significant implications in AKU and precision medicine strategies for SAA-related diseases.
Collapse
Affiliation(s)
- Alfonso Trezza
- ONE-HEALTH Laboratory, Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 53100 Siena, Italy; (A.T.); (B.R.); (A.V.); (R.B.); (L.P.); (B.M.); (D.B.); (O.S.)
| | - Bianca Roncaglia
- ONE-HEALTH Laboratory, Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 53100 Siena, Italy; (A.T.); (B.R.); (A.V.); (R.B.); (L.P.); (B.M.); (D.B.); (O.S.)
| | - Anna Visibelli
- ONE-HEALTH Laboratory, Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 53100 Siena, Italy; (A.T.); (B.R.); (A.V.); (R.B.); (L.P.); (B.M.); (D.B.); (O.S.)
| | - Roberta Barletta
- ONE-HEALTH Laboratory, Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 53100 Siena, Italy; (A.T.); (B.R.); (A.V.); (R.B.); (L.P.); (B.M.); (D.B.); (O.S.)
| | - Luana Peruzzi
- ONE-HEALTH Laboratory, Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 53100 Siena, Italy; (A.T.); (B.R.); (A.V.); (R.B.); (L.P.); (B.M.); (D.B.); (O.S.)
| | - Barbara Marzocchi
- ONE-HEALTH Laboratory, Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 53100 Siena, Italy; (A.T.); (B.R.); (A.V.); (R.B.); (L.P.); (B.M.); (D.B.); (O.S.)
| | - Daniela Braconi
- ONE-HEALTH Laboratory, Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 53100 Siena, Italy; (A.T.); (B.R.); (A.V.); (R.B.); (L.P.); (B.M.); (D.B.); (O.S.)
| | - Ottavia Spiga
- ONE-HEALTH Laboratory, Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 53100 Siena, Italy; (A.T.); (B.R.); (A.V.); (R.B.); (L.P.); (B.M.); (D.B.); (O.S.)
| | - Annalisa Santucci
- ONE-HEALTH Laboratory, Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 53100 Siena, Italy; (A.T.); (B.R.); (A.V.); (R.B.); (L.P.); (B.M.); (D.B.); (O.S.)
- MetabERN, Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 53100 Siena, Italy
| |
Collapse
|
3
|
Chang Y, Liu Y, Zou Y, Ye RD. Recent Advances in Studies of Serum Amyloid A: Implications in Inflammation, Immunity and Tumor Metastasis. Int J Mol Sci 2025; 26:987. [PMID: 39940756 PMCID: PMC11817213 DOI: 10.3390/ijms26030987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/19/2025] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
Research on serum amyloid A (SAA) has seen major advancement in recent years with combined approaches of structural analysis and genetically altered mice. Initially identified as an acute-phase reactant, SAA is now recognized as a major player in host defense, inflammation, lipid metabolism and tumor metastasis. SAA binding and the neutralization of LPS attenuate sepsis in mouse models. SAA also displays immunomodulatory functions in Th17 differentiation and macrophage polarization, contributing to a pro-metastatic tumor microenvironment. In spite of the progress, the regulatory mechanisms for these diverse functions of SAA remain unclear. This review provides a brief summary of recent advances in SAA research on immunity, inflammation, tumor microenvironment and in vivo models.
Collapse
Affiliation(s)
- Yixin Chang
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Yezhou Liu
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Yuanrui Zou
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Richard D. Ye
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
- The Chinese University of Hong Kong, Shenzhen Futian Biomedical Innovation R&D Center, Shenzhen 518000, China
| |
Collapse
|
4
|
Mascolo R, Bizzi E, Martelli M, Facoetti C, Colazzo G, Barone F, Brucato A. Old and New Biomarkers in Idiopathic Recurrent Acute Pericarditis (IRAP): Prognosis and Outcomes. Curr Cardiol Rep 2025; 27:17. [PMID: 39798014 PMCID: PMC11724782 DOI: 10.1007/s11886-024-02170-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/07/2024] [Indexed: 01/13/2025]
Abstract
PURPOSE OF REVIEW To outline the latest discoveries regarding the utility and reliability of serum biomarkers in idiopathic recurrent acute pericarditis (IRAP), considering recent findings on its pathogenesis. The study highlights the predictive role of these biomarkers in potential short- (cardiac tamponade, recurrences) and long-term complications (constrictive pericarditis, death). RECENT FINDINGS The pathogenesis of pericarditis has been better defined in recent years, focusing on the autoinflammatory pathway. New studies have demonstrated the pivotal role of the classical inflammatory biomarkers in distinguishing pericarditis phenotypes (high-grade vs. low-grade inflammation) and in defining outcomes of this condition. Pericarditis involves intense inflammatory activity, which causes elevation of different markers, such as C-reactive protein, erythrocyte sedimentation rate, neutrophils and platelets, serum amyloid A and D-Dimer. Conversely, lymphocytes are often reduced, as well as hemoglobin during the acute phase. Cardiac troponins T and I are elevated in up to 30% of cases. A Biomarker for CRP-negative cases is needed. Other markers have been proposed for diagnosis and prognosis in IRAP, such as anti-heart antibodies and anti-intercalated disk antibodies, but we need further studies to validate them.
Collapse
Affiliation(s)
- Ruggiero Mascolo
- Division of Internal Medicine, Fatebenefratelli Hospital, ASST Fatebenefratelli Sacco, University of Milan, Piazzale Principessa Clotilde, 3, Milan, 20121, Italy.
| | - Emanuele Bizzi
- Division of Internal Medicine, Fatebenefratelli Hospital, ASST Fatebenefratelli Sacco, University of Milan, Piazzale Principessa Clotilde, 3, Milan, 20121, Italy
| | - Martina Martelli
- Division of Internal Medicine, Fatebenefratelli Hospital, ASST Fatebenefratelli Sacco, University of Milan, Piazzale Principessa Clotilde, 3, Milan, 20121, Italy
| | - Chiara Facoetti
- Division of Internal Medicine, Fatebenefratelli Hospital, ASST Fatebenefratelli Sacco, University of Milan, Piazzale Principessa Clotilde, 3, Milan, 20121, Italy
| | - Giulia Colazzo
- Division of Internal Medicine, Fatebenefratelli Hospital, ASST Fatebenefratelli Sacco, University of Milan, Piazzale Principessa Clotilde, 3, Milan, 20121, Italy
| | - Fabio Barone
- Division of Internal Medicine, Fatebenefratelli Hospital, ASST Fatebenefratelli Sacco, University of Milan, Piazzale Principessa Clotilde, 3, Milan, 20121, Italy
| | - Antonio Brucato
- Division of Internal Medicine, Fatebenefratelli Hospital, ASST Fatebenefratelli Sacco, University of Milan, Piazzale Principessa Clotilde, 3, Milan, 20121, Italy
- Department of Biomedical and Clinical Sciences, University of Milan, Via G.B. Grassi, 74, Milan, 20157, Italy
| |
Collapse
|
5
|
Sahin D, Di Matteo A, Emery P. Biomarkers in the diagnosis, prognosis and management of rheumatoid arthritis: A comprehensive review. Ann Clin Biochem 2025; 62:3-21. [PMID: 39242085 PMCID: PMC11707974 DOI: 10.1177/00045632241285843] [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] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic, systemic, autoimmune condition that primarily affects the joints and periarticular soft tissues. In the past two decades, the discovery of new biomarkers has contributed to advances in the understanding of the pathogenesis and natural history of RA. These biomarkers, including genetic, clinical, serological and imaging biomarkers, play a key role in the different stages and aspects of RA, from the so called 'pre-clinical RA', which is characterized by subclinical pathological events, such as autoimmunity and inflammation, to diagnosis (including differential diagnosis), treatment decision making and disease monitoring.This review will provide an overview on the current role of traditional and newer biomarkers in the main aspects of RA management, from the identification of individuals 'at-risk' of RA who are likely to progress to clinically evident disease, to 'early' diagnosis of RA, prognosis, precision medicine, and prediction of response to treatment.
Collapse
Affiliation(s)
- Didem Sahin
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals, NHS Trust, Leeds, UK
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | - Andrea Di Matteo
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals, NHS Trust, Leeds, UK
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | - Paul Emery
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals, NHS Trust, Leeds, UK
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| |
Collapse
|
6
|
Mastroeni P, Geminiani M, Olmastroni T, Frusciante L, Trezza A, Visibelli A, Santucci A. An in vitro cell model for exploring inflammatory and amyloidogenic events in alkaptonuria. J Cell Physiol 2024; 239:e31449. [PMID: 39351877 DOI: 10.1002/jcp.31449] [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: 06/27/2024] [Revised: 09/04/2024] [Accepted: 09/19/2024] [Indexed: 12/18/2024]
Abstract
Alkaptonuria (AKU) is a progressive systemic inherited metabolic disorder primarily affecting the osteoarticular system, characterized by the degeneration of cartilage induced by ochronosis, ultimately leading to early osteoarthritis (OA). However, investigating AKU pathology in human chondrocytes, which is crucial for understanding the disease, encounters challenges due to limited availability and donor variability. To overcome this obstacle, an in vitro model has been established using homogentisic acid (HGA) to simulate AKU conditions. This model employed immortalized C20/A4 human chondrocytes and serves as a dependable platform for studying AKU pathogenesis. Significantly, the model demonstrates the accumulation of ochronotic pigment in HGA-treated cells, consistent with findings from previous studies. Furthermore, investigations into inflammatory processes during HGA exposure revealed notable oxidative stress, as indicated by elevated levels of reactive oxygen species and lipid peroxidation. Additionally, the model demonstrated HGA-induced inflammatory responses, evidenced by increased production of nitric oxide, overexpression of inducible nitric oxide synthase, and cyclooxygenase-2. These findings underscore the model's utility in studying inflammation associated with AKU. Moreover, analysis of serum amyloid A and serum amyloid P proteins revealed a potential interaction, corroborating evidence of amyloid fibril formation. This hypothesis was further supported by Congo red staining, which showed fibril formation exclusively in HGA-treated cells. Overall, the C20/A4 cell model provided valuable insights into AKU pathogenesis, emphasizing its potential for facilitating drug development and therapeutic interventions.
Collapse
Affiliation(s)
| | - Michela Geminiani
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, Siena, Italy
| | - Tommaso Olmastroni
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, Siena, Italy
| | - Luisa Frusciante
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, Siena, Italy
| | - Alfonso Trezza
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, Siena, Italy
| | - Anna Visibelli
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, Siena, Italy
| | - Annalisa Santucci
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, Siena, Italy
- MetabERN, Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, Siena, Italy
| |
Collapse
|
7
|
Nikitina M, Khramtsov P, Devyatov S, Valeev R, Eryomina M, Chukavin A, Rayev M. The development of a method to produce diagnostic reagents using LaNiO 3 nanospheres and their application in nanozyme-linked immunosorbent assay for the colorimetric screening of C-reactive protein with high sensitivity. Analyst 2024; 149:5657-5667. [PMID: 39474875 DOI: 10.1039/d4an01160k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
LaNiO3 perovskite nanoparticles, especially nanospheres (LNNS), show great promise in biomedical assays due to their peroxidase-like catalytic properties. However, LNNS-based diagnostic reagents have not been tested in nanozyme enzyme-linked immunosorbent assay (NLISA) or other enzyme-linked immunosorbent assays, and there is limited data on their synthesis. To fill this gap, it is necessary to develop a method for creating LNNS conjugates with monoclonal antibodies and to investigate the reproducibility, scalability, and applicability of these diagnostic reagents in NLISA. We have successfully developed a method for producing novel diagnostic reagents utilizing LaNiO3 nanospheres. Our research demonstrates the application of these nanospheres in a NLISA specifically designed for the detection of C-reactive protein (CRP) in real serum samples. This method is both reproducible and scalable, allowing for the efficient production of nanospheres that are functionalized with monoclonal antibodies targeting CRP, with a mean diameter of approximately 270 nm. Based on the promising results obtained from our experiments, we have developed and optimized a sandwich-format NLISA for CRP detection. This assay achieved a lower limit of detection at 0.178 μg L-1, with a dynamic range from 12.5 to 0.195 μg L-1 and a linear detection range extending from 0.195 to 6.25 μg L-1, showcasing its potential for clinical applications. The new NLISA method, utilizing LaNiO3 nanospheres in a sandwich format for the detection of CRP, significantly enhances sensitivity compared to similar use horseradish peroxidase-based ELISA. In this study for the first time, the functionalization of lanthanum nickelate nanospheres with recognition elements has been demonstrated. This advancement also sheds light on the technological challenges involved in synthesizing diagnostic reagents, identifying areas that need further exploration.
Collapse
Affiliation(s)
- Maria Nikitina
- Institute of Ecology and Genetics of Microorganisms, Urals Branch of RAS, Perm, Russia.
- Biology Faculty, Perm State University, Perm, Russia
| | - Pavel Khramtsov
- Institute of Ecology and Genetics of Microorganisms, Urals Branch of RAS, Perm, Russia.
- Biology Faculty, Perm State University, Perm, Russia
| | | | - Rishat Valeev
- Udmurt Federal Research Center, Ural Branch of RAS, Izhevsk, Russia
| | - Marina Eryomina
- Udmurt Federal Research Center, Ural Branch of RAS, Izhevsk, Russia
| | - Andrey Chukavin
- Udmurt Federal Research Center, Ural Branch of RAS, Izhevsk, Russia
| | - Mikhail Rayev
- Institute of Ecology and Genetics of Microorganisms, Urals Branch of RAS, Perm, Russia.
- Biology Faculty, Perm State University, Perm, Russia
- Udmurt Federal Research Center, Ural Branch of RAS, Izhevsk, Russia
| |
Collapse
|
8
|
Hoste L, Meertens B, Ogunjimi B, Sabato V, Guerti K, van der Hilst J, Bogie J, Joos R, Claes K, Debacker V, Janssen F, Tavernier SJ, Jacques P, Callens S, Dehoorne J, Haerynck F. Identification of a 5-Plex Cytokine Signature that Differentiates Patients with Multiple Systemic Inflammatory Diseases. Inflammation 2024:10.1007/s10753-024-02183-3. [PMID: 39528768 DOI: 10.1007/s10753-024-02183-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
Patients with non-infectious systemic inflammation may suffer from one of many diseases, including hyperinflammation (HI), autoinflammatory disorders (AID), and systemic autoimmune disease (AI). Despite their clinical overlap, the pathophysiology and patient management differ between these disorders. We aimed to investigate blood biomarkers able to discriminate between patient groups. We included 44 patients with active clinical and/or genetic systemic inflammatory disease (9 HI, 27 AID, 8 systemic AI) and 16 healthy controls. We quantified 55 serum proteins and combined multiple machine learning algorithms to identify five proteins (CCL26, CXCL10, ICAM-1, IL-27, and SAA) that maximally separated patient groups. High ICAM-1 was associated with HI. AID was characterized by an increase in SAA and decrease in CXCL10 levels. A trend for higher CXCL10 and statistically lower SAA was observed in patients with systemic AI. Principal component analysis and unsupervised hierarchical clustering confirmed separation of disease groups. Logistic regression modelling revealed a high statistical significance for HI (P = 0.001), AID, and systemic AI (P < 0.0001). Predictive accuracy was excellent for systemic AI (AUC 0.94) and AID (0.91) and good for HI (0.81). Further research is needed to validate findings in a larger prospective cohort. Results will contribute to a better understanding of the pathophysiology of systemic inflammatory disorders and can improve diagnosis and patient management.
