1
|
Schmoch T, Gallenstein N, Peters V, Bartosova M, Uhle F, Kummer L, Mair A, Krauser U, Feisst M, Nawroth PP, Weigand MA, Schmitt CP, Brenner T. Anserine reduces mortality in experimental sepsis by preventing methylglyoxal-induced capillary leakage. EBioMedicine 2025; 114:105644. [PMID: 40107203 PMCID: PMC11995882 DOI: 10.1016/j.ebiom.2025.105644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND We previously identified methylglyoxal as a biomarker for early identification and outcome prediction in human sepsis. We hypothesised that methylglyoxal causally impacts disease severity, and the methylglyoxal-scavenging dipeptide anserine can attenuate the detrimental effects of methylglyoxal. METHODS Using a translational approach, secondary analyses of two observational trials were performed to test the initial hypotheses. Afterwards, these results were re-evaluated in different murine models of experimental sepsis in vivo. The detrimental effects of methylglyoxal as well as the underlying mechanisms were further assessed in vitro using transendothelial electrical resistance measurements, fluorescence-activated cell sorting analyses, cytokine assays, gene expression analyses, and enzyme activity assays, as well as immunofluorescence and immunohistochemistry staining. FINDINGS The secondary analyses confirmed methylglyoxal as an independent marker associated with increased mortality within the first 48 h after sepsis onset and high catecholamine and fluid requirements in the first 24 h after sepsis onset. In the sepsis models, methylglyoxal-derived carbonyl stress significantly contributed to the development of capillary leakage by disrupting endothelial barrier-forming proteins. Mechanistically, a pathway involving the receptor of advanced glycation end products and mitogen-activated protein kinase was identified. The methylglyoxal-scavenging dipeptide anserine (β-alanyl-N-methylhistidine) reduced methylglyoxal-induced advanced glycation end-product formation and disruptions of junctional complexes in vitro. Moreover, anserine reduced capillary leakage and mortality in vivo. INTERPRETATION Methylglyoxal causally contributes to capillary leak formation and mortality in experimental sepsis, which can be mitigated by anserine. Therefore, anserine represents an innovative therapeutic option for the treatment of septic shock. FUNDING German Research Foundation (grant number BR 4144/2-1).
Collapse
Affiliation(s)
- Thomas Schmoch
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany; Medical Faculty Heidelberg, Department of Anesthesiology, Heidelberg University, Heidelberg, Germany; Department of Anesthesiology and Intensive Care Medicine, Hôpitaux Robert Schuman - Hôpital Kirchberg, Luxembourg City, Luxembourg.
| | - Nadia Gallenstein
- Medical Faculty Heidelberg, Department of Anesthesiology, Heidelberg University, Heidelberg, Germany.
| | - Verena Peters
- Medical Faculty Heidelberg, Department of Pediatrics I, Center for Paediatric and Adolescent Medicine, Heidelberg University, Heidelberg, Germany
| | - Maria Bartosova
- Medical Faculty Heidelberg, Department of Pediatrics I, Center for Paediatric and Adolescent Medicine, Heidelberg University, Heidelberg, Germany
| | - Florian Uhle
- Medical Faculty Heidelberg, Department of Anesthesiology, Heidelberg University, Heidelberg, Germany
| | - Laura Kummer
- Medical Faculty Heidelberg, Department of Anesthesiology, Heidelberg University, Heidelberg, Germany
| | - Anian Mair
- Medical Faculty Heidelberg, Department of Anesthesiology, Heidelberg University, Heidelberg, Germany
| | - Ute Krauser
- Medical Faculty Heidelberg, Department of Anesthesiology, Heidelberg University, Heidelberg, Germany
| | - Manuel Feisst
- Institute of Medical Biometry, Heidelberg University, Heidelberg, Germany
| | - Peter P Nawroth
- Medical Faculty Heidelberg, Department of Medicine I and Clinical Chemistry, Heidelberg University, Heidelberg, Germany
| | - Markus A Weigand
- Medical Faculty Heidelberg, Department of Anesthesiology, Heidelberg University, Heidelberg, Germany
| | - Claus Peter Schmitt
- Medical Faculty Heidelberg, Department of Pediatrics I, Center for Paediatric and Adolescent Medicine, Heidelberg University, Heidelberg, Germany
| | - Thorsten Brenner
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany; Medical Faculty Heidelberg, Department of Anesthesiology, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
2
|
Prantner D, Vogel SN. Intracellular methylglyoxal accumulation in classically activated mouse macrophages is mediated by HIF-1α. J Leukoc Biol 2025; 117:qiae215. [PMID: 39360990 DOI: 10.1093/jleuko/qiae215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 09/30/2024] [Indexed: 03/30/2025] Open
Abstract
Approximately one million cases of sepsis in the United States occur annually. The early phase of sepsis features dramatic changes in host metabolism and inflammation. While examining the effects of metabolic pathways on inflammation, we discovered that the highly reactive glycolytic metabolite, methylglyoxal (MG), accumulates intracellularly during classical activation of macrophages. Herein, we explored the role of glycolysis and the master regulator of glycolysis, Hypoxia-Inducing Factor-1α (HIF-1α), in inflammation and MG accumulation in mouse and human macrophages. To determine how HIF-1α regulates the inflammatory response of macrophages, we correlated HIF-1α stabilization with proinflammatory gene expression and MG-adduct accumulation in WT vs HIF1a-deficient macrophages treated with LPS or LPS + IFN-γ. A nearly complete loss of HIF-1α protein expression in response to the hypoxia mimetic, cobalt chloride, confirmed the phenotype of the HIF1a-deficient macrophages. Moreover, absence of HIF-1α was also associated with decreased MG accumulation. Increasing the glucose concentration in cultured macrophages was sufficient to cause accumulation of endogenous MG-adducts which correlated with increased Tnf and Il1b expression during classical activation. The use of the MG antagonist, aminoguanidine, led to a significant decrease in Tnf and Il1b expression in both mouse macrophages and the THP-1 human macrophage cell line. Although off-target effects cannot be ruled out, these results are consistent with the possibility that MG regulates cytokine expression in classically activated macrophages. Collectively, this work suggests that HIF-1α stabilization is upstream of MG accumulation and that targeting the activity of HIF-1α in macrophages may be therapeutic during sepsis by limiting endogenous MG accumulation.
Collapse
Affiliation(s)
- Daniel Prantner
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 West Baltimore St., Suite 380, Baltimore, MD 21201, USA
| | - Stefanie N Vogel
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 West Baltimore St., Suite 380, Baltimore, MD 21201, USA
| |
Collapse
|
3
|
Vizuete AFK, Gonçalves CA. Is Methylglyoxal a Potential Biomarker for the Warburg Effect Induced by the Lipopolysaccharide Neuroinflammation Model? Neurochem Res 2024; 49:1823-1837. [PMID: 38727985 DOI: 10.1007/s11064-024-04142-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/26/2024] [Accepted: 05/02/2024] [Indexed: 06/02/2024]
Abstract
Methylglyoxal (MG) is considered a classical biomarker of diabetes mellitus and its comorbidities. However, a role for this compound in exacerbated immune responses, such as septicemia, is being increasingly observed and requires clarification, particularly in the context of neuroinflammatory responses. Herein, we used two different approaches (in vivo and acute hippocampal slice models) to investigate MG as a biomarker of neuroinflammation and the neuroimmunometabolic shift to glycolysis in lipopolysaccharide (LPS) inflammation models. Our data reinforce the hypothesis that LPS-induced neuroinflammation stimulates the cerebral innate immune response by increasing IL-1β, a classical pro-inflammatory cytokine, and the astrocyte reactive response, via elevating S100B secretion and GFAP levels. Acute neuroinflammation promotes an early neuroimmunometabolic shift to glycolysis by elevating glucose uptake, lactate release, PFK1, and PK activities. We observed high serum and cerebral MG levels, in association with a reduction in glyoxalase 1 detoxification activity, and a close correlation between serum and hippocampus MG levels with the systemic and neuroinflammatory responses to LPS. Findings strongly suggest a role for MG in immune responses.
Collapse
Affiliation(s)
- Adriana Fernanda Kuckartz Vizuete
- Laboratory of Calcium-Binding Proteins in the CNS, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS) Ramio Barcelos, 2600-Anexo, Porto Alegre, RS, 90035-003, Brazil.
