1
|
Ferreira BL, Sousa MB, Leite GGF, Brunialti MKC, Nishiduka ES, Tashima AK, van der Poll T, Salomão R. Glucose metabolism is upregulated in the mononuclear cell proteome during sepsis and supports endotoxin-tolerant cell function. Front Immunol 2022; 13:1051514. [PMID: 36466921 PMCID: PMC9718365 DOI: 10.3389/fimmu.2022.1051514] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 11/01/2022] [Indexed: 07/24/2023] Open
Abstract
Metabolic adaptations shape immune cell function. In the acute response, a metabolic switch towards glycolysis is necessary for mounting a proinflammatory response. During the clinical course of sepsis, both suppression and activation of immune responses take place simultaneously. Leukocytes from septic patients present inhibition of cytokine production while other functions such as phagocytosis and production of reactive oxygen species (ROS) are preserved, similarly to the in vitro endotoxin tolerance model, where a first stimulation with lipopolysaccharide (LPS) affects the response to a second stimulus. Here, we sought to investigate how cellular metabolism is related to the modulation of immune responses in sepsis and endotoxin tolerance. Proteomic analysis in peripheral blood mononuclear cells (PBMCs) from septic patients obtained at intensive care unit admission showed an upregulation of proteins related to glycolysis, the pentose phosphate pathway (PPP), production of ROS and nitric oxide, and downregulation of proteins in the tricarboxylic acid cycle and oxidative phosphorylation compared to healthy volunteers. Using the endotoxin-tolerance model in PBMCs from healthy subjects, we observed increased lactate production in control cells upon LPS stimulation, while endotoxin-tolerant cells presented inhibited tumor necrosis factor-α and lactate production along with preserved phagocytic capacity. Inhibition of glycolysis and PPP led to impairment of phagocytosis and cytokine production both in control and in endotoxin-tolerant cells. These data indicate that glucose metabolism supports leukocyte functions even in a condition of endotoxin tolerance.
Collapse
Affiliation(s)
- Bianca Lima Ferreira
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Mônica Bragança Sousa
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | - Milena Karina Colo Brunialti
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Erika Sayuri Nishiduka
- Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Alexandre Keiji Tashima
- Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Tom van der Poll
- Center of Experimental and Molecular Medicine, Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam, Netherlands
- Division of Infectious Diseases, Amsterdam Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Reinaldo Salomão
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
2
|
Harmon MBA, Scicluna BP, Wiewel MA, Schultz MJ, Horn J, Cremer OL, van der Poll T, Joost Wiersinga W, Juffermans NP. Patients with hypothermic sepsis have a unique gene expression profile compared to patients with fever and sepsis. J Cell Mol Med 2022; 26:1896-1904. [PMID: 35934940 PMCID: PMC8980902 DOI: 10.1111/jcmm.17156] [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: 04/12/2021] [Revised: 09/13/2021] [Accepted: 11/25/2021] [Indexed: 11/30/2022] Open
Abstract
The pathophysiology of hypothermia during sepsis is unclear. Using genomic profiling of blood leukocytes, we aimed to determine if hypothermia is associated with a different gene expression profile compared to fever during sepsis. Patients with sepsis and either hypothermia or fever within 24 hours after ICU admission were included in the study (n = 168). Hypothermia was defined as body temperature below 36 °C. Fever was defined as body temperature equal to or above 38.3°C. We compared blood gene expression (whole-genome transcriptome in leukocytes) in hypothermic septic compared to febrile septic patients in an unmatched analysis and matched for APACHE IV score and the presence of shock. In total, 67 septic patients were hypothermic and 101 patients were febrile. Hypothermia was associated with a distinct gene expression profile in both unmatched and matched analyses. There were significant differences related to the up- and downregulation of canonical signalling pathways. In the matched analysis, the top upregulated gene was cold-inducible mRNA binding protein (CIRBP) which plays a role in cold-induced suppression of cell proliferation. In addition, we found three signalling pathways significantly upregulated in hypothermic patients compared to febrile patients; tryptophan degradation X, phenylalanine degradation IV and putrescine degradation III. In conclusion, there are distinct signalling pathways and genes associated with hypothermia, including tryptophan degradation and CIRBP expression, providing a possible link to the modulation of body temperature and early immunosuppression. Future studies may focus on the canonical signalling pathways presented in this paper to further investigate spontaneous hypothermia in sepsis.