Collapse
Affiliation(s)
- Levi Hoste
- Primary Immune Deficiency Research Laboratory, Department of Internal Diseases and Pediatrics, Centre for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Division of Pediatric Pulmonology, Infectious Diseases and Inborn Errors of Immunity ,Ghent University Hospital, European Reference Network for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases Network (ERN-RITA) Center, Ghent, Belgium
| | - Bram Meertens
- Primary Immune Deficiency Research Laboratory, Department of Internal Diseases and Pediatrics, Centre for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Division of Pediatric Pulmonology, Infectious Diseases and Inborn Errors of Immunity ,Ghent University Hospital, European Reference Network for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases Network (ERN-RITA) Center, Ghent, Belgium
| | - Benson Ogunjimi
- Rheumatology Department, Antwerp Hospital Network, Antwerp, Belgium
- Division of Pediatric Rheumatology, Antwerp University Hospital, Edegem, Belgium
- Antwerp Center for Pediatric Rheumatology and Autoinflammatory Diseases, Antwerp, Belgium
- Division of Pediatric Rheumatology, Brussels University Hospital, Jette, Belgium
- Antwerp Center for Translational Immunology and Virology (ACTIV), Vaccine and Infectious Disease Institute (VAXINFECTIO) ,Centre for Health Economics Research and Modeling Infectious Diseases (CHERMID), University of Antwerp, Antwerp, Belgium
| | - Vito Sabato
- Department of Immunology, Allergology, and Rheumatology, Antwerp University Hospital, Edegem, Belgium
| | - Khadija Guerti
- Department of Clinical Chemistry, Antwerp University Hospital, Edegem, Belgium
| | - Jeroen van der Hilst
- Department of Infectious Diseases and Immune Pathology, Jessa General Hospital, Hasselt, Belgium
- Limburg Clinical Research Center, Hasselt University, Hasselt, Belgium
| | - Jeroen Bogie
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Centre, Hasselt University, Hasselt, Belgium
| | - Rik Joos
- Division of Pediatric Rheumatology, Antwerp University Hospital, Edegem, Belgium
- Department of Immunology, Allergology, and Rheumatology, Antwerp University Hospital, Edegem, Belgium
- Department of Pediatric Rheumatology, Ghent University Hospital, European Reference Network for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases (ERN-RITA) Center, Ghent, Belgium
| | - Karlien Claes
- Primary Immune Deficiency Research Laboratory, Department of Internal Diseases and Pediatrics, Centre for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Division of Pediatric Pulmonology, Infectious Diseases and Inborn Errors of Immunity ,Ghent University Hospital, European Reference Network for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases Network (ERN-RITA) Center, Ghent, Belgium
| | - Veronique Debacker
- Primary Immune Deficiency Research Laboratory, Department of Internal Diseases and Pediatrics, Centre for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Fleur Janssen
- Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Simon J Tavernier
- Primary Immune Deficiency Research Laboratory, Department of Internal Diseases and Pediatrics, Centre for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Ghent, Belgium
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Unit of Molecular Signal Transduction in Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Peggy Jacques
- Department of Rheumatology, University Hospital Ghent, Ghent, Belgium
| | - Steven Callens
- Department of General Internal Medicine, Ghent University Hospital, Ghent, Belgium
| | - Joke Dehoorne
- Department of Pediatric Rheumatology, Ghent University Hospital, European Reference Network for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases (ERN-RITA) Center, Ghent, Belgium
| | - Filomeen Haerynck
- Primary Immune Deficiency Research Laboratory, Department of Internal Diseases and Pediatrics, Centre for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Ghent, Belgium.
- Department of Internal Medicine and Pediatrics, Division of Pediatric Pulmonology, Infectious Diseases and Inborn Errors of Immunity ,Ghent University Hospital, European Reference Network for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases Network (ERN-RITA) Center, Ghent, Belgium.
| |
Collapse
|
9
|
Ali AA, Darwish WS. Acute phase proteins patterns as biomarkers in bacterial infection: Recent insights. Open Vet J 2024; 14:2539-2550. [PMID: 39545194 PMCID: PMC11560262 DOI: 10.5455/ovj.2024.v14.i10.4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 09/07/2024] [Indexed: 11/17/2024] Open
Abstract
Escherichia coli is a bacterium with command and pathogenic variants. It has been implicated in the induction of several inflammatory conditions. Finding a biomarker for infection began many years ago. The challenge of using acute phase proteins (APPs) as biomarkers for infection is a promising target for many researchers in this field. Many APPs have been studied for their roles as biomarkers of E. coli infection. The following review aims to highlight recent trials that have approved the use of adiponectin, amyloid A, ceruloplasmin, C-reactive protein, Haptoglobin, and Pentraxin 3 as biomarkers for E. coli infection and assess the obtained results. In conclusion, despite the existing approaches for the use of APPs as biomarkers in E. coli infection, we recommend more precise studies to enable these markers to be more specific and applicable in clinical fields. APPs could be markers for systemic inflammatory conditions, regardless of the causative agent.
Collapse
Affiliation(s)
- Amer Al Ali
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha, Saudi Arabia
| | - Wageh Sobhy Darwish
- Department of Food Hygiene, Safety and Technology, Faculty of Veterinary Medicine, Zagazig University, Zagazig City, Egypt
| |
Collapse
|
10
|
Zhang S, Yang Y, Zhang L, Liu Y, Guo Z, Wu J, Zhou W, Hong Z, Zhang W. Identification and Validation of a Prognostic Signature Based on Fibroblast Immune-related Genes to Predict the Prognosis and Therapeutic Response of renal clear cell carcinoma by Integrated Analysis of Single-Cell and Bulk RNA Sequencing Data. J Cancer 2024; 15:5942-5955. [PMID: 39440053 PMCID: PMC11493018 DOI: 10.7150/jca.100194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 08/24/2024] [Indexed: 10/25/2024] Open
Abstract
Background: The importance of fibroblasts in cancer progression is becoming more acknowledged, particularly the significance of their immune-related genes. However, the precise roles these genes play in fibroblasts throughout tumor development remains unclear. Exploring how these genes function in advancing kidney renal clear cell carcinoma (KIRC) could provide answers to these uncertainties. Material and method: The Cancer Genome Atlas (TCGA) database served as the source of data for KIRC patients. We distinguished fibroblast immune-related genes (FIGs), which are used to construct risk score. Further analysis conducted including enrichment analysis, assessment of tumor mutation burden (TMB), evaluation of tumor microenvironment (TME), analysis of immune cell infiltration, and drug sensitivity prediction. Result: The risk score using 6 FIGs effectively predicts the outcomes for KIRC patients. Nomogram which is based on the risk score and clinical data, demonstrated superior predictive performance compared to the version without the risk score. Enrichment analysis identified that coagulation pathway predominates in high-risk group, the protein secretion pathway is prevalent in low-risk patients' cohort. The adverse prognosis in high-risk patient cohort could be linked to an elevated TMB. TME analysis showed that high-risk group's tumor tissues contain more immune and stromal cells. Furthermore, the amount of regulatory T cells increases with the risk score. Low-risk group response better to immunotherapy. Finally, RT-qPCR confirmed the differential expression of FIGs in KIRC patients. Conclusion: This risk score and nomogram are valuable tools assessing KIRC patients' prognosis. Poorer prognosis in high-risk categories may have relationship with activation of coagulation pathway and a higher TMB.
Collapse
Affiliation(s)
- Shuwen Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China, 330006
- Queen Mary School, Jiangxi medical college, Nanchang University, Nanchang, China, 330006
| | - Yuqian Yang
- Queen Mary School, Jiangxi medical college, Nanchang University, Nanchang, China, 330006
- Department of Urology Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China, 330006
| | - Liyi Zhang
- Queen Mary School, Jiangxi medical college, Nanchang University, Nanchang, China, 330006
- Department of Urology Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China, 330006
| | - Yijiang Liu
- Department of Urology Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China, 330006
- Jiangxi medical college, Nanchang University, Nanchang, China, 330006
| | - Zihun Guo
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China, 330006
| | - Jiajun Wu
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China, 330006
| | - Weijun Zhou
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China, 330006
| | - Zhengdong Hong
- Department of Urology Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China, 330006
| | - Wenxiong Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China, 330006
| |
Collapse
|
11
|
Atosuo J, Karhuvaara O, Suominen E, Virtanen J, Vilén L, Nuutila J. The role of gamma globulin, complement component 1q, factor B, properdin, body temperature, C-reactive protein and serum amyloid alpha to the activity and the function of the human complement system and its pathways. J Immunol Methods 2024; 531:113709. [PMID: 38862098 DOI: 10.1016/j.jim.2024.113709] [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: 02/07/2024] [Revised: 05/20/2024] [Accepted: 06/08/2024] [Indexed: 06/13/2024]
Abstract
The complement system plays a crucial role in orchestrating the activation and regulation of inflammation within the human immune system. Three distinct activation pathways-classical, lectin, and alternative-converge to form the common lytic pathway, culminating in the formation of the membrane-attacking complex that disrupts the structure of pathogens. Dysregulated complement system activity can lead to tissue damage, autoimmune diseases, or immune deficiencies. In this study, the antimicrobial activity of human serum was investigated by using a bioluminescent microbe probe, Escherichia coli (pEGFPluxABCDEamp). This probe has previously been used to determine the antimicrobial activity of complement system and the polymorphonuclear neutrophils. In this study, blocking antibodies against key serum activators and components, including IgG, complement component 1q, factor B, and properdin, were utilized. The influence of body temperature and acute phase proteins, such as C reactive protein (CRP) and serum amyloid alpha (SAA), on the complement system was also examined. The study reveals the critical factors influencing complement system activity and pathway function. Alongside crucial factors like C1q and IgG, alternative pathway components factor B and properdin played pivotal roles. Results indicated that the alternative pathway accounted for approximately one third of the overall serum antimicrobial activity, and blocking this pathway disrupted the entire complement system. Contrary to expectations, elevated body temperature during inflammation did not enhance the antimicrobial activity of human serum. CRP demonstrated complement activation properties, but at higher physiological concentrations, it exhibited antagonistic tendencies, dampening the response. On the other hand, SAA enhanced the serum's activity. Overall, this study sheds a light on the critical factors affecting both complement system activity and pathway functionality, emphasizing the importance of a balanced immune response.
Collapse
Affiliation(s)
- Janne Atosuo
- Laboratory of Immunochemistry, Department of Life Technologies, Faculty of Technology, University of Turku, 20140, Finland.
| | - Outi Karhuvaara
- Laboratory of Immunochemistry, Department of Life Technologies, Faculty of Technology, University of Turku, 20140, Finland.
| | - Eetu Suominen
- Laboratory of Immunochemistry, Department of Life Technologies, Faculty of Technology, University of Turku, 20140, Finland.
| | - Julia Virtanen
- Laboratory of Immunochemistry, Department of Life Technologies, Faculty of Technology, University of Turku, 20140, Finland
| | - Liisa Vilén
- Department of Occupational Medicine, Clinical Department, Faculty of Medicine, 20140, University of Turku, Finland.
| | - Jari Nuutila
- Laboratory of Immunochemistry, Department of Life Technologies, Faculty of Technology, University of Turku, 20140, Finland.
| |
Collapse
|
12
|
Kudo A, Osedo H, Aisyah R, Yazawa N, Saliu TP, Miyata K, Kumrungsee T, Yanaka N. Serum Amyloid A3 Promoter-Luciferase Reporter Mice Are Useful for Early Drug-Induced Nephrotoxicity Detection. Int J Mol Sci 2024; 25:5124. [PMID: 38791162 PMCID: PMC11120996 DOI: 10.3390/ijms25105124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/27/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
Early detection of drug-induced kidney injury is essential for drug development. In this study, multiple low-dose aristolochic acid (AA) and cisplatin (Cis) injections increased renal mRNA levels of inflammation, fibrosis, and renal tubule injury markers. We applied a serum amyloid A3 (Saa3) promoter-driven luciferase reporter (Saa3 promoter-luc mice) to these two tubulointerstitial nephritis models and performed in vivo bioluminescence imaging to monitor early renal pathologies. The bioluminescent signals from renal tissues with AA or CIS injections were stronger than those from normal kidney tissues obtained from normal mice. To verify whether the visualized bioluminescence signal was specifically generated by the injured kidney, we performed in vivo bioluminescence analysis after opening the stomachs of Saa3 promoter-luc mice, and the Saa3-mediated bioluminescent signal was specifically detected in the injured kidney. This study showed that Saa3 promoter activity is a potent non-invasive indicator for the early detection of drug-induced nephrotoxicity.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Noriyuki Yanaka
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8528, Japan (T.P.S.)
| |
Collapse
|
13
|
Nady A, Reichheld SE, Sharpe S. Structural studies of a serum amyloid A octamer that is primed to scaffold lipid nanodiscs. Protein Sci 2024; 33:e4983. [PMID: 38659173 PMCID: PMC11043621 DOI: 10.1002/pro.4983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/26/2024]
Abstract
Serum amyloid A (SAA) is a highly conserved acute-phase protein that plays roles in activating multiple pro-inflammatory pathways during the acute inflammatory response and is commonly used as a biomarker of inflammation. It has been linked to beneficial roles in tissue repair through improved clearance of lipids and cholesterol from sites of damage. In patients with chronic inflammatory diseases, elevated levels of SAA may contribute to increased severity of the underlying condition. The majority of circulating SAA is bound to lipoproteins, primarily high-density lipoprotein (HDL). Interaction with HDL not only stabilizes SAA but also alters its functional properties, likely through altered accessibility of protein-protein interaction sites on SAA. While high-resolution structures for lipid-free, or apo-, forms of SAA have been reported, their relationship with the HDL-bound form of the protein, and with other possible mechanisms of SAA binding to lipids, has not been established. Here, we have used multiple biophysical techniques, including SAXS, TEM, SEC-MALS, native gel electrophoresis, glutaraldehyde crosslinking, and trypsin digestion to characterize the lipid-free and lipid-bound forms of SAA. The SAXS and TEM data show the presence of soluble octamers of SAA with structural similarity to the ring-like structures reported for lipid-free ApoA-I. These SAA octamers represent a previously uncharacterized structure for lipid-free SAA and are capable of scaffolding lipid nanodiscs with similar morphology to those formed by ApoA-I. The SAA-lipid nanodiscs contain four SAA molecules and have similar exterior dimensions as the lipid-free SAA octamer, suggesting that relatively few conformational rearrangements may be required to allow SAA interactions with lipid-containing particles such as HDL. This study suggests a new model for SAA-lipid interactions and provides new insight into how SAA might stabilize protein-lipid nanodiscs or even replace ApoA-I as a scaffold for HDL particles during inflammation.