- Pos Graduate Program in Biochemistry, Institute of Basic Health Sciences, UFRGS, Porto Alegre, RS, Brazil.
| | - Carlos-Alberto Gonçalves
- Laboratory of Calcium-Binding Proteins in the CNS, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS) Ramio Barcelos, 2600-Anexo, Porto Alegre, RS, 90035-003, Brazil
- Pos Graduate Program in Biochemistry, Institute of Basic Health Sciences, UFRGS, Porto Alegre, RS, Brazil
- Department of Biochemistry, Institute of Basic Health Sciences, UFRGS, Porto Alegre, RS, Brazil
| |
Collapse
|
4
|
Greppmair S, Liebchen U. [Treatment of sepsis on the pulse of time : Proven standards and current trends]. DIE ANAESTHESIOLOGIE 2024; 73:1-3. [PMID: 38226994 DOI: 10.1007/s00101-023-01366-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/20/2023] [Indexed: 01/17/2024]
Affiliation(s)
- Sebastian Greppmair
- Klinik für Anaesthesiologie, LMU Klinikum, LMU München, Marchioninistr. 15, 81377, München, Deutschland
| | - Uwe Liebchen
- Klinik für Anaesthesiologie, LMU Klinikum, LMU München, Marchioninistr. 15, 81377, München, Deutschland.
| |
Collapse
|
5
|
Bénard A, Hansen FJ, Uhle F, Klösch B, Czubayko F, Mittelstädt A, Jacobsen A, David P, Podolska MJ, Anthuber A, Swierzy I, Schaack D, Mühl-Zürbes P, Steinkasserer A, Weyand M, Weigand MA, Brenner T, Krautz C, Grützmann R, Weber GF. Interleukin-3 protects against viral pneumonia in sepsis by enhancing plasmacytoid dendritic cell recruitment into the lungs and T cell priming. Front Immunol 2023; 14:1140630. [PMID: 36911737 PMCID: PMC9996195 DOI: 10.3389/fimmu.2023.1140630] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 02/02/2023] [Indexed: 02/25/2023] Open
Abstract
Rationale Sepsis, a global health burden, is often complicated by viral infections leading to increased long-term morbidity and mortality. Interleukin-3 (IL-3) has been identified as an important mediator amplifying acute inflammation in sepsis; however, its function in the host response to viral infections during sepsis remains elusive. Objectives To investigate the role of IL-3 during viral pneumonia in sepsis. Methods We included septic patients from two different cohorts and used in vitro and in vivo assays. The obtained data were substantiated using a second model (SARS-CoV-2 infections). Measurements and main results Low plasma IL-3 levels were associated with increased herpes simplex virus (HSV) airway infections in septic patients, resulting in reduced overall survival. Likewise, Il-3-deficient septic mice were more susceptible to pulmonary HSV-1 infection and exhibited higher pulmonary inflammation than control mice. Mechanistically, IL-3 increases innate antiviral immunity by promoting the recruitment of circulating plasmacytoid dendritic cells (pDCs) into the airways and by enhancing pDC-mediated T cell activation upon viral stimulation. Interestingly, the ability of IL-3 to improve adaptive immunity was confirmed in patients with SARS-CoV-2 infections. Conclusion Our study identifies IL-3 as a predictive disease marker for viral reactivation in sepsis and reveals that IL-3 improves antiviral immunity by enhancing the recruitment and the function of pDCs.
Collapse
Affiliation(s)
- Alan Bénard
- Department of Surgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Frederik J. Hansen
- Department of Surgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Florian Uhle
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Bettina Klösch
- Department of Surgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Franziska Czubayko
- Department of Surgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Anke Mittelstädt
- Department of Surgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Anne Jacobsen
- Department of Surgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Paul David
- Department of Surgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Malgorzata J. Podolska
- Department of Surgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Anna Anthuber
- Department of Surgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Izabela Swierzy
- Department of Surgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Dominik Schaack
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Petra Mühl-Zürbes
- Department of Immune Modulation, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Alexander Steinkasserer
- Department of Immune Modulation, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Michael Weyand
- Department of Cardiac Surgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Markus A. Weigand
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Thorsten Brenner
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Christian Krautz
- Department of Surgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Robert Grützmann
- Department of Surgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Georg F. Weber
- Department of Surgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
6
|
Kim CJ. Current Status of Antibiotic Stewardship and the Role of Biomarkers in Antibiotic Stewardship Programs. Infect Chemother 2022; 54:674-698. [PMID: 36596680 PMCID: PMC9840952 DOI: 10.3947/ic.2022.0172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 12/19/2022] [Indexed: 12/27/2022] Open
Abstract
The importance of antibiotic stewardship is increasingly emphasized in accordance with the increasing incidences of multidrug-resistant organisms and accompanying increases in disease burden. This review describes the obstacles in operating an antibiotic stewardship program (ASP), and whether the use of biomarkers within currently available resources can help. Surveys conducted around the world have shown that major obstacles to ASPs are shortages of time and personnel, lack of appropriate compensation for ASP operation, and lack of guidelines or appropriate manuals. Sufficient investment, such as the provision of full-time equivalent ASP practitioners, and adoption of computerized clinical decision systems are useful measures to improve ASP within an institution. However, these methods are not easy in terms of both time commitments and cost. Some biomarkers, such as C-reactive protein, procalcitonin, and presepsin are promising tools in ASP due to their utility in diagnosis and forecasting the prognosis of sepsis. Recent studies have demonstrated the usefulness of algorithmic approaches based on procalcitonin level to determine the initiation or discontinuation of antibiotics, which would be helpful in decreasing antibiotics use, resulting in more appropriate antibiotics use.
Collapse
Affiliation(s)
- Chung-Jong Kim
- Department of Internal Medicine, College of Medicine, Ewha Womans University, Seoul, Korea
| |
Collapse
|
7
|
Biermann AR, Hogan DA. Transcriptional Response of Candida auris to the Mrr1 Inducers Methylglyoxal and Benomyl. mSphere 2022; 7:e0012422. [PMID: 35473297 PMCID: PMC9241502 DOI: 10.1128/msphere.00124-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 03/18/2022] [Indexed: 11/20/2022] Open
Abstract
Candida auris is an urgent threat to human health due to its rapid spread in health care settings and its repeated development of multidrug resistance. Diseases that increase risk for C. auris infection, such as diabetes, kidney failure, or immunocompromising conditions, are associated with elevated levels of methylglyoxal (MG), a reactive dicarbonyl compound derived from several metabolic processes. In other Candida species, expression of MG reductase enzymes that catabolize and detoxify MG are controlled by Mrr1, a multidrug resistance-associated transcription factor, and MG induces Mrr1 activity. Here, we used transcriptomics and genetic assays to determine that C. auris MRR1a contributes to MG resistance, and that the main Mrr1a targets are an MG reductase and MDR1, which encodes a drug efflux protein. The C. auris Mrr1a regulon is smaller than Mrr1 regulons described in other species. In addition to MG, benomyl (BEN), a known Mrr1 stimulus, induces C. auris Mrr1 activity, and characterization of the MRR1a-dependent and -independent transcriptional responses revealed substantial overlap in genes that were differentially expressed in response to each compound. Additionally, we found that an MRR1 allele specific to one C. auris phylogenetic clade, clade III, encodes a hyperactive Mrr1 variant, and this activity correlated with higher MG resistance. C. auris MRR1a alleles were functional in Candida lusitaniae and were inducible by BEN, but not by MG, suggesting that the two Mrr1 inducers act via different mechanisms. Together, the data presented in this work contribute to the understanding of Mrr1 activity and MG resistance in C. auris. IMPORTANCE Candida auris is a fungal pathogen that has spread since its identification in 2009 and is of concern due to its high incidence of resistance against multiple classes of antifungal drugs. In other Candida species, the transcription factor Mrr1 plays a major role in resistance against azole antifungals and other toxins. More recently, Mrr1 has been recognized to contribute to resistance to methylglyoxal (MG), a toxic metabolic product that is often elevated in different disease states. MG can activate Mrr1 and its induction of Mdr1 which can protect against diverse challenges. The significance of this work lies in showing that MG is also an inducer of Mrr1 in C. auris, and that one of the major pathogenic C. auris lineages has an activating Mrr1 mutation that confers protection against MG.