Collapse
Affiliation(s)
- Matthew B. A. Harmon
- Department of Intensive CareAmsterdam University Medical Centerslocation Academic Medical CentreUniversity of AmsterdamAmsterdamThe Netherlands
- Laboratory of Experimental Intensive Care and AnesthesiologyAmsterdam University Medical Centerslocation Academic Medical CentreUniversity of AmsterdamAmsterdamThe Netherlands
| | - Brendon P. Scicluna
- Center for Experimental & Molecular MedicineAmsterdam University Medical Centerslocation Academic Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
| | - Maryse A. Wiewel
- Center for Experimental & Molecular MedicineAmsterdam University Medical Centerslocation Academic Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
| | - Marcus J. Schultz
- Department of Intensive CareAmsterdam University Medical Centerslocation Academic Medical CentreUniversity of AmsterdamAmsterdamThe Netherlands
- Laboratory of Experimental Intensive Care and AnesthesiologyAmsterdam University Medical Centerslocation Academic Medical CentreUniversity of AmsterdamAmsterdamThe Netherlands
- Mahidol Oxford Research UnitMahidol UniversityBangkokThailand
- Nuffield Department of MedicineUniversity of OxfordOxfordUK
| | - Janneke Horn
- Department of Intensive CareAmsterdam University Medical Centerslocation Academic Medical CentreUniversity of AmsterdamAmsterdamThe Netherlands
- Laboratory of Experimental Intensive Care and AnesthesiologyAmsterdam University Medical Centerslocation Academic Medical CentreUniversity of AmsterdamAmsterdamThe Netherlands
| | - Olaf L. Cremer
- Department of Intensive Care MedicineUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Tom van der Poll
- Center for Experimental & Molecular MedicineAmsterdam University Medical Centerslocation Academic Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
| | - W. Joost Wiersinga
- Center for Experimental & Molecular MedicineAmsterdam University Medical Centerslocation Academic Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
| | - Nicole P. Juffermans
- Department of Intensive CareAmsterdam University Medical Centerslocation Academic Medical CentreUniversity of AmsterdamAmsterdamThe Netherlands
- Laboratory of Experimental Intensive Care and AnesthesiologyAmsterdam University Medical Centerslocation Academic Medical CentreUniversity of AmsterdamAmsterdamThe Netherlands
| | | |
Collapse
|
3
|
Stothers CL, Burelbach KR, Owen AM, Patil NK, McBride MA, Bohannon JK, Luan L, Hernandez A, Patil TK, Williams DL, Sherwood ER. β-Glucan Induces Distinct and Protective Innate Immune Memory in Differentiated Macrophages. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:2785-2798. [PMID: 34740960 PMCID: PMC8612974 DOI: 10.4049/jimmunol.2100107] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 09/27/2021] [Indexed: 12/14/2022]
Abstract
Bacterial infections are a common and deadly threat to vulnerable patients. Alternative strategies to fight infection are needed. β-Glucan, an immunomodulator derived from the fungal cell wall, provokes resistance to infection by inducing trained immunity, a phenomenon that persists for weeks to months. Given the durability of trained immunity, it is unclear which leukocyte populations sustain this effect. Macrophages have a life span that surpasses the duration of trained immunity. Thus, we sought to define the contribution of differentiated macrophages to trained immunity. Our results show that β-glucan protects mice from Pseudomonas aeruginosa infection by augmenting recruitment of innate leukocytes to the site of infection and facilitating local clearance of bacteria, an effect that persists for more than 7 d. Adoptive transfer of macrophages, trained using β-glucan, into naive mice conferred a comparable level of protection. Trained mouse bone marrow-derived macrophages assumed an antimicrobial phenotype characterized by enhanced phagocytosis and reactive oxygen species production in parallel with sustained enhancements in glycolytic and oxidative metabolism, increased mitochondrial mass, and membrane potential. β-Glucan induced broad transcriptomic changes in macrophages consistent with early activation of the inflammatory response, followed by sustained alterations in transcripts associated with metabolism, cellular differentiation, and antimicrobial function. Trained macrophages constitutively secreted CCL chemokines and robustly produced proinflammatory cytokines and chemokines in response to LPS challenge. Induction of the trained phenotype was independent of the classic β-glucan receptors Dectin-1 and TLR-2. These findings provide evidence that β-glucan induces enhanced protection from infection by driving trained immunity in macrophages.