Collapse
Affiliation(s)
- Asal Nady
- Molecular Medicine ProgramThe Hospital for Sick ChildrenTorontoCanada
- Department of BiochemistryUniversity of TorontoTorontoCanada
| | - Sean E. Reichheld
- Molecular Medicine ProgramThe Hospital for Sick ChildrenTorontoCanada
| | - Simon Sharpe
- Molecular Medicine ProgramThe Hospital for Sick ChildrenTorontoCanada
- Department of BiochemistryUniversity of TorontoTorontoCanada
| |
Collapse
|
14
|
Huang X, An X, Gao X, Wang N, Liu J, Zhang Y, Qi G, Zhang C. Serum amyloid A facilitates expansion of CD4 + T cell and CD19 + B cell subsets implicated in the severity of myasthenia gravis patients. J Neurochem 2024; 168:224-237. [PMID: 38214332 DOI: 10.1111/jnc.16047] [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: 11/05/2023] [Revised: 12/12/2023] [Accepted: 12/29/2023] [Indexed: 01/13/2024]
Abstract
Serum amyloid A (SAA) is a clinically useful inflammatory marker involved in the pathogenesis of autoimmune diseases. This study aimed to explore the SAA levels in a cohort of patients with myasthenia gravis (MG) in relation to disease-related clinical parameters and myasthenic crisis (MC) and elucidate the effects of SAA on immune response. A total of 82 MG patients including 50 new-onset MG patients and 32 MC patients were enrolled in this study. Baseline data and laboratory parameters of all enrolled MG patients were routinely recorded through electronic medical systems. SAA levels were measured by enzyme-linked immunosorbent assay (ELISA) kit. CD4+ T and CD19+ B cell subsets were analyzed by flow cytometry. In vitro, human recombinant SAA (Apo-SAA) was applied to stimulate peripheral blood mononuclear cells (PBMCs) from MG patients to observe the effect on T and B cell differentiation. Our results indicated that SAA levels in new-onset MG patients were higher than those in controls and were positively correlated with QMG score, MGFA classification, plasmablast cells, IL-6, and IL-17 levels. Subgroup analysis revealed that SAA levels were increased in generalized MG (GMG) patients than in ocular MG (OMG), as well as elevated in late-onset MG (LOMG) than in early-onset MG (EOMG) and higher in MGFA III/IV compared with MGFA I/II. The ROC curve demonstrated that SAA showed good diagnostic value for MC, especially when combined with NLR. In vitro, Apo-SAA promoted the Th1 cells, Th17 cells, plasmablast cells, and plasma cells differentiation in MG PBMCs. The present findings suggested that SAA was increased in MG patients and promoted expansion of CD4+ T cell and CD19+ B cell subsets, which implicated in the severity of MG patients.
Collapse
Affiliation(s)
- Xiaoyu Huang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Xueting An
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Xue Gao
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Ningning Wang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Jia Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yong Zhang
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Guoyan Qi
- Center of Treatment of Myasthenia Gravis Hebei Province, First Hospital of Shijiazhuang, Shijiazhuang, Hebei Province, China
| | - Chao Zhang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
15
|
Ozbek DA, Koc SC, Özkan NE, Kablan SE, Yet I, Uner M, Ozlu N, Nemutlu E, Lay I, Ayhan AS, Yildirim T, Arici M, Yilmaz SR, Erdem Y, Altun B. A comparative urinary proteomic and metabolomic analysis between renal aa amyloidosis and membranous nephropathy with clinicopathologic correlations. J Proteomics 2024; 293:105064. [PMID: 38154551 DOI: 10.1016/j.jprot.2023.105064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 11/20/2023] [Accepted: 12/18/2023] [Indexed: 12/30/2023]
Abstract
Urinary omics has become a powerful tool for elucidating pathophysiology of glomerular diseases. However, no urinary omics analysis has been performed yet on renal AA amyloidosis. Here, we performed a comparative urine proteomic and metabolomic analysis between recently diagnosed renal AA amyloidosis (AA) and membranous nephropathy (MN) patients. Urine samples of 22 (8 AA, 8 MN and 6 healthy control) patients were analyzed with nLC-MS/MS and GC/MS for proteomic and metabolomic studies, respectively. Pathological specimens were scored for glomerulosclerosis and tubulointerstitial fibrosis grades. Functional enrichment analysis between AA and control groups showed enrichment in cell adhesion related sub-domains. Uromodulin (UMOD) was lower, whereas ribonuclease 1 (RNase1) and α-1-microglobulin/bikunin precursor (AMBP) were higher in AA compared to MN group. Correlations were demonstrated between UMOD-proteinuria (r = -0.48, p = 0.03) and AMBP-eGFR (r = -0.69, p = 0.003) variables. Metabolomic analysis showed myo-inositol and urate were higher in AA compared to MN group. A positive correlation was detected between RNase1 and urate independent of eGFR values (r = 0.63, p = 0.01). Enrichment in cell adhesion related domains suggested a possible increased urinary shear stress due to amyloid fibrils. UMOD, AMBP and myo-inositol were related with tubulointerstitial damage, whereas RNase1 and urate were believed to be related with systemic inflammation in AA amyloidosis. SIGNIFICANCE: Urinary omics studies have become a standard tool for biomarker studies. However, no urinary omics analysis has been performed yet on renal AA amyloidosis. Here, we performed a comparative urinary omics analysis between recently diagnosed renal AA amyloidosis (AA), membranous nephropathy (MN) patients and healthy controls. Pathological specimens were scored with glomerulosclerosis (G) and tubulointerstitial fibrosis (IF) grades to consolidate the results of the omics studies and correlation analyzes. Functional enrichment analysis showed enrichment in cell adhesion related sub-domains due to downregulation of cadherins; which could be related with increased urinary shear stress due to amyloid deposition and disruption of tissue micro-architecture. In comparative proteomic analyzes UMOD was lower, whereas RNase1 and AMBP were higher in AA compared to MN group. Whereas in metabolomic analyzes; myo-inositol, urate and maltose were higher in AA compared to MN group. Correlations were demonstrated between UMOD-proteinuria (r = -0.48, p = 0.03), AMBP-eGFR (r = -0.69, p = 0.003) and between RNase1-Urate independent of eGFR values (r = 0.63, p = 0.01). This study is the first comprehensive urinary omics analysis focusing on renal AA Amyloidosis to the best of our knowledge. Based on physiologic roles and clinicopathologic correlations of the molecules; UMOD, AMBP and myo-inositol were related with tubulointerstitial damage, whereas RNase1 and urate were believed to be increased with systemic inflammation and endothelial damage in AA amyloidosis.
Collapse
Affiliation(s)
- Deniz Aral Ozbek
- Hacettepe University Faculty of Medicine, Department of Internal Medicine, Ankara, Turkey.
| | - Sila Cankurtaran Koc
- Hacettepe University Faculty of Medicine, Department of Nephrology, Ankara, Turkey
| | - Nazlı Ezgi Özkan
- Koc University Research Center for Translational Medicine, Istanbul, Turkey
| | - Sevilay Erdogan Kablan
- Hacettepe University Faculty of Pharmacy, Department of Analytical Chemistry, Ankara, Turkey
| | - Idil Yet
- Hacettepe University Graduate School of Health Sciences, Department of Bioinformatics, Ankara, Turkey
| | - Meral Uner
- Hacettepe University Faculty of Medicine, Department of Pathology, Ankara, Turkey
| | - Nurhan Ozlu
- Koc University Research Center for Translational Medicine, Istanbul, Turkey
| | - Emirhan Nemutlu
- Hacettepe University Faculty of Pharmacy, Department of Analytical Chemistry, Ankara, Turkey
| | - Incilay Lay
- Hacettepe University Faculty of Medicine, Department of Biochemistry, Ankara, Turkey
| | - Arzu Saglam Ayhan
- Hacettepe University Faculty of Medicine, Department of Pathology, Ankara, Turkey
| | - Tolga Yildirim
- Hacettepe University Faculty of Medicine, Department of Nephrology, Ankara, Turkey
| | - Mustafa Arici
- Hacettepe University Faculty of Medicine, Department of Nephrology, Ankara, Turkey
| | - Seref Rahmi Yilmaz
- Hacettepe University Faculty of Medicine, Department of Nephrology, Ankara, Turkey
| | - Yunus Erdem
- Hacettepe University Faculty of Medicine, Department of Nephrology, Ankara, Turkey
| | - Bulent Altun
- Hacettepe University Faculty of Medicine, Department of Nephrology, Ankara, Turkey
| |
Collapse
|
16
|
Vogli M, Peters A, Wolf K, Thorand B, Herder C, Koenig W, Cyrys J, Maestri E, Marmiroli N, Karrasch S, Zhang S, Pickford R. Long-term exposure to ambient air pollution and inflammatory response in the KORA study. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 912:169416. [PMID: 38123091 DOI: 10.1016/j.scitotenv.2023.169416] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/27/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023]
Abstract
Previous studies consistently showed an association between fine atmospheric particulate matter (PM2.5) and cardiovascular diseases. Concerns about adverse health effects of ultrafine particles (UFP) are growing but long-term studies are still scarce. In this study, we examined the association between long-term exposure to ambient air pollutants and blood biomarkers of inflammation and coagulation, including fibrinogen, high-sensitivity C-reactive protein (hs-CRP), serum amyloid A (SAA) adiponectin and interleukin-6 (IL-6), measured in the German KORA-S4 cohort study (1999-2001). IL-6 was available for older participants only, who were therefore considered as a subsample. Annual mean concentrations of UFP (as particle number concentration), particulate matter in different particles sizes (PM10, PMcoarse, PM2.5, PM2.5 absorbance), ozone (O3), and nitrogen oxides (NO2, NOX) were estimated by land-use regression models and assigned to participants' home addresses. We performed a multiple linear regression between each pollutant and each biomarker with adjustment for confounders. Per 1 interquartile range (IQR, 1945 particles/cm3) increase of UFP, fibrinogen increased by 0.70 % (0.04; 1.37) and hs-CRP increased by 3.16 % (-0.52; 6.98). Adiponectin decreased by -2.53 % (-4.78; -0.24) per 1 IQR (1.4 μg/m3) increase of PM2.5. Besides, PM2.5 was associated with increased IL-6 in the subsample. In conclusion, we observed that long-term exposure to air pollutants, including both fine and ultrafine particles, was associated with higher concentrations of pro-inflammatory and lower concentrations of an anti-inflammatory blood biomarkers, which is consistent with an increased risk for cardiovascular disease observed for long-term exposure to air pollutants.
Collapse
Affiliation(s)
- Megi Vogli
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute for Medical Information Processing, Biometry and Epidemiology, Medical Faculty, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; Munich Heart Alliance, German Center for Cardiovascular Health (DZHK e.V., partner-site Munich), Munich, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Kathrin Wolf
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Barbara Thorand
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute for Medical Information Processing, Biometry and Epidemiology, Medical Faculty, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Christian Herder
- German Center for Diabetes Research (DZD), Neuherberg, Germany; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Wolfgang Koenig
- German Research Center for Cardiovascular Disease, Partner Site of Munich Heart Alliance, Munich, Germany; Deutsches Herzzentrum München, Technische Universität München, Munich, Germany; Institute of Epidemiology and Medical Biometry, University of Ulm, Ulm, Germany
| | - Josef Cyrys
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Elena Maestri
- Department of Chemistry, Life Science and Environmental Sustainability, University of Parma, Parco Area delle Scienze, 43124 Parma, Italy; National Interuniversity Consortium for Environmental Sciences (CINSA), Parco Area delle Scienze, 43124 Parma, Italy
| | - Nelson Marmiroli
- Department of Chemistry, Life Science and Environmental Sustainability, University of Parma, Parco Area delle Scienze, 43124 Parma, Italy; National Interuniversity Consortium for Environmental Sciences (CINSA), Parco Area delle Scienze, 43124 Parma, Italy
| | - Stefan Karrasch
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute and Clinic for Occupational, Social and Environmental Medicine, University Hospital LMU Munich, Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Siqi Zhang
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Regina Pickford
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany.
| |
Collapse
|
17
|
Zeng L, Yu G, Yang K, He Q, Hao W, Xiang W, Long Z, Chen H, Tang X, Sun L. Exploring the mechanism of Celastrol in the treatment of rheumatoid arthritis based on systems pharmacology and multi-omics. Sci Rep 2024; 14:1604. [PMID: 38238321 PMCID: PMC10796403 DOI: 10.1038/s41598-023-48248-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 11/23/2023] [Indexed: 01/22/2024] Open
Abstract
To explore the molecular network mechanism of Celastrol in the treatment of rheumatoid arthritis (RA) based on a novel strategy (integrated systems pharmacology, proteomics, transcriptomics and single-cell transcriptomics). Firstly, the potential targets of Celastrol and RA genes were predicted through the database, and the Celastrol-RA targets were obtained by taking the intersection. Then, transcriptomic data and proteomic data of Celastrol treatment of RA were collected. Subsequently, Celastrol-RA targets, differentially expressed genes, and differentially expressed proteins were imported into Metascape for enrichment analysis, and related networks were constructed. Finally, the core targets of Celastrol-RA targets, differentially expressed genes, and differentially expressed proteins were mapped to synoviocytes of RA mice to find potential cell populations for Celastrol therapy. A total of 195 Celastrol-RA targets, 2068 differential genes, 294 differential proteins were obtained. The results of enrichment analysis showed that these targets, genes and proteins were mainly related to extracellular matrix organization, TGF-β signaling pathway, etc. The results of single cell sequencing showed that the main clusters of these targets, genes, and proteins could be mapped to RA synovial cells. For example, Mmp9 was mainly distributed in Hematopoietic cells, especially in Ptprn+fibroblast. The results of molecular docking also suggested that Celastrol could stably combine with molecules predicted by network pharmacology. In conclusion, this study used systems pharmacology, transcriptomics, proteomics, single-cell transcriptomics to reveal that Celastrol may regulate the PI3K/AKT signaling pathway by regulating key targets such as TNF and IL6, and then play an immune regulatory role.
Collapse
Affiliation(s)
- Liuting Zeng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China
| | - Ganpeng Yu
- People's Hospital of Ningxiang City, Ningxiang, China
| | - Kailin Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Qi He
- Department of Rehabilitation Medicine, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Wensa Hao
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wang Xiang
- Department of Rheumatology, The First People's Hospital Changde City, Changde, China
| | - Zhiyong Long
- Department of Rehabilitation Medicine, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Hua Chen
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Xiaojun Tang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China.
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China.
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Anhui, China.
| |
Collapse
|
18
|
Maller J, Morgan T, Morita M, McCarthy F, Jung Y, Svensson KJ, Elias JE, Macaubas C, Mellins E. Extracellular vesicles in systemic juvenile idiopathic arthritis. J Leukoc Biol 2023; 114:387-403. [PMID: 37201912 PMCID: PMC10602196 DOI: 10.1093/jleuko/qiad059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 04/28/2023] [Accepted: 05/11/2023] [Indexed: 05/20/2023] Open
Abstract
Systemic juvenile idiopathic arthritis is a chronic pediatric inflammatory disease of unknown etiology, characterized by fever, rash, hepatosplenomegaly, serositis, and arthritis. We hypothesized that intercellular communication, mediated by extracellular vesicles, contributes to systemic juvenile idiopathic arthritis pathogenesis and that the number and cellular sources of extracellular vesicles would differ between inactive and active states of systemic juvenile idiopathic arthritis and healthy controls. We evaluated plasma from healthy pediatric controls and patients with systemic juvenile idiopathic arthritis with active systemic flare or inactive disease. We isolated extracellular vesicles by size exclusion chromatography and determined total extracellular vesicle abundance and size distribution using microfluidic resistive pulse sensing. Cell-specific extracellular vesicle subpopulations were measured by nanoscale flow cytometry. Isolated extracellular vesicles were validated using a variety of ways, including nanotracking and cryo-electron microscopy. Extracellular vesicle protein content was analyzed in pooled samples using mass spectrometry. Total extracellular vesicle concentration did not significantly differ between controls and patients with systemic juvenile idiopathic arthritis. Extracellular vesicles with diameters <200 nm were the most abundant, including the majority of cell-specific extracellular vesicle subpopulations. Patients with systemic juvenile idiopathic arthritis had significantly higher levels of extracellular vesicles from activated platelets, intermediate monocytes, and chronically activated endothelial cells, with the latter significantly more elevated in active systemic juvenile idiopathic arthritis relative to inactive disease and controls. Protein analysis of isolated extracellular vesicles from active patients showed a proinflammatory profile, uniquely expressing heat shock protein 47, a stress-inducible protein. Our findings indicate that multiple cell types contribute to altered extracellular vesicle profiles in systemic juvenile idiopathic arthritis. The extracellular vesicle differences between systemic juvenile idiopathic arthritis disease states and healthy controls implicate extracellular vesicle-mediated cellular crosstalk as a potential driver of systemic juvenile idiopathic arthritis disease activity.