Collapse
Affiliation(s)
- Amy R. Biermann
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Deborah A. Hogan
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| |
Collapse
|
8
|
Zhang X, Schalkwijk CG, Wouters K. Immunometabolism and the modulation of immune responses and host defense: A role for methylglyoxal? Biochim Biophys Acta Mol Basis Dis 2022; 1868:166425. [DOI: 10.1016/j.bbadis.2022.166425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 04/14/2022] [Accepted: 04/25/2022] [Indexed: 11/26/2022]
|
9
|
Methylglyoxal Levels in Human Colorectal Precancer and Cancer: Analysis of Tumor and Peritumor Tissue. Life (Basel) 2021; 11:life11121319. [PMID: 34947850 PMCID: PMC8708054 DOI: 10.3390/life11121319] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/05/2021] [Accepted: 11/26/2021] [Indexed: 12/23/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers worldwide and its incidence is increasing; therefore, an understanding of its oncogenic mechanisms is critical for improving its treatment and management. Methylglyoxal (MGO) has a highly reactive aldehyde group and has been suggested to play a role in oncogenesis. However, no standardized data are currently available on MGO levels in colorectal precancerous and cancerous lesions. We collected 40 matched colorectal tumor and peritumor tissues from patients with low-grade dysplasia (LGD), high-grade dysplasia (HGD), and invasive cancer (IC). MGO levels increased between LGD, HGD, and IC tumor tissues (215.25 ± 39.69, 267.45 ± 100.61, and 587.36 ± 123.19 μg/g protein, respectively; p = 0.014). The MGO levels in peritumor tissue increased and were significantly higher than MGO levels in tumor tissue (197.99 ± 49.40, 738.09 ± 247.87, 933.41 ± 164.83 μg/g protein, respectively; p = 0.002). Tumor tissue MGO levels did not correlate with age, sex, underlying disease, or smoking status. These results suggest that MGO levels fluctuate in progression of CRC and warrants further research into its underlying mechanisms and function in tumor biology.
Collapse
|
10
|
Systemic inflammation down-regulates glyoxalase-1 expression: an experimental study in healthy males. Biosci Rep 2021; 41:229081. [PMID: 34156474 PMCID: PMC8411911 DOI: 10.1042/bsr20210954] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/11/2021] [Accepted: 06/15/2021] [Indexed: 01/25/2023] Open
Abstract
Background: Hypoxia and inflammation are hallmarks of critical illness, related
to multiple organ failure. A possible mechanism leading to multiple organ
failure is hypoxia- or inflammation-induced down-regulation of the detoxifying
glyoxalase system that clears dicarbonyl stress. The dicarbonyl methylglyoxal
(MGO) is a highly reactive agent produced by metabolic pathways such as
anaerobic glycolysis and gluconeogenesis. MGO leads to protein damage and
ultimately multi-organ failure. Whether detoxification of MGO into D-lactate by
glyoxalase functions appropriately under conditions of hypoxia and inflammation
is largely unknown. We investigated the effect of inflammation and hypoxia on
the MGO pathway in humans in vivo. Methods: After prehydration with glucose 2.5% solution, ten healthy males
were exposed to hypoxia (arterial saturation 80–85%) for 3.5 h
using an air-tight respiratory helmet, ten males to experimental endotoxemia
(LPS 2 ng/kg i.v.), ten males to LPS+hypoxia and ten males to none of these
interventions (control group). Serial blood samples were drawn, and glyoxalase-1
mRNA expression, MGO, methylglyoxal-derived hydroimidazolone-1 (MG-H1),
D-lactate and L-lactate levels, were measured serially. Results: Glyoxalase-1 mRNA expression decreased in the LPS (β
(95%CI); -0.87 (-1.24; -0.50) and the LPS+hypoxia groups; -0.78 (-1.07;
-0.48) (P<0.001). MGO was equal between groups, whereas
MG-H1 increased over time in the control group only
(P=0.003). D-Lactate was increased in all four groups.
L-Lactate was increased in all groups, except in the control group. Conclusion: Systemic inflammation downregulates glyoxalase-1 mRNA expression in
humans. This is a possible mechanism leading to cell damage and multi-organ
failure in critical illness with potential for intervention.
Collapse
|
11
|
Schmoch T, Westhoff JH, Decker SO, Skarabis A, Hoffmann GF, Dohna-Schwake C, Felderhoff-Müser U, Skolik C, Feisst M, Klose C, Bruckner T, Luntz S, Weigand MA, Sohn K, Brenner T. Next-generation sequencing diagnostics of bacteremia in pediatric sepsis. Medicine (Baltimore) 2021; 100:e26403. [PMID: 34160425 PMCID: PMC8238315 DOI: 10.1097/md.0000000000026403] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 06/03/2021] [Indexed: 01/04/2023] Open
Abstract
INTRODUCTION Sepsis and septic shock are the most severe forms of infection affecting predominantly elderly people, preterm and term neonates, and young infants. Even in high-income countries sepsis causes about 8% of admissions to pediatric intensive care units (PICUs). Early diagnosis, rapid anti-infective treatment, and prompt hemodynamic stabilization are crucial for patient survival. In this context, it is essential to identify the causative pathogen as soon as possible to optimize antimicrobial treatment. To date, culture-based diagnostic procedures (e.g., blood cultures) represent the standard of care. However, they have 2 major problems: on the one hand, in the case of very small sample volumes (and thus usually in children), they are not sufficiently sensitive. On the other hand, with a time-to-result of 2 to 5 days, blood cultures need a relatively long time for the anti-infective therapy to be calculated. To overcome these problems, culture-independent molecular diagnostic procedures such as unbiased sequence analysis of circulating cell-free DNA (cfDNA) from plasma samples of septic patients by next-generation sequencing (NGS) have been tested successfully in adult septic patients. However, these results still need to be transferred to the pediatric setting. METHODS The Next GeneSiPS-Trial is a prospective, observational, non-interventional, multicenter study used to assess the diagnostic performance of an NGS-based approach for the identification of causative pathogens in (preterm and term) neonates (d1-d28, n = 50), infants (d29 to <1 yr, n = 50), and toddlers (1 yr to <5 yr, n = 50) with suspected or proven severe sepsis or septic shock (according to the pediatric sepsis definition) by the use of the quantitative sepsis indicating quantifier (SIQ) score in comparison to standard of care (culture-based) microbiological diagnostics. Potential changes in anti-infective treatment regimens based on these NGS results will be estimated retrospectively by a panel of 3 independent clinical specialists. DISCUSSION Neonates, infants, and young children are significantly affected by sepsis. Fast and more sensitive diagnostic approaches are urgently needed. This prospective, observational, non-interventional, multicenter study seeks to evaluate an NGS-based approach in critically ill children suffering from sepsis. TRIAL REGISTRATION DRKS-ID: DRKS00015705 (registered October 24, 2018). https://www.drks.de/drks_web/navigate.do?navigationId=trial.HTML&TRIAL_ID=DRKS00015705.
Collapse
Affiliation(s)
- Thomas Schmoch
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University Duisburg-Essen, Essen
- Department of Anesthesiology, Heidelberg University Hospital
| | - Jens H. Westhoff
- Department of Pediatrics I, University Children's Hospital Heidelberg, Heidelberg
| | | | - Annabell Skarabis
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University Duisburg-Essen, Essen
| | - Georg F. Hoffmann
- Department of Pediatrics I, University Children's Hospital Heidelberg, Heidelberg
| | - Christian Dohna-Schwake
- Department of Pediatrics I, Neonatology, Pediatric Intensive Care, Pediatric Neurology, University Hospital Essen, University Duisburg-Essen, Essen
| | - Ursula Felderhoff-Müser
- Department of Pediatrics I, Neonatology, Pediatric Intensive Care, Pediatric Neurology, University Hospital Essen, University Duisburg-Essen, Essen
| | | | - Manuel Feisst
- Institute of Medical Biometry, University of Heidelberg
| | | | | | - Steffen Luntz
- Coordination Centre for Clinical Trials (KKS), Ruprecht-Karls-University, Heidelberg
| | | | - Kai Sohn
- Fraunhofer Institute for Interfacial Engineering and Biotechnology, Stuttgart, Germany
| | - Thorsten Brenner
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University Duisburg-Essen, Essen
| |
Collapse
|
12
|
Prantner D, Nallar S, Richard K, Spiegel D, Collins KD, Vogel SN. Classically activated mouse macrophages produce methylglyoxal that induces a TLR4- and RAGE-independent proinflammatory response. J Leukoc Biol 2021; 109:605-619. [PMID: 32678947 PMCID: PMC7855181 DOI: 10.1002/jlb.3a0520-745rr] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 01/05/2023] Open
Abstract
The highly reactive compound methylglyoxal (MG) can cause direct damage to cells and tissues by reacting with cellular macromolecules. MG has been identified as a biomarker associated with increased sepsis-induced mortality. Patients undergoing septic shock have significantly elevated circulating MG levels compared to postoperative patients and healthy controls. Furthermore, MG has been implicated in the development of type II diabetes mellitus and Alzheimer's disease. Because MG is generated during glycolysis, we hypothesized that MG may be produced by classically activated (M1) macrophages, possibly contributing to the inflammatory response. LPS and IFN-γ-treated macrophages acquired an M1 phenotype (as evidenced by M1 markers and enhanced glycolysis) and formed MG adducts, MG-H1, MG-H2, and MG-H3, which were detected using antibodies specific for MG-modified proteins (methylglyoxal 5-hydro-5-methylimidazolones). MG adducts were also increased in the lungs of LPS-treated mice. Macrophages treated with LPS and IFN-γ also exhibited decreased expression of glyoxalase 1 (Glo1), an enzyme that metabolizes MG. Concentrations of exogenous, purified MG > 0.5 mM were toxic to macrophages; however, a nontoxic dose of 0.3 mM induced TNF-α and IL-1β, albeit to a lesser extent than LPS stimulation. Despite prior evidence that MG adducts may signal through "receptor for advanced glycation endproducts" (RAGE), MG-mediated cell death and cytokine induction by exogenous MG was RAGE-independent in primary macrophages. Finally, RAGE-deficient mice did not exhibit a significant survival advantage following lethal LPS injection. Overall, our evidence suggests that MG may be produced by M1 macrophages during sepsis, following IFN-γ-dependent down-regulation of Glo1, contributing to over-exuberant inflammation.
