51
|
Hemed-Shaked M, Cowman MK, Kim JR, Huang X, Chau E, Ovadia H, Amar KO, Eshkar-Sebban L, Melamed M, Lev LB, Kedar E, Armengol J, Alemany J, Beyth S, Okon E, Kanduc D, Elgavish S, Wallach-Dayan SB, Cohen SJ, Naor D. MTADV 5-MER peptide suppresses chronic inflammations as well as autoimmune pathologies and unveils a new potential target-Serum Amyloid A. J Autoimmun 2021; 124:102713. [PMID: 34390919 DOI: 10.1016/j.jaut.2021.102713] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/21/2021] [Accepted: 07/24/2021] [Indexed: 11/17/2022]
Abstract
Despite the existence of potent anti-inflammatory biological drugs e.g., anti-TNF and anti IL-6 receptor antibodies, for treating chronic inflammatory and autoimmune diseases, these are costly and not specific. Cheaper oral available drugs remain an unmet need. Expression of the acute phase protein Serum Amyloid A (SAA) is dependent on release of pro-inflammatory cytokines IL-1, IL-6 and TNF-α during inflammation. Conversely, SAA induces pro-inflammatory cytokine secretion, including Th17, leading to a pathogenic vicious cycle and chronic inflammation. 5- MER peptide (5-MP) MTADV (methionine-threonine-alanine-aspartic acid-valine), also called Amilo-5MER, was originally derived from a sequence of a pro-inflammatory CD44 variant isolated from synovial fluid of a Rheumatoid Arthritis (RA) patient. This human peptide displays an efficient anti-inflammatory effects to ameliorate pathology and clinical symptoms in mouse models of RA, Inflammatory Bowel Disease (IBD) and Multiple Sclerosis (MS). Bioinformatics and qRT-PCR revealed that 5-MP, administrated to encephalomyelytic mice, up-regulates genes contributing to chronic inflammation resistance. Mass spectrometry of proteins that were pulled down from an RA synovial cell extract with biotinylated 5-MP, showed that it binds SAA. 5-MP disrupted SAA assembly, which is correlated with its pro-inflammatory activity. The peptide MTADV (but not scrambled TMVAD) significantly inhibited the release of pro-inflammatory cytokines IL-6 and IL-1β from SAA-activated human fibroblasts, THP-1 monocytes and peripheral blood mononuclear cells. 5-MP suppresses the pro-inflammatory IL-6 release from SAA-activated cells, but not from non-activated cells. 5-MP could not display therapeutic activity in rats, which are SAA deficient, but does inhibit inflammations in animal models of IBD and MS, both are SAA-dependent, as shown by others in SAA knockout mice. In conclusion, 5-MP suppresses chronic inflammation in animal models of RA, IBD and MS, which are SAA-dependent, but not in animal models, which are SAA-independent.
Collapse
Affiliation(s)
- Maayan Hemed-Shaked
- The Lautenberg Center of Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Mary K Cowman
- Department of Biomedical Engineering, Tandon School of Engineering, New York University, New York, NY, USA
| | - Jin Ryoun Kim
- Othmer-Jacobs Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, Brooklyn, USA
| | - Xiayun Huang
- Othmer-Jacobs Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, Brooklyn, USA
| | - Edward Chau
- Othmer-Jacobs Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, Brooklyn, USA
| | - Haim Ovadia
- Department of Neurology, Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Keren-Or Amar
- The Lautenberg Center of Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Lora Eshkar-Sebban
- The Lautenberg Center of Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Michal Melamed
- The Lautenberg Center of Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Libat Bar Lev
- The Lautenberg Center of Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Eli Kedar
- The Lautenberg Center of Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | | | | | - Shaul Beyth
- Orthopedic Surgery Department, Hadassah University Hospital, Jerusalem, Israel
| | - Eli Okon
- Department of Pathology, Hadassah University Hospital, Jerusalem, Israel
| | - Darja Kanduc
- Biotechnologies and Biopharmaceutics, University of Bari, Bari, 70126, Italy
| | - Sharona Elgavish
- Bioinformatics Unit of the Hebrew University of Jerusalem and Hadassah Medical Center, Israel
| | - Shulamit B Wallach-Dayan
- Lung Cellular and Molecular Biology Laboratory, Institute of Pulmonary Medicine, Hadassah-Hebrew University Medical Center, Israel
| | - Shmuel Jaffe Cohen
- The Lautenberg Center of Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - David Naor
- The Lautenberg Center of Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel.
| |
Collapse
|
52
|
Sudo G, Aoki H, Yamamoto E, Takasawa A, Niinuma T, Yoshido A, Kitajima H, Yorozu A, Kubo T, Harada T, Ishiguro K, Kai M, Katanuma A, Yamano HO, Osanai M, Nakase H, Suzuki H. Activated macrophages promote invasion by early colorectal cancer via an interleukin 1β-serum amyloid A1 axis. Cancer Sci 2021; 112:4151-4165. [PMID: 34293235 PMCID: PMC8486202 DOI: 10.1111/cas.15080] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/06/2021] [Accepted: 07/14/2021] [Indexed: 01/15/2023] Open
Abstract
Submucosal invasion and lymph node metastasis are important issues affecting treatment options for early colorectal cancer (CRC). In this study, we aimed to unravel the molecular mechanism underlying the invasiveness of early CRCs. We performed RNA‐sequencing (RNA‐seq) with poorly differentiated components (PORs) and their normal counterparts isolated from T1 CRC tissues and detected significant upregulation of serum amyloid A1 (SAA1) in PORs. Immunohistochemical analysis revealed that SAA1 was specifically expressed in PORs at the invasive front of T1b CRCs. Upregulation of SAA1 in CRC cells promoted cell migration and invasion. Coculture experiments using CRC cell lines and THP‐1 cells suggested that interleukin 1β (IL‐1β) produced by macrophages induces SAA1 expression in CRC cells. Induction of SAA1 and promotion of CRC cell migration and invasion by macrophages were inhibited by blocking IL‐1β. These findings were supported by immunohistochemical analysis of primary T1 CRCs showing accumulation of M1‐like/M2‐like macrophages at SAA1‐positive invasive front regions. Moreover, SAA1 produced by CRC cells stimulated upregulation of matrix metalloproteinase‐9 in macrophages. Our data suggest that tumor‐associated macrophages at the invasive front of early CRCs promote cancer cell migration and invasion through induction of SAA1 and that SAA1 may be a predictive biomarker and a useful therapeutic target.
Collapse
Affiliation(s)
- Gota Sudo
- Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hironori Aoki
- Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo, Japan.,Center for Gastroenterology, Teine-Keijinkai Hospital, Sapporo, Japan
| | - Eiichiro Yamamoto
- Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Akira Takasawa
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takeshi Niinuma
- Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Ayano Yoshido
- Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hiroshi Kitajima
- Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Akira Yorozu
- Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Toshiyuki Kubo
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Taku Harada
- Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo, Japan.,Center for Gastroenterology, Teine-Keijinkai Hospital, Sapporo, Japan
| | - Kazuya Ishiguro
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Masahiro Kai
- Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Akio Katanuma
- Center for Gastroenterology, Teine-Keijinkai Hospital, Sapporo, Japan
| | - Hiro-O Yamano
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Makoto Osanai
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hiroshi Nakase
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hiromu Suzuki
- Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
53
|
Davis TA, Conradie D, Shridas P, de Beer FC, Engelbrecht AM, de Villiers WJS. Serum Amyloid A Promotes Inflammation-Associated Damage and Tumorigenesis in a Mouse Model of Colitis-Associated Cancer. Cell Mol Gastroenterol Hepatol 2021; 12:1329-1341. [PMID: 34217896 PMCID: PMC8463861 DOI: 10.1016/j.jcmgh.2021.06.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 06/20/2021] [Accepted: 06/21/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND & AIMS Identifying new approaches to lessen inflammation, as well as the associated malignant consequences, remains crucial to improving the lives and prognosis of patients diagnosed with inflammatory bowel diseases. Although it previously has been suggested as a suitable biomarker for monitoring disease activity in patients diagnosed with Crohn's disease, the role of the acute-phase protein serum amyloid A (SAA) in inflammatory bowel disease remains unclear. In this study, we aimed to assess the role of SAA in colitis-associated cancer. METHODS We established a model of colitis-associated cancer in wild-type and SAA double-knockout (Saa1/2-/-) mice by following the azoxymethane/dextran sulfate sodium protocol. Disease activity was monitored throughout the study while colon and tumor tissues were harvested for subsequent use in cytokine analyses, Western blot, and immunohistochemistry +experiments. RESULTS We observed attenuated disease activity in mice deficient for Saa1/2 as evidenced by decreased weight loss, increased stool consistency, decreased rectal bleeding, and decreased colitis-associated tissue damage. Macrophage infiltration, including CD206+ M2-like macrophages, also was attenuated in SAA knockout mice, while levels of interleukin 4, interleukin 10, and tumor necrosis factor-ɑ were decreased in the distal colon. Mice deficient for SAA also showed a decreased tumor burden, and tumors were found to have increased apoptotic activity coupled with decreased expression for markers of proliferation. CONCLUSION Based on these findings, we conclude that SAA has an active role in inflammatory bowel disease and that it could serve as a therapeutic target aimed at decreasing chronic inflammation and the associated risk of developing colitis-associated cancer.
Collapse
Affiliation(s)
| | | | - Preetha Shridas
- Department of Internal Medicine, University of Kentucky, Lexington, Kentucky
| | - Frederick C de Beer
- Department of Internal Medicine, University of Kentucky, Lexington, Kentucky
| | - Anna-Mart Engelbrecht
- Department of Physiological Sciences; African Cancer Institute, Department of Global Health
| | - Willem J S de Villiers
- African Cancer Institute, Department of Global Health; Department of Internal Medicine, Stellenbosch University, Stellenbosch, South Africa.
| |
Collapse
|
54
|
Serum Amyloid A Proteins and Their Impact on Metastasis and Immune Biology in Cancer. Cancers (Basel) 2021; 13:cancers13133179. [PMID: 34202272 PMCID: PMC8267706 DOI: 10.3390/cancers13133179] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/19/2021] [Accepted: 06/22/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary The liver responds to systemic inflammation and injury in a coordinated manner, called the acute phase response. While this normal physiological response aims to restore homeostasis, malignant transformation coopts this biology to increase the risk for metastasis, immune evasion, and therapeutic resistance. In this Review, we discuss the importance of acute phase response proteins in regulating cancer biology and treatment efficacy. We also consider potential strategies to intervene on acute phase biology as an approach to improve outcomes in cancer. Abstract Cancer triggers the systemic release of inflammatory molecules that support cancer cell metastasis and immune evasion. Notably, this biology shows striking similarity to an acute phase response that is coordinated by the liver. Consistent with this, a role for the liver in defining cancer biology is becoming increasingly appreciated. Understanding the mechanisms that link acute phase biology to metastasis and immune evasion in cancer may reveal vulnerable pathways and novel therapeutic targets. Herein, we discuss a link between acute phase biology and cancer with a focus on serum amyloid A proteins and their involvement in regulating the metastatic cascade and cancer immunobiology.
Collapse
|
55
|
Abouelasrar Salama S, Gouwy M, Van Damme J, Struyf S. The turning away of serum amyloid A biological activities and receptor usage. Immunology 2021; 163:115-127. [PMID: 33315264 PMCID: PMC8114209 DOI: 10.1111/imm.13295] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/24/2020] [Accepted: 12/04/2020] [Indexed: 12/21/2022] Open
Abstract
Serum amyloid A (SAA) is an acute-phase protein (APP) to which multiple immunological functions have been attributed. Regardless, the true biological role of SAA remains poorly understood. SAA is remarkably conserved in mammalian evolution, thereby suggesting an important biological function. Since its discovery in the 1970s, the majority of researchers have investigated SAA using recombinant forms made available through bacterial expression. Nevertheless, recent studies indicate that these recombinant forms of SAA are unreliable. Indeed, commercial SAA variants have been shown to be contaminated with bacterial products including lipopolysaccharides and lipoproteins. As such, biological activities and receptor usage (TLR2, TLR4) revealed through the use of commercial SAA variants may not reflect the inherent nature of this APP. Within this review, we discuss the biological effects of SAA that have been demonstrated through more solid experimental approaches. SAA takes part in the innate immune response via the recruitment of leucocytes and executes, through pathogen recognition, antimicrobial activity. Knockout animal models implicate SAA in a range of functions, such as regulation of T-cell-mediated responses and monopoiesis. Moreover, through its structural motifs, not only does SAA function as an extracellular matrix protein, but it also binds extracellular matrix proteins. Finally, we here also provide an overview of definite SAA receptor-mediated functions and highlight those that are yet to be validated. The role of FPR2 in SAA-mediated leucocyte recruitment has been confirmed; nevertheless, SAA has been linked to a range of other receptors including CD36, SR-BI/II, RAGE and P2RX7.
Collapse
Affiliation(s)
- Sara Abouelasrar Salama
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| | - Mieke Gouwy
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| | - Jo Van Damme
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| | - Sofie Struyf
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| |
Collapse
|
56
|
Pieri M, Ciotti M, Nuccetelli M, Perrone MA, Caliò MT, Lia MS, Minieri M, Bernardini S. Serum Amyloid A Protein as a useful biomarker to predict COVID-19 patients severity and prognosis. Int Immunopharmacol 2021; 95:107512. [PMID: 33735714 PMCID: PMC7923863 DOI: 10.1016/j.intimp.2021.107512] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/13/2021] [Accepted: 02/13/2021] [Indexed: 12/15/2022]
Abstract
Coronavirus Disease 2019 (COVID-19) can present with different grades of severity from mild to critical. Evaluation of biomarkers predicting severity is crucial to identify patients at high risk of disease progression and poor prognosis. Serum Amyloid A (SAA) is an acute-phase protein mainly produced by the liver in response to pro-inflammatory cytokines. In this study, we investigated SAA levels at admission (T1) and after 15 days (T2) of hospitalization in two groups of patients: survivors and non-survivors. At T1, the non-survivors showed higher SAA level than survivors (74 mg/dL vs 48.75 mg/dL). At T2, the survivor group value decreased to 6.55 mg/dL, the non-survivor group still showed high levels (51.1 mg/dL). The SAA level in control group was 0.35 mg/dL. Furthermore, a cut-off value of 63 mg/dL able to discriminate survivors from non-survivors was established by ROC curve analysis at T1. At T2, the cut-off decreased to 30.9 mg/dL. A similar decreasing trend was observed for D-Dimer, hsCRP, IL-6 and procalcitonin levels. The results of this retrospective study suggest that SAA is a good marker of COVID-19 disease alone and/or in combination with other inflammatory biomarkers. Identification of reliable prognostic analytes is of great clinical relevance, as it would improve patient management besides being costs saving.