Collapse
Affiliation(s)
- Justine Maller
- Department of Pediatrics, Stanford University School of Medicine, 269 Campus Drive, CCSR Rm 2105c, Stanford, CA 94305, United States
| | - Terry Morgan
- Departments of Pathology and Biomedical Engineering, Oregon Health & Sciences University, 3181 SW Sam Jackson Portland, OR 97239, United States
| | - Mayu Morita
- Departments of Pathology and Biomedical Engineering, Oregon Health & Sciences University, 3181 SW Sam Jackson Portland, OR 97239, United States
| | - Frank McCarthy
- Chan Zuckerberg Biohub, 265 Campus Drive, Palo Alto, CA 94305, United States
| | - Yunshin Jung
- Department of Pathology, Stanford University School of Medicine, 300 Pasteur Dr, Edwards R238, Stanford, CA 94305, United States
| | - Katrin J Svensson
- Department of Pathology, Stanford University School of Medicine, 300 Pasteur Dr, Edwards R238, Stanford, CA 94305, United States
- Stanford Diabetes Research Center, Stanford, CA 94305, United States
- Stanford Cardiovascular Institute, Stanford, CA 94305, United States
| | - Joshua E Elias
- Chan Zuckerberg Biohub, 265 Campus Drive, Palo Alto, CA 94305, United States
| | - Claudia Macaubas
- Department of Pediatrics, Stanford University School of Medicine, 269 Campus Drive, CCSR Rm 2105c, Stanford, CA 94305, United States
- Department of Pediatrics, Program in Immunology, Stanford University School of Medicine, 269 Campus Drive, CCSR Rm 2105c, Stanford, CA 94305, United States
| | - Elizabeth Mellins
- Department of Pediatrics, Stanford University School of Medicine, 269 Campus Drive, CCSR Rm 2105c, Stanford, CA 94305, United States
- Department of Pediatrics, Program in Immunology, Stanford University School of Medicine, 269 Campus Drive, CCSR Rm 2105c, Stanford, CA 94305, United States
| |
Collapse
|
19
|
Abdullah M, Ali A, Usman M, Naz A, Qureshi JA, Bajaber MA, Zhang X. Post COVID-19 complications and follow up biomarkers. NANOSCALE ADVANCES 2023; 5:5705-5716. [PMID: 37881715 PMCID: PMC10597564 DOI: 10.1039/d3na00342f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 09/11/2023] [Indexed: 10/27/2023]
Abstract
Millions of people were infected by the coronavirus disease (COVID-19) epidemic, which left a huge burden on the care of post COVID-19 survivors around the globe. The self-reported COVID-19 symptoms were experienced by an estimated 1.3 million people in the United Kingdom (2% of the population), and these symptoms persisted for about 4 weeks from the beginning of the infection. The symptoms most frequently reported were exhaustion, shortness of breath, muscular discomfort, joint pain, headache, cough, chest pain, cognitive impairment, memory loss, anxiety, sleep difficulties, diarrhea, and a decreased sense of smell and taste in post-COVID-19 affected people. The post COVID-19 complications were frequently related to the respiratory, cardiac, nervous, psychological and musculoskeletal systems. The lungs, liver, kidneys, heart, brain and other organs had been impaired by hypoxia and inflammation in post COVID-19 individuals. The upregulation of substance "P" (SP) and various cytokines such as tumor necrosis factor-alpha (TNF-α), interleukin 6 (IL-6), interleukin 10 (IL-10), interleukin 1 beta (IL-1β), angiotensin-converting enzyme 2 (ACE2) and chemokine C-C motif ligand 3 (CCL3) has muddled respiratory, cardiac, neuropsychiatric, dermatological, endocrine, musculoskeletal, gastrointestinal, renal and genitourinary complications in post COVID-19 people. To prevent these complications from worsening, it was therefore important to study how these biomarkers were upregulated and block their receptors.
Collapse
Affiliation(s)
- Muhammad Abdullah
- Institute of Molecular Biology and Biotechnology, University of Lahore Pakistan
| | - Amjed Ali
- University Institute of Physical Therapy, University of Lahore Pakistan
| | - Muhammad Usman
- Department of Bioinformatics, School of Medical Informatics and Engineering, Xuzhou Medical University Xuzhou China
| | - Anam Naz
- Institute of Molecular Biology and Biotechnology, University of Lahore Pakistan
| | - Javed Anver Qureshi
- Institute of Molecular Biology and Biotechnology, University of Lahore Pakistan
| | - Majed A Bajaber
- Department of Chemistry, Faculty of Science, King Khalid University P.O. Box 9004 Abha 61413 Saudi Arabia
| | - Xiao Zhang
- Department of Bioinformatics, School of Medical Informatics and Engineering, Xuzhou Medical University Xuzhou China
| |
Collapse
|
20
|
Ejma-Multański A, Wajda A, Paradowska-Gorycka A. Cell Cultures as a Versatile Tool in the Research and Treatment of Autoimmune Connective Tissue Diseases. Cells 2023; 12:2489. [PMID: 37887333 PMCID: PMC10605903 DOI: 10.3390/cells12202489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 10/28/2023] Open
Abstract
Cell cultures are an important part of the research and treatment of autoimmune connective tissue diseases. By culturing the various cell types involved in ACTDs, researchers are able to broaden the knowledge about these diseases that, in the near future, may lead to finding cures. Fibroblast cultures and chondrocyte cultures allow scientists to study the behavior, physiology and intracellular interactions of these cells. This helps in understanding the underlying mechanisms of ACTDs, including inflammation, immune dysregulation and tissue damage. Through the analysis of gene expression patterns, surface proteins and cytokine profiles in peripheral blood mononuclear cell cultures and endothelial cell cultures researchers can identify potential biomarkers that can help in diagnosing, monitoring disease activity and predicting patient's response to treatment. Moreover, cell culturing of mesenchymal stem cells and skin modelling in ACTD research and treatment help to evaluate the effects of potential drugs or therapeutics on specific cell types relevant to the disease. Culturing cells in 3D allows us to assess safety, efficacy and the mechanisms of action, thereby aiding in the screening of potential drug candidates and the development of novel therapies. Nowadays, personalized medicine is increasingly mentioned as a future way of dealing with complex diseases such as ACTD. By culturing cells from individual patients and studying patient-specific cells, researchers can gain insights into the unique characteristics of the patient's disease, identify personalized treatment targets, and develop tailored therapeutic strategies for better outcomes. Cell culturing can help in the evaluation of the effects of these therapies on patient-specific cell populations, as well as in predicting overall treatment response. By analyzing changes in response or behavior of patient-derived cells to a treatment, researchers can assess the response effectiveness to specific therapies, thus enabling more informed treatment decisions. This literature review was created as a form of guidance for researchers and clinicians, and it was written with the use of the NCBI database.
Collapse
Affiliation(s)
- Adam Ejma-Multański
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland; (A.W.); (A.P.-G.)
| | | | | |
Collapse
|
21
|
Hu X, Xu J, Li P, Zheng H. Correlation of Serum Clara Cell Secretory Protein 16, Plasma Fibrinogen and Serum Amyloid A with the Severity of Acute Exacerbated COPD and Their Combination in Prognosis Assessment. Int J Chron Obstruct Pulmon Dis 2023; 18:1949-1957. [PMID: 37700931 PMCID: PMC10493104 DOI: 10.2147/copd.s410917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 06/25/2023] [Indexed: 09/14/2023] Open
Abstract
Introduction Chronic obstructive pulmonary disease (COPD) has tremendous detrimental effects on patients' quality of life, lung function, disease progression and socioeconomic burden. This study aimed to investigate new serum biomarkers for COPD detection. Three recently emerging biomarkers, including Clara cell secretory protein⁃16 (CC16), plasma fibrinogen (FIB) and serum amyloid A (SAA), were investigated for their potential in stratifying the severity of COPD. Methods A total of 220 patients with AECOPD were recruited. Multivariate logistical regression was used to analyze odds ratios of an array of characteristic of patients, including age, global initiative for chronic obstructive lung disease (GOLD), diabetes mellitus, heart diseases, PaCO2, CC16, FIB, and SAA. Correlations of CC16, FIB and SAA levels to each other, GOLD, and PaCO2 were also measured using Spearman correlation. Receiver operating characteristic (ROC)/curve analysis was used to assess sensitivity and specificity of CC16, FIB, SAA and the combination of the three markers in identifying AECOPD patients with poor prognosis. Results Our data suggested that age, GOLD, diabetes mellitus, heart diseases, PaCO2, CC16, FIB, and SAA are all significant risk factors for poor prognosis of AECOPD. CC16, FIB and SAA were positively correlated to each other and to GOLD and PaCO2 levels. CC16, FIB and SAA all had a high sensitivity and specificity in identifying patients with a poor prognosis. CC16, FIB and SAA are new markers with potentially high predictive value in AECOPD. Discussion Our data support further development of these biomarkers to improve clinical management of AECOPD through providing more accurate prognosis of AECOPD patients that enable timely adjustment of treatment plans.
Collapse
Affiliation(s)
- Xiaojuan Hu
- Department of Pulmonary and Critical Care Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, 200434, People’s Republic of China
| | - Jin Xu
- Department of Pulmonary and Critical Care Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, 200434, People’s Republic of China
| | - Pei Li
- Department of Nephrology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050004, People’s Republic of China
| | - Hui Zheng
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, 200032, People’s Republic of China
| |
Collapse
|
22
|
Ma W, Zhu J, Bai L, Zhao P, Li F, Zhang S. The role of neutrophil extracellular traps and proinflammatory damage-associated molecular patterns in idiopathic inflammatory myopathies. Clin Exp Immunol 2023; 213:202-208. [PMID: 37289984 PMCID: PMC10361739 DOI: 10.1093/cei/uxad059] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 04/13/2023] [Accepted: 05/25/2023] [Indexed: 06/10/2023] Open
Abstract
Idiopathic inflammatory myopathies (IIMs) are a group of systemic autoimmune diseases characterized by immune-mediated muscle injury. Abnormal neutrophil extracellular traps (NETs) can be used as a biomarker of IIM disease activity, but the mechanism of NET involvement in IIMs needs to be elucidated. Important components of NETs, including high-mobility group box 1, DNA, histones, extracellular matrix, serum amyloid A, and S100A8/A9, act as damage-associated molecular patterns (DAMPs) to promote inflammation in IIMs. NETs can act on different cells to release large amounts of cytokines and activate the inflammasome, which can subsequently aggravate the inflammatory response. Based on the idea that NETs may be proinflammatory DAMPs of IIMs, we describe the role of NETs, DAMPs, and their interaction in the pathogenesis of IIMs and discuss the possible targeted treatment strategies in IIMs.
Collapse
Affiliation(s)
- Wenlan Ma
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Jiarui Zhu
- Department of Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, China
| | - Ling Bai
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Peipei Zhao
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Feifei Li
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Sigong Zhang
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
23
|
Evren G, Er E, Yalcinkaya EE, Horzum N, Odaci D. Electrospun Nanofibers including Organic/Inorganic Nanohybrids: Polystyrene- and Clay-Based Architectures in Immunosensor Preparation for Serum Amyloid A. BIOSENSORS 2023; 13:673. [PMID: 37504072 PMCID: PMC10377714 DOI: 10.3390/bios13070673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/14/2023] [Accepted: 06/19/2023] [Indexed: 07/29/2023]
Abstract
Diagnostic techniques based on biomolecules have application potential that can be realized in many fields, such as disease diagnosis, bioprocess imaging, food/beverage industries, and environmental pollutant imaging. Successful surface immobilization of biomolecules is critical to increasing the stabilization, sensitivity, and selectivity of biomolecules used in bioassay systems. Nanofibers are good candidates for the immobilization of biomolecules owing to many advantages such as morphology and pore size. In this study, montmorillonite (MMT) clay is modified with poly(amidoamine) (PAMAM) generation 3 (PAMAMG3) and added to polystyrene (PS) solutions, following which PS/MMT-PAMAMG3 nanofibers are obtained using the electrospinning method. The nanofibers are obtained by testing PS% (wt%) and MMT-PAMAMG3% (wt%) ratios and characterized with scanning electron microscopy. Antiserum amyloid A antibody (Anti-SAA) is then conjugated to the nanofibers on the electrode surface via covalent bonds using a zero-length cross linker. Finally, the obtained selective surface is used for electrochemical determination of serum amyloid A (SAA) levels. The linear range of PS/MMT-PAMAM/Anti-SAA is between 1 and 200 ng/mL SAA, and the detection limit is 0.57 ng/mL SAA. The applicability of PS/MMT-PAMAMG3/Anti-SAA is investigated by taking measurements in synthetic saliva and serum both containing SAA.
Collapse
Affiliation(s)
- Gizem Evren
- Department of Biochemistry, Faculty of Science, Ege University, Bornova, Izmir 35100, Turkey
| | - Eray Er
- Department of Biochemistry, Faculty of Science, Ege University, Bornova, Izmir 35100, Turkey
| | - Esra Evrim Yalcinkaya
- Department of Chemistry, Faculty of Science, Ege University, Bornova, Izmir 35100, Turkey
| | - Nesrin Horzum
- Department of Engineering Sciences, Izmir Katip Celebi University, Cigli, Izmir 35620, Turkey
| | - Dilek Odaci
- Department of Biochemistry, Faculty of Science, Ege University, Bornova, Izmir 35100, Turkey
| |
Collapse
|
24
|
Popescu DM, Gheorghe DN, Turcu-Stiolica A, Soancă A, Roman A, Ionele CM, Ciucă EM, Boldeanu VM, Boldeanu L, Pitru A, Șurlin P. Evaluation of Pentraxin 3 and Serum Amyloid A in the Gingival Crevicular Fluid of Patients with Periodontal Disease and Obesity. J Clin Med 2023; 12:3523. [PMID: 37240630 PMCID: PMC10219408 DOI: 10.3390/jcm12103523] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 04/25/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND Pentraxin 3 (PTX3) is associated with periodontal tissue inflammation, a condition that precedes alveolar bone resorption. It is also elevated in obese tissues and is a useful biomarker of proinflammatory status. Serum amyloid A (SAA) is a proinflammatory and lipolytic adipokine. Adipocytes strongly express SAA, which suggests that it may have a significant role in the production of free fatty acids and local and systemic inflammation. MATERIALS AND METHODS We statistically analyzed the gingival crevicular fluid (GCF) values of PTX3 and SAA in patients with periodontal disease, who were diagnosed with obesity, and compared them with the values of inflammatory markers from patients diagnosed with one of the diseases and with healthy patients. RESULTS The patients with obesity and periodontitis had significantly higher levels of PTX3 and SAA than the patients diagnosed with either obesity or periodontitis. CONCLUSIONS These two markers are involved in the association between the two pathologies, as evidenced by the correlations between these levels and some clinical parameters.