Collapse
Affiliation(s)
- Daniel Prantner
- Dept. of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, MD
| | - Shreeram Nallar
- Dept. of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, MD
| | - Katharina Richard
- Dept. of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, MD
| | - David Spiegel
- Department of Chemistry, Yale University, New Haven, CT
| | - Kim D. Collins
- Dept. of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, MD
- Institute of Marine and Environmental Technology (IMET), University of Maryland, Baltimore, Baltimore, MD
| | - Stefanie N. Vogel
- Dept. of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, MD
| |
Collapse
|
13
|
Tang Y, Zhao Y, Wang P, Sang S. Simultaneous Determination of Multiple Reactive Carbonyl Species in High Fat Diet-Induced Metabolic Disordered Mice and the Inhibitory Effects of Rosemary on Carbonyl Stress. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:1123-1131. [PMID: 33464893 DOI: 10.1021/acs.jafc.0c07748] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
As potential endogenous biomarkers, reactive carbonyl species (RCS) have gained abundant attention for monitoring oxidative and carbonyl stress. However, there is no accurate method to evaluate multiple RCS in biological samples. In this study, a 2,4-dinitrophenylhydrazine (DNPH) derivatization-based LC-MS method was developed and validated to quantitate eight RCS: malondialdehyde (MDA), acrolein (ACR), 4-hydroxy-2-nonenal (4-HNE), 4-oxo-2-nonenal (4-ONE), methylglyoxal (MGO), glyoxal (GO), 3-deoxyglucosone (3-DG), and 2-keto-d-glucose (2-Keto). Subsequently, the method was applied to assess the RCS in low fat (LF), high fat (HF), and HF plus rosemary extract (RE) diet-fed mouse samples. The quantitative results on RCS levels indicated that the HF diet significantly increased the total RCS levels in mouse urine, plasma, and kidney with an average rate of 280.69%, 153.87%, and 61.30%, respectively. The RE administration significantly inhibited the elevated RCS levels induced by the HF diet, especially for MDA, 4-ONE, 4-HNE, and 2-Keto in mouse plasma, and ACR and 2-Keto in mouse kidney. This is the first study to simultaneously measure eight RCS in biological samples and demonstrate that RE was able to eliminate the accumulation of the HF diet-induced RCS.
Collapse
Affiliation(s)
- Yao Tang
- Laboratory for Functional Foods and Human Health, Center for Excellence in Post-Harvest Technologies, North Carolina Agricultural and Technical State University, North Carolina Research Campus, 500 Laureate Way, Kannapolis, North Carolina 28081, United States
| | - Yantao Zhao
- Laboratory for Functional Foods and Human Health, Center for Excellence in Post-Harvest Technologies, North Carolina Agricultural and Technical State University, North Carolina Research Campus, 500 Laureate Way, Kannapolis, North Carolina 28081, United States
| | - Pei Wang
- Laboratory for Functional Foods and Human Health, Center for Excellence in Post-Harvest Technologies, North Carolina Agricultural and Technical State University, North Carolina Research Campus, 500 Laureate Way, Kannapolis, North Carolina 28081, United States
| | - Shengmin Sang
- Laboratory for Functional Foods and Human Health, Center for Excellence in Post-Harvest Technologies, North Carolina Agricultural and Technical State University, North Carolina Research Campus, 500 Laureate Way, Kannapolis, North Carolina 28081, United States
| |
Collapse
|
14
|
Biermann AR, Demers EG, Hogan DA. Mrr1 regulation of methylglyoxal catabolism and methylglyoxal-induced fluconazole resistance in Candida lusitaniae. Mol Microbiol 2020; 115:116-130. [PMID: 33319423 DOI: 10.1111/mmi.14604] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/26/2020] [Accepted: 09/05/2020] [Indexed: 12/11/2022]
Abstract
Transcription factor Mrr1, best known for its regulation of Candida azole resistance genes such as MDR1, regulates other genes that are poorly characterized. Among the other Mrr1-regulated genes are putative methylglyoxal reductases. Methylglyoxal (MG) is a toxic metabolite that is elevated in diabetes, uremia, and sepsis, which are diseases that increase the risk for candidiasis, and MG serves as a regulatory signal in diverse organisms. Our studies in Clavispora lusitaniae, also known as Candida lusitaniae, showed that Mrr1 regulates expression of two paralogous MG reductases, MGD1 and MGD2, and that both participate in MG resistance and MG catabolism. Exogenous MG increased Mrr1-dependent expression of MGD1 and MGD2 as well as expression of MDR1, which encodes an efflux pump that exports fluconazole. MG improved growth in the presence of fluconazole and this was largely Mrr1-dependent with contributions from a secondary transcription factor, Cap1. Increased fluconazole resistance was also observed in mutants lacking Glo1, a Mrr1-independent MG catabolic enzyme. Isolates from other Candida species displayed heterogeneity in MG resistance and MG stimulation of azole resistance. We propose endogenous and host-derived MG can induce MDR1 and other Mrr1-regulated genes causing increased drug resistance, which may contribute to some instances of fungal treatment failure.
Collapse
Affiliation(s)
- Amy R Biermann
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Elora G Demers
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Deborah A Hogan
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| |
Collapse
|
15
|
Prantner D, Nallar S, Vogel SN. The role of RAGE in host pathology and crosstalk between RAGE and TLR4 in innate immune signal transduction pathways. FASEB J 2020; 34:15659-15674. [PMID: 33131091 DOI: 10.1096/fj.202002136r] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/07/2020] [Accepted: 10/13/2020] [Indexed: 12/15/2022]
Abstract
Although the innate immune receptor protein, Receptor for Advanced Glycation End products (RAGE), has been extensively studied, there has been renewed interest in RAGE for its potential role in sepsis, along with a host of other inflammatory diseases of chronic, noninfectious origin. In contrast to other innate immune receptors, for example, Toll-like receptors (TLRs), that recognize ligands derived from pathogenic organisms that are collectively known as "pathogen-associated molecular patterns" (PAMPs) or host-derived "damage-associated molecular patterns" (DAMPs), RAGE has been shown to recognize a broad collection of DAMPs exclusively. Historically, these DAMPs have been shown to be pro-inflammatory in nature. Early studies indicated that the adaptor molecule, MyD88, might be important for this change. More recent studies have explored further the mechanisms underlying this inflammatory change. Overall, the newer results have shown that there is extensive crosstalk between RAGE and TLRs. The three canonical RAGE ligands, Advanced Glycation End products (AGEs), HMGB1, and S100 proteins, have all been shown to activate both TLRs and RAGE to varying degrees in order to induce inflammation in in vitro models. As with any field that delves deeply into innate signaling, obstacles of reagent purity may be a cause of some of the discrepancies in the literature, and we have found that commercial antibodies that have been widely used exhibit a high degree of nonspecificity. Nonetheless, the weight of published evidence has led us to speculate that RAGE may be physically interacting with TLRs on the cell surface to elicit inflammation via MyD88-dependent signaling.