Collapse
Affiliation(s)
- Massimo Pieri
- Department of Experimental Medicine, University of Tor Vergata, Rome, Italy.
| | | | | | | | | | - Maria Stella Lia
- Department of Experimental Medicine, University of Tor Vergata, Rome, Italy
| | - Marilena Minieri
- Department of Experimental Medicine, University of Tor Vergata, Rome, Italy; Tor Vergata University Hospital, Rome, Italy
| | - Sergio Bernardini
- Department of Experimental Medicine, University of Tor Vergata, Rome, Italy; Tor Vergata University Hospital, Rome, Italy; IFCC Emerging Technologies Division, Milan, Italy
| |
Collapse
|
57
|
Horst EA, Kvidera SK, Baumgard LH. Invited review: The influence of immune activation on transition cow health and performance-A critical evaluation of traditional dogmas. J Dairy Sci 2021; 104:8380-8410. [PMID: 34053763 DOI: 10.3168/jds.2021-20330] [Citation(s) in RCA: 143] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/15/2021] [Indexed: 12/11/2022]
Abstract
The progression from gestation into lactation represents the transition period, and it is accompanied by marked physiological, metabolic, and inflammatory adjustments. The entire lactation and a cow's opportunity to have an additional lactation are heavily dependent on how successfully she adapts during the periparturient period. Additionally, a disproportionate amount of health care and culling occurs early following parturition. Thus, lactation maladaptation has been a heavily researched area of dairy science for more than 50 yr. It was traditionally thought that excessive adipose tissue mobilization in large part dictated transition period success. Further, the magnitude of hypocalcemia has also been assumed to partly control whether a cow effectively navigates the first few months of lactation. The canon became that adipose tissue released nonesterified fatty acids (NEFA) and the resulting hepatic-derived ketones coupled with hypocalcemia lead to immune suppression, which is responsible for transition disorders (e.g., mastitis, metritis, retained placenta, poor fertility). In other words, the dogma evolved that these metabolites and hypocalcemia were causal to transition cow problems and that large efforts should be enlisted to prevent increased NEFA, hyperketonemia, and subclinical hypocalcemia. However, despite intensive academic and industry focus, the periparturient period remains a large hurdle to animal welfare, farm profitability, and dairy sustainability. Thus, it stands to reason that there are alternative explanations to periparturient failures. Recently, it has become firmly established that immune activation and the ipso facto inflammatory response are a normal component of transition cow biology. The origin of immune activation likely stems from the mammary gland, tissue trauma during parturition, and the gastrointestinal tract. If inflammation becomes pathological, it reduces feed intake and causes hypocalcemia. Our tenet is that immune system utilization of glucose and its induction of hypophagia are responsible for the extensive increase in NEFA and ketones, and this explains why they (and the severity of hypocalcemia) are correlated with poor health, production, and reproduction outcomes. In this review, we argue that changes in circulating NEFA, ketones, and calcium are simply reflective of either (1) normal homeorhetic adjustments that healthy, high-producing cows use to prioritize milk synthesis or (2) the consequence of immune activation and its sequelae.
Collapse
Affiliation(s)
- E A Horst
- Department of Animal Science, Iowa State University, Ames 50011
| | - S K Kvidera
- Department of Animal Science, Iowa State University, Ames 50011
| | - L H Baumgard
- Department of Animal Science, Iowa State University, Ames 50011.
| |
Collapse
|
58
|
Karlsson MJ, Costa Svedman F, Tebani A, Kotol D, Höiom V, Fagerberg L, Edfors F, Uhlén M, Egyhazi Brage S, Maddalo G. Inflammation and Apolipoproteins Are Potential Biomarkers for Stratification of Cutaneous Melanoma Patients for Immunotherapy and Targeted Therapy. Cancer Res 2021; 81:2545-2555. [PMID: 33574091 DOI: 10.1158/0008-5472.can-20-2000] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/22/2020] [Accepted: 02/09/2021] [Indexed: 11/16/2022]
Abstract
Malignant cutaneous melanoma is one of the most common cancers in young adults. During the last decade, targeted and immunotherapies have significantly increased the overall survival of patients with malignant cutaneous melanoma. Nevertheless, disease progression is common, and a lack of predictive biomarkers of patient response to therapy hinders individualized treatment strategies. To address this issue, we performed a longitudinal study using an unbiased proteomics approach to identify and quantify proteins in plasma both before and during treatment from 109 patients treated with either targeted or immunotherapy. Linear modeling and machine learning approaches identified 43 potential prognostic and predictive biomarkers. A reverse correlation between apolipoproteins and proteins related to inflammation was observed. In the immunotherapy group, patients with low pretreatment expression of apolipoproteins and high expression of inflammation markers had shorter progression-free survival. Similarly, increased expression of LDHB during treatment elicited a significant impact on response to immunotherapy. Overall, we identified potential common and treatment-specific biomarkers in malignant cutaneous melanoma, paving the way for clinical use of these biomarkers following validation on a larger cohort. SIGNIFICANCE: This study identifies a potential biomarker panel that could improve the selection of therapy for patients with cutaneous melanoma.
Collapse
Affiliation(s)
- Max J Karlsson
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | | | - Abdellah Tebani
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - David Kotol
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Veronica Höiom
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Linn Fagerberg
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Fredrik Edfors
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Mathias Uhlén
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | | | - Gianluca Maddalo
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden.
| |
Collapse
|
59
|
Zinellu A, Paliogiannis P, Carru C, Mangoni AA. Serum amyloid A concentrations, COVID-19 severity and mortality: An updated systematic review and meta-analysis. Int J Infect Dis 2021; 105:668-674. [PMID: 33737133 PMCID: PMC7959678 DOI: 10.1016/j.ijid.2021.03.025] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/01/2021] [Accepted: 03/10/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND OBJECTIVES An excessive inflammatory response in patients with coronavirus disease 2019 (COVID-19) is associated with high disease severity and mortality. Specific acute phase reactants might be useful for risk stratification. A systematic review and meta-analysis was conducted of studies on serum amyloid A (SAA) in patients with COVID-19. METHODS The PubMed, Web of Science, and Scopus databases were searched, covering the period January 2020 to December 2020, for studies reporting SAA concentrations, COVID-19 severity, and survival status. RESULTS Nineteen studies involving 5617 COVID-19 patients were included in the meta-analysis. Pooled results showed that SAA concentrations were significantly higher in patients with severe disease and non-survivors (standard mean difference (SMD) 1.20, 95% confidence interval 0.91-1.49, P < 0.001). Extreme between-study heterogeneity was observed (I2 = 92.4%, P < 0.001). In the sensitivity analysis, the effect size was not significantly affected when each study was removed in turn (range 1.10-1.29). The Begg test (P = 0.030), but not the Egger test (P = 0.385), revealed the presence of publication bias. Pooled SMD values were significantly and positively associated with sex (t = 2.20, P = 0.047) and aspartate aminotransferase (t = 3.44, P = 0.014). CONCLUSIONS SAA concentrations were significantly and positively associated with higher COVID-19 severity and mortality. This acute phase reactant might assist with risk stratification and monitoring in this group.
Collapse
Affiliation(s)
- Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Panagiotis Paliogiannis
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Ciriaco Carru
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy; Quality Control Unit, University Hospital (AOUSS), Sassari, Italy
| | - Arduino A Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Adelaide, Australia.
| |
Collapse
|
60
|
Jiang Y, Pin L, Shi W, Huang Q, Wang L, Liu H. SAA1 regulates pro-labour mediators in term labour by activating YAP pathway. Mol Cell Biochem 2021; 476:2791-2801. [PMID: 33719002 DOI: 10.1007/s11010-021-04125-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 03/03/2021] [Indexed: 01/22/2023]
Abstract
Term labour is associated with activation of inflammation which results in myometrial contractility, cervical ripening and decidual/membrane rupture. Serum amyloid A1 (SAA1) is an acute response protein, whose role and underlying regulatory mechanisms in human labour remain unknown. In this study, we found that the mRNA and protein expression of SAA1 in human myometrium at term was increased in labouring tissues compared to non-labouring tissues. In addition, the expression of SAA1 was significantly increased in human primary myometrial cells treated with the pro-inflammatory cytokines interleukin-1 beta (IL-1β) or tumour necrosis factor-alpha (TNF-α). Knockdown of SAA1 using siRNA (siSAA1) resulted in a significant reduction in the expression and secretion of pro-inflammatory cytokines (IL8, IL6), chemokines (CXCL5, CCL2), adhesion molecules (ICAM1, ICAM5) and contraction-associated factors (COX2, PGE2). Mechanistically, the effects of SAA1 were mediated through activation of the Yes-associated protein (YAP) pathway. There was a decrease in the protein expression of phosphorylated YAP (pYAP) after treatment of siSAA1-transfected human primary myometrial cells with IL-1β or TNF-α. Moreover, enhanced expression of YAP reversed the effect of siSAA1 on pro-labour mediators. In conclusion, these experiments demonstrated that SAA1 accelerates the inflammatory response associated with parturition by activating YAP pathway, which may be a novel understanding of the molecular mechanism of labour onset.
Collapse
Affiliation(s)
- Yanmin Jiang
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, No.9, Jinsui Road, Tianhe District, Guangzhou, 510623, China
| | - Li Pin
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, No.9, Jinsui Road, Tianhe District, Guangzhou, 510623, China
| | - Weiqun Shi
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, No.9, Jinsui Road, Tianhe District, Guangzhou, 510623, China
| | - Qian Huang
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, No.9, Jinsui Road, Tianhe District, Guangzhou, 510623, China
| | - Lele Wang
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, No.9, Jinsui Road, Tianhe District, Guangzhou, 510623, China
| | - Huishu Liu
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, No.9, Jinsui Road, Tianhe District, Guangzhou, 510623, China.
| |
Collapse
|
61
|
Abstract
PURPOSE OF REVIEW Serum amyloid A (SAA) is a highly sensitive acute phase reactant that has been linked to a number of chronic inflammatory diseases. During a systemic inflammatory response, liver-derived SAA is primarily found on high-density lipoprotein (HDL). The purpose of this review is to discuss recent literature addressing the pathophysiological functions of SAA and the significance of its association with HDL. RECENT FINDINGS Studies in gene-targeted mice establish that SAA contributes to atherosclerosis and some metastatic cancers. Accumulating evidence indicates that the lipidation state of SAA profoundly affects its bioactivities, with lipid-poor, but not HDL-associated, SAA capable of inducing inflammatory responses in vitro and in vivo. Factors that modulate the equilibrium between lipid-free and HDL-associated SAA have been identified. HDL may serve to limit SAA's bioactivities in vivo. Understanding the factors leading to the release of systemic SAA from HDL may provide insights into chronic disease mechanisms.
Collapse
Affiliation(s)
- Nancy R Webb
- Department of Pharmacology and Nutritional Sciences, Saha Cardiovascular Research Center, and Barnstable Brown Diabetes Center, University of Kentucky, 553 Wethington Building, 900 South Limestone, Lexington, KY, 40536-0200, USA.
| |
Collapse
|
62
|
Serum Amyloid A Is a Biomarker of Disease Activity and Health-Related Quality-of-Life in Patients with Antineutrophil Cytoplasmic Antibody-Associated Vasculitis. DISEASE MARKERS 2020; 2020:8847306. [PMID: 33488845 PMCID: PMC7787824 DOI: 10.1155/2020/8847306] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 11/26/2020] [Accepted: 12/04/2020] [Indexed: 11/17/2022]
Abstract
Serum amyloid A (SAA) is one of the acute phase proteins synthesized in hepatocytes and secreted by various inflammation or infectious stimuli. We investigated the clinical implication of measuring SAA in patients with antineutrophil cytoplasmic antibody- (ANCA-) associated vasculitis (AAV). Seventy-five patients who had been classified as AAV and enrolled in our prospective observational cohort for AAV patients were included. Clinical and laboratory data were obtained on the day of blood sampling, and SAA was measured by ELISA kits. Birmingham Vasculitis Activity Score (BVAS) and Short-Form 36-Item Health Survey (SF-36) were assessed for disease activity and health-related quality-of-life (HRQoL) measures. We stratified patients into having high BVAS when the BVAS was over the median values, and those with either low SF-36 PCS or low SF-36 MCS were defined as having poor HRQoL. Multivariate logistic regression analysis was conducted to estimate independent predictors of high BVAS. The relative risk (RR) was analyzed using the contingency tables and the chi-squared test. SAA was positively correlated with BVAS (r = 0.642) and FFS (r = 0.367) and was inversely correlated with both the SF-36 physical component summary (r = -0.456) and mental component summary scores (r = -0.394). Furthermore, SAA was significantly correlated with acute phase reactants ESR (r = 0.611) and CRP (r = 0.629). Patients with high BVAS exhibited significantly higher SAA than those with low BVAS (1317.1 ng/mL vs. 423.1 ng/mL). In multivariable logistic regression analysis, serum albumin (odds ratio (OR) 0.132) and SAA > 1173.6 ng/mL (OR 15.132) were independently associated with high BVAS. The risk of having high BVAS and poor HRQoL in patients with SAA > 1173.6 ng/mL was higher than in those with SAA ≤ 1173.6 ng/mL (RR 3.419 and 1.493). Our results suggest that SAA might be a useful biomarker in assessing disease activity and HRQoL in AAV.
Collapse
|
63
|
Structural Basis for Vital Function and Malfunction of Serum Amyloid A: an Acute-Phase Protein that Wears Hydrophobicity on Its Sleeve. Curr Atheroscler Rep 2020; 22:69. [PMID: 32968930 PMCID: PMC7511256 DOI: 10.1007/s11883-020-00888-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW This review addresses normal and pathologic functions of serum amyloid A (SAA), an enigmatic biomarker of inflammation and protein precursor of AA amyloidosis, a life-threatening complication of chronic inflammation. SAA is a small, highly evolutionarily conserved acute-phase protein whose plasma levels increase up to one thousand-fold in inflammation, infection, or after trauma. The advantage of this dramatic but transient increase is unclear, and the complex role of SAA in immune response is intensely investigated. This review summarizes recent advances in our understanding of the structure-function relationship of this intrinsically disordered protein, outlines its newly emerging beneficial roles in lipid transport and inflammation control, and discusses factors that critically influence its misfolding in AA amyloidosis. RECENT FINDINGS High-resolution structures of lipid-free SAA in crystals and fibrils have been determined by x-ray crystallography and electron cryo-microscopy. Low-resolution structural studies of SAA-lipid complexes, together with biochemical, cell-based, animal model, genetic, and clinical studies, have provided surprising new insights into a wide range of SAA functions. An emerging vital role of SAA is lipid encapsulation to remove cell membrane debris from sites of injury. The structural basis for this role has been proposed. The lysosomal origin of AA amyloidosis has solidified, and its molecular and cellular mechanisms have emerged. Recent studies have revealed molecular underpinnings for understanding complex functions of this Cambrian protein in lipid transport, immune response, and amyloid formation. These findings help guide the search for much-needed targeted therapies to block the protein deposition in AA amyloidosis.