Collapse
Affiliation(s)
- Dora Maria Popescu
- Department of Periodontology, Faculty of Dental Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (D.M.P.); (D.N.G.)
| | - Dorin Nicolae Gheorghe
- Department of Periodontology, Faculty of Dental Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (D.M.P.); (D.N.G.)
| | - Adina Turcu-Stiolica
- Department of Pharmacoeconomics and Statistical Analysis, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Andrada Soancă
- Department of Periodontology, Faculty of Dental Medicine, Iuliu Hațieganu University of Medicine and Pharmacy Cluj-Napoca, Victor Babeș St., No. 15, 400012 Cluj-Napoca, Romania
| | - Alexandra Roman
- Department of Periodontology, Faculty of Dental Medicine, Iuliu Hațieganu University of Medicine and Pharmacy Cluj-Napoca, Victor Babeș St., No. 15, 400012 Cluj-Napoca, Romania
| | - Claudiu Marinel Ionele
- Department of Gastroenterology and Hepatology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Eduard Mihai Ciucă
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Virgil Mihail Boldeanu
- Department of Immunology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Lidia Boldeanu
- Department of Microbiology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Allma Pitru
- Department of Oral Pathology, Faculty of Dental Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Petra Șurlin
- Department of Periodontology, Faculty of Dental Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (D.M.P.); (D.N.G.)
| |
Collapse
|
25
|
Chen R, Chen Q, Zheng J, Zeng Z, Chen M, Li L, Zhang S. Serum amyloid protein A in inflammatory bowel disease: from bench to bedside. Cell Death Discov 2023; 9:154. [PMID: 37164984 PMCID: PMC10172326 DOI: 10.1038/s41420-023-01455-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/18/2023] [Accepted: 04/27/2023] [Indexed: 05/12/2023] Open
Abstract
Inflammatory bowel diseases (IBD) is featured by gastrointestinal inflammation and a disease course with alternating recurrence and remission. The global burden caused by IBD has significantly boosted in recent years, necessitating treatment optimization. Serum amyloid A (SAA) is a class of 104 amino acid conservative acute-phase proteins, which is essential in immune-mediated inflammatory processes, like IBD. The SAA monomeric structure is composed of four α-helical regions and a C-terminal amorphous tail. Its disordered structure enables multiple bindings to different ligands and permits multiple functions. It has been proven that SAA has dual roles in the inflammatory process. SAA stimulates the pro-inflammatory cytokine expression and promotes the pathogenic differentiation of TH17 cells. In addition, SAA can remove toxic lipids produced during inflammatory responses and membrane debris from dead cells, redirect HDL, and recycle cholesterol for tissue repair. In IBD, SAA acts on gut epithelium barriers, induces T-cell differentiation, and promotes phagocytosis of Gram-negative bacteria. Owing to the tight connection between SAA and IBD, several clinical studies have taken SAA for a biomarker for diagnosis, assessing disease activity, and predicting prognosis in IBD. Furthermore, 5-MER peptide, a drug specifically targeting SAA, has shown anti-inflammatory effects in some SAA-dependent animal models, providing novel insights into the therapeutic targets of IBD.
Collapse
Affiliation(s)
- Rirong Chen
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qia Chen
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jieqi Zheng
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhirong Zeng
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Minhu Chen
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li Li
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Shenghong Zhang
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
26
|
Sulimai N, Brown J, Lominadze D. Vascular Effects on Cerebrovascular Permeability and Neurodegeneration. Biomolecules 2023; 13:648. [PMID: 37189395 PMCID: PMC10136045 DOI: 10.3390/biom13040648] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/29/2023] [Accepted: 04/02/2023] [Indexed: 05/17/2023] Open
Abstract
Neurons and glial cells in the brain are protected by the blood brain barrier (BBB). The local regulation of blood flow is determined by neurons and signal conducting cells called astrocytes. Although alterations in neurons and glial cells affect the function of neurons, the majority of effects are coming from other cells and organs of the body. Although it seems obvious that effects beginning in brain vasculature would play an important role in the development of various neuroinflammatory and neurodegenerative pathologies, significant interest has only been directed to the possible mechanisms involved in the development of vascular cognitive impairment and dementia (VCID) for the last decade. Presently, the National Institute of Neurological Disorders and Stroke applies considerable attention toward research related to VCID and vascular impairments during Alzheimer's disease. Thus, any changes in cerebral vessels, such as in blood flow, thrombogenesis, permeability, or others, which affect the proper vasculo-neuronal connection and interaction and result in neuronal degeneration that leads to memory decline should be considered as a subject of investigation under the VCID category. Out of several vascular effects that can trigger neurodegeneration, changes in cerebrovascular permeability seem to result in the most devastating effects. The present review emphasizes the importance of changes in the BBB and possible mechanisms primarily involving fibrinogen in the development and/or progression of neuroinflammatory and neurodegenerative diseases resulting in memory decline.
Collapse
Affiliation(s)
- Nurul Sulimai
- Department of Surgery, College of Medicine, University of South Florida Morsani, Tampa, FL 33612, USA; (N.S.); (J.B.)
| | - Jason Brown
- Department of Surgery, College of Medicine, University of South Florida Morsani, Tampa, FL 33612, USA; (N.S.); (J.B.)
| | - David Lominadze
- Department of Surgery, College of Medicine, University of South Florida Morsani, Tampa, FL 33612, USA; (N.S.); (J.B.)
- Department of Molecular Pharmacology and Physiology, College of Medicine, University of South Florida Morsani, Tampa, FL 33612, USA
| |
Collapse
|
27
|
Almusalami EM, Lockett A, Ferro A, Posner J. Serum amyloid A—A potential therapeutic target for hyper-inflammatory syndrome associated with COVID-19. Front Med (Lausanne) 2023; 10:1135695. [PMID: 37007776 PMCID: PMC10060655 DOI: 10.3389/fmed.2023.1135695] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 02/24/2023] [Indexed: 03/18/2023] Open
Abstract
Serum amyloid-A (SAA) is associated with inflammatory disorders such as rheumatoid arthritis, Familial Mediterranean Fever, sarcoidosis, and vasculitis. There is accumulating evidence that SAA is a reliable biomarker for these autoinflammatory and rheumatic diseases and may contribute to their pathophysiology. Hyperinflammatory syndrome associated with COVID-19 is a complex interaction between infection and autoimmunity and elevation of SAA is strongly correlated with severity of the inflammation. In this review we highlight the involvement of SAA in these different inflammatory conditions, consider its potential role and discuss whether it could be a potential target for treatment of the hyperinflammatory state of COVID-19 with many potential advantages and fewer adverse effects. Additional studies linking SAA to the pathophysiology of COVID-19 hyper-inflammation and autoimmunity are needed to establish the causal relationship and the therapeutic potential of inhibitors of SAA activity.
Collapse
Affiliation(s)
- Eman M. Almusalami
- Centre for Pharmaceutical Medicine Research, King’s College London, London, United Kingdom
- *Correspondence: Eman M. Almusalami,
| | - Anthony Lockett
- Centre for Pharmaceutical Medicine Research, King’s College London, London, United Kingdom
| | - Albert Ferro
- Centre for Pharmaceutical Medicine Research, King’s College London, London, United Kingdom
- School of Cardiovascular and Metabolic Medicine and Sciences, British Heart Foundation Centre for Research Excellence, King’s College London, London, United Kingdom
| | - John Posner
- Centre for Pharmaceutical Medicine Research, King’s College London, London, United Kingdom
| |
Collapse
|
28
|
Adherence to the Mediterranean Diet Association with Serum Inflammatory Factors Stress Oxidative and Appetite in COVID-19 Patients. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59020227. [PMID: 36837428 PMCID: PMC9968085 DOI: 10.3390/medicina59020227] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 01/18/2023] [Accepted: 01/22/2023] [Indexed: 01/27/2023]
Abstract
Background and Objectives: The Mediterranean diet's bioactive components are suggested to strengthen the immune system and to exert anti-inflammatory actions. This study investigated the association between adherence to the Mediterranean diet with serum inflammatory factors, total antioxidant capacity, appetite, and symptoms of COVID-19 patients. Materials and Methods: This cross-sectional study was conducted among 600 Iranian COVID-19 patients selected by a simple random method. The ten-item Mediterranean diet adherence questionnaire was used to assess diet adherence. At the beginning of the study, 5 cc of blood was taken from all patients for measurement of serum interleukin 1β) IL-1β), tumor necrosis factor (TNF-α), malondialdehyde (MDA), high sensitivity C-reactive protein (hs-CRP) and total antioxidant capacity (TAC). A human ELISA kit with serial number 950.090.096 produced by the Diaclone Company was used to test this cytokine using the sandwich ELISA method. Results: One hundred and five patients presented a high adherence and 495 patients presented a low adherence to the Mediterranean diet. The incidence of fever, cough, diarrhea, taste changes, and pneumonia severity index were significantly lower in patients who adhered to the Mediterranean diet more than other patients. Serum levels of tumor necrosis factor (5.7 ± 2.1 vs. 6.9 ± 2.8 p = 0.02), interleukin 1 beta (3.2 ± 0.02 vs. 4.9 ± 0.01 p = 0.02), high-sensitivity C-reactive protein (17.08 ± 4.2 vs. 19.8 ± 2.5 p = 0.03), and malondialdehyde (5.7 ± 0.2 vs. 6.2 ± 0.3 p = 0.02) were significantly lower in patients who adhered more to the Mediterranean diet than other patients. Conclusion: The Mediterranean diet can improve the symptoms and elevated serum inflammatory factors in COVID-19 patients, so clinical trial studies are suggested to confirm this effect.
Collapse
|
29
|
Sun Z, Li Y, Chang F, Jiang K. Utility of serum amyloid A as a potential prognostic biomarker of aneurysmal subarachnoid hemorrhage. Front Neurol 2023; 13:1099391. [PMID: 36712452 PMCID: PMC9878451 DOI: 10.3389/fneur.2022.1099391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 12/28/2022] [Indexed: 01/15/2023] Open
Abstract
Objectives Inflammation plays a vital role in the aneurysmal subarachnoid hemorrhage (aSAH), while serum amyloid A (SAA) has been identified as an inflammatory biomarker. The present study aimed to elucidate the relationship between SAA concentrations and prognosis in aSAH. Methods From prospective analyses of patients admitted to our department between March 2016 and August 2022, aSAH patients with complete medical records were evaluated. Meanwhile, the healthy control group consisted of the age and sex matched individuals who came to our hospital for healthy examination between March 2018 and August 2022. SAA level was measured by enzyme-linked immunosorbent assay kit (Invitrogen Corp). The Glasgow Outcome Scale (GOS) was used to classify patients into good (GOS score of 4 or 5) and poor (GOS score of 1, 2, or 3) outcome. Results 456 patients were enrolled in the study, thereinto, 200 (43.86%) patients had a poor prognosis at the 3-months follow-up. Indeed, the SAA of poor outcome group were significantly increased compared to good outcome group and healthy control group [36.44 (32.23-41.00) vs. 28.99 (14.67-34.12) and 5.64 (3.43-7.45), P < 0.001]. In multivariate analyses, SAA served for independently predicting the poor outcome after aICH at 3 months [OR:1.129 (95% CI, 1.081-1.177), P < 0.001]. After adjusting the underlying confounding factors, the odds ratio (OR) of depression after aSAH was 2.247 (95% CI: 1.095-4.604, P = 0.021) for the highest tertile of SAA relative to the lowest tertile. With an AUC of 0.807 (95% CI, 0.623-0.747), SAA demonstrated an obviously better discriminatory ability relative to CRP, WBC, and IL-6. SAA as an indicator for predicting poor outcome after aSAH had an optimal cut-off value of 30.28, and the sensitivity and specificity were 61.9 and 78.7%, respectively. Conclusions Elevated level of SAA was associated with poor outcome at 3 months, suggesting that SAA might be a useful inflammatory markers to predict prognosis after aSAH.
Collapse
Affiliation(s)
- Zhongbo Sun
- Department of Neurosurgery, First Affiliated Hospital of Anhui University of Science and Technology (First People's Hospital of Huainan), Huainan, China
| | - Yaqiang Li
- Department of Neurosurgery, First Affiliated Hospital of Anhui University of Science and Technology (First People's Hospital of Huainan), Huainan, China,Department of Neurology, People's Hospital of Lixin County, Bozhou, China,*Correspondence: Yaqiang Li ✉
| | - Fu Chang
- Department of Neurosurgery, First Affiliated Hospital of Anhui University of Science and Technology (First People's Hospital of Huainan), Huainan, China
| | - Ke Jiang
- Department of Neurosurgery, First Affiliated Hospital of Anhui University of Science and Technology (First People's Hospital of Huainan), Huainan, China
| |
Collapse
|
30
|
Meng S, Wang T, Zhao Q, Hu Q, Chen Y, Li H, Liu C, Liu D, Hong X. Proteomics Analysis of Plasma-Derived Exosomes Unveils the Aberrant Complement and Coagulation Cascades in Dermatomyositis/Polymyositis. J Proteome Res 2023; 22:123-137. [PMID: 36507906 PMCID: PMC9830643 DOI: 10.1021/acs.jproteome.2c00532] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Dermatomyositis and polymyositis (DM/PM) are systemic autoimmune diseases characterized by proximal muscle weakness. The underlying pathogenetic mechanism of this disease remains under-researched. Here, using proteomics analysis, a great overlap of differentially expressed plasma exosomal proteins involved in the complement and coagulation cascade pathway, including FGA, FGB, FGG, C1QB, C1QC, and VWF, was identified in DM/PM patients versus healthy controls. Correlation analysis showed that the expression levels of complement-associated proteins (C1QB and C1QC) correlated positively with CRP, ESR, and platelet count. ROC curve analysis demonstrated that complement and coagulation cascade-associated proteins could be strong predictors for DM/PM. In addition, we also identified several other proteins that were differentially expressed in DM and PM. The selected candidate proteins were further validated by parallel reaction monitoring (PRM) and enzyme-linked immunosorbent assay (ELISA). Together, our findings indicate that these exosome-derived proteins might participate in microvascular damage in DM/PM through the activation of the complement and coagulation cascade pathway and function as biomarkers for the clinical diagnosis of DM/PM.