Collapse
Affiliation(s)
- Daniel Prantner
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Shreeram Nallar
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Stefanie N Vogel
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
| |
Collapse
|
16
|
|
17
|
Griggs RB, Yermakov LM, Drouet DE, Nguyen DVM, Susuki K. Methylglyoxal Disrupts Paranodal Axoglial Junctions via Calpain Activation. ASN Neuro 2019; 10:1759091418766175. [PMID: 29673258 PMCID: PMC5944142 DOI: 10.1177/1759091418766175] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Nodes of Ranvier and associated paranodal and juxtaparanodal domains along myelinated axons are essential for normal function of the peripheral and central nervous systems. Disruption of these domains as well as increases in the reactive carbonyl species methylglyoxal are implicated as a pathophysiology common to a wide variety of neurological diseases. Here, using an ex vivo nerve exposure model, we show that increasing methylglyoxal produces paranodal disruption, evidenced by disorganized immunostaining of axoglial cell-adhesion proteins, in both sciatic and optic nerves from wild-type mice. Consistent with previous studies showing that increase of methylglyoxal can alter intracellular calcium homeostasis, we found upregulated activity of the calcium-activated protease calpain in sciatic nerves after methylglyoxal exposure. Methylglyoxal exposure altered clusters of proteins that are known as calpain substrates: ezrin in Schwann cell microvilli at the perinodal area and zonula occludens 1 in Schwann cell autotypic junctions at paranodes. Finally, treatment with the calpain inhibitor calpeptin ameliorated methylglyoxal-evoked ezrin loss and paranodal disruption in both sciatic and optic nerves. Our findings strongly suggest that elevated methylglyoxal levels and subsequent calpain activation contribute to the disruption of specialized axoglial domains along myelinated nerve fibers in neurological diseases.
Collapse
Affiliation(s)
- Ryan B Griggs
- 1 Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, USA
| | - Leonid M Yermakov
- 1 Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, USA
| | - Domenica E Drouet
- 1 Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, USA
| | - Duc V M Nguyen
- 1 Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, USA
| | - Keiichiro Susuki
- 1 Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, USA
| |
Collapse
|
18
|
Mostofa MG, Ghosh A, Li ZG, Siddiqui MN, Fujita M, Tran LSP. Methylglyoxal - a signaling molecule in plant abiotic stress responses. Free Radic Biol Med 2018; 122:96-109. [PMID: 29545071 DOI: 10.1016/j.freeradbiomed.2018.03.009] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 02/16/2018] [Accepted: 03/06/2018] [Indexed: 01/03/2023]
Abstract
Abiotic stresses are the most common harmful factors, adversely affecting all aspects of plants' life. Plants have to elicit appropriate responses against multifaceted effects of abiotic stresses by reprogramming various cellular processes. Signaling molecules play vital roles in sensing environmental stimuli to modulate gene expression, metabolism and physiological processes in plants to cope with the adverse effects. Methylglyoxal (MG), a dicarbonyl compound, is known to accumulate in cells as a byproduct of various metabolic pathways, including glycolysis. Several works in recent years have demonstrated that MG could play signaling roles via Ca2+, reactive oxygen species (ROS), K+ and abscisic acid. Recently, global gene expression profiling has shown that MG could induce signaling cascades, and an overlap between MG-responsive and stress-responsive signaling events might exist in plants. Once overaccumulated in cells, MG can provoke detrimental effects by generating ROS, forming advanced glycation end products and inactivating antioxidant systems. Plants are also equipped with MG-detoxifying glyoxalase system to save cellular organelles from MG toxicity. Since MG has regulatory functions in plant growth and development, and glyoxalase system is an integral component of abiotic stress adaptation, an in-depth understanding on MG metabolism and glyoxalase system will help decipher mechanisms underlying plant responses to abiotic stresses. Here, we provide a comprehensive update on the current knowledge of MG production and detoxification in plants, and highlight the putative functions of glyoxalase system in mediating plant defense against abiotic stresses. We particularly emphasize on the dual roles of MG and its connection with glutathione-related redox regulation, which is crucial for plant defense and adaptive responses under changing environmental conditions.
Collapse
Affiliation(s)
- Mohammad Golam Mostofa
- Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur 1706, Bangladesh.
| | - Ajit Ghosh
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh.
| | - Zhong-Guang Li
- School of Life Sciences, Yunnan Normal University, Kunming 650500, PR China.
| | - Md Nurealam Siddiqui
- Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur 1706, Bangladesh.
| | - Masayuki Fujita
- Laboratory of Plant Stress Responses, Department of Applied Biological Science, Faculty of Agriculture, Kagawa University, Miki, Kagawa 761-0795, Japan.
| | - Lam-Son Phan Tran
- Plant Stress Research Group & Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, 700000, Vietnam; Signaling Pathway Research Unit, RIKEN Center for Sustainable Resource Science, 1-7-22, Suehiro-cho, Tsurumi, Yokohama 230-0045, Japan.
| |
Collapse
|
19
|
Riedinger C, Mendler M, Schlotterer A, Fleming T, Okun J, Hammes HP, Herzig S, Nawroth PP. High-glucose toxicity is mediated by AICAR-transformylase/IMP cyclohydrolase and mitigated by AMP-activated protein kinase in Caenorhabditis elegans. J Biol Chem 2018; 293:4845-4859. [PMID: 29414769 PMCID: PMC5880143 DOI: 10.1074/jbc.m117.805879] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 01/29/2018] [Indexed: 11/06/2022] Open
Abstract
The enzyme AICAR-transformylase/IMP cyclohydrolase (ATIC) catalyzes the last two steps of purine de novo synthesis. It metabolizes 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR), which is an AMP analogue, leading to activation of AMP-activated kinase (AMPK). We investigated whether the AICAR-ATIC pathway plays a role in the high glucose (HG)-mediated DNA damage response and AICAR-mediated AMPK activation, explaining the detrimental effects of glucose on neuronal damage and shortening of the lifespan. HG up-regulated the expression and activity of the Caenorhabditis elegans homologue of ATIC, C55F2.1 (atic-1), and increased the levels of reactive oxygen species and methylglyoxal-derived advanced glycation end products. Overexpression of atic-1 decreased the lifespan and head motility and increased neuronal damage under both standard and HG conditions. Inhibition of atic-1 expression, by RNAi, under HG was associated with increased lifespan and head motility and reduced neuronal damage, reactive oxygen species, and methylglyoxal-derived advanced glycation end product accumulation. This effect was independent of an effect on DNA damage or antioxidant defense pathways, such as superoxide dismutase (sod-3) or glyoxalase-1 (glod-4), but was dependent on AMPK and accumulation of AICAR. Through AMPK, AICAR treatment also reduced the negative effects of HG. The mitochondrial inhibitor rotenone abolished the AICAR/AMPK-induced amelioration of HG effects, pointing to mitochondria as a prime target of the glucotoxic effects in C. elegans We conclude that atic-1 is involved in glucotoxic effects under HG conditions, either by blocked atic-1 expression or via AICAR and AMPK induction.
Collapse
Affiliation(s)
- Christin Riedinger
- Department of Medicine I and Clinical Chemistry, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Michael Mendler
- Department of Medicine I and Clinical Chemistry, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Andrea Schlotterer
- Department of Medicine I and Clinical Chemistry, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Thomas Fleming
- Department of Medicine I and Clinical Chemistry, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Jürgen Okun
- Department of Pediatrics, Dietmar Hopp Metabolism Centre, 69120 Heidelberg, Germany
| | - Hans-Peter Hammes
- V. Medical Hospital, University Hospital Mannheim, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Stephan Herzig
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany; Joint Heidelberg Institute for Diabetes and Cancer Translational Diabetes Program, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; German Center for Diabetes Research, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Peter P Nawroth
- Department of Medicine I and Clinical Chemistry, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; Joint Heidelberg Institute for Diabetes and Cancer Translational Diabetes Program, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; German Center for Diabetes Research, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany.
| |
Collapse
|
20
|
Polydatin Prevents Methylglyoxal-Induced Apoptosis through Reducing Oxidative Stress and Improving Mitochondrial Function in Human Umbilical Vein Endothelial Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:7180943. [PMID: 29057033 PMCID: PMC5615983 DOI: 10.1155/2017/7180943] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 07/10/2017] [Accepted: 08/06/2017] [Indexed: 11/20/2022]
Abstract
Methylglyoxal (MGO), an active metabolite of glucose, has been reported to induce vascular cell apoptosis in diabetic complication. Polydatin (PD), a small natural compound from Polygonum cuspidatum, has a number of biological functions, such as antioxidative, anti-inflammatory, and nephroprotective properties. However, the protective effects of PD on MGO-induced apoptosis in endothelial cells remain to be elucidated. In this study, human umbilical vein endothelial cells (HUVECs) were used to explore the effects of PD on MGO-induced cell apoptosis and the possible mechanism involved. HUVECs were pretreated with PD for 2 h, followed by stimulation with MGO. Then cell apoptosis, reactive oxygen species (ROS) generation, mitochondrial membrane potential (MMP) impairment, mitochondrial morphology alterations, and Akt phosphorylation were assessed. The results demonstrated that PD significantly prevented MGO-induced HUVEC apoptosis. PD pretreatment also significantly inhibited MGO-induced ROS production, MMP impairment, mitochondrial morphology changes, and Akt dephosphorylation. These results and the experiments involving N-acetyl cysteine (antioxidant), Cyclosporin A (mitochondrial protector), and LY294002 (Akt inhibitor) suggest that PD prevents MGO-induced HUVEC apoptosis, at least in part, through inhibiting oxidative stress, maintaining mitochondrial function, and activating Akt pathway. All of these data indicate the potential application of PD for the treatment of diabetic vascular complication.