Collapse
|
64
|
Mao K, Geng W, Liao Y, Luo P, Zhong H, Ma P, Xu J, Zhang S, Tan Q, Jin Y. Identification of robust genetic signatures associated with lipopolysaccharide-induced acute lung injury onset and astaxanthin therapeutic effects by integrative analysis of RNA sequencing data and GEO datasets. Aging (Albany NY) 2020; 12:18716-18740. [PMID: 32969837 PMCID: PMC7585091 DOI: 10.18632/aging.104042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 08/19/2020] [Indexed: 01/24/2023]
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are life-threatening clinical conditions predominantly arising from uncontrolled inflammatory reactions. It has been found that the administration of astaxanthin (AST) can exert protective effects against lipopolysaccharide (LPS)-induced ALI; however, the robust genetic signatures underlying LPS induction and AST treatment remain obscure. Here we performed a statistical meta-analysis of five publicly available gene expression datasets from LPS-induced ALI mouse models, conducted RNA-sequencing (RNA-seq) to screen differentially expressed genes (DEGs) in response to LPS administration and AST treatment, and integrative analysis to determine robust genetic signatures associated with LPS-induced ALI onset and AST administration. Both the meta-analyses and our experimental data identified a total of 198 DEGs in response to LPS administration, and 11 core DEGs (Timp1, Ly6i, Cxcl13, Irf7, Cxcl5, Ccl7, Isg15, Saa3, Saa1, Tgtp1, and Gbp11) were identified to be associated with AST therapeutic effects. Further, the 11 core DEGs were verified by quantitative real-time PCR (qRT-PCR) and immunohistochemistry (IHC), and functional enrichment analysis revealed that these genes are primarily associated with neutrophils and chemokines. Collectively, these findings unearthed the robust genetic signatures underlying LPS administration and the molecular targets of AST for ameliorating ALI/ARDS which provide directions for further research.
Collapse
Affiliation(s)
- Kaimin Mao
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Wei Geng
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Yuhan Liao
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Ping Luo
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Hua Zhong
- College of Life Sciences, Wuhan University, Hubei Province, Wuhan, 430072, China
| | - Pei Ma
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Juanjuan Xu
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Shuai Zhang
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Qi Tan
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Yang Jin
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| |
Collapse
|
65
|
Lin A, Liu J, Gong P, Chen Y, Zhang H, Zhang Y, Yu Y. Serum amyloid A inhibits astrocyte migration via activating p38 MAPK. J Neuroinflammation 2020; 17:254. [PMID: 32861245 PMCID: PMC7456509 DOI: 10.1186/s12974-020-01924-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 08/13/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The accumulation of astrocytes around senile plaques is one of the pathological characteristics in Alzheimer's disease (AD). Serum amyloid A (SAA), known as a major acute-phase protein, colocalizes with senile plaques in AD patients. Here, we demonstrate the role of SAA in astrocyte migration. METHODS The effects of SAA on astrocyte activation and accumulation around amyloid β (Aβ) deposits were detected in APP/PS1 transgenic mice mated with Saa3-/- mice. SAA expression, astrocyte activation, and colocalization with Aβ deposits were evaluated in mice using immunofluorescence staining and/or Western blotting. The migration of primary cultures of mouse astrocytes and human glioma U251 cells was examined using Boyden chamber assay and scratch-would assay. The actin and microtubule networks, protrusion formation, and Golgi apparatus location in astrocytes were determined using scratch-would assay and immunofluorescence staining. RESULTS Saa3 expression was significantly induced in aged APP/PS1 transgenic mouse brain. Saa3 deficiency exacerbated astrocyte activation and increased the number of astrocytes around Aβ deposits in APP/PS1 mice. In vitro studies demonstrated that SAA inhibited the migration of primary cultures of astrocytes and U251 cells. Mechanistic studies showed that SAA inhibited astrocyte polarization and protrusion formation via disrupting actin and microtubule reorganization and Golgi reorientation. Inhibition of the p38 MAPK pathway abolished the suppression of SAA on astrocyte migration and polarization. CONCLUSIONS These results suggest that increased SAA in the brain of APP/PS1 mice inhibits the migration of astrocytes to amyloid plaques by activating the p38 MAPK pathway.
Collapse
Affiliation(s)
- Aihua Lin
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jin Liu
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China.,State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ping Gong
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yanqing Chen
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Haibo Zhang
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yan Zhang
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yang Yu
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
66
|
Szeto A, Cecati M, Ahmed R, McCabe PM, Mendez AJ. Oxytocin reduces adipose tissue inflammation in obese mice. Lipids Health Dis 2020; 19:188. [PMID: 32819381 PMCID: PMC7441653 DOI: 10.1186/s12944-020-01364-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 08/10/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Obesity and adipose tissue expansion is characterized by a chronic state of systemic inflammation that contributes to disease. The neuropeptide, oxytocin, working through its receptor has been shown to attenuate inflammation in sepsis, wound healing, and cardiovascular disease. The current study examined the effects of chronic oxytocin infusions on adipose tissue inflammation in a murine model of obesity, the leptin receptor-deficient (db/db) mouse. METHODS The effect of obesity on oxytocin receptor protein and mRNA expression in adipose tissue was evaluated by Western blotting and real-time polymerase chain reaction. Mice were implanted with osmotic minipumps filled with oxytocin or vehicle for 8 weeks. At study endpoint adipose tissue inflammation was assessed by measurement of cytokine and adipokine mRNA tissue levels, adipocyte size and macrophage infiltration via histopathology, and plasma levels of adiponectin and serum amyloid A as markers of systemic inflammation. RESULTS The expression of adipose tissue oxytocin receptor was increased in obese db/db mice compared to lean controls. In adipose tissue oxytocin infusion reduced adipocyte size, macrophage infiltration, IL-6 and TNFα mRNA expression, and increased the expression of the anti-inflammatory adipokine, adiponectin. In plasma, oxytocin infusion reduced the level of serum amyloid A, a marker of systemic inflammation, and increased circulating adiponectin. CONCLUSIONS In an animal model of obesity and diabetes chronic oxytocin treatment led to a reduction in visceral adipose tissue inflammation and plasma markers of systemic inflammation, which may play a role in disease progression.
Collapse
Affiliation(s)
- Angela Szeto
- Department of Psychology, University of Miami, PO Box 248185, Coral Gables, FL, 33124, USA
| | - Monia Cecati
- Department of Medicine and Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 N.W. 10th Avenue, Miami, FL, 33136, USA
| | - Raisa Ahmed
- Department of Psychology, University of Miami, PO Box 248185, Coral Gables, FL, 33124, USA
| | - Philip M McCabe
- Department of Psychology, University of Miami, PO Box 248185, Coral Gables, FL, 33124, USA
| | - Armando J Mendez
- Department of Medicine and Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 N.W. 10th Avenue, Miami, FL, 33136, USA.
| |
Collapse
|
67
|
Boff D, Oliveira VLS, Queiroz Junior CM, Galvão I, Batista NV, Gouwy M, Menezes GB, Cunha TM, Verri Junior WA, Proost P, Teixeira MM, Amaral FA. Lipoxin A 4 impairs effective bacterial control and potentiates joint inflammation and damage caused by Staphylococcus aureus infection. FASEB J 2020; 34:11498-11510. [PMID: 32741032 DOI: 10.1096/fj.201802830rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 05/12/2020] [Accepted: 05/15/2020] [Indexed: 02/07/2023]
Abstract
Staphylococcus aureus is the main cause of septic arthritis in humans, a disease associated with high morbidity and mortality. Inflammation triggered in response to infection is fundamental to control bacterial growth but may cause permanent tissue damage. Here, we evaluated the role of Lipoxin A4 (LXA4 ) in S aureus-induced arthritis in mice. Septic arthritis was induced by S aureus injection into tibiofemoral joints. At different time points, we evaluated cell recruitment and bacterial load in the joint, the production of pro-inflammatory molecules, and LXA4 in inflamed tissue and analyzed joint damage and dysfunction. LXA4 was investigated using genetically modified mice or by pharmacological blockade of its synthesis and receptor. CD11c+ cells were evaluated in lymph nodes by confocal microscopy and flow cytometry and dendritic cell chemotaxis using the Boyden chamber. Absence or pharmacological blockade of 5-lipoxygenase (5-LO) reduced joint inflammation and dysfunction and was associated with better control of infection at 4 to 7 days after the infection. There was an increase in LXA4 in joints of S aureus-infected mice and administration of LXA4 reversed the phenotype in 5-LO-/- mice. Blockade or absence of the LXA4 receptor FPR2 has a phenotype similar to 5-LO-/- mice. Mechanistically, LXA4 appeared to control migration and function of dendritic cells, cells shown to be crucial for adequate protective responses in the model. Thus, after the first days of infection when symptoms become evident therapies that inhibit LXA4 synthesis or action could be useful for treatment of S aureus-induced arthritis.
Collapse
Affiliation(s)
- Daiane Boff
- Immunopharmacology, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Vivian Louise Soares Oliveira
- Immunopharmacology, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Celso M Queiroz Junior
- Department of Morphology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Izabela Galvão
- Immunopharmacology, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Nathalia Vieira Batista
- Immunopharmacology, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mieke Gouwy
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Gustavo Batista Menezes
- Departamento de Morfologia, Centro de Biologia Gastrointestinal, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Thiago Mattar Cunha
- Department of Pharmacology, Faculdade de Medicina de Ribeirao Preto, Center for Research in Inflammatory Diseases, Universidade de São Paulo, São Paulo, Brazil
| | | | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Mauro Martins Teixeira
- Immunopharmacology, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Flávio Almeida Amaral
- Immunopharmacology, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
68
|
Clinical Value of Serum Amyloid-A Protein, High-density Lipoprotein Cholesterol and Apolipoprotein-A1 in the Diagnosis and Follow-up of Neonatal Sepsis. Pediatr Infect Dis J 2020; 39:749-755. [PMID: 32251257 DOI: 10.1097/inf.0000000000002682] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND To evaluate the performance of serum amyloid-A (SAA), high-density lipoprotein cholesterol (HDL-C) and apolipoprotein-A1 (Apo-A1) levels in the identification and monitoring of neonatal sepsis. METHODS This prospective study included 113 full-term septic neonates (postnatal age 4-28 days) admitted to the Special Care Neonatal Unit of a University Hospital from January 1, 2016, to April 30, 2019, and 68 healthy neonates (controls). Blood samples were drawn serially in septic neonates at enrollment and on days 1, 3 and 7, and once in controls, for SAA, HDL-C and Apo-A1 determination. RESULTS At enrollment, SAA levels were significantly higher in septic neonates in comparison with controls (median 50.7 vs. 3.5 mg/L; P < 0.0001); HDL-C and Apo-A1 levels were significantly lower in patients than in controls (P < 0.001 and P < 0.006, respectively). SAA levels were higher in culture-positive compared with culture-negative sepsis (median 202.0 vs. 14.2 mg/L; P < 0.0001). HDL-C and Apo-A1 levels did not differ significantly between culture-positive and culture-negative sepsis. Receiver operating characteristic curve analysis of SAA levels at enrollment resulted in significant areas under the curve (AUC) for detecting sepsis {AUC = 0.929 [95% confidence interval: 0.885-0.973]; P < 0.0001} and also for discriminating between culture-positive and culture-negative sepsis [AUC = 0.933 (95% confidence interval: 0.882-0.984); P < 0.0001]. The combination of HDL-C and Apo-A1 with SAA increased its diagnostic performance. Furthermore, serial SAA levels following enrollment could indicate clinical response in septic neonates. CONCLUSIONS SAA seems to be a useful biomarker for identification and monitoring of neonatal sepsis, and also for discriminating between culture-positive and culture-negative sepsis. HDL-C and Apo-A1 could be used as complementary markers.
Collapse
|
69
|
Chait A, den Hartigh LJ, Wang S, Goodspeed L, Babenko I, Altemeier WA, Vaisar T. Presence of serum amyloid A3 in mouse plasma is dependent on the nature and extent of the inflammatory stimulus. Sci Rep 2020; 10:10397. [PMID: 32587356 PMCID: PMC7316782 DOI: 10.1038/s41598-020-66898-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 05/26/2020] [Indexed: 11/09/2022] Open
Abstract
Serum amyloid A3 (Saa3) derives mainly from extrahepatic tissue and is not detected in plasma from moderately inflamed obese mice. In contrast, it is present in plasma from mice acutely inflamed by injection of high dose of lipopolysaccharide (LPS). To reconcile these differences, we evaluated whether different acute inflammatory stimuli could affect the presence of Saa3 in plasma. Saa3 appeared dose dependently in plasma after LPS injection. In contrast, only very low levels were detected after sterile inflammation with silver nitrate despite levels of Saa1 and Saa2 being comparable to high dose LPS. Saa3 was not detected in plasma following casein administration. Although most Saa3 was found in HDL, a small amount was not lipoprotein associated. Gene expression and proteomic analysis of liver and adipose tissue suggested that a major source of Saa3 in plasma after injection of LPS was adipose tissue rather than liver. We conclude that Saa3 only appears in plasma after induction of acute inflammation by some but not all inflammatory stimuli. These findings are consistent with the observation that Saa3 is not detectable in plasma in more moderate chronic inflammatory states such as obesity.