Collapse
Affiliation(s)
- Shuhui Meng
- Department
of Rheumatology and Immunology, The Second Clinical Medical College
of Jinan University, The First Affiliated Hospital of Southern University
of Science and Technology, Shenzhen People’s
Hospital, Shenzhen 518020, Guangdong, China
| | - Tingting Wang
- Department
of Rheumatology and Immunology, The Second Clinical Medical College
of Jinan University, The First Affiliated Hospital of Southern University
of Science and Technology, Shenzhen People’s
Hospital, Shenzhen 518020, Guangdong, China,Integrated
Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou 510632, China
| | - Qianqian Zhao
- Department
of Rheumatology and Immunology, The Second Clinical Medical College
of Jinan University, The First Affiliated Hospital of Southern University
of Science and Technology, Shenzhen People’s
Hospital, Shenzhen 518020, Guangdong, China,Integrated
Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou 510632, China
| | - Qiu Hu
- Department
of Rheumatology and Immunology, The Second Clinical Medical College
of Jinan University, The First Affiliated Hospital of Southern University
of Science and Technology, Shenzhen People’s
Hospital, Shenzhen 518020, Guangdong, China
| | - Yulan Chen
- Department
of Rheumatology and Immunology, The Second Clinical Medical College
of Jinan University, The First Affiliated Hospital of Southern University
of Science and Technology, Shenzhen People’s
Hospital, Shenzhen 518020, Guangdong, China
| | - Heng Li
- Department
of Rheumatology and Immunology, The Second Clinical Medical College
of Jinan University, The First Affiliated Hospital of Southern University
of Science and Technology, Shenzhen People’s
Hospital, Shenzhen 518020, Guangdong, China,Integrated
Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou 510632, China
| | - Cuilian Liu
- Department
of Rheumatology and Immunology, The Second Clinical Medical College
of Jinan University, The First Affiliated Hospital of Southern University
of Science and Technology, Shenzhen People’s
Hospital, Shenzhen 518020, Guangdong, China
| | - Dongzhou Liu
- Department
of Rheumatology and Immunology, The Second Clinical Medical College
of Jinan University, The First Affiliated Hospital of Southern University
of Science and Technology, Shenzhen People’s
Hospital, Shenzhen 518020, Guangdong, China,
| | - Xiaoping Hong
- Department
of Rheumatology and Immunology, The Second Clinical Medical College
of Jinan University, The First Affiliated Hospital of Southern University
of Science and Technology, Shenzhen People’s
Hospital, Shenzhen 518020, Guangdong, China,
| |
Collapse
|
31
|
Gomes AC, Sousa DM, Oliveira TC, Fonseca Ó, Pinto RJ, Silvério D, Fernandes AI, Moreira AC, Silva T, Teles MJ, Pereira L, Saraiva M, Lamghari M, Gomes MS. Serum amyloid A proteins reduce bone mass during mycobacterial infections. Front Immunol 2023; 14:1168607. [PMID: 37153579 PMCID: PMC10161249 DOI: 10.3389/fimmu.2023.1168607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 04/11/2023] [Indexed: 05/09/2023] Open
Abstract
Introduction Osteopenia has been associated to several inflammatory conditions, including mycobacterial infections. How mycobacteria cause bone loss remains elusive, but direct bone infection may not be required. Methods Genetically engineered mice and morphometric, transcriptomic, and functional analyses were used. Additionally, inflammatory mediators and bone turnover markers were measured in the serum of healthy controls, individuals with latent tuberculosis and patients with active tuberculosis. Results and discussion We found that infection with Mycobacterium avium impacts bone turnover by decreasing bone formation and increasing bone resorption, in an IFNγ- and TNFα-dependent manner. IFNγ produced during infection enhanced macrophage TNFα secretion, which in turn increased the production of serum amyloid A (SAA) 3. Saa3 expression was upregulated in the bone of both M. avium- and M. tuberculosis-infected mice and SAA1 and 2 proteins (that share a high homology with murine SAA3 protein) were increased in the serum of patients with active tuberculosis. Furthermore, the increased SAA levels seen in active tuberculosis patients correlated with altered serum bone turnover markers. Additionally, human SAA proteins impaired bone matrix deposition and increased osteoclastogenesis in vitro. Overall, we report a novel crosstalk between the cytokine-SAA network operating in macrophages and bone homeostasis. These findings contribute to a better understanding of the mechanisms of bone loss during infection and open the way to pharmacological intervention. Additionally, our data and disclose SAA proteins as potential biomarkers of bone loss during infection by mycobacteria.
Collapse
Affiliation(s)
- Ana Cordeiro Gomes
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IMBC – Instituto de Biologia Molecular e Celular, Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
- *Correspondence: Ana Cordeiro Gomes,
| | - Daniela Monteiro Sousa
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB – Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | | | - Óscar Fonseca
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Mestrado em Bioquímica Clínica, Universidade de Aveiro, , Aveiro, Portugal
| | - Ricardo J. Pinto
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
- IPATIMUP – Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
| | - Diogo Silvério
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IMBC – Instituto de Biologia Molecular e Celular, Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Ana Isabel Fernandes
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IMBC – Instituto de Biologia Molecular e Celular, Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Ana C. Moreira
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IMBC – Instituto de Biologia Molecular e Celular, Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Tânia Silva
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Maria José Teles
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- CHUSJ – Centro Hospitalar de São João, Porto, Portugal
- EPIUnit, ISPUP - Instituto de Saúde Pública da Universidade do Porto, Porto, Portugal
| | - Luísa Pereira
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IPATIMUP – Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
| | - Margarida Saraiva
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IMBC – Instituto de Biologia Molecular e Celular, Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Meriem Lamghari
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB – Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Maria Salomé Gomes
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| |
Collapse
|
32
|
Karatemiz G, Esatoglu SN, Gurcan M, Ozguler Y, Yurdakul S, Hamuryudan V, Fresko I, Melikoglu M, Seyahi E, Ugurlu S, Ozdogan H, Yazici H, Hatemi G. Frequency of AA amyloidosis has decreased in Behçet's syndrome: a retrospective study with long-term follow-up and a systematic review. Rheumatology (Oxford) 2022; 62:9-18. [PMID: 35657376 DOI: 10.1093/rheumatology/keac223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 03/23/2022] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE A decline in the frequency of AA amyloidosis secondary to RA and infectious diseases has been reported. We aimed to determine the change in the frequency of AA amyloidosis in our Behçet's syndrome (BS) patients and to summarize the clinical characteristics of and outcomes for our patients, and also those identified by a systematic review. METHODS We identified patients with amyloidosis in our BS cohort (as well as their clinical and laboratory features, treatment, and outcome) through a chart review. The primary end points were end-stage renal disease and death. The prevalence of AA amyloidosis was estimated separately for patients registered during 1976-2000 and those registered during 2001-2017, in order to determine whether there was any change in the frequency. We searched PubMed and EMBASE for reports on BS patients with AA amyloidosis. Risk of bias was assessed using the Grading of Recommendations Assessment, Development and Evaluation (GRADE) tool. RESULTS The prevalence of AA amyloidosis was 0.62% (24/3820) in the earlier cohort and declined to 0.054% (3/5590) in the recent cohort. The systematic review revealed 82 cases in 42 publications. The main features of patients were male predominance and a high frequency of vascular involvement. One-third of patients died within 6 months after diagnosis of amyloidosis. CONCLUSION The frequency of AA amyloidosis has decreased in patients with BS, which is similar to the decrease observed for AA amyloidosis due to other inflammatory and infectious causes. However, AA amyloidosis is a rare, but potentially fatal complication of BS.
Collapse
Affiliation(s)
- Guzin Karatemiz
- Division of Rheumatology, Department of Internal Medicine, Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Sinem Nihal Esatoglu
- Division of Rheumatology, Department of Internal Medicine, Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Mert Gurcan
- Division of Rheumatology, Department of Internal Medicine, Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Yesim Ozguler
- Division of Rheumatology, Department of Internal Medicine, Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Sebahattin Yurdakul
- Division of Rheumatology, Department of Internal Medicine, Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Vedat Hamuryudan
- Division of Rheumatology, Department of Internal Medicine, Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Izzet Fresko
- Division of Rheumatology, Department of Internal Medicine, Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Melike Melikoglu
- Division of Rheumatology, Department of Internal Medicine, Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Emire Seyahi
- Division of Rheumatology, Department of Internal Medicine, Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Serdal Ugurlu
- Division of Rheumatology, Department of Internal Medicine, Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Huri Ozdogan
- Division of Rheumatology, Department of Internal Medicine, Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Hasan Yazici
- Division of Rheumatology, Department of Internal Medicine, Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Gulen Hatemi
- Division of Rheumatology, Department of Internal Medicine, Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul, Turkey
| |
Collapse
|
33
|
Proteomics profiles of blood glucose-related proteins involved in a Chinese longevity cohort. Clin Proteomics 2022; 19:45. [PMID: 36463101 PMCID: PMC9719669 DOI: 10.1186/s12014-022-09382-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 11/23/2022] [Indexed: 12/07/2022] Open
Abstract
BACKGROUND High blood glucose level is one of the main characteristics of diabetes mellitus. Based on previous studies, it is speculated longevity families may have certain advantages in blood glucose regulation. However, limited information on these items has been reported. The purpose of this study was to profile differences of plasma proteomics between longevity subjects (with normal fructosamine (FUN) level) and non-longevity area participants (with exceeding standard FUN level). METHODS In this study, a TMT-based proteomics analysis was used to profile differences of plasma proteomics between longevity subjects (with normal FUN level) and non-longevity area participants (with exceeding standard FUN level). Results were validated by Luminex detection. RESULTS A total of 155 differentially expressed proteins (DEPs) were identified between these two groups. The DEPs related to blood glucose regulation were mainly involved in glycolysis/gluconeogenesis, pyruvate metabolism and propanoate metabolism, and most of the DEPs were contained in carbohydrate metabolism, PI3K-Akt pathway, glucagon signaling pathway and inflammatory response. Validation by Luminex detection confirmed that CD163 was down-regulated, and SPARC, PARK 7 and IGFBP-1 were up-regulated in longevity participants. CONCLUSIONS This study not only highlighted carbohydrate metabolism, PI3K-Akt pathway, glucagon signaling pathway and inflammatory response may play important roles in blood glucose regulation, but also indicated that YWHAZ, YWHAB, YWHAG, YWHAE, CALM3, CRP, SAA2, PARK 7, IGFBP1 and VNN1 may serve as potential biomarkers for predicting abnormal blood glucose levels.
Collapse
|
34
|
Effects of Nitisinone on Oxidative and Inflammatory Markers in Alkaptonuria: Results from SONIA1 and SONIA2 Studies. Cells 2022; 11:cells11223668. [PMID: 36429096 PMCID: PMC9688277 DOI: 10.3390/cells11223668] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/11/2022] [Accepted: 11/16/2022] [Indexed: 11/22/2022] Open
Abstract
Nitisinone (NTBC) was recently approved to treat alkaptonuria (AKU), but there is no information on its impact on oxidative stress and inflammation, which are observed in AKU. Therefore, serum samples collected during the clinical studies SONIA1 (40 AKU patients) and SONIA2 (138 AKU patients) were tested for Serum Amyloid A (SAA), CRP and IL-8 by ELISA; Advanced Oxidation Protein Products (AOPP) by spectrophotometry; and protein carbonyls by Western blot. Our results show that NTBC had no significant effects on the tested markers except for a slight but statistically significant effect for NTBC, but not for the combination of time and NTBC, on SAA levels in SONIA2 patients. Notably, the majority of SONIA2 patients presented with SAA > 10 mg/L, and 30 patients in the control group (43.5%) and 40 patients (58.0%) in the NTBC-treated group showed persistently elevated SAA > 10 mg/L at each visit during SONIA2. Higher serum SAA correlated with lower quality of life and higher morbidity. Despite no quantitative differences in AOPP, the preliminary analysis of protein carbonyls highlighted patterns that deserve further investigation. Overall, our results suggest that NTBC cannot control the sub-clinical inflammation due to increased SAA observed in AKU, which is also a risk factor for developing secondary amyloidosis.
Collapse
|
35
|
Ailioaie LM, Ailioaie C, Litscher G. Biomarkers in Systemic Juvenile Idiopathic Arthritis, Macrophage Activation Syndrome and Their Importance in COVID Era. Int J Mol Sci 2022; 23:12757. [PMID: 36361547 PMCID: PMC9655921 DOI: 10.3390/ijms232112757] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 08/30/2023] Open
Abstract
Systemic juvenile idiopathic arthritis (sJIA) and its complication, macrophage activation syndrome (sJIA-MAS), are rare but sometimes very serious or even critical diseases of childhood that can occasionally be characterized by nonspecific clinical signs and symptoms at onset-such as non-remitting high fever, headache, rash, or arthralgia-and are biologically accompanied by an increase in acute-phase reactants. For a correct positive diagnosis, it is necessary to rule out bacterial or viral infections, neoplasia, and other immune-mediated inflammatory diseases. Delays in diagnosis will result in late initiation of targeted therapy. A set of biomarkers is useful to distinguish sJIA or sJIA-MAS from similar clinical entities, especially when arthritis is absent. Biomarkers should be accessible to many patients, with convenient production and acquisition prices for pediatric medical laboratories, as well as being easy to determine, having high sensitivity and specificity, and correlating with pathophysiological disease pathways. The aim of this review was to identify the newest and most powerful biomarkers and their synergistic interaction for easy and accurate recognition of sJIA and sJIA-MAS, so as to immediately guide clinicians in correct diagnosis and in predicting disease outcomes, the response to treatment, and the risk of relapses. Biomarkers constitute an exciting field of research, especially due to the heterogeneous nature of cytokine storm syndromes (CSSs) in the COVID era. They must be selected with utmost care-a fact supported by the increasingly improved genetic and pathophysiological comprehension of sJIA, but also of CSS-so that new classification systems may soon be developed to define homogeneous groups of patients, although each with a distinct disease.
Collapse
Affiliation(s)
- Laura Marinela Ailioaie
- Department of Medical Physics, Alexandru Ioan Cuza University, 11 Carol I Boulevard, 700506 Iasi, Romania
| | - Constantin Ailioaie
- Department of Medical Physics, Alexandru Ioan Cuza University, 11 Carol I Boulevard, 700506 Iasi, Romania
| | - Gerhard Litscher
- Research Unit of Biomedical Engineering in Anesthesia and Intensive Care Medicine, Research Unit for Complementary and Integrative Laser Medicine, Traditional Chinese Medicine (TCM) Research Center Graz, Department of Anesthesiology and Intensive Care Medicine, Medical University of Graz, Auenbruggerplatz 39, 8036 Graz, Austria
| |
Collapse
|
36
|
Fedotov SA, Khrabrova MS, Anpilova AO, Dobronravov VA, Rubel AA. Noninvasive Diagnostics of Renal Amyloidosis: Current State and Perspectives. Int J Mol Sci 2022; 23:ijms232012662. [PMID: 36293523 PMCID: PMC9604123 DOI: 10.3390/ijms232012662] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 10/17/2022] [Accepted: 10/18/2022] [Indexed: 11/05/2022] Open
Abstract
Amyloidoses is a group of diseases characterized by the accumulation of abnormal proteins (called amyloids) in different organs and tissues. For systemic amyloidoses, the disease is related to increased levels and/or abnormal synthesis of certain proteins in the organism due to pathological processes, e.g., monoclonal gammopathy and chronic inflammation in rheumatic arthritis. Treatment of amyloidoses is focused on reducing amyloidogenic protein production and inhibition of its aggregation. Therapeutic approaches critically depend on the type of amyloidosis, which underlines the importance of early differential diagnostics. In fact, the most accurate diagnostics of amyloidosis and its type requires analysis of a biopsy specimen from the disease-affected organ. However, absence of specific symptoms of amyloidosis and the invasive nature of biomaterial sampling causes the late diagnostics of these diseases, which leads to a delayed treatment, and significantly reduces its efficacy and patient survival. The establishment of noninvasive diagnostic methods and discovery of specific amyloidosis markers are essential for disease detection and identification of its type at earlier stages, which enables timely and targeted treatment. This review focuses on current approaches to the diagnostics of amyloidoses, primarily with renal involvement, and research perspectives in order to design new specific tests for early diagnosis.