Collapse
|
21
|
Wu CC, Lan HM, Han ST, Chaou CH, Yeh CF, Liu SH, Li CH, Blaney GN, Liu ZY, Chen KF. Comparison of diagnostic accuracy in sepsis between presepsin, procalcitonin, and C-reactive protein: a systematic review and meta-analysis. Ann Intensive Care 2017; 7:91. [PMID: 28875483 PMCID: PMC5585118 DOI: 10.1186/s13613-017-0316-z] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 08/14/2017] [Indexed: 02/06/2023] Open
Abstract
Background The soluble cluster of differentiation 14 (or presepsin) is a free fragment of glycoprotein expressed on monocytes and macrophages. Although many studies have been conducted recently, the diagnostic performance of presepsin for sepsis remains debated. We performed a systematic review and meta-analysis of the available literature to assess the accuracy of presepsin for the diagnosis of sepsis in adult patients and compared the performance between presepsin, C-reactive protein (CRP), and procalcitonin (PCT). Methods A comprehensive systemic search was conducted in PubMed, EMBASE, and Google Scholar for studies that evaluated the diagnostic accuracy of presepsin for sepsis until January 2017. The hierarchical summary receiver operating characteristic method was used to pool individual sensitivity, specificity, diagnostic odds ratio (DOR), positive likelihood ratio (PLR), negative likelihood ratio (NLR), and area under the receiver operating characteristic curve (AUC). Results Eighteen studies, comprising 3470 patients, met our inclusion criteria. The pooled diagnosis sensitivity and specificity of presepsin for sepsis were 0.84 (95% CI 0.80–0.87) and 0.76 (95% CI 0.67–0.82), respectively. Furthermore, the pooled DOR, PLR, NLR, and AUC were 16 (95% CI 10–25), 3.4 (95% CI 2.5–4.6), 0.22 (95% CI 0.17–0.27), and 0.88 (95% CI 0.85–0.90), respectively. Significant heterogeneity was found in both sensitivities (Cochrane Q = 137.43, p < 0.001, I2 = 87.63%) and specificities (Cochrane Q = 180.76, p < 0.001, I2 = 90.60%). Additionally, we found no significant difference between presepsin and PCT (AUC 0.87 vs. 0.86) or CRP (AUC 0.85 vs. 0.85). However, for studies conducted in ICU, the pooled sensitivity of presepsin was found to be higher than PCT (0.88, 95% CI 0.82–0.92 vs. 0.75, 95% CI 0.68–0.81), while the pooled specificity of presepsin was lower than PCT (0.58, 95% CI 0.42–0.73 vs. 0.75, 95% CI 0.65–0.83). Conclusion Based on the results of our meta-analysis, presepsin is a promising marker for diagnosis of sepsis as PCT or CRP, but its results should be interpreted more carefully and cautiously since too few studies were included and those studies had high heterogeneity between them. In addition, continuing re-evaluation during the course of sepsis is advisable.
Collapse
Affiliation(s)
- Chin-Chieh Wu
- Department of Emergency Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Hao-Min Lan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shih-Tsung Han
- Department of Emergency Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chung-Hsien Chaou
- Department of Emergency Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chun-Fu Yeh
- Division of Infectious Diseases, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Su-Hsun Liu
- School of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Family Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chih-Huang Li
- Department of Emergency Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Gerald N Blaney
- Department of Emergency Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Zhen-Ying Liu
- Department of Emergency Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Kuan-Fu Chen
- Department of Emergency Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan. .,Department of Emergency Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan. .,Clinical Informatics and Medical Statistics Research Center, Chang Gung University, 5 Fu-Shin Street, Gueishan Village, Taoyuan, 333, Taiwan. .,Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung, Taiwan.
| |
Collapse
|
22
|
Wang WC, Chou CK, Chuang MC, Li YC, Lee JA. Elevated levels of liver methylglyoxal and d
-lactate in early-stage hepatitis in rats. Biomed Chromatogr 2017; 32. [DOI: 10.1002/bmc.4039] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 05/22/2017] [Accepted: 06/20/2017] [Indexed: 01/29/2023]
Affiliation(s)
- Wen-Chuang Wang
- Department of Pathology; Chia-Yi Christian Hospital; Chia-Yi City Taiwan
| | - Chu-Kuang Chou
- Division of Gastroenterology and Hepatology, Department of Internal Medicine; Chia-Yi Christian Hospital; Chia-Yi City Taiwan
- Department of Internal Medicine National Taiwan University Hospital; Taipei City Taiwan
| | - Ming-Cheng Chuang
- School of Pharmacy, College of Pharmacy; Taipei Medical University; Taipei Taiwan
| | - Yi-Chieh Li
- School of Pharmacy, College of Pharmacy; Taipei Medical University; Taipei Taiwan
| | - Jen-Ai Lee
- School of Pharmacy, College of Pharmacy; Taipei Medical University; Taipei Taiwan
| |
Collapse
|
23
|
Schmoch T, Uhle F, Siegler BH, Fleming T, Morgenstern J, Nawroth PP, Weigand MA, Brenner T. The Glyoxalase System and Methylglyoxal-Derived Carbonyl Stress in Sepsis: Glycotoxic Aspects of Sepsis Pathophysiology. Int J Mol Sci 2017; 18:E657. [PMID: 28304355 PMCID: PMC5372669 DOI: 10.3390/ijms18030657] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 03/08/2017] [Accepted: 03/14/2017] [Indexed: 01/08/2023] Open
Abstract
Sepsis remains one of the leading causes of death in intensive care units. Although sepsis is caused by a viral, fungal or bacterial infection, it is the dysregulated generalized host response that ultimately leads to severe dysfunction of multiple organs and death. The concomitant profound metabolic changes are characterized by hyperglycemia, insulin resistance, and profound transformations of the intracellular energy supply in both peripheral and immune cells. A further hallmark of the early phases of sepsis is a massive formation of reactive oxygen (ROS; e.g., superoxide) as well as nitrogen (RNS; e.g., nitric oxide) species. Reactive carbonyl species (RCS) form a third crucial group of highly reactive metabolites, which until today have been not the focus of interest in sepsis. However, we previously showed in a prospective observational clinical trial that patients suffering from septic shock are characterized by significant methylglyoxal (MG)-derived carbonyl stress, with the glyoxalase system being downregulated in peripheral blood mononuclear cells. In this review, we give a detailed insight into the current state of research regarding the metabolic changes that entail an increased MG-production in septicemia. Thus, we point out the special role of the glyoxalase system in the context of sepsis.
Collapse
Affiliation(s)
- Thomas Schmoch
- Department of Anesthesiology, Heidelberg University Hospital, 69120 Heidelberg, Germany.
| | - Florian Uhle
- Department of Anesthesiology, Heidelberg University Hospital, 69120 Heidelberg, Germany.
| | - Benedikt H Siegler
- Department of Anesthesiology, Heidelberg University Hospital, 69120 Heidelberg, Germany.
| | - Thomas Fleming
- Department of Medicine I and Clinical Chemistry, Heidelberg University Hospital, 69120 Heidelberg, Germany.
| | - Jakob Morgenstern
- Department of Medicine I and Clinical Chemistry, Heidelberg University Hospital, 69120 Heidelberg, Germany.
| | - Peter P Nawroth
- Department of Medicine I and Clinical Chemistry, Heidelberg University Hospital, 69120 Heidelberg, Germany.
| | - Markus A Weigand
- Department of Anesthesiology, Heidelberg University Hospital, 69120 Heidelberg, Germany.
| | - Thorsten Brenner
- Department of Anesthesiology, Heidelberg University Hospital, 69120 Heidelberg, Germany.