Collapse
Affiliation(s)
- Alan Chait
- Divisions of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle, WA, 98109, USA.
| | - Laura J den Hartigh
- Divisions of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle, WA, 98109, USA
| | - Shari Wang
- Divisions of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle, WA, 98109, USA
| | - Leela Goodspeed
- Divisions of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle, WA, 98109, USA
| | - Ilona Babenko
- Divisions of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle, WA, 98109, USA
| | - William A Altemeier
- Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, WA, 98109, USA
| | - Tomas Vaisar
- Divisions of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle, WA, 98109, USA
| |
Collapse
|
70
|
Lin X, Zhang H, Boyce BF, Xing L. Ubiquitination of interleukin-1α is associated with increased pro-inflammatory polarization of murine macrophages deficient in the E3 ligase ITCH. J Biol Chem 2020; 295:11764-11775. [PMID: 32587089 DOI: 10.1074/jbc.ra120.014298] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/21/2020] [Indexed: 01/02/2023] Open
Abstract
Macrophages play critical roles in homeostasis and inflammation. Macrophage polarization to either a pro-inflammatory or anti-inflammatory status is controlled by activating inflammatory signaling pathways. Ubiquitination is a posttranslational modification that regulates these inflammatory signaling pathways. However, the influence of protein ubiquitination on macrophage polarization has not been well studied. We hypothesized that the ubiquitination status of key proteins in inflammatory pathways contributes to macrophage polarization, which is regulated by itchy E3 ubiquitin ligase (ITCH), a negative regulator of inflammation. Using ubiquitin proteomics, we found that ubiquitination profiles are different among polarized murine macrophage subsets. Interestingly, interleukin-1α (IL-1α), an important pro-inflammatory mediator, was specifically ubiquitinated in lipopolysaccharide-induced pro-inflammatory macrophages, which was enhanced in ITCH-deficient macrophages. The ITCH-deficient macrophages had increased levels of the mature form of IL-1α and exhibited pro-inflammatory polarization, and reduced deubiquitination of IL-1α protein. Finally, IL-1α neutralization attenuated pro-inflammatory polarization of the ITCH-deficient macrophages. In conclusion, ubiquitination of IL-1α is associated with increased pro-inflammatory polarization of macrophages deficient in the E3 ligase ITCH.
Collapse
Affiliation(s)
- Xi Lin
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Hengwei Zhang
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Brendan F Boyce
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA.,Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Lianping Xing
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA .,Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
71
|
Abstract
From a clinical perspective, there is a need for a reliable and comprehensive list of diseases causing AA amyloidosis. This list could guide clinicians in the evaluation of patients with AA amyloidosis in whom an obvious cause is lacking. In this systematic review, a PubMed, Embase and Web of Science literature search were performed on causes of AA amyloidosis published in the last four decades. Initially, 4066 unique titles were identified, but only 795 full-text articles and letters were finally selected for analysis. Titles were excluded because of non-AA type of amyloidosis, language, no full-text publication or irrelevance. Hundred and fifty diseases were initially reported to be associated with the development of AA amyloidosis. The presence of AA amyloid was proven in 208 articles (26% of all) of which 140 (67%) showed a strong association with an underlying disease process. Disease associations were categorized and 48 were listed as strong, 19 as weak, 23 as unclear, and 60 as unlikely. Most newly described diseases are not really unexpected because they often cause longstanding inflammation. Based on the spectrum of identified causes, a pragmatic diagnostic approach is proposed for the AA amyloidosis patient in whom an obvious underlying disease is lacking.
Collapse
Affiliation(s)
- Anne Floor Brunger
- Departments of Rheumatology & Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.,Amyloidosis Center of Expertise, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Hans L A Nienhuis
- Amyloidosis Center of Expertise, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.,Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Johan Bijzet
- Departments of Rheumatology & Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.,Amyloidosis Center of Expertise, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Bouke P C Hazenberg
- Departments of Rheumatology & Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.,Amyloidosis Center of Expertise, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
72
|
Kuret T, Sodin-Šemrl S, Mrak-Poljšak K, Čučnik S, Lakota K, Erman A. Interleukin-1β Induces Intracellular Serum Amyloid A1 Expression in Human Coronary Artery Endothelial Cells and Promotes its Intercellular Exchange. Inflammation 2020; 42:1413-1425. [PMID: 31011929 DOI: 10.1007/s10753-019-01003-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Serum amyloid A (SAA) is an acute-phase protein with important, pathogenic role in the development of atherosclerosis. Since dysfunctional endothelium represents a key early step in atherogenesis, we aimed to determine whether induced human coronary artery endothelial cells (HCAEC) modulate SAA1/2/4 expression and influence intracellular location and intercellular transport of SAA1. HCAEC were stimulated with 1 ng/ml IL-1β, 10 ng/ml IL-6, and/or 1 μM dexamethasone for 24 h. QPCR, Western blots, ELISA, and immunofluorescent labeling were performed for detection of SAA1/2/4 mRNA and protein levels, respectively. In SAA1 transport experiments, FITC- or Cy3-labeled SAA1 were added to HCAEC separately, for 24 h, followed by a combined incubation of SAA1-FITC and SAA1-Cy3 positive cells, with IL-1β and analysis by flow cytometry. IL-1β upregulated SAA1 (119.9-fold, p < 0.01) and SAA2 (9.3-fold; p < 0.05) mRNA expression levels, while mRNA expression of SAA4 was not affected. Intracellular SAA1 was found mainly as a monomer, while SAA2 and SAA4 formed octamers as analyzed by Western blots. Within HCAEC, SAA1/2/4 located mostly to the perinuclear area and tunneling membrane nanotubes. Co-culturing of SAA1-FITC and SAA1-Cy3 positive cells for 48 h showed a significantly higher percentage of double positive cells in IL-1β-stimulated (mean ± SD; 60 ± 4%) vs. non-stimulated cells (48 ± 2%; p < 0.05). IL-1β induces SAA1 expression in HCAEC and promotes its intercellular exchange, suggesting that direct communication between cells in inflammatory conditions could ultimately lead to faster development of atherosclerosis in coronary arteries.
Collapse
Affiliation(s)
- Tadeja Kuret
- Department of Rheumatology, University Medical Centre Ljubljana, Vodnikova 62, SI-1000, Ljubljana, Slovenia.
- Faculty of Pharmacy, Chair of Clinical Biochemistry, University of Ljubljana, Aškerčeva 7, SI-1000, Ljubljana, Slovenia.
| | - Snežna Sodin-Šemrl
- Department of Rheumatology, University Medical Centre Ljubljana, Vodnikova 62, SI-1000, Ljubljana, Slovenia
- Faculty of Mathematics, Natural Sciences and Information Technologies, University of Primorska, Glagoljaška 8, SI-6000, Koper, Slovenia
| | - Katjuša Mrak-Poljšak
- Department of Rheumatology, University Medical Centre Ljubljana, Vodnikova 62, SI-1000, Ljubljana, Slovenia
| | - Saša Čučnik
- Department of Rheumatology, University Medical Centre Ljubljana, Vodnikova 62, SI-1000, Ljubljana, Slovenia
- Faculty of Pharmacy, Chair of Clinical Biochemistry, University of Ljubljana, Aškerčeva 7, SI-1000, Ljubljana, Slovenia
| | - Katja Lakota
- Department of Rheumatology, University Medical Centre Ljubljana, Vodnikova 62, SI-1000, Ljubljana, Slovenia
- Faculty of Mathematics, Natural Sciences and Information Technologies, University of Primorska, Glagoljaška 8, SI-6000, Koper, Slovenia
| | - Andreja Erman
- Faculty of Medicine, Institute of Cell Biology, University of Ljubljana, Vrazov trg 2, SI-1000, Ljubljana, Slovenia
| |
Collapse
|
73
|
|
74
|
Abstract
As normal constituents of blood serum, the Serum Amyloid A (SAA) proteins are small (104 amino acids in humans) and remarkably well-conserved in mammalian evolution. They are synthesized prominently, but not exclusively, in the liver. Fragments of SAA can associate into insoluble fibrils (called "amyloid") characteristic of "secondary" amyloid disease in which they can interrupt normal physiology and lead to organ failure. SAA proteins comprise a family of molecules, two members of which (SAA1 and SAA2) are (along with C-reactive protein, CRP) the most prominent members of the acute phase response (APR) during which their serum levels rise dramatically after trauma, infection and other stimuli. Biologic function (s) of SAA are unresolved but features are consistent with a prominent role in primordial host defense (including the APR ). SAA proteins are lipophilic and contribute to high density lipoproteins (HDL) and cholesterol transport. SAA proteins interact with specific receptors and have been implicated in tissue remodeling through metalloproteinases, local tissue changes in atherosclerosis, cancer metastasis, lung inflammation, maternal-fetal health and intestinal physiology. Molecular details of some of these are emerging.
Collapse
Affiliation(s)
- George H Sack
- Departments of Biological Chemistry and Medicine, The Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Physiology 615, Baltimore, MD, 21205, USA.
| |
Collapse
|
75
|
Wei Y, Wang S, Wang D, Liu C. Expression and clinical significance of serum amyloid A and interleukin-6 in patients with acute exacerbation of chronic obstructive pulmonary disease. Exp Ther Med 2019; 19:2089-2094. [PMID: 32104270 DOI: 10.3892/etm.2019.8366] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 11/14/2019] [Indexed: 02/07/2023] Open
Abstract
Expression and clinical significance of serum amyloid A (SAA) and interleukin-6 (IL-6) in patients with acute exacerbation of chronic obstructive pulmonary disease (AECOPD) were investigated. Fifty-one patients with AECOPD (acute group, AG) and 51 patients with stable chronic obstructive pulmonary disease (stable group, SG) admitted to Qingdao Eighth People's Hospital were selected. The expression levels of SAA and IL-6 in peripheral blood of patients in the two groups were detected and compared before and after treatment. Pearson analysis was used in the correlation between SAA and IL-6 and Receiver operating characteristic (ROC) curve was employd to analyze the predictive value of SAA and IL-6 for AECOPD progression. Logistic regression analysis was used to analyze the risk factors of AECOPD patients. The expression levels of SAA and IL-6 of patients in AG were significantly higher than those in SG (P<0.05). Pearson analysis showed that SAA was positively correlated with IL-6 expression (P<0.05). ROC curve analysis showed that AUC predicted by SAA and IL-6 for AECOPD progress was 0.789 and 0.762 (P<0.05). Logistic regression analysis showed that SAA and IL-6 were prediction indexes of AECOPD progression. The levels of SAA and IL-6 were significantly increased during AECOPD and effectively predicted the progress of AECOPD and is a risk factor affecting AECOPD patients.
Collapse
Affiliation(s)
- Yongtao Wei
- Clinical Laboratory, Qingdao Eighth People's Hospital, Qingdao, Shandong 266100, P.R. China
| | - Songxia Wang
- Blood Transfusion Department, Qingdao Eighth People's Hospital, Qingdao, Shandong 266100, P.R. China
| | - Dongping Wang
- Clinical Laboratory, Qingdao Eighth People's Hospital, Qingdao, Shandong 266100, P.R. China
| | - Cheng Liu
- Clinical Laboratory, Qingdao Eighth People's Hospital, Qingdao, Shandong 266100, P.R. China
| |
Collapse
|
76
|
Fan Y, Zhang G, Vong CT, Ye RD. Serum amyloid A3 confers protection against acute lung injury in Pseudomonas aeruginosa-infected mice. Am J Physiol Lung Cell Mol Physiol 2019; 318:L314-L322. [PMID: 31851532 DOI: 10.1152/ajplung.00309.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Pseudomonas aeruginosa is a gram-negative bacterium associated with serious illnesses, including ventilator-associated pneumonia and various sepsis syndromes in humans. Understanding the host immune mechanisms against P. aeruginosa is, therefore, of clinical importance. The present study identified serum amyloid A3 (SAA3) as being highly inducible in mouse bronchial epithelium following P. aeruginosa infection. Genetic deletion of Saa3 rendered mice more susceptible to P. aeruginosa infection with decreased neutrophil superoxide anion production, and ex vivo treatment of mouse neutrophils with recombinant SAA3 restored the ability of neutrophils to produce superoxide anions. The SAA3-deficient mice showed exacerbated inflammatory responses, which was characterized by pronounced neutrophil infiltration, elevated expression of TNF-α, KC/CXCL1, and MIP-2/CXCL2 in bronchoalveolar lavage fluid (BALF), and increased lung microvascular permeability compared with their wild-type littermates. BALF neutrophils from Saa3 knockout mice exhibited reduced superoxide anion production compared with neutrophils from wild-type mice. Adoptive transfer of SAA3-treated neutrophils to Saa3 knockout mice ameliorated P. aeruginosa-induced acute lung injury. These findings demonstrate that SAA3 not only serves as a biomarker for infection and inflammation, but also plays a protective role against P. aeruginosa infection-induced lung injury in part through augmentation of neutrophil bactericidal functions.
Collapse
Affiliation(s)
- Yu Fan
- State Key Laboratory for Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau Special Administrative Region, China
| | - Gufang Zhang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Chi Teng Vong
- State Key Laboratory for Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau Special Administrative Region, China
| | - Richard D Ye
- State Key Laboratory for Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau Special Administrative Region, China.,School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, China
| |
Collapse
|
77
|
VAN Roekel EH, Dugué PA, Jung CH, Joo JE, Makalic E, Wong EEM, English DR, Southey MC, Giles GG, Lynch BM, Milne RL. Physical Activity, Television Viewing Time, and DNA Methylation in Peripheral Blood. Med Sci Sports Exerc 2019; 51:490-498. [PMID: 30376510 DOI: 10.1249/mss.0000000000001827] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Physical activity may affect health via DNA methylation. The epigenetic influences of sedentary behaviors such as television viewing are unknown. We performed a genomewide study of DNA methylation in peripheral blood in relation to physical activity and television viewing time. METHODS DNA methylation was measured using the Illumina Infinium HumanMethylation450K BeadChip array in blood samples collected at baseline (N = 5513) and follow-up (N = 1249) from participants in the Melbourne Collaborative Cohort Study. At baseline, times per week of leisure-time physical activity were self-reported. At follow-up, the International Physical Activity Questionnaire was used to assess MET-hours per week of total and leisure-time physical activity and hours per day of television viewing time. Linear mixed models were used to assess associations between physical activity and television viewing measures and DNA methylation at individual CpG sites, adjusted for potential confounders and batch effects. RESULTS At follow-up, total physical activity was associated with DNA methylation at cg10266336 (P = 6.0 × 10), annotated to the SAA2 gene. Weaker evidence of associations (P < 1.0 × 10) were observed for an additional 14 CpG sites with total physical activity, for 7 CpG sites with leisure-time physical activity, and for 9 CpG sites with television viewing time. Changes in leisure-time physical activity between baseline and follow-up were associated with methylation changes (P < 0.05) at four of the seven CpG sites with weaker evidence of cross-sectional associations with leisure-time physical activity. CONCLUSION Physical activity and television viewing may be associated with blood DNA methylation, a potential pathway to chronic disease development. Further research using accelerometer data and larger sample sizes is warranted.