Collapse
Affiliation(s)
- Sergei A. Fedotov
- Laboratory of Amyloid Biology, St. Petersburg State University, St. Petersburg 199034, Russia
- Pavlov Institute of Physiology, Russian Academy of Sciences, St. Petersburg 199034, Russia
| | - Maria S. Khrabrova
- Laboratory of Amyloid Biology, St. Petersburg State University, St. Petersburg 199034, Russia
- Research Institute of Nephrology, Pavlov University, St. Petersburg 197101, Russia
| | - Anastasia O. Anpilova
- Laboratory of Amyloid Biology, St. Petersburg State University, St. Petersburg 199034, Russia
- Research Institute of Nephrology, Pavlov University, St. Petersburg 197101, Russia
| | | | - Aleksandr A. Rubel
- Laboratory of Amyloid Biology, St. Petersburg State University, St. Petersburg 199034, Russia
- Department of Genetics and Biotechnology, St. Petersburg State University, St. Petersburg 199034, Russia
- Correspondence: ; Tel.: +7-812-428-40-09
| |
Collapse
|
37
|
Practical Significance of Biomarkers in Axial Spondyloarthritis: Updates on Diagnosis, Disease Activity, and Prognosis. Int J Mol Sci 2022; 23:ijms231911561. [PMID: 36232862 PMCID: PMC9570274 DOI: 10.3390/ijms231911561] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/20/2022] [Accepted: 09/22/2022] [Indexed: 11/26/2022] Open
Abstract
Axial spondyloarthritis (axSpA) is a chronic inflammatory disease that can lead to ankylosis by secondary ossification of inflammatory lesions, with progressive disability and a significant impact on quality of life. It is also a risk factor for the occurrence of comorbidities, especially cardiovascular diseases (CVDs), mood disorders, osteoporosis, and malignancies. Early diagnosis and treatment are needed to prevent or decrease functional decline and to improve the patient's prognosis. In respect of axSpA, there is an unmet need for biomarkers that can help to diagnose the disease, define disease activity and prognosis, and establish personalized treatment approaches. The aim of this review was to summarize the available information regarding the most promising biomarkers for axSpA. We classified and identified six core categories of biomarkers: (i) systemic markers of inflammation; (ii) molecules involved in bone homeostasis; (iii) HLA-B27 and newer genetic biomarkers; (iv) antibody-based biomarkers; (v) microbiome biomarkers; and (vi) miscellaneous biomarkers. Unfortunately, despite efforts to validate new biomarkers, few of them are used in clinical practice; however, we believe that these studies provide useful data that could aid in better disease management.
Collapse
|
38
|
Lv Z, Duan S, Zhou M, Gu M, Li S, Wang Y, Xia Q, Xu D, Mao Y, Dong W, Jiang L. Mouse Bone Marrow Mesenchymal Stem Cells Inhibit Sepsis-Induced Lung Injury in Mice via Exosomal SAA1. Mol Pharm 2022; 19:4254-4263. [PMID: 36173129 DOI: 10.1021/acs.molpharmaceut.2c00542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Sepsis is a global disease burden, and approximately 40% of cases develop acute lung injury (ALI). Bone marrow mesenchymal stromal cells (BMSCs) and their exosomes are widely used in treating a variety of diseases including sepsis. As an acute phase protein, serum amyloid A1 (SAA1) regulates inflammation and immunity. However, the role of SAA1 in BMSCs-exosomes in septic lung injury remains to be elucidated. Exosomes derived from serum and BMSCs were isolated by ultracentrifugation. SAA1 was silenced or overexpressed in mouse BMSCs using lentiviral plasmids, containing either SAA1-targeting short interfering RNAs or SAA1 cDNA. Sepsis was induced by cecal ligation and puncture (CLP). LPS was used to induce ALI in mice. Mouse alveolar macrophages were isolated by flow cytometry. Levels of SAA1, endotoxin, TNF-α, and IL-6 were measured using commercial kits. LPS internalization was monitored by immunostaining. RT-qPCR or immunoblots were performed to test gene and protein expressions. Serum exosomes of patients with sepsis-induced lung injury had significantly higher levels of SAA1, endotoxin, TNF-α, and IL-6. Overexpression of SAA1 in BMSCs inhibited CLP- or LPS-induced lung injury and decreased CLP- or LPS-induced endotoxin, TNF-α, and IL-6 levels. Administration of the SAA1 blocking peptide was found to partially inhibit SAA1-induced LPS internalization by mouse alveolar macrophages and reverse the protective effect of SAA1. In conclusion, BMSCs inhibit sepsis-induced lung injury through exosomal SAA1. These results highlight the importance of BMSCs, exosomes, and SAA1, which may provide novel directions for the treatment of septic lung injury.
Collapse
Affiliation(s)
- Zhou Lv
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No.1665, Kongjiang Road, Yangpu District, Shanghai 200092, China
| | - Shuxian Duan
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No.1665, Kongjiang Road, Yangpu District, Shanghai 200092, China
| | - Miao Zhou
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No.1665, Kongjiang Road, Yangpu District, Shanghai 200092, China
| | - Minglu Gu
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No.1665, Kongjiang Road, Yangpu District, Shanghai 200092, China
| | - Siyuan Li
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No.1665, Kongjiang Road, Yangpu District, Shanghai 200092, China
| | - Yan Wang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No.1665, Kongjiang Road, Yangpu District, Shanghai 200092, China
| | - Qin Xia
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No.1665, Kongjiang Road, Yangpu District, Shanghai 200092, China
| | - Dunfeng Xu
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No.1665, Kongjiang Road, Yangpu District, Shanghai 200092, China
| | - Yanfei Mao
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No.1665, Kongjiang Road, Yangpu District, Shanghai 200092, China
| | - Wenwen Dong
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No.1665, Kongjiang Road, Yangpu District, Shanghai 200092, China
| | - Lai Jiang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No.1665, Kongjiang Road, Yangpu District, Shanghai 200092, China
| |
Collapse
|
39
|
Thorne J, Clark D, Geldenhuys L, More K, Vinson A, Tennankore K. Serum Amyloid A Protein–Associated Kidney Disease: Presentation, Diagnosis, and Management. Kidney Med 2022; 4:100504. [PMID: 35879979 PMCID: PMC9307948 DOI: 10.1016/j.xkme.2022.100504] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Serum amyloid A protein (AA) amyloidosis, also known as secondary amyloidosis, is a known consequence of chronic inflammation and results from several conditions including inflammatory arthritis, periodic fever syndromes, and chronic infection. AA amyloidosis can lead to multiorgan dysfunction, including changes in glomerular filtration rate and proteinuria. Definitive diagnosis requires tissue biopsy, and management of AA amyloid kidney disease is primarily focused on treating the underlying inflammatory condition to stabilize glomerular filtration rate, reduce proteinuria, and slow potential progression to kidney failure. In this narrative review, we describe the causes, pathophysiology, presentation, and pathologic diagnosis of AA amyloid kidney disease using an illustrative case of biopsy-proven AA amyloid kidney disease in a patient with long-standing rheumatoid arthritis who had a favorable response to interleukin 6 inhibition. We conclude the review with a description of established and more novel therapies for AA amyloidosis including published cases of use of tocilizumab (an interleukin 6 inhibitor) in biopsy-proven AA amyloid kidney disease.
Collapse
Affiliation(s)
- Jordan Thorne
- Department of Medicine, Dalhousie University and Nova Scotia Health
- Address for Correspondence: Jordan Thorne, MD, Department of Medicine, Dalhousie University and Nova Scotia Health, 1276 South Park St, Halifax, NS B3H 2Y9, Canada.
| | - David Clark
- Department of Medicine, Dalhousie University and Nova Scotia Health
- Division of Nephrology, Nova Scotia Health
| | - Laurette Geldenhuys
- Division of Nephrology, Nova Scotia Health
- Department of Pathology, Dalhousie University and Nova Scotia Health
| | - Keigan More
- Department of Medicine, Dalhousie University and Nova Scotia Health
- Division of Nephrology, Nova Scotia Health
| | - Amanda Vinson
- Department of Medicine, Dalhousie University and Nova Scotia Health
- Division of Nephrology, Nova Scotia Health
| | - Karthik Tennankore
- Department of Medicine, Dalhousie University and Nova Scotia Health
- Division of Nephrology, Nova Scotia Health
| |
Collapse
|
40
|
Hong Y, Chu Z, Kong J, Li Q, Li N, Liu L, Wu T, Liu J, Ge D, Li J, Peng G. IL-17A aggravates asthma-induced intestinal immune injury by promoting neutrophil trafficking. J Leukoc Biol 2022; 112:425-435. [PMID: 35815539 DOI: 10.1002/jlb.3ma0622-426rr] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 06/10/2022] [Indexed: 11/06/2022] Open
Abstract
With the concept of the gut-lung axis reinforced in recent years, emerging evidence has shown that intestinal homeostasis is vital for lung health. Nevertheless, the impacts of lung homeostasis on intestinal tracts and their mechanism are rarely studied. Our results showed that papain-induced asthmatic mice exhibited apparent colonic injuries compared with controls, including increased intestinal permeability, neutrophil and Th17 infiltration in the colonic lamina propria. Moreover, the intranasal administration of papain aggravated such colonic injuries in mice with dextran sulfate sodium-induced colitis, as evidenced by increased occult blood scores, shortened colon length, and accumulated neutrophils. The level of IL-17A was also higher in the serum of asthmatic mice than wild-type mice. Interestingly, the pathologic scores, the proportion of Th17 cells, and neutrophil infiltration in the colon were markedly reduced after IL-17A blocking. Similarly, longer length, lower pathologic scores, and fewer neutrophils were also observed in the colon of IL-17-deficient asthmatic mice. More importantly, we demonstrated that severe gastrointestinal symptoms could accompany clinical asthmatics. The frequencies of Th17 cells and the mRNA expression of IL-17A in the peripheral blood of these patients were significantly enhanced. Besides, the gastrointestinal symptom rating scale scores positively correlated with the frequencies of Th17 in asthmatics. These findings enlighten that IL-17A aggravates asthma-induced intestinal immune injury by promoting neutrophil trafficking, which facilitates the exploration of new potential biomarkers to treat asthma.
Collapse
Affiliation(s)
- Yanfei Hong
- Department of Immunology and Microbiology, School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Zhulang Chu
- Experimental Teaching Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jingwei Kong
- Department of Immunology and Microbiology, School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Qiuyi Li
- Department of Immunology and Microbiology, School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Na Li
- Department of Immunology and Microbiology, School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Liting Liu
- Zengcheng Traditional Chinese Medicine Hospital, Guangzhou, China
| | - Tong Wu
- Department of Respiratory Medicine, Dong Zhi Men Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jiajing Liu
- Department of Immunology and Microbiology, School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Dongyu Ge
- Scientific Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jie Li
- Department of Respiratory Medicine, Dong Zhi Men Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Guiying Peng
- Department of Immunology and Microbiology, School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
41
|
Shan LL, Wang YL, Qiao TC, Bian YF, Huo YJ, Guo C, Liu QY, Yang ZD, Li ZZ, Liu MY, Han Y. Association of Serum Interleukin-8 and Serum Amyloid A With Anxiety Symptoms in Patients With Cerebral Small Vessel Disease. Front Neurol 2022; 13:938655. [PMID: 35923828 PMCID: PMC9341200 DOI: 10.3389/fneur.2022.938655] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 06/15/2022] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE Cerebral small vessel disease (CSVD) is a clinical syndrome caused by pathological changes in small vessels. Anxiety is a common symptom of CSVD. Previous studies have reported the association between inflammatory factors and anxiety in other diseases, but this association in patients with CSVD remains uncovered. Our study aimed to investigate whether serum inflammatory factors correlated with anxiety in patients with CSVD. METHODS A total of 245 CSVD patients confirmed using brain magnetic resonance imaging (MRI) were recruited from December 2019 to December 2021. Hamilton Anxiety Rating Scale (HAMA) was used to assess the anxiety symptoms of CSVD patients. Patients with HAMA scores ≥7 were considered to have anxiety symptoms. The serum levels of interleukin-1β (IL-1β), IL-2R, IL-6, IL-8, IL-10, tumor necrosis factor-α (TNF-α), serum amyloid A (SAA), C-reactive protein (CRP), high-sensitivity C-reactive protein (hs-CRP) and erythrocyte sedimentation rate (ESR) were detected. We compared levels of inflammatory factors between the anxiety and non-anxiety groups. Logistic regression analyses examined the correlation between inflammatory factors and anxiety symptoms. We further performed a gender subgroup analysis to investigate whether this association differed by gender. RESULTS In the fully adjusted multivariate logistic regression analysis model, we found that lower levels of IL-8 were linked to a higher risk of anxiety symptoms. Moreover, higher levels of SAA were linked to a lower risk of anxiety symptoms. Our study identified sex-specific effects, and the correlation between IL-8 and anxiety symptoms remained significant among males, while the correlation between SAA and anxiety symptoms remained significant among females. CONCLUSIONS In this study, we found a suggestive association between IL-8, SAA, and anxiety symptoms in CSVD participants. Furthermore, IL-8 and SAA may have a sex-specific relationship with anxiety symptoms.
Collapse
Affiliation(s)
- Li-Li Shan
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yi-Lin Wang
- Georgetown Preparatory School, North Bethesda, MD, United States
| | - Tian-Ci Qiao
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yue-Feng Bian
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ya-Jing Huo
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Cen Guo
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qian-Yun Liu
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zi-Dong Yang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Ze-Zhi Li
- Department of Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ming-Yuan Liu
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Han
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
42
|
Wu JH, Imadojemu S, Caplan AS. The Evolving Landscape of Cutaneous Sarcoidosis: Pathogenic Insight, Clinical Challenges, and New Frontiers in Therapy. Am J Clin Dermatol 2022; 23:499-514. [PMID: 35583850 DOI: 10.1007/s40257-022-00693-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2022] [Indexed: 12/13/2022]
Abstract
Sarcoidosis is a multisystem disorder of unknown etiology characterized by accumulation of granulomas in affected tissue. Cutaneous manifestations are among the most common extrapulmonary manifestations in sarcoidosis and can lead to disfiguring disease requiring chronic therapy. In many patients, skin disease may be the first recognized manifestation of sarcoidosis, necessitating a thorough evaluation for systemic involvement. Although the precise etiology of sarcoidosis and the pathogenic mechanisms leading to granuloma formation, persistence, or resolution remain unclear, recent research has led to significant advances in our understanding of this disease. This article reviews recent advances in epidemiology, sarcoidosis clinical assessment with a focus on the dermatologist's role, disease pathogenesis, and new therapies in use and under investigation for cutaneous and systemic sarcoidosis.
Collapse
Affiliation(s)
- Julie H Wu
- Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, 240 East 38th Street, 11th Floor, New York, NY, 10016, USA
| | - Sotonye Imadojemu
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Avrom S Caplan
- Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, 240 East 38th Street, 11th Floor, New York, NY, 10016, USA.
- New York University Sarcoidosis Program, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
43
|
Smoldovskaya OV, Voloshin SA, Novikov AA, Aleksandrova EN, Feyzkhanova GU, Rubina AY. Adaptation of Microarray Assay for Serum Amyloid a Analysis in Human Serum. Mol Biol 2022; 56:290-296. [PMID: 35440828 PMCID: PMC9009981 DOI: 10.1134/s0026893322020145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/08/2021] [Accepted: 09/08/2021] [Indexed: 11/24/2022]
Abstract
Serum amyloid A is an inflammatory biomarker whose concentration changes during infectious and inflammatory diseases. SAA’s tendency for aggregation and complex formation makes it difficult to determine its concentration in samples, especially when there is an increased level of it. Immunofluorescence SAA determination on a microarray was adapted for SAA quantification in human serum. Both the procedure and the diluent for the calibrator samples were chosen to obtain a dynamic range between 1 and 100 μg/mL. Mixtures of animal (rabbit, goat, mouse) sera with recombinant antigen diluted in certain concentrations were used for the calibrator samples. The method was tested using serum samples from 15 patients with rheumatoid arthritis or ankylosing spondylitis and 9 healthy donors. The results obtained on the microarray demonstrated a good correlation with the results determined by ELISA (Pearson’s correlation coefficient is 0.93). The method developed could be a convenient tool for assessing SAA levels in a number of diseases, such as rheumatoid arthritis or infections of various etiologies, characterized by a significant increase in the level of this protein in the blood. The use of a microarray for the analysis allows the determination of the SAA concentration simultaneously with other inflammatory biomarkers.