| |
Collapse
|
24
|
van Bussel BCT, van de Poll MCG, Schalkwijk CG, Bergmans DCJJ. Increased Dicarbonyl Stress as a Novel Mechanism of Multi-Organ Failure in Critical Illness. Int J Mol Sci 2017; 18:ijms18020346. [PMID: 28178202 PMCID: PMC5343881 DOI: 10.3390/ijms18020346] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Revised: 01/16/2017] [Accepted: 02/03/2017] [Indexed: 01/09/2023] Open
Abstract
Molecular pathological pathways leading to multi-organ failure in critical illness are progressively being unravelled. However, attempts to modulate these pathways have not yet improved the clinical outcome. Therefore, new targetable mechanisms should be investigated. We hypothesize that increased dicarbonyl stress is such a mechanism. Dicarbonyl stress is the accumulation of dicarbonyl metabolites (i.e., methylglyoxal, glyoxal, and 3-deoxyglucosone) that damages intracellular proteins, modifies extracellular matrix proteins, and alters plasma proteins. Increased dicarbonyl stress has been shown to impair the renal, cardiovascular, and central nervous system function, and possibly also the hepatic and respiratory function. In addition to hyperglycaemia, hypoxia and inflammation can cause increased dicarbonyl stress, and these conditions are prevalent in critical illness. Hypoxia and inflammation have been shown to drive the rapid intracellular accumulation of reactive dicarbonyls, i.e., through reduced glyoxalase-1 activity, which is the key enzyme in the dicarbonyl detoxification enzyme system. In critical illness, hypoxia and inflammation, with or without hyperglycaemia, could thus increase dicarbonyl stress in a way that might contribute to multi-organ failure. Thus, we hypothesize that increased dicarbonyl stress in critical illness, such as sepsis and major trauma, contributes to the development of multi-organ failure. This mechanism has the potential for new therapeutic intervention in critical care.
Collapse
Affiliation(s)
- Bas C T van Bussel
- Department of Intensive Care, Maastricht University Medical Centre +, Maastricht 6229 HX, The Netherlands.
| | - Marcel C G van de Poll
- Department of Intensive Care, Maastricht University Medical Centre +, Maastricht 6229 HX, The Netherlands.
- Department of Surgery, and NUTRIM School for Nutrition and Translational Research, Maastricht University Medical Centre +, Maastricht 6229 HX, The Netherlands.
| | - Casper G Schalkwijk
- Department of Internal Medicine, and CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre +, Maastricht 6229 HX, The Netherlands.
| | - Dennis C J J Bergmans
- Department of Intensive Care, Maastricht University Medical Centre +, Maastricht 6229 HX, The Netherlands.
| |
Collapse
|
25
|
Brenner T, Uhle F, Fleming T, Wieland M, Schmoch T, Schmitt F, Schmidt K, Zivkovic AR, Bruckner T, Weigand MA, Hofer S. Soluble TREM-1 as a diagnostic and prognostic biomarker in patients with septic shock: an observational clinical study. Biomarkers 2016; 22:63-69. [PMID: 27319606 DOI: 10.1080/1354750x.2016.1204005] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVES The impact of TREM-1-mediated inflammation was investigated in different inflammatory settings. METHODS Secondary analyses of an observational clinical pilot study, including 60 patients with septic shock, 30 postoperative controls and 30 healthy volunteers. RESULTS Plasma levels of sTREM-1 were found to identify patients with septic shock more effectively than procalcitonin and C-reactive protein. Moreover, sTREM-1 was identified to be an early predictor for survival in patients with septic shock. CONCLUSION Due to its diagnostic as well as prognostic value in sepsis syndrome, implementation of sTREM-1 measurements in routine diagnostics should be taken into account.
Collapse
Affiliation(s)
- Thorsten Brenner
- a Department of Anesthesiology , Heidelberg University Hospital , Heidelberg , Germany
| | - Florian Uhle
- a Department of Anesthesiology , Heidelberg University Hospital , Heidelberg , Germany
| | - Thomas Fleming
- b Department of Medicine I and Clinical Chemistry , Heidelberg University Hospital , Heidelberg , Germany
| | - Matthias Wieland
- a Department of Anesthesiology , Heidelberg University Hospital , Heidelberg , Germany
| | - Thomas Schmoch
- a Department of Anesthesiology , Heidelberg University Hospital , Heidelberg , Germany
| | - Felix Schmitt
- a Department of Anesthesiology , Heidelberg University Hospital , Heidelberg , Germany
| | - Karsten Schmidt
- a Department of Anesthesiology , Heidelberg University Hospital , Heidelberg , Germany
| | - Aleksandar R Zivkovic
- a Department of Anesthesiology , Heidelberg University Hospital , Heidelberg , Germany
| | - Thomas Bruckner
- c Institute of Medical Biometry and Informatics , University of Heidelberg , Heidelberg , Germany
| | - Markus A Weigand
- a Department of Anesthesiology , Heidelberg University Hospital , Heidelberg , Germany
| | - Stefan Hofer
- a Department of Anesthesiology , Heidelberg University Hospital , Heidelberg , Germany
| |
Collapse
|
26
|
Grumaz S, Stevens P, Grumaz C, Decker SO, Weigand MA, Hofer S, Brenner T, von Haeseler A, Sohn K. Next-generation sequencing diagnostics of bacteremia in septic patients. Genome Med 2016; 8:73. [PMID: 27368373 PMCID: PMC4930583 DOI: 10.1186/s13073-016-0326-8] [Citation(s) in RCA: 240] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 06/10/2016] [Indexed: 02/08/2023] Open
Abstract
Background Bloodstream infections remain one of the major challenges in intensive care units, leading to sepsis or even septic shock in many cases. Due to the lack of timely diagnostic approaches with sufficient sensitivity, mortality rates of sepsis are still unacceptably high. However a prompt diagnosis of the causative microorganism is critical to significantly improve outcome of bloodstream infections. Although various targeted molecular tests for blood samples are available, time-consuming blood culture-based approaches still represent the standard of care for the identification of bacteria. Methods Here we describe the establishment of a complete diagnostic workflow for the identification of infectious microorganisms from seven septic patients based on unbiased sequence analyses of free circulating DNA from plasma by next-generation sequencing. Results We found significant levels of DNA fragments derived from pathogenic bacteria in samples from septic patients. Quantitative evaluation of normalized read counts and introduction of a sepsis indicating quantifier (SIQ) score allowed for an unambiguous identification of Gram-positive as well as Gram-negative bacteria that exactly matched with blood cultures from corresponding patient samples. In addition, we also identified species from samples where blood cultures were negative. Reads of non-human origin also comprised fragments derived from antimicrobial resistance genes, showing that, in principle, prediction of specific types of resistance might be possible. Conclusions The complete workflow from sample preparation to species identification report could be accomplished in roughly 30 h, thus making this approach a promising diagnostic platform for critically ill patients suffering from bloodstream infections. Electronic supplementary material The online version of this article (doi:10.1186/s13073-016-0326-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Silke Grumaz
- Fraunhofer IGB, Nobelstr. 12, 70569, Stuttgart, Germany
| | - Philip Stevens
- IGVP, University of Stuttgart, Nobelstr. 12, 70569, Stuttgart, Germany.,Center for Integrative Bioinformatics Vienna, Max F. Perutz Laboratories, University of Vienna, Medical University of Vienna, Vienna, Austria
| | | | - Sebastian O Decker
- Department of Anesthesiology, Heidelberg University Hospital, Im Neuenheimer Feld 110, 69120, Heidelberg, Germany
| | - Markus A Weigand
- Department of Anesthesiology, Heidelberg University Hospital, Im Neuenheimer Feld 110, 69120, Heidelberg, Germany
| | - Stefan Hofer
- Department of Anesthesiology, Heidelberg University Hospital, Im Neuenheimer Feld 110, 69120, Heidelberg, Germany
| | - Thorsten Brenner
- Department of Anesthesiology, Heidelberg University Hospital, Im Neuenheimer Feld 110, 69120, Heidelberg, Germany
| | - Arndt von Haeseler
- Center for Integrative Bioinformatics Vienna, Max F. Perutz Laboratories, University of Vienna, Medical University of Vienna, Vienna, Austria.,Bioinformatics and Computational Biology, Faculty of Computer Science, University of Vienna, Vienna, Austria
| | - Kai Sohn
- Fraunhofer IGB, Nobelstr. 12, 70569, Stuttgart, Germany. .,IGVP, University of Stuttgart, Nobelstr. 12, 70569, Stuttgart, Germany.