Collapse
Affiliation(s)
- Eline H VAN Roekel
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, AUSTRALIA.,Department of Epidemiology, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, THE NETHERLANDS
| | - Pierre-Antoine Dugué
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, AUSTRALIA.,Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, AUSTRALIA
| | - Chol-Hee Jung
- Melbourne Bioinformatics, The University of Melbourne, Parkville, AUSTRALIA
| | - Jihoon E Joo
- Genetic Epidemiology Laboratory, Department of Clinical Pathology, The University of Melbourne, Parkville, AUSTRALIA
| | - Enes Makalic
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, AUSTRALIA
| | - E E Ming Wong
- Genetic Epidemiology Laboratory, Department of Clinical Pathology, The University of Melbourne, Parkville, AUSTRALIA.,Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, AUSTRALIA
| | - Dallas R English
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, AUSTRALIA.,Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, AUSTRALIA
| | - Melissa C Southey
- Genetic Epidemiology Laboratory, Department of Clinical Pathology, The University of Melbourne, Parkville, AUSTRALIA.,Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, AUSTRALIA
| | - Graham G Giles
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, AUSTRALIA.,Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, AUSTRALIA
| | - Brigid M Lynch
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, AUSTRALIA.,Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, AUSTRALIA.,Physical Activity Laboratory, Baker Heart and Diabetes Institute, Melbourne, AUSTRALIA
| | - Roger L Milne
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, AUSTRALIA.,Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, AUSTRALIA
| |
Collapse
|
78
|
Bioassay for Endothelial Damage Mediators Retrieved by Hemoadsorption. Sci Rep 2019; 9:14522. [PMID: 31601835 PMCID: PMC6787199 DOI: 10.1038/s41598-019-50517-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 09/05/2019] [Indexed: 12/13/2022] Open
Abstract
Hemoadsorption devices are used to treat septic shock by adsorbing inflammatory cytokines and as yet incompletely defined danger and pathogen associated molecular patterns. In an ideal case, hemoadsorption results in immediate recovery of microvascular endothelial cells’ (mEC) function and rapid recovery from catecholamine-dependency and septic shock. We here tested a single device, which consists of polystyrene-divinylbenzene core particles of 450 μm diameter with a high affinity for hydrophobic compounds. The current study aimed at the proof of concept that endothelial-specific damage mediators are adsorbed and can be recovered from hemoadsorption devices. Because of excellent clinical experience, we tested protein fractions released from a hemoadsorber in a novel endothelial bioassay. Video-based, long-term imaging of mEC proliferation and cell death were evaluated and combined with apoptosis and ATP measurements. Out of a total of 39 fractions recovered from column fractionation, we identified 3 fractions that caused i) inhibition of mEC proliferation, ii) increased cell death and iii) induction of apoptosis in mEC. When adding these 3 fractions to mEC, their ATP contents were reduced. These fractions contained proteins of approximately 15 kDa, and high amounts of nucleic acid, which was at least in part oxidized. The efficacy for endothelial cell damage prevention by hemoadsorption can be addressed by a novel endothelial bioassay and long-term video observation procedures. Protein fractionation of the hemoadsorption devices used is feasible to study and define endothelial damage ligands on a molecular level. The results suggest a significant effect by circulating nucleic acids – bound to an as yet undefined protein, which may constitute a major danger-associated molecular pattern (DAMP) in the exacerbation of inflammation when patients experience septic shock. Hemoadsorption devices may thus limit endothelial damage, through the binding of nucleic acid-bearing aggregates and thus contribute to improved endothelial barrier function.
Collapse
|
79
|
Deshayes S, Fellahi S, Bastard JP, Launay JM, Callebert J, Fraisse T, Buob D, Boffa JJ, Giurgea I, Dupont C, Jegou S, Straube M, Karras A, Aouba A, Grateau G, Sokol H, Georgin-Lavialle S. Specific changes in faecal microbiota are associated with familial Mediterranean fever. Ann Rheum Dis 2019; 78:1398-1404. [PMID: 31377728 DOI: 10.1136/annrheumdis-2019-215258] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 07/13/2019] [Accepted: 07/18/2019] [Indexed: 12/27/2022]
Abstract
OBJECTIVES Familial Mediterranean fever (FMF) can be complicated by AA amyloidosis (AAA), though it remains unclear why only some patients develop amyloidosis. We examined the gut microbiota composition and inflammatory markers in patients with FMF complicated or not by AAA. METHODS We analysed the gut microbiota of 34 patients with FMF without AAA, 7 patients with FMF with AAA, 19 patients with AAA of another origin, and 26 controls using 16S ribosomal RNA gene sequencing with the Illumina MiSeq platform. Associations between bacterial taxa and clinical phenotypes were evaluated using multivariate association with linear models statistical method. Blood levels of interleukin (IL)-1β, IL-6, tumour necrosis factor-α and adipokines were assessed by ELISA; indoleamine 2,3-dioxygenase (IDO) activity was determined by high-performance liquid chromatography. RESULTS Compared with healthy subjects, specific changes in faecal microbiota were observed in FMF and AAA groups. Several operational taxonomic units (OTUs) were associated with FMF. Moreover, two OTUs were over-represented in FMF-related AAA compared with FMF without AAA. Additionally, higher adiponectin levels and IDO activity were observed in FMF-related AAA compared with FMF without AAA (p<0.05). CONCLUSION The presence of specific changes in faecal microbiota in FMF and in FMF-related AAA suggests that intestinal microorganisms may play a role in the pathogenesis of these diseases. These findings may offer an opportunity to use techniques for gut microbiota manipulation.
Collapse
Affiliation(s)
- Samuel Deshayes
- Service de Médecine Interne, Normandie Univ, UNICAEN, CHU de Caen Normandie, 14000 Caen, France
- Service de Médecine Interne, Centre de référence des maladies auto-inflammatoires et des amyloses inflammatoires (CEREMAIA), Sorbonne Université, Assistance Publique des Hôpitaux de Paris, Hôpital Tenon, Paris, France
- Service de Gastroentérologie, Centre de Recherche Saint-Antoine, CRSA, Sorbonne Université, Inserm, AP-HP, Hôpital Saint-Antoine, F-75012 Paris, France
| | - Soraya Fellahi
- UF Biomarqueurs Inflammatoires et Métaboliques, Service de Biochimie, Assistance Publique des Hôpitaux de Paris, Hôpital Tenon, Paris, France
- Centre de Recherche Saint-Antoine, IHU ICAN, Sorbonne Universités, UPMC Université Paris 06, INSERM UMRS 938, Paris, France
| | - Jean-Philippe Bastard
- UF Biomarqueurs Inflammatoires et Métaboliques, Service de Biochimie, Assistance Publique des Hôpitaux de Paris, Hôpital Tenon, Paris, France
- Centre de Recherche Saint-Antoine, IHU ICAN, Sorbonne Universités, UPMC Université Paris 06, INSERM UMRS 938, Paris, France
| | - Jean-Marie Launay
- Service de Biochimie, INSERM UMR S942, Assistance Publique des Hôpitaux de Paris, Hôpital Lariboisière, Paris, France
| | - Jacques Callebert
- Service de Biochimie, INSERM UMR S942, Assistance Publique des Hôpitaux de Paris, Hôpital Lariboisière, Paris, France
| | - Thibault Fraisse
- Service de Médecine Interne, Centre de référence des maladies auto-inflammatoires et des amyloses inflammatoires (CEREMAIA), Sorbonne Université, Assistance Publique des Hôpitaux de Paris, Hôpital Tenon, Paris, France
| | - David Buob
- Service d'Anatomopathologie, Assistance Publique des Hôpitaux de Paris, Hôpital Tenon, Paris, France
| | - Jean-Jacques Boffa
- INSERM 1155, Sorbonne Université, AP-HP, Hôpital Tenon, F-75020 Paris, France
| | - Irina Giurgea
- Service de Génétique Médicale, Assistance Publique des Hôpitaux de Paris, Hôpital Trousseau, Paris, France
| | - Charlotte Dupont
- INSERM équipe Lipodystrophies génétiques et acquises. Service de biologiede la reproduction-CECOS, Sorbonne Université, Saint Antoine Research Center, AP-HP, Hôpital Tenon, F-75020 Paris, France
| | - Sarah Jegou
- Service de Gastroentérologie, Centre de Recherche Saint-Antoine, CRSA, Sorbonne Université, Inserm, AP-HP, Hôpital Saint-Antoine, F-75012 Paris, France
| | - Marjolène Straube
- Service de Gastroentérologie, Centre de Recherche Saint-Antoine, CRSA, Sorbonne Université, Inserm, AP-HP, Hôpital Saint-Antoine, F-75012 Paris, France
| | - Alexandre Karras
- Service de Néphrologie, Assistance Publique des Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France
| | - Achille Aouba
- Service de Médecine Interne, Normandie Univ, UNICAEN, CHU de Caen Normandie, 14000 Caen, France
| | - Gilles Grateau
- Service de Médecine Interne, Centre de référence des maladies auto-inflammatoires et des amyloses inflammatoires (CEREMAIA), Sorbonne Université, Assistance Publique des Hôpitaux de Paris, Hôpital Tenon, Paris, France
| | - Harry Sokol
- Service de Gastroentérologie, Centre de Recherche Saint-Antoine, CRSA, Sorbonne Université, Inserm, AP-HP, Hôpital Saint-Antoine, F-75012 Paris, France
- MICALIS Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
- Service de Gastroentérologie, Assistance Publique des Hôpitaux de Paris, Hôpital Saint-Antoine, Paris, France
| | - Sophie Georgin-Lavialle
- Service de Médecine Interne, Centre de référence des maladies auto-inflammatoires et des amyloses inflammatoires (CEREMAIA), Sorbonne Université, Assistance Publique des Hôpitaux de Paris, Hôpital Tenon, Paris, France
| |
Collapse
|
80
|
Randeria SN, Thomson GJA, Nell TA, Roberts T, Pretorius E. Inflammatory cytokines in type 2 diabetes mellitus as facilitators of hypercoagulation and abnormal clot formation. Cardiovasc Diabetol 2019; 18:72. [PMID: 31164120 PMCID: PMC6549308 DOI: 10.1186/s12933-019-0870-9] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 05/16/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The global burden of type 2 diabetes mellitus (T2DM), together with the presence of cardiovascular risk in this population, is reaching pandemic levels. A prominent feature of T2DM is chronic and systemic inflammation, with the accompanying presence of circulating and dysregulated inflammatory biomarkers; which in turn is associated with abnormal clot formation. METHODS Here, we investigate the correlation between abnormal blood clotting, using thromboelastography (TEG), clot ultrastructure using scanning electron microscopy (SEM) and the presence of a dysregulated inflammatory cytokine profile, by examining various circulating biomarkers. RESULTS Our results show that many biomarkers, across TEG, cytokine and lipid groups, were greatly dysregulated in the T2DM sample. Furthermore, our T2DM sample's coagulation profiles were significantly more hypercoagulable when compared to our heathy sample, and ultrastructural analysis confirmed a matted and denser clot structure in the T2DM sample. CONCLUSIONS We suggest that dysregulated circulating molecules may in part be responsible for a hypercoagulable state and vascular dysfunction in the T2DM sample. We propose further that a personalized approach could be of great value when planning treatment and tracking the patient health status after embarking on a treatment regimes, and that looking to novel inflammatory and vascular biomarkers might be crucial.
Collapse
Affiliation(s)
- Shehan N Randeria
- Department of Physiological Sciences, Stellenbosch University, Private Bag X1, Matieland, 7602, South Africa
| | - Greig J A Thomson
- Department of Physiological Sciences, Stellenbosch University, Private Bag X1, Matieland, 7602, South Africa
| | - Theo A Nell
- Department of Physiological Sciences, Stellenbosch University, Private Bag X1, Matieland, 7602, South Africa
| | - Timothy Roberts
- Department of Physiological Sciences, Stellenbosch University, Private Bag X1, Matieland, 7602, South Africa.
- Department of Biochemistry, Institute of Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St, Liverpool, L69 7ZB, UK.
| | - Etheresia Pretorius
- Department of Physiological Sciences, Stellenbosch University, Private Bag X1, Matieland, 7602, South Africa.
| |
Collapse
|
81
|
Evidence for the important role of inflammation in xenotransplantation. JOURNAL OF INFLAMMATION-LONDON 2019; 16:10. [PMID: 31148951 PMCID: PMC6537172 DOI: 10.1186/s12950-019-0213-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 05/02/2019] [Indexed: 12/17/2022]
Abstract
There is increasing evidence of a sustained state of systemic inflammation after pig-to-nonhuman primate (NHP) xenotransplantation (that has been termed systemic inflammation in xenograft recipients [SIXR]). Increases in inflammatory markers, e.g., C-reactive protein, histones, serum amyloid A, D-dimer, cytokines, chemokines, and a decrease in free triiodothyronine, have been demonstrated in the recipient NHPs. The complex interactions between inflammation, coagulation, and the immune response are well-recognized, but the role of inflammation in xenograft recipients is not fully understood. The evidence suggests that inflammation can promote the activation of coagulation and the adaptive immune response, but the exact mechanisms remain uncertain. If prolonged xenograft survival is to be achieved, anti-inflammatory strategies (e.g., the administration of anti-inflammatory agents, and/or the generation of genetically-engineered organ-source pigs that are protected from the effect of inflammation) may be necessary to prevent, control, or negate the effect of the systemic inflammation that develops in xenograft recipients. This may allow for a reduction in the intensity of exogenous immunosuppressive therapy. If immunological tolerance to a xenograft is to be obtained, then control of inflammation may be essential.
Collapse
|
82
|
Jumeau C, Awad F, Assrawi E, Cobret L, Duquesnoy P, Giurgea I, Valeyre D, Grateau G, Amselem S, Bernaudin JF, Karabina SA. Expression of SAA1, SAA2 and SAA4 genes in human primary monocytes and monocyte-derived macrophages. PLoS One 2019; 14:e0217005. [PMID: 31100086 PMCID: PMC6524798 DOI: 10.1371/journal.pone.0217005] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Accepted: 05/02/2019] [Indexed: 12/28/2022] Open
Abstract
Circulating serum amyloid A (SAA) is increased in various inflammatory conditions. The human SAA protein family comprises the acute phase SAA1/SAA2, known to activate a large set of innate and adaptive immune cells, and the constitutive SAA4. The liver synthesis of SAA1/SAA2 is well-established but there is still an open debate on extrahepatic SAA expression especially in macrophages. We aimed to investigate the ability of human primary monocytes and monocyte-derived macrophages to express SAA1, SAA2 and SAA4 at both the transcriptional and protein levels, as previous studies almost exclusively dealt with monocytic cell lines. Monocytes and derived macrophages from healthy donors were stimulated under various conditions. In parallel with SAA, pro-inflammatory IL1A, IL1B and IL6 cytokine expression was assessed. While LPS alone was non-effective, a combined LPS/dexamethasone treatment induced SAA1 and to a lesser extent SAA2 transcription in human monocytes and macrophages. In contrast, as expected, pro-inflammatory cytokine expression was strongly induced following stimulation with LPS, an effect which was dampened in the presence of dexamethasone. Furthermore, in monocytes polarized towards a pro-inflammatory M1 phenotype, SAA expression in response to LPS/dexamethasone was potentiated; a result mainly seen for SAA1. However, a major discrepancy was observed between SAA mRNA and intracellular protein levels under the experimental conditions used. Our results demonstrate that human monocytes and macrophages can express SAA genes, mainly SAA1 in response to an inflammatory environment. While SAA is considered as a member of a large cytokine network, its expression in the monocytes-macrophages in response to LPS-dexamethasone is strikingly different from that observed for classic pro-inflammatory cytokines. As monocytes-macrophages are major players in chronic inflammatory diseases, it may be hypothesized that SAA production from macrophages may contribute to the local inflammatory microenvironment, especially when macrophages are compactly organized in granulomas as in sarcoidosis.