Collapse
Affiliation(s)
- O. V. Smoldovskaya
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - S. A. Voloshin
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - A. A. Novikov
- Moscow Clinical Scientific Center A.S. Loginov, 111123 Moscow, Russia
| | | | - G. U. Feyzkhanova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - A. Yu. Rubina
- Moscow Clinical Scientific Center A.S. Loginov, 111123 Moscow, Russia
| |
Collapse
|
44
|
AA amyloidosis without systemic inflammation: when clinical evidence validates predictions of experimental medicine. Kidney Int 2022; 101:219-221. [PMID: 35065689 DOI: 10.1016/j.kint.2021.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/02/2021] [Accepted: 12/07/2021] [Indexed: 11/20/2022]
Abstract
Amyloid A (AA) amyloidosis is a well-known consequence of chronic inflammatory diseases in which elevated plasma concentrations of serum amyloid A result in amyloid aggregation and organ damage. In this issue, Sikora et al. report, for the first time, an inherited form of AA amyloidosis occurring in the absence of systemic inflammation. This finding may provide novel insights into the pathogenesis of AA amyloidosis, allowing researchers to further dissect the role of inflammation from that of serum amyloid A.
Collapse
|
45
|
Feyzkhanova GU, Voloshin SA, Novikov AA, Aleksandrova EN, Smoldovskaya OV, Rubina AY. Analysis of rheumatoid factor and acute phase proteins using microarrays in patients with rheumatoid arthritis. Klin Lab Diagn 2022; 67:43-47. [PMID: 35077069 DOI: 10.51620/0869-2084-2022-67-1-43-47] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
One of the biomarkers of biggest clinical importance in rheumatoid arthritis (RA) is rheumatoid factor (IgM RF). The rheumatoid factor has insufficient sensitivity and specificity, therefore, to increase the diagnostic information of the test, acute phase proteins were used as concomitant biomarkers. Using biological microchips, we measured IgM RF, C-reactive protein (CRP) and Serum amyloid protein A (SAA) in patients with RA (n = 60), ankylosing spondylitis (AS) (n=55), systemic lupus erythematosus (SLE) (n=20) and healthy donors (HD) (n=9). It was shown that the medians of IgM RF concentrations are significantly higher (p<0.01) in patients with RA compared to patients suffering from other diseases and healthy donors. CRP and SAA were also significantly increased (p<0.05) in patients with RA and AS compared with SLE and HD. It has been shown that the complex determination of three biomarkers in differentiating RA patients with the comparison group had a higher diagnostic sensitivity than the isolated determination of IgM RF, while the addition of SAA makes the greatest contribution to improving the diagnostic characteristics of the biomarker panel: the use of a logistic regression model based on IgM RF and SAA allowed to increase the diagnostic sensitivity of the analysis from 58.3% to 65%. Thus, the developed microarray-based method can be used to detect and elucidate the diagnostic characteristics of RA biomarkers; however, further use requires validation of the obtained results on an expanded sampling.
Collapse
Affiliation(s)
- G U Feyzkhanova
- FSBIS Engelhardt Institute of Molecular Biology, Russian Academy of Sciences
| | - S A Voloshin
- FSBIS Engelhardt Institute of Molecular Biology, Russian Academy of Sciences
| | - A A Novikov
- SBIHC Moscow Clinical Scientific Center n.a. A.S. Loginov
| | | | - O V Smoldovskaya
- FSBIS Engelhardt Institute of Molecular Biology, Russian Academy of Sciences
| | - A Yu Rubina
- FSBIS Engelhardt Institute of Molecular Biology, Russian Academy of Sciences
| |
Collapse
|
46
|
Saliu TP, Yazawa N, Hashimoto K, Miyata K, Kudo A, Horii M, Kamesawa M, Kumrungsee T, Yanaka N. Serum Amyloid A3 Promoter-Driven Luciferase Activity Enables Visualization of Diabetic Kidney Disease. Int J Mol Sci 2022; 23:ijms23020899. [PMID: 35055081 PMCID: PMC8779903 DOI: 10.3390/ijms23020899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 12/10/2022] Open
Abstract
The early detection of diabetic nephropathy (DN) in mice is necessary for the development of drugs and functional foods. The purpose of this study was to identify genes that are significantly upregulated in the early stage of DN progression and develop a novel model to non-invasively monitor disease progression within living animals using in vivo imaging technology. Streptozotocin (STZ) treatment has been widely used as a DN model; however, it also exhibits direct cytotoxicity to the kidneys. As it is important to distinguish between DN-related and STZ-induced nephropathy, in this study, we compared renal responses induced by the diabetic milieu with two types of STZ models: multiple low-dose STZ injections with a high-fat diet and two moderate-dose STZ injections to induce DN. We found 221 genes whose expression was significantly altered during DN development in both models and identified serum amyloid A3 (Saa3) as a candidate gene. Next, we applied the Saa3 promoter-driven luciferase reporter (Saa3-promoter luc mice) to these two STZ models and performed in vivo bioluminescent imaging to monitor the progression of renal pathology. In this study, to further exclude the possibility that the in vivo bioluminescence signal is related to renal cytotoxicity by STZ treatment, we injected insulin into Saa3-promoter luc mice and showed that insulin treatment could downregulate renal inflammatory responses with a decreased signal intensity of in vivo bioluminescence imaging. These results strongly suggest that Saa3 promoter activity is a potent non-invasive indicator that can be used to monitor DN progression and explore therapeutic agents and functional foods.
Collapse
|
47
|
Zhou J, Dai Y, Lin Y, Chen K. Association between serum amyloid A and rheumatoid arthritis: A systematic review and meta-analysis. Semin Arthritis Rheum 2021; 52:151943. [PMID: 35027248 DOI: 10.1016/j.semarthrit.2021.12.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/12/2021] [Accepted: 12/20/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUNDS Consistent correlation of serum amyloid A (SAA) to rheumatoid arthritis (RA) is not completely established. The present study is to systematically summarize their relationship. METHODS Publications up to may 2021 were examined using key terms in the PubMed, Cochrane Library, Embase and China national knowledge infrastructure (CNKI) databases. RESULTS The total 33 studies, involving in 3524 RA cases and 3537 normal participants, were included. The pooled result indicated that the SAA level in the RA group was markedly higher than that in the control group [standardized mean difference (SMD) = 0.80, 95% CI (0.51, 1.08)]. By stratified analyses, the concentration of SAA was found to be gradually increased with the aggravation of RA. Additionally, the meta-analysis of correlation demonstrated that SAA levels were positively associated with the levels of disease activity score 28 (DAS28) [r = 0.55, 95% CI (0.15, 0.94)], erythrocyte sedimentation rate (ESR) [r = 0.65, 95% CI (0.53, 0.76)], C-reactive protein (CRP) [r = 0.92, 95% CI (0.57, 1.57)], rheumatoid factor (RF) [r = 0.24, 95% CI (0.09, 0.39)], interleukin 4 (IL-4) [r = 0.54, 95% CI (0.30, 0.78)], interleukin 6 (IL-6) [r = 0.46, 95% CI (0.27, 0.65)], interleukin 10 (IL-10) [r = 0.53, 95% CI (0.29, 0.77)], interleukin 17 (IL-17) [r = 0.52, 95% CI (0.27, 0.77)], and anti-cyclic citrullinated peptide antibody (A-CCP) [r = 0.32, 95% CI (0.15, 0.50)], but inversely linked with the levels of hemoglobin [r=-0.51, 95% CI (-0.84, -0.18)]. Furthermore, the allele of SAA 1.3 was actively related with increased risks of RA [OR=1.30, 95% CI (1.02, 1.65)] and of RA with amyloidosis [OR=2.06, 95% CI (1.63, 2.60)]. Besides, the genotype of SAA 1.3/1.3 was positively connected with the risks of RA [OR=1.56, 95% CI (1.00, 2.43)] and of RA with amyloidosis [OR=4.47, 95% CI (2.70, 7.41)]. CONCLUSIONS High levels of SAA might be associated with elevated risk of RA, and the concentration of SAA might be gradually increased with the aggravation of RA. Moreover, high levels of SAA might play a vital role in RA by enhancing the levels of DAS28, ESR, CRP, RF, IL-4, IL-6, IL-10, IL-17 and A-CCP, or by attenuating hemoglobin levels. More importantly, the allele of SAA 1.3 and genotype of SAA 1.3/1.3 might be the risk factor of RA and of RA with amyloidosis.
Collapse
Affiliation(s)
- Jielin Zhou
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, Anhui 230032,China
| | - Yu Dai
- Department of Surgery, Suzhou Hospital of Anhui Medical University, Suzhou, Anhui 234000, China
| | - Yan Lin
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
| | - Keyang Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, Anhui 230032,China; Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China.
| |
Collapse
|
48
|
Nienhuis WA, Grutters JC. Potential therapeutic targets to prevent organ damage in chronic pulmonary sarcoidosis. Expert Opin Ther Targets 2021; 26:41-55. [PMID: 34949145 DOI: 10.1080/14728222.2022.2022123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Sarcoidosis is a granulomatous inflammatory disease with high chances of reduced quality of life, irreversible organ damage, and reduced life expectancy when vital organs are involved. Any organ system can be affected, and the lungs are most often affected. There is no preventive strategy as the exact etiology is unknown, and complex immunogenetic and environmental factors determine disease susceptibility and phenotype. Present-day treatment options originated from clinical practice and are effective in many patients. However, a substantial percentage of patients suffer from unacceptable side effects or still develop refractory, threatening pulmonary or extrapulmonary disease. AREAS COVERED As non-caseating granulomas, the pathological hallmark of disease, are assigned to divergent activation and regulation of the immune system, targets in relation to the possible triggers of granuloma formation and their sequelae were searched and reviewed. EXPERT OPINION :The immunopathogenesis underlying sarcoidosis has been a dynamic field of study. Several recent new insights give way to promising new therapeutic targets, such as certain antigenic triggers (e.g. from Aspergillus nidulans), mTOR, JAK-STAT and PPARγ pathways, the NRP2 receptor and MMP-12, which await further exploration. Clinical and trigger related phenotyping, and molecular endotyping in sarcoidosis will likely hold the key for precision medicine in the future.
Collapse
Affiliation(s)
- W A Nienhuis
- ILD Center of Excellence, Department of Pulmonology, St Antonius Hospital, Nieuwegein, The Netherlands
| | - J C Grutters
- ILD Center of Excellence, Department of Pulmonology, St Antonius Hospital, Nieuwegein, The Netherlands.,Division of Hearth and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
49
|
Saiki O, Uda H. Ratio of serum amyloid A to C-reactive protein is constant in the same patients but differs greatly between patients with inflammatory diseases. Scand J Immunol 2021; 95:e13121. [PMID: 34796986 DOI: 10.1111/sji.13121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 11/13/2021] [Accepted: 11/17/2021] [Indexed: 11/28/2022]
Abstract
C-reactive protein (CRP) is commonly monitored to track the activity of inflammation and has become the gold standard in the management of all inflammatory diseases. Indeed, serum amyloid A (SAA) have seemed to correlate moderately with CRP, but the discrepancy of CRP and SAA levels has often been reported, especially in rheumatoid arthritis. Then, we examined CRP reflects a real magnitude of inflammation in patients with rheumatic and infectious inflammatory diseases. A total of 414 patients with infectious and non-infectious inflammatory diseases were enrolled. At initial visit, each patient underwent a clinical assessment and had also laboratory tests such as SAA and CRP. In each patient, we carried out a longitudinal analysis of CRP and SAA levels. We determined the inter-individual correlation between SAA and CRP and also clarified intra-individual changes of SAA/CRP ratio. SAA and CRP levels changed approximately linearly over time within individuals irrespective of rheumatic and infectious inflammatory diseases. However, SAA/CRP ratios differed dramatically between patients (from 0.117 to 50.8, median 5.71). In patients with high SAA/CRP ratio (>8.44), SAA is a better predictor of inflammation than CRP. In contrast, CRP is a better predictor in patients with low ratio (<3.52). Our results suggest that the SAA/CRP ratio differed greatly between individuals but was constant in intra-individuals. Low CRP levels could be accompanied by SAA levels predicting any degree of inflammation, implying that CRP is not reflecting a real magnitude of inflammation. To evaluate the real magnitude of inflammation, to access the SAA/CRP ratio in advance is essential.
Collapse
Affiliation(s)
- Osamu Saiki
- Department of Rheumatology, Higashiosaka City Medical Center, Higashiosaka City, Japan
| | - Hiroshi Uda
- Department of Rheumatology, Higashiosaka City Medical Center, Higashiosaka City, Japan
| |
Collapse
|
50
|
Barros B, Oliveira M, Morais S. Firefighters' occupational exposure: Contribution from biomarkers of effect to assess health risks. ENVIRONMENT INTERNATIONAL 2021; 156:106704. [PMID: 34161906 DOI: 10.1016/j.envint.2021.106704] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 06/04/2021] [Accepted: 06/07/2021] [Indexed: 06/13/2023]
Abstract
Firefighting is physically and physiologically exhausting besides encompassing exposure to toxic fire emissions. Biomonitoring studies from the past five years have been significantly contributing to characterize the occupational-related health effects in this group of professionals and to improve risk assessment. Therefore, this study gathers and critically discusses the most characterized biomarkers of effect (oxidative stress, DNA and protein damage, stress hormones, inflammation, and vascular, lung, and liver injury), including those potentially more promising to be explored in future studies, and their relation with health outcomes. Various studies proved an association between exposures to fire emissions and/or heat and significantly altered values of biomarkers of inflammation (soluble adhesion molecules, tumor necrosis factor, interleukins, and leucocyte count), vascular damage and tissue injury (pentraxin-3, vascular endothelial growth factor, and cardiac troponin T) in firefighting forces. Moreover, preliminary data of DNA damage in blood, urinary mutagenicity and 8-isoprostaglandin in exhaled breath condensate suggest that these biomarkers of oxidative stress should be further explored. However, most of the reported studies are based on cross-sectional designs, which limit full identification and characterization of the risk factors and their association with development of work-related diseases. Broader studies based on longitudinal designs and strongly supported by the analysis of several types of biomarkers in different biological fluids are further required to gain deeper insights into the firefighters occupational related health hazards and contribute to implementation of new or improved surveillance programs.
Collapse
Affiliation(s)
- Bela Barros
- REQUIMTE-LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, Rua Dr. António Bernardino de Almeida 431, 4249-015 Porto
| | - Marta Oliveira
- REQUIMTE-LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, Rua Dr. António Bernardino de Almeida 431, 4249-015 Porto
| | - Simone Morais
- REQUIMTE-LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, Rua Dr. António Bernardino de Almeida 431, 4249-015 Porto.
| |
Collapse
|