| |
Collapse
|
27
|
RAGE-mediated inflammation in patients with septic shock. J Surg Res 2016; 202:315-27. [PMID: 27229106 DOI: 10.1016/j.jss.2016.01.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 01/08/2016] [Accepted: 01/13/2016] [Indexed: 01/11/2023]
Abstract
BACKGROUND The receptor for advanced glycation end-products (RAGE)-pathway is described to be a crucial component of the innate immune response in sepsis. The aims of the present study were, therefore, to delineate the kinetics of membrane-bound RAGE expression, to quantify its soluble isoforms, and to determine the extent of metabolic (e.g., AGE-CML) as well as immunologic (e.g., S100A8/A9) ligands in different inflammatory settings in humans. MATERIALS AND METHODS The presented data result from secondary analyses of an observational clinical pilot study, including patients with septic shock (n = 60), postoperative controls (n = 30), and healthy volunteers (n = 30). Surface-bound expression of RAGE by peripheral blood leukocytes was determined by flow cytometry. In addition, plasma levels of sRAGE, esRAGE, AGE-CML, S100A8/A9, S100A8/A9-CML, RBP, RBP-CML, HSA-CML, HMBG-1, and ß-Amyloid were measured using ELISA. RESULTS In patients with septic shock, RAGE expression was significantly increased in comparison to both control groups, which was paralleled by a significant increase in sRAGE plasma levels. Formation of AGE-CML was shown to be dependent on the availability of the unmodified protein. However, the total amount of AGE-CML did not differ significantly between septic patients and healthy volunteers at early stages or was even lower in patients with sepsis at later stages. In contrast, immunologic ligands (e.g., S100A8/A9) were shown to be significantly elevated in septic patients within the entire study period. CONCLUSIONS Activation of the RAGE-pathway was shown to be of relevance in patients with septic shock, mainly driven by an increase in immunologic (e.g., S100A8/A9) rather than metabolic ligands (e.g., CML-derived AGE-formation).
Collapse
|
28
|
Role of the RAGE Axis during the Immune Response after Severe Trauma: A Prospective Pilot Study. Mediators Inflamm 2015; 2015:691491. [PMID: 26880860 PMCID: PMC4736010 DOI: 10.1155/2015/691491] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 11/24/2015] [Accepted: 12/09/2015] [Indexed: 01/07/2023] Open
Abstract
Background. Severe traumatization induces a complex pathophysiology, driven by the patient's own immune system. The initial activation is a result of damage-associated molecular patterns, which are released from disrupted and dying cells and recognized by immune receptors, for example, RAGE. In this study we aimed to evaluate the contribution of the RAGE axis to early and late immune responses. Methods. We enrolled 16 patients with severe trauma together with 10 patients after major abdominal surgery and 10 healthy volunteers. Blood samples were taken on admission and every 48 h for a total of 8 days. Plasma concentrations of various RAGE ligands as well as RAGE isoforms and IL-6 were measured by ELISA. Monocyte surface expression of RAGE and HLA-DR was assessed by flow cytometry. Results. High and transient levels of IL-6 and methylglyoxal characterize the early immune response after trauma, whereas samples from later time points provide evidence for a secondary release of RAGE ligands. Conclusion. Our results provide evidence for a persisting activation of the RAGE axis while classical mediators like IL-6 disappear early. Considering the immunocompromised phenotype of the monocytes, the RAGE ligands might be substantial contributors to the well-known secondary stage of impaired immune responsiveness in trauma patients.
Collapse
|
29
|
Becze Z, Molnár Z, Fazakas J. Can procalcitonin levels indicate the need for adjunctive therapies in sepsis? Int J Antimicrob Agents 2015; 46 Suppl 1:S13-8. [PMID: 26621136 DOI: 10.1016/j.ijantimicag.2015.11.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
After decades of extensive experimental and clinical research, septic shock and the related multiple organ dysfunction still remain the leading cause of mortality in intensive care units (ICUs) worldwide. Defining sepsis is a difficult task, but what is even more challenging is differentiating infection-induced from non-infection-induced systemic inflammatory response-related multiple organ dysfunction. As conventional signs of infection are often unreliable in intensive care, biomarkers are used, of which one of the most frequently investigated is procalcitonin. Early stabilisation of vital functions via adequate supportive therapy and antibiotic treatment has resulted in substantial improvements in outcome over the last decades. However, there are certain patients who may need extra help, hence modulation of the immune system and the host's response may also be an important therapeutic approach in these situations. Polyclonal intravenous immunoglobulins have been used in critical care for decades. A relatively new potential approach could be attenuation of the overwhelming cytokine storm by specific cytokine adsorbents. Both interventions have been applied in daily practice on a large scale, with firm pathophysiological rationale but weak evidence supported by clinical trials. The purpose of this review is to give an overview on the pathophysiology of sepsis as well as the role and interpretation of biomarkers and their potential use in assisting adjunctive therapies in sepsis in the future.
Collapse
Affiliation(s)
- Zsolt Becze
- Department of ENT, Siófok District Hospital, Siófok, Hungary.
| | - Zsolt Molnár
- Department of Anesthesiology and Intensive Therapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - János Fazakas
- Department of Transplantation and Surgery, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
30
|
Burke-Gaffney A, Creagh-Brown BC. Clinical solutions: not always what they seem? CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2015; 19:213. [PMID: 25947007 PMCID: PMC4423094 DOI: 10.1186/s13054-015-0870-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Anne Burke-Gaffney
- Vascular Biology, Cardiovascular Sciences, National Heart and Lung Institute Division, Faculty of Medicine, Imperial College London, Dovehouse Street, London, SW3 6LY, UK.
| | - Benedict C Creagh-Brown
- Intensive Care and Respiratory Medicine, The Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford, GU2 7XX, UK. .,Surrey Perioperative Anaesthesia and Critical care collaborative Research group (SPACeR), Faculty of Health and Medical Science, Duke of Kent Building, University of Surrey, Guildford, Surrey, GU2 7TE, UK.
| |
Collapse
|
31
|
Weber GF, Chousterman BG, He S, Fenn AM, Nairz M, Anzai A, Brenner T, Uhle F, Iwamoto Y, Robbins CS, Noiret L, Maier SL, Zönnchen T, Rahbari NN, Schölch S, Klotzsche-von Ameln A, Chavakis T, Weitz J, Hofer S, Weigand MA, Nahrendorf M, Weissleder R, Swirski FK. Interleukin-3 amplifies acute inflammation and is a potential therapeutic target in sepsis. Science 2015; 347:1260-5. [PMID: 25766237 PMCID: PMC4376966 DOI: 10.1126/science.aaa4268] [Citation(s) in RCA: 265] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Sepsis is a frequently fatal condition characterized by an uncontrolled and harmful host reaction to microbial infection. Despite the prevalence and severity of sepsis, we lack a fundamental grasp of its pathophysiology. Here we report that the cytokine interleukin-3 (IL-3) potentiates inflammation in sepsis. Using a mouse model of abdominal sepsis, we showed that innate response activator B cells produce IL-3, which induces myelopoiesis of Ly-6C(high) monocytes and neutrophils and fuels a cytokine storm. IL-3 deficiency protects mice against sepsis. In humans with sepsis, high plasma IL-3 levels are associated with high mortality even after adjusting for prognostic indicators. This study deepens our understanding of immune activation, identifies IL-3 as an orchestrator of emergency myelopoiesis, and reveals a new therapeutic target for treating sepsis.
Collapse
Affiliation(s)
- Georg F Weber
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA. Department of Visceral, Thoracic and Vascular Surgery, Technische Universität Dresden, Dresden, Germany.
| | - Benjamin G Chousterman
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Shun He
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ashley M Fenn
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Manfred Nairz
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Atsushi Anzai
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Thorsten Brenner
- Department of Anesthesiology, University of Heidelberg, Heidelberg, Germany
| | - Florian Uhle
- Department of Anesthesiology, University of Heidelberg, Heidelberg, Germany
| | - Yoshiko Iwamoto
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Clinton S Robbins
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lorette Noiret
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sarah L Maier
- Department of Visceral, Thoracic and Vascular Surgery, Technische Universität Dresden, Dresden, Germany
| | - Tina Zönnchen
- Department of Visceral, Thoracic and Vascular Surgery, Technische Universität Dresden, Dresden, Germany
| | - Nuh N Rahbari
- Department of Visceral, Thoracic and Vascular Surgery, Technische Universität Dresden, Dresden, Germany
| | - Sebastian Schölch
- Department of Visceral, Thoracic and Vascular Surgery, Technische Universität Dresden, Dresden, Germany
| | - Anne Klotzsche-von Ameln
- Department of Clinical Pathobiochemistry and Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
| | - Triantafyllos Chavakis
- Department of Clinical Pathobiochemistry and Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
| | - Jürgen Weitz
- Department of Visceral, Thoracic and Vascular Surgery, Technische Universität Dresden, Dresden, Germany
| | - Stefan Hofer
- Department of Anesthesiology, University of Heidelberg, Heidelberg, Germany
| | - Markus A Weigand
- Department of Anesthesiology, University of Heidelberg, Heidelberg, Germany
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA. Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Filip K Swirski
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|