Collapse
Affiliation(s)
- Claire Jumeau
- Sorbonne Université, INSERM, UMR_S 933, Assistance Publique Hôpitaux de Paris, Hôpital Trousseau, Service de Génétique et d’Embryologie médicale, Paris, France
| | - Fawaz Awad
- Sorbonne Université, INSERM, UMR_S 933, Assistance Publique Hôpitaux de Paris, Hôpital Trousseau, Service de Génétique et d’Embryologie médicale, Paris, France
| | - Eman Assrawi
- Sorbonne Université, INSERM, UMR_S 933, Assistance Publique Hôpitaux de Paris, Hôpital Trousseau, Service de Génétique et d’Embryologie médicale, Paris, France
| | - Laetitia Cobret
- Sorbonne Université, INSERM, UMR_S 933, Assistance Publique Hôpitaux de Paris, Hôpital Trousseau, Service de Génétique et d’Embryologie médicale, Paris, France
| | - Philippe Duquesnoy
- Sorbonne Université, INSERM, UMR_S 933, Assistance Publique Hôpitaux de Paris, Hôpital Trousseau, Service de Génétique et d’Embryologie médicale, Paris, France
| | - Irina Giurgea
- Sorbonne Université, INSERM, UMR_S 933, Assistance Publique Hôpitaux de Paris, Hôpital Trousseau, Service de Génétique et d’Embryologie médicale, Paris, France
| | - Dominique Valeyre
- Assistance Publique Hôpitaux de Paris, Hôpital Avicenne, Service de Pneumologie, Bobigny, France
- Université Paris 13, INSERM UMR 1272, Laboratoire ‘Hypoxie & Poumon’, Bobigny, France
| | - Gilles Grateau
- Sorbonne Université, INSERM, UMR_S 933, Assistance Publique Hôpitaux de Paris, Hôpital Trousseau, Service de Génétique et d’Embryologie médicale, Paris, France
- Assistance Publique Hôpitaux de Paris, Hôpital Tenon, Service de médecine interne, Paris, France
| | - Serge Amselem
- Sorbonne Université, INSERM, UMR_S 933, Assistance Publique Hôpitaux de Paris, Hôpital Trousseau, Service de Génétique et d’Embryologie médicale, Paris, France
| | - Jean-François Bernaudin
- Sorbonne Université, INSERM, UMR_S 933, Assistance Publique Hôpitaux de Paris, Hôpital Trousseau, Service de Génétique et d’Embryologie médicale, Paris, France
- Assistance Publique Hôpitaux de Paris, Hôpital Avicenne, Service de Pneumologie, Bobigny, France
- Université Paris 13, INSERM UMR 1272, Laboratoire ‘Hypoxie & Poumon’, Bobigny, France
| | - Sonia-Athina Karabina
- Sorbonne Université, INSERM, UMR_S 933, Assistance Publique Hôpitaux de Paris, Hôpital Trousseau, Service de Génétique et d’Embryologie médicale, Paris, France
| |
Collapse
|
83
|
Lu Y, Wang WS, Lin YK, Lu JW, Li WJ, Zhang CY, Sun K. Enhancement of cortisol-induced SAA1 transcription by SAA1 in the human amnion. J Mol Endocrinol 2019; 62:149-158. [PMID: 30817315 DOI: 10.1530/jme-18-0263] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 02/22/2019] [Indexed: 12/15/2022]
Abstract
Our previous studies have demonstrated that human fetal membranes are capable of de novo synthesis of serum amyloid A1 (SAA1), an acute phase protein of inflammation, wherein SAA1 may participate in parturition by inducing a number of inflammation mediators including interleukine-1β, interleukine-6 and prostaglandin E2. However, the regulation of SAA1 expression in the fetal membranes remains largely unknown. In the current study, we examined the regulation of SAA1 expression by cortisol, a crucial steroid produced locally in the fetal membranes at parturition, and the interaction between cortisol and SAA1 in the feed-forward induction of SAA1 expression in human amnion fibroblasts. Results showed that cortisol-induced SAA1 expression in a concentration-dependent manner, which was greatly enhanced by SAA1 despite modest induction of SAA1 expression by itself. Mechanism studies revealed that the induction of SAA1 expression by cortisol and SAA1 was blocked by either the transcription factor STAT3 antagonist AZD0530 or siRNA-mediated knockdown of STAT3. Furthermore, cortisol- and SAA1-induced STAT3 phosphorylation in a sequential order with the induction by SAA1 preceding the induction by cortisol. However, combination of cortisol and SAA1 failed to further intensify the phosphorylation of STAT3. Consistently, cortisol and SAA1 increased the enrichment of STAT3 at the SAA1 promoter. Taking together, this study has demonstrated that cortisol and SAA1 can reinforce each other in the induction of SAA1 expression through sequential phosphorylation of STAT3. The enhancement of cortisol-induced SAA1 expression by SAA1 may lead to excessive SAA1 accumulation resulting in parturition-associated inflammation in the fetal membranes.
Collapse
Affiliation(s)
- Yi Lu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, People's Republic of China
| | - Wang-Sheng Wang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, People's Republic of China
| | - Yi-Kai Lin
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, People's Republic of China
| | - Jiang-Wen Lu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, People's Republic of China
| | - Wen-Jiao Li
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, People's Republic of China
| | - Chu-Yue Zhang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, People's Republic of China
| | - Kang Sun
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, People's Republic of China
| |
Collapse
|
84
|
Zhang Y, Zhang J, Sheng H, Li H, Wang R. Acute phase reactant serum amyloid A in inflammation and other diseases. Adv Clin Chem 2019; 90:25-80. [PMID: 31122611 DOI: 10.1016/bs.acc.2019.01.002] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Acute-phase reactant serum amyloid A (A-SAA) plays an important role in acute and chronic inflammation and is used in clinical laboratories as an indicator of inflammation. Although both A-SAA and C-reactive protein (CRP) are acute-phase proteins, the detection of A-SAA is more conclusive than the detection of CRP in patients with viral infections, severe acute pancreatitis, and rejection reactions to kidney transplants. A-SAA has greater clinical diagnostic value in patients who are immunosuppressed, patients with cystic fibrosis who are treated with corticoids, and preterm infants with late-onset sepsis. Nevertheless, for the assessment of the inflammation status and identification of viral infection in other pathologies, such as bacterial infections, the combinatorial use of A-SAA and other acute-phase proteins (APPs), such as CRP and procalcitonin (PCT), can provide more information and sensitivity than the use of any of these proteins alone, and the information generated is important in guiding antibiotic therapy. In addition, A-SAA-associated diseases and the diagnostic value of A-SAA are discussed. However, the relationship between different A-SAA isotypes and their human diseases are mostly derived from research laboratories with limited clinical samples. Thus, further clinical evaluations are necessary to confirm the clinical significance of each A-SAA isotype. Furthermore, the currently available A-SAA assays are based on polyclonal antibodies, which lack isotype specificity and are associated with many inflammatory diseases. Therefore, these assays are usually used in combination with other biomarkers in the clinic.
Collapse
Affiliation(s)
- Yan Zhang
- Shanghai R&D Center, DiaSys Diagnostic Systems (Shanghai) Co., Ltd., Shanghai, China
| | - Jie Zhang
- Shanghai R&D Center, DiaSys Diagnostic Systems (Shanghai) Co., Ltd., Shanghai, China
| | - Huiming Sheng
- Department of Laboratory Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haichuan Li
- C.N. Maternity & Infant Health Hospital, Shanghai, China
| | - Rongfang Wang
- Shanghai R&D Center, DiaSys Diagnostic Systems (Shanghai) Co., Ltd., Shanghai, China.
| |
Collapse
|
85
|
Kapurniotu A, Gokce O, Bernhagen J. The Multitasking Potential of Alarmins and Atypical Chemokines. Front Med (Lausanne) 2019; 6:3. [PMID: 30729111 PMCID: PMC6351468 DOI: 10.3389/fmed.2019.00003] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 01/04/2019] [Indexed: 12/19/2022] Open
Abstract
When the human genome was sequenced, it came as a surprise that it contains “only” 21,306 protein-coding genes. However, complexity and diversity are multiplied by alternative splicing, non-protein-coding transcripts, or post-translational modifications (PTMs) on proteome level. Here, we discuss how the multi-tasking potential of proteins can substantially enhance the complexity of the proteome further, while at the same time offering mechanisms for the fine-regulation of cell responses. Discoveries over the past two decades have led to the identification of “surprising” and previously unrecognized functionalities of long known cytokines, inflammatory mediators, and intracellular proteins that have established novel molecular networks in physiology, inflammation, and cardiovascular disease. In this mini-review, we focus on alarmins and atypical chemokines such as high-mobility group box protein-1 (HMGB-1) and macrophage migration-inhibitory factor (MIF)-type proteins that are prototypical examples of these classes, featuring a remarkable multitasking potential that allows for an elaborate fine-tuning of molecular networks in the extra- and intracellular space that may eventually give rise to novel “task”-based precision medicine intervention strategies.
Collapse
Affiliation(s)
- Aphrodite Kapurniotu
- Division of Peptide Biochemistry, Technische Universität München, Freising, Germany
| | - Ozgun Gokce
- System Neuroscience Laboratory, Institute for Stroke and Dementia Research, Klinikum der Universität München, Munich, Germany
| | - Jürgen Bernhagen
- Department of Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany.,Munich Heart Alliance, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
86
|
Pretorius L, Kell DB, Pretorius E. Iron Dysregulation and Dormant Microbes as Causative Agents for Impaired Blood Rheology and Pathological Clotting in Alzheimer's Type Dementia. Front Neurosci 2018; 12:851. [PMID: 30519157 PMCID: PMC6251002 DOI: 10.3389/fnins.2018.00851] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 10/30/2018] [Indexed: 12/13/2022] Open
Abstract
Alzheimer’s disease and other similar dementias are debilitating neurodegenerative disorders whose etiology and pathogenesis remain largely unknown, even after decades of research. With the anticipated increase in prevalence of Alzheimer’s type dementias among the more susceptible aging population, the need for disease-modifying treatments is urgent. While various hypotheses have been put forward over the last few decades, we suggest that Alzheimer’s type dementias are triggered by external environmental factors, co-expressing in individuals with specific genetic susceptibilities. These external stressors are defined in the Iron Dysregulation and Dormant Microbes (IDDM) hypothesis, previously put forward. This hypothesis is consistent with current literature in which serum ferritin levels of individuals diagnosed with Alzheimer’s disease are significantly higher compared those of age- and gender-matched controls. While iron dysregulation contributes to oxidative stress, it also causes microbial reactivation and virulence of the so-called dormant blood (and tissue) microbiome. Dysbiosis (changes in the microbiome) or previous infections can contribute to the dormant blood microbiome (atopobiosis1), and also directly promotes systemic inflammation via the amyloidogenic formation and shedding of potent inflammagens such as lipopolysaccharides. The simultaneous iron dysregulation and microbial aberrations affect the hematological system, promoting fibrin amylodiogenesis, and pathological clotting. Systemic inflammation and oxidative stress can contribute to blood brain barrier permeability and the ensuing neuro-inflammation, characteristic of Alzheimer’s type dementias. While large inter-individual variability exists, especially concerning disease pathogenesis, the IDDM hypothesis acknowledges primary causative factors which can be targeted for early diagnosis and/or for prevention of disease progression.
Collapse
Affiliation(s)
- Lesha Pretorius
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Douglas B Kell
- School of Chemistry, The University of Manchester, Manchester, United Kingdom.,The Manchester Institute of Biotechnology, The University of Manchester, Manchester, United Kingdom
| | - Etheresia Pretorius
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
87
|
Abstract
Secondary, AA, amyloidosis is a rare systemic complication that can develop in any long-term inflammatory disorder, and is characterized by the extracellular deposition of fibrils derived from serum amyloid A (SAA) protein. SAA is an acute-phase reactant synthetized largely by hepatocytes under the transcriptional regulation of proinflammatory cytokines. The kidney is the major involved organ with proteinuria as first clinical manifestation; renal biopsy is the commonest diagnostic investigation. Targeted anti-inflammatory treatment promotes normalization of circulating SAA levels preventing amyloid deposition and renal damage. Novel therapies aimed at promoting clearance of existing amyloid deposits soon may be an effective treatment approach.
Collapse
Affiliation(s)
- Riccardo Papa
- Autoinflammatory Diseases and Immunodeficiencies Centre, Pediatric and Rheumatology Clinic, Giannina Gaslini Institute, University of Genoa, Via Gerolamo Gaslini 5, Genova 16147, Italy.
| | - Helen J Lachmann
- National Amyloidosis Centre, Royal Free Campus, University College Medical School, Rowland Hill Street, London NW3 2PF, UK
| |
Collapse
|
88
|
Abstract
Serum amyloid A (SAA) proteins were isolated and named over 50 years ago. They are small (104 amino acids) and have a striking relationship to the acute phase response with serum levels rising as much as 1000-fold in 24 hours. SAA proteins are encoded in a family of closely-related genes and have been remarkably conserved throughout vertebrate evolution. Amino-terminal fragments of SAA can form highly organized, insoluble fibrils that accumulate in “secondary” amyloid disease. Despite their evolutionary preservation and dynamic synthesis pattern SAA proteins have lacked well-defined physiologic roles. However, considering an array of many, often unrelated, reports now permits a more coordinated perspective. Protein studies have elucidated basic SAA structure and fibril formation. Appreciating SAA’s lipophilicity helps relate it to lipid transport and metabolism as well as atherosclerosis. SAA’s function as a cytokine-like protein has become recognized in cell-cell communication as well as feedback in inflammatory, immunologic, neoplastic and protective pathways. SAA likely has a critical role in control and possibly propagation of the primordial acute phase response. Appreciating the many cellular and molecular interactions for SAA suggests possibilities for improved understanding of pathophysiology as well as treatment and disease prevention.
Collapse
Affiliation(s)
- George H Sack
- Departments of Biological Chemistry and Medicine, The Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Physiology 615, Baltimore, MD, 21205, USA.
| |
Collapse
|
89
|
Kell DB, Pretorius E. No effects without causes: the Iron Dysregulation and Dormant Microbes hypothesis for chronic, inflammatory diseases. Biol Rev Camb Philos Soc 2018; 93:1518-1557. [PMID: 29575574 PMCID: PMC6055827 DOI: 10.1111/brv.12407] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 02/12/2018] [Accepted: 02/15/2018] [Indexed: 12/11/2022]
Abstract
Since the successful conquest of many acute, communicable (infectious) diseases through the use of vaccines and antibiotics, the currently most prevalent diseases are chronic and progressive in nature, and are all accompanied by inflammation. These diseases include neurodegenerative (e.g. Alzheimer's, Parkinson's), vascular (e.g. atherosclerosis, pre-eclampsia, type 2 diabetes) and autoimmune (e.g. rheumatoid arthritis and multiple sclerosis) diseases that may appear to have little in common. In fact they all share significant features, in particular chronic inflammation and its attendant inflammatory cytokines. Such effects do not happen without underlying and initially 'external' causes, and it is of interest to seek these causes. Taking a systems approach, we argue that these causes include (i) stress-induced iron dysregulation, and (ii) its ability to awaken dormant, non-replicating microbes with which the host has become infected. Other external causes may be dietary. Such microbes are capable of shedding small, but functionally significant amounts of highly inflammagenic molecules such as lipopolysaccharide and lipoteichoic acid. Sequelae include significant coagulopathies, not least the recently discovered amyloidogenic clotting of blood, leading to cell death and the release of further inflammagens. The extensive evidence discussed here implies, as was found with ulcers, that almost all chronic, infectious diseases do in fact harbour a microbial component. What differs is simply the microbes and the anatomical location from and at which they exert damage. This analysis offers novel avenues for diagnosis and treatment.
Collapse
Affiliation(s)
- Douglas B. Kell
- School of ChemistryThe University of Manchester, 131 Princess StreetManchesterLancsM1 7DNU.K.
- The Manchester Institute of BiotechnologyThe University of Manchester, 131 Princess StreetManchesterLancsM1 7DNU.K.
- Department of Physiological SciencesStellenbosch University, Stellenbosch Private Bag X1Matieland7602South Africa
| | - Etheresia Pretorius
- Department of Physiological SciencesStellenbosch University, Stellenbosch Private Bag X1Matieland7602South Africa
| |
Collapse
|
90
|
Yu MH, Chen MH, Han F, Li Q, Sun RH, Tu YX. Prognostic value of the biomarkers serum amyloid A and nitric oxide in patients with sepsis. Int Immunopharmacol 2018; 62:287-292. [PMID: 30048858 DOI: 10.1016/j.intimp.2018.07.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 06/29/2018] [Accepted: 07/21/2018] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Sepsis is a major cause of mortality among critically ill patients in the intensive care unit (ICU). Alterations in serum amyloid A (SAA) and nitric oxide (NO) levels have been associated with mortality in critically ill patients. In the present study, we investigated the predictive value of SAA and/or NO compared to traditional predictive markers such as C-reactive protein (CRP) and Acute Physiology and Chronic Health Evaluation II (APACHE II) score. METHODS 100 adult patients with sepsis and 25 without sepsis were enrolled in a prospective, randomized study in our ICU. The APACHE II score was calculated, and their peripheral venous blood SAA, NO and CRP levels were evaluated on days 1, 3, and 7 after sepsis was diagnosed. The patients were sorted based on incidence of septic shock into septic shock (A) and non-septic shock (B) groups. Comparative analyses of altered levels of these indicators between the two groups were performed, and correlations between SAA, NO, and the more traditional APACHE II score were probed. Patients were sorted based on survival status into death (D) and survival (S) groups based on death endpoint within 28 days after admission. RESULTS We observed that the difference in APACHE II score, SAA and CRP levels were statistically significantly (p < 0.05) between groups A and B on days 1, 3 and 7 post-diagnosis, while inter-group NO level significantly differed (p < 0.05) on days 1 and 3 post-diagnosis, no apparent difference was observed on day 7 post-diagnosis. For groups D and S, SAA, CRP and NO levels significantly differed (p < 0.05) on days 3 and 7 post-diagnosis, with no apparent difference on day 1. APACHE II score was significantly different on day 7 (p < 0.05), however the difference on days 1 and 3 were non-significant. We also demonstrated a positive correlation between APACHE II scores, SAA levels on days 1, 3, and 7, as well as NO levels on days 1 and 3. In addition, for the D and S groups, SAA at all time points, NO on day 3 and CRP on day 7 positively correlated with increased death events. CONCLUSION The dynamic monitoring of SAA and NO serum levels with APACHE II scores better reflect the severity of sepsis than traditional indicators like CRP and may serve as independent prognosticators of sepsis in critically ill patients, shorten time to diagnosis confirmation and improve therapeutic decision-making.
Collapse
Affiliation(s)
- Mei-Hong Yu
- Department of Critical Care Medicine, Chun'an First People's Hospital (Zhejiang Provincial People's Hospital Chun'an Branch), Hangzhou 311700, Zhejiang Province, China
| | - Min-Hua Chen
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang Province, China
| | - Fang Han
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang Province, China
| | - Qian Li
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang Province, China
| | - Ren-Hua Sun
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang Province, China
| | - Yue-Xing Tu
- Department of Critical Care Medicine, Chun'an First People's Hospital (Zhejiang Provincial People's Hospital Chun'an Branch), Hangzhou 311700, Zhejiang Province, China; Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang Province, China.
| |
Collapse
|
91
|
Gouwy M, De Buck M, Abouelasrar Salama S, Vandooren J, Knoops S, Pörtner N, Vanbrabant L, Berghmans N, Opdenakker G, Proost P, Van Damme J, Struyf S. Matrix Metalloproteinase-9-Generated COOH-, but Not NH 2-Terminal Fragments of Serum Amyloid A1 Retain Potentiating Activity in Neutrophil Migration to CXCL8, With Loss of Direct Chemotactic and Cytokine-Inducing Capacity. Front Immunol 2018; 9:1081. [PMID: 29915572 PMCID: PMC5994419 DOI: 10.3389/fimmu.2018.01081] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 04/30/2018] [Indexed: 12/21/2022] Open
Abstract
Serum amyloid A1 (SAA1) is a prototypic acute phase protein, induced to extremely high levels by physical insults, including inflammation and infection. Human SAA and its NH2-terminal part have been studied extensively in the context of amyloidosis. By contrast, little is known about COOH-terminal fragments of SAA. Intact SAA1 chemoattracts leukocytes via the G protein-coupled receptor formyl peptide receptor like 1/formyl peptide receptor 2 (FPR2). In addition to direct leukocyte activation, SAA1 induces chemokine production by signaling through toll-like receptor 2. We recently discovered that these induced chemokines synergize with intact SAA1 to chemoattract leukocytes in vitro and in vivo. Gelatinase B or matrix metalloproteinase-9 (MMP-9) is also induced by SAA1 during infection and inflammation and processes many substrates in the immune system. We demonstrate here that MMP-9 rapidly cleaves SAA1 at a known consensus sequence that is also present in gelatins. Processing of SAA1 by MMP-9 at an accessible loop between two alpha helices yielded predominantly three COOH-terminal fragments: SAA1(52–104), SAA1(57–104), and SAA1(58–104), with a relative molecular mass of 5,884.4, 5,327.3, and 5,256.3, respectively. To investigate the effect of proteolytic processing on the biological activity of SAA1, we chemically synthesized the COOH-terminal SAA fragments SAA1(52–104) and SAA1(58–104) and the complementary NH2-terminal peptide SAA1(1–51). In contrast to intact SAA1, the synthesized SAA1 peptides did not induce interleukin-8/CXCL8 in monocytes or fibroblasts. Moreover, these fragments possessed no direct chemotactic activity for neutrophils, as observed for intact SAA1. However, comparable to intact SAA1, SAA1(58–104) cooperated with CXCL8 in neutrophil activation and migration, whereas SAA1(1–51) lacked this potentiating activity. This cooperative interaction between the COOH-terminal SAA1 fragment and CXCL8 in neutrophil chemotaxis was mediated by FPR2. Hence, proteolytic cleavage of SAA1 by MMP-9 fine tunes the inflammatory capacity of this acute phase protein in that only the synergistic interactions with chemokines remain to prolong the duration of inflammation.
Collapse
Affiliation(s)
- Mieke Gouwy
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| | - Mieke De Buck
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| | - Sara Abouelasrar Salama
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| | - Jennifer Vandooren
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| | - Sofie Knoops
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| | - Noëmie Pörtner
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| | - Lotte Vanbrabant
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| | - Nele Berghmans
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| | - Ghislain Opdenakker
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| | - Jo Van Damme
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| | - Sofie Struyf
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| |
Collapse
|
92
|
The ectoenzyme-side of matrix metalloproteinases (MMPs) makes inflammation by serum amyloid A (SAA) and chemokines go round. Immunol Lett 2018; 205:1-8. [PMID: 29870759 DOI: 10.1016/j.imlet.2018.06.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 05/16/2018] [Accepted: 06/01/2018] [Indexed: 12/18/2022]
Abstract
During an inflammatory response, a large number of distinct mediators appears in the affected tissues or in the blood circulation. These include acute phase proteins such as serum amyloid A (SAA), cytokines and chemokines and proteolytic enzymes. Although these molecules are generated within a cascade sequence in specific body compartments allowing for independent action, their co-appearance in space and time during acute or chronic inflammation points toward important mutual interactions. Pathogen-associated molecular patterns lead to fast induction of the pro-inflammatory endogenous pyrogens, which are evoking the acute phase response. Interleukin-1, tumor necrosis factor-α and interferons simultaneously trigger different cell types, including leukocytes, endothelial cells and fibroblasts for tissue-specific or systemic production of chemokines and matrix metalloproteinases (MMPs). In addition, SAA induces chemokines and both stimulate secretion of MMPs from multiple cell types. As a consequence, these mediators may cooperate to enhance the inflammatory response. Indeed, SAA synergizes with chemokines to increase chemoattraction of monocytes and granulocytes. On the other hand, MMPs post-translationally modify chemokines and SAA to reduce their activity. Indeed, MMPs internally cleave SAA with loss of its cytokine-inducing and direct chemotactic potential whilst retaining its capacity to synergize with chemokines in leukocyte migration. Finally, MMPs truncate chemokines at their NH2- or COOH-terminal end, resulting in reduced or enhanced chemotactic activity. Therefore, the complex interactions between chemokines, SAA and MMPs either maintain or dampen the inflammatory response.
Collapse
|
93
|
Facci L, Barbierato M, Zusso M, Skaper SD, Giusti P. Serum amyloid A primes microglia for ATP-dependent interleukin-1β release. J Neuroinflammation 2018; 15:164. [PMID: 29803222 PMCID: PMC5970445 DOI: 10.1186/s12974-018-1205-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 05/15/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Acute-phase response is a systemic reaction to environmental/inflammatory insults and involves production of acute-phase proteins, including serum amyloid A (SAA). Interleukin-1β (IL-1β), a master regulator of neuroinflammation produced by activated inflammatory cells of the myeloid lineage, in particular microglia, plays a key role in the pathogenesis of acute and chronic diseases of the peripheral nervous system and CNS. IL-1β release is promoted by ATP acting at the purinergic P2X7 receptor (P2X7R) in cells primed with toll-like receptor (TLR) ligands. METHODS Purified (> 99%) microglia cultured from neonatal rat cortex and cerebellum were first primed with the putative TLR4/TLR2 agonist SAA (recombinant human Apo-SAA) or the established TLR4 agonist lipopolysaccharide (LPS) followed by addition of ATP. Expression of genes for the NLRP3 inflammasome, IL-1β, tumor necrosis factor-α (TNF-α), and SAA1 was measured by quantitative real-time polymerase chain reaction (q-PCR). Intracellular and extracellular amounts of IL-1β were determined by ELISA. RESULTS Apo-SAA stimulated, in a time-dependent manner, the expression of NLRP3, IL-1β, and TNF-α in cortical microglia, and produced a concentration-dependent increase in the intracellular content of IL-1β in these cells. A 2-h 'priming' of the microglia with Apo-SAA followed by addition of ATP for 1 h, resulting in a robust release of IL-1β into the culture medium, with a concomitant reduction in its intracellular content. The selective P2X7R antagonist A740003 blocked ATP-dependent release of IL-1β. Microglia prepared from rat cerebellum displayed similar behaviors. As with LPS, Apo-SAA upregulated SAA1 and TLR2 mRNA, and downregulated that of TLR4. LPS was less efficacious than Apo-SAA, perhaps reflecting an action of the latter at TLR4 and TLR2. The TLR4 antagonist CLI-095 fully blocked the action of LPS, but only partially that of Apo-SAA. Although the TLR2 antagonist CU-CPT22 was inactive against Apo-SAA, it also failed to block the TLR2 agonist Pam3CSK4. CONCLUSIONS Microglia are central to the inflammatory process and a major source of IL-1β when activated. P2X7R-triggered IL-1β maturation and export is thus likely to represent an important contributor to this cytokine pool. Given that SAA is detected in Alzheimer disease and multiple sclerosis brain, together with IL-1β-immunopositive microglia, these findings propose a link between P2X7R, SAA, and IL-1β in CNS pathophysiology.
Collapse
Affiliation(s)
- Laura Facci
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo "E. Meneghetti" 2, 35131, Padua, Italy
| | - Massimo Barbierato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo "E. Meneghetti" 2, 35131, Padua, Italy
| | - Morena Zusso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo "E. Meneghetti" 2, 35131, Padua, Italy
| | - Stephen D Skaper
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo "E. Meneghetti" 2, 35131, Padua, Italy.
| | - Pietro Giusti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo "E. Meneghetti" 2, 35131, Padua, Italy
| |
Collapse
|
94
|
Kuret T, Lakota K, Mali P, Čučnik S, Praprotnik S, Tomšič M, Sodin-Semrl S. Naturally occurring antibodies against serum amyloid A reduce IL-6 release from peripheral blood mononuclear cells. PLoS One 2018; 13:e0195346. [PMID: 29617422 PMCID: PMC5884545 DOI: 10.1371/journal.pone.0195346] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 03/20/2018] [Indexed: 12/14/2022] Open
Abstract
Serum amyloid A (SAA) is a sensitive inflammatory marker rapidly increased in response to infection, injury or trauma during the acute phase. Resolution of the acute phase and SAA reduction are well documented, however the exact mechanism remains elusive. Two inducible SAA proteins, SAA1 and SAA2, with their variants could contribute to systemic inflammation. While unconjugated human variant SAA1α is already commercially available, the variants of SAA2 are not. Antibodies against SAA have been identified in apparently healthy blood donors (HBDs) in smaller, preliminary studies. So, our objective was to detect anti-SAA and anti-SAA1α autoantibodies in the sera of 300 HBDs using ELISA, characterize their specificity and avidity. Additionally, we aimed to determine the presence of anti-SAA and anti-SAA1α autoantibodies in intravenous immunoglobulin (IVIg) preparations and examine their effects on released IL-6 from SAA/SAA1α-treated peripheral blood mononuclear cells (PBMCs). Autoantibodies against SAA and SAA1α had a median (IQR) absorbance OD (A450) of 0.655 (0.262–1.293) and 0.493 (0.284–0.713), respectively. Both anti-SAA and anti-SAA1α exhibited heterogeneous to high avidity and reached peak levels between 41–50 years, then diminished with age in the oldest group (51–67 years). Women consistently exhibited significantly higher levels than men. Good positive correlation was observed between anti-SAA and anti-SAA1α. Both anti-SAA and anti-SAA1α were detected in IVIg, their fractions subsequently isolated, and shown to decrease IL-6 protein levels released from SAA/SAA1α-treated PBMCs. In conclusion, naturally occurring antibodies against SAA and anti-SAA1α could play a physiological role in down-regulating their antigen and proinflammatory cytokines leading to the resolution of the acute phase and could be an important therapeutic option in patients with chronic inflammatory diseases.
Collapse
Affiliation(s)
- Tadeja Kuret
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Katja Lakota
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Mathematics, Natural Science and Information Technologies, University of Primorska, Koper, Slovenia
| | - Polonca Mali
- Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | - Saša Čučnik
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Sonja Praprotnik
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Matija Tomšič
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Snezna Sodin-Semrl
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Mathematics, Natural Science and Information Technologies, University of Primorska, Koper, Slovenia
- * E-mail:
| |
Collapse
|
95
|
Wang Y, Huang H, Sun R, Chen B, Han F, Li Q, Ni Y, Li X, Liu J, Mou X, Tu Y. Serum amyloid a induces M2b-like macrophage polarization during liver inflammation. Oncotarget 2017; 8:109238-109246. [PMID: 29312604 PMCID: PMC5752517 DOI: 10.18632/oncotarget.22652] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 09/24/2017] [Indexed: 11/25/2022] Open
Abstract
Hepatitis causes hepatic cell injury, regeneration and different levels of fibrogenesis, and severe liver fibrogenesis progresses into cirrhosis with liver dysfunction. Serum amyloid A (SAA) is an acute phase protein that is predominantly secreted by hepatocytes during early injury or infection. Nevertheless, the relationship of SAA and development of cirrhosis as well as the underlying molecular mechanisms is largely unknown. Here, we found that macrophages are the major SAA-binding cells in the injured liver. in vitro, macrophages treated with SAA exhibited high production of IL-10 but low production of IL-12, as features for M2 macrophages. Moreover, these polarized M2 macrophages by SAA also produced IL-1, IL-6 and TNFa, characteristics for an M2b subtype, rather than an alternative M2a or fibrogenic M2c subtype. In a mouse model of carbon tetrachloride (CCl4)-induced hepatic fibrogenesis/cirrhosis, anti-SAA sera were used to block the effects of SAA, resulting in increases in the severity of hepatic fibrosis, suggesting an overall anti-fibrogenic effect of SAA. Isolated macrophages from mouse liver showed that anti-SAA appeared to alter the polarization of macrophages from M2b to M2c, suggesting that SAA may induce M2b-like macrophage polarization during liver inflammation, which prevents the liver from fibrogenesis.
Collapse
Affiliation(s)
- Yibin Wang
- Department of Cardiology, Chunan First People’s Hospital, Hangzhou 311700, China
| | - Haijun Huang
- Department of Infectious Diseases, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Renhua Sun
- ICU Department, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Bingyu Chen
- Centre of Laboratory Medicine, Chunan First People’s Hospital, Hangzhou 311700, China
- Department of Transfusion Medicine, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Fang Han
- ICU Department, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Qian Li
- ICU Department, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Yin Ni
- ICU Department, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Xi Li
- Centre of Laboratory Medicine, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Jingquan Liu
- ICU Department, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Xiaozhou Mou
- Clinical Research Institute, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medicine College, Hangzhou 310014, China
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Hangzhou 310014, China
| | - Yuexing Tu
- ICU Department, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou 310014, China
| |
Collapse
|
96
|
Konuma T, Kohara C, Watanabe E, Mizukami M, Nagai E, Oiwa-Monna M, Tanoue S, Isobe M, Kato S, Tojo A, Takahashi S. Cytokine Profiles of Pre-Engraftment Syndrome after Single-Unit Cord Blood Transplantation for Adult Patients. Biol Blood Marrow Transplant 2017; 23:1932-1938. [DOI: 10.1016/j.bbmt.2017.07.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 07/16/2017] [Indexed: 10/19/2022]
|
97
|
Mao JY, Sun JT, Yang K, Shen WF, Lu L, Zhang RY, Tong X, Liu Y. Serum amyloid A enrichment impairs the anti-inflammatory ability of HDL from diabetic nephropathy patients. J Diabetes Complications 2017; 31:1538-1543. [PMID: 28760652 DOI: 10.1016/j.jdiacomp.2017.07.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 07/06/2017] [Accepted: 07/07/2017] [Indexed: 01/13/2023]
Abstract
AIMS Impaired anti-inflammatory ability of high-density lipoprotein (HDL) has been demonstrated in patients with type-2 diabetes mellitus (T2DM). However, whether HDL from patients with diabetic nephropathy (DN) suffers additional damage remains unknown. This study compared the anti-inflammatory capacities of HDL from healthy controls, T2DM patients with normal renal function, and T2DM patients with DN. MATERIALS AND METHODS HDL was isolated from healthy controls (n=33) and T2DM patients with normal renal function (n=21), chronic kidney disease (CKD) (n=27), and end-stage renal disease (ESRD) (n=27). Human peripheral blood mononuclear cells (PBMCs) from healthy volunteers were pretreated with HDL (100μg/mL) for 1h, then incubated with lipopolysaccharide (LPS) (50ng/mL) for 24h. The anti-inflammatory ability of HDL was measured as the secretion of TNF-α in LPS-activated monocytes. RESULTS The anti-inflammatory ability of HDL was gradually impaired as kidney function declined. Serum amyloid A (SAA) concentration in HDLDN significantly increased and was positively correlated with the impaired anti-inflammatory ability of HDL (Pearson r=0.315, P=0.006). Furthermore, HDL supplemented with SAA significantly increased TNF-α release from PBMCs compared with that from control HDL. CONCLUSIONS These findings identified an impaired anti-inflammatory capacity of HDL from DN patients, which might be attributable to SAA enrichment.
Collapse
MESH Headings
- Adult
- Aged
- Cells, Cultured
- China/epidemiology
- Cross-Sectional Studies
- Diabetes Mellitus, Type 2/complications
- Diabetic Angiopathies/epidemiology
- Diabetic Angiopathies/immunology
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/pathology
- Diabetic Nephropathies/immunology
- Diabetic Nephropathies/metabolism
- Diabetic Nephropathies/pathology
- Diabetic Nephropathies/physiopathology
- Female
- Hospitals, University
- Humans
- Kidney/physiopathology
- Kidney Failure, Chronic/complications
- Kidney Failure, Chronic/immunology
- Kidney Failure, Chronic/metabolism
- Kidney Failure, Chronic/pathology
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/immunology
- Leukocytes, Mononuclear/metabolism
- Leukocytes, Mononuclear/pathology
- Lipopolysaccharides/toxicity
- Lipoproteins, HDL/blood
- Lipoproteins, HDL/isolation & purification
- Lipoproteins, HDL/metabolism
- Male
- Middle Aged
- Outpatient Clinics, Hospital
- Renal Insufficiency, Chronic/complications
- Renal Insufficiency, Chronic/immunology
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Risk Factors
- Serum Amyloid A Protein/analysis
- Serum Amyloid A Protein/metabolism
- Severity of Illness Index
- Vasculitis/complications
- Vasculitis/immunology
- Vasculitis/metabolism
- Vasculitis/pathology
Collapse
Affiliation(s)
- Jing Yan Mao
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jia Teng Sun
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ke Yang
- Institute of Cardiovascular Disease, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wei Feng Shen
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Institute of Cardiovascular Disease, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lin Lu
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Rui Yan Zhang
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xuemei Tong
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Yan Liu
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| |
Collapse
|
98
|
Expression and Differential Responsiveness of Central Nervous System Glial Cell Populations to the Acute Phase Protein Serum Amyloid A. Sci Rep 2017; 7:12158. [PMID: 28939905 PMCID: PMC5610307 DOI: 10.1038/s41598-017-12529-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 09/12/2017] [Indexed: 02/07/2023] Open
Abstract
Acute-phase response is a systemic reaction to environmental/inflammatory insults and involves hepatic production of acute-phase proteins, including serum amyloid A (SAA). Extrahepatically, SAA immunoreactivity is found in axonal myelin sheaths of cortex in Alzheimer's disease and multiple sclerosis (MS), although its cellular origin is unclear. We examined the responses of cultured rat cortical astrocytes, microglia and oligodendrocyte precursor cells (OPCs) to master pro-inflammatory cytokine tumour necrosis factor (TNF)-α and lipopolysaccaride (LPS). TNF-α time-dependently increased Saa1 (but not Saa3) mRNA expression in purified microglia, enriched astrocytes, and OPCs (as did LPS for microglia and astrocytes). Astrocytes depleted of microglia were markedly less responsive to TNF-α and LPS, even after re-addition of microglia. Microglia and enriched astrocytes showed complementary Saa1 expression profiles following TNF-α or LPS challenge, being higher in microglia with TNF-α and higher in astrocytes with LPS. Recombinant human apo-SAA stimulated production of both inflammatory mediators and its own mRNA in microglia and enriched, but not microglia-depleted astrocytes. Co-ultramicronized palmitoylethanolamide/luteolin, an established anti-inflammatory/ neuroprotective agent, reduced Saa1 expression in OPCs subjected to TNF-α treatment. These last data, together with past findings suggest that co-ultramicronized palmitoylethanolamide/luteolin may be a novel approach in the treatment of inflammatory demyelinating disorders like MS.
Collapse
|
99
|
Saulnier PJ, Dieter BP, Tanamas SK, McPherson SM, Wheelock KM, Knowler WC, Looker HC, Meek RL, Nelson RG, Tuttle KR. Association of Serum Amyloid A with Kidney Outcomes and All-Cause Mortality in American Indians with Type 2 Diabetes. Am J Nephrol 2017; 46:276-284. [PMID: 28934744 DOI: 10.1159/000481269] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 08/30/2017] [Indexed: 12/30/2022]
Abstract
BACKGROUND Serum amyloid A (SAA) induces inflammation and apoptosis in kidney cells and is found to be causing the pathologic changes that are associated with diabetic kidney disease (DKD). Higher serum SAA concentrations were previously associated with increased risk of end-stage renal disease (ESRD) and death in persons with type 2 diabetes and advanced DKD. We explored the prognostic value of SAA in American Indians with type 2 diabetes without DKD or with early DKD. METHODS SAA concentration was measured in serum samples obtained at the start of follow-up. Multivariate proportional hazards models were employed to examine the magnitude of the risk of ESRD or death across tertiles of SAA concentration after adjustment for traditional risk factors. The C statistic was used to assess the additional predictive value of SAA relative to traditional risk factors. RESULTS Of 256 participants (mean ± SD glomerular filtration rate [iothalamate] = 148 ± 45 mL/min, and median [interquartile range] urine albumin/creatinine = 39 [14-221] mg/g), 76 developed ESRD and 125 died during a median follow-up period of 15.2 and 15.7 years, respectively. After multivariable proportional hazards regression, participants in the 2 highest SAA tertiles together exhibited a 53% lower risk of ESRD (hazard ratio [HR] 0.47, 95% CI 0.29-0.78), and a 30% lower risk of death (HR 0.70, 95% CI 0.48-1.02), compared with participants in the lowest SAA tertile, although the lower risk of death was not statistically significant. Addition of SAA to the ESRD model increased the C statistic from 0.814 to 0.815 (p = 0.005). CONCLUSIONS Higher circulating SAA concentration is associated with a reduced risk of ESRD in American Indians with type 2 diabetes.
Collapse
|
100
|
Su Q, Weindl G. Glucocorticoids and Toll-like receptor 2 cooperatively induce acute-phase serum amyloid A. Pharmacol Res 2017; 128:145-152. [PMID: 28941781 DOI: 10.1016/j.phrs.2017.09.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 08/08/2017] [Accepted: 09/18/2017] [Indexed: 12/12/2022]
Abstract
Serum amyloid A (SAA) is a highly conserved acute-phase protein and extrahepatic produced SAA1/2 contributes to cutaneous inflammation. Prolonged systemic or topical treatment with glucocorticoids can provoke skin diseases such as steroid-induced acne. Glucocorticoids increase Toll-like receptor 2 (TLR2) expression, however, an inflammatory mediator linked to this side effect remains elusive. We report that TLR2 agonists in combination with dexamethasone substantially increase SAA expression and production in human keratinocytes and epithelial cells. Dexamethasone-mediated SAA1 induction depends on the glucocorticoid receptor (GR). In response to Propionibacterium acnes, TLR2-activated signal transducer and activator of transcription 3 (STAT3) and nuclear factor κB (NF-κB) signaling pathways are critically involved in dexamethasone-induced SAA1 production. The formation of transcription factor complexes between GR or p300 and phospho-STAT3 was confirmed by co-immunoprecipitation in dexamethasone- and P. acnes-stimulated keratinocytes. Furthermore, dexamethasone and P. acnes-increased TLR2 and mitogen-activated protein kinase phosphatase-1 (MKP-1) contribute to induction of SAA1 and 2. Likewise, tumor necrosis factor (TNF) induces SAA1 in combination with dexamethasone. GR, transcription factors STAT3 and NF-κB, but not MKP-1, mediate TNF- and dexamethasone-induced SAA1. Conclusively, we provide evidence that glucocorticoids promote SAA1 production under infectious and sterile inflammatory conditions which may provide significant insights to the pathogenesis of steroid-induced acne.
Collapse
Affiliation(s)
- Qi Su
- Institute of Pharmacy (Pharmacology and Toxicology), Freie Universität Berlin, Berlin, Germany
| | - Günther Weindl
- Institute of Pharmacy (Pharmacology and Toxicology), Freie Universität Berlin, Berlin, Germany.
| |
Collapse
|