Collapse
Affiliation(s)
- Cody L Stothers
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN;
| | - Katherine R Burelbach
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN; and
| | - Allison M Owen
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN; and
| | - Naeem K Patil
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN; and
| | - Margaret A McBride
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Julia K Bohannon
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN; and
| | - Liming Luan
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN; and
| | - Antonio Hernandez
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN; and
| | - Tazeen K Patil
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN; and
| | - David L Williams
- Center for Inflammation, Infectious Disease and Immunity, Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN
| | - Edward R Sherwood
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN; and
| |
Collapse
|
4
|
Santos SS, Brunialti MKC, Soriano FG, Szabo C, Salomão R. Repurposing of Clinically Approved Poly-(ADP-Ribose) Polymerase Inhibitors for the Therapy of Sepsis. Shock 2021; 56:901-909. [PMID: 34115723 DOI: 10.1097/shk.0000000000001820] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT Sepsis' pathogenesis involves multiple mechanisms that lead to a dysregulation of the host's response. Significant efforts have been made in search of interventions that can reverse this situation and increase patient survival. Poly (ADP-polymerase) (PARP) is a constitutive nuclear and mitochondrial enzyme, which functions as a co-activator and co-repressor of gene transcription, thus regulating the production of inflammatory mediators. Several studies have already demonstrated an overactivation of PARP1 in various human pathophysiological conditions and that its inhibition has benefits in regulating intracellular processes. The PARP inhibitor olaparib, originally developed for cancer therapy, paved the way for the expansion of its clinical use for nononcological indications. In this review we discuss sepsis as one of the possible indications for the use of olaparib and other clinically approved PARP inhibitors as modulators of the inflammatory response and cellular dysfunction. The benefit of olaparib and other clinically approved PARP inhibitors has already been demonstrated in several experimental models of human diseases, such as neurodegeneration and neuroinflammation, acute hepatitis, skeletal muscle disorders, aging and acute ischemic stroke, protecting, for example, from the deterioration of the blood-brain barrier, restoring the cellular levels of NAD+, improving mitochondrial function and biogenesis and, among other effects, reducing oxidative stress and pro-inflammatory mediators, such as TNF-α, IL1-β, IL-6, and VCAM1. These data demonstrated that repositioning of clinically approved PARP inhibitors may be effective in protecting against hemodynamic dysfunction, metabolic dysfunction, and multiple organ failure in patients with sepsis. Age and gender affect the response to PARP inhibitors, the mechanisms underlying the lack of many protective effects in females and aged animals should be further investigated and be cautiously considered in designing clinical trials.
Collapse
Affiliation(s)
- Sidnéia Sousa Santos
- Division of Infectious Diseasses, Paulista School of Medicine, Federal University of Sao Paulo, Brazil
| | | | - Francisco Garcia Soriano
- Laboratory of Medical Research, Faculty of Medicine of the University of São Paulo-USP, São Paulo, Brazil
| | - Csaba Szabo
- Chair of Pharmacology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Reinaldo Salomão
- Division of Infectious Diseasses, Paulista School of Medicine, Federal University of Sao Paulo, Brazil
| |
Collapse
|
5
|
Abstract
Immunosuppression is the most commonly used concept to qualify the immune status of patients with either sterile systemic inflammatory response syndrome (SIRS) or sepsis. In this review we attempt to demonstrate that the concept of immunosuppression is an oversimplification of the complex anti-inflammatory response that occurs in patients dealing with a severe sterile or infectious insult. Particularly, the immune status of leukocytes varies greatly depending on the compartment from where they are derived from. Furthermore, although certain functions of immune cells present in the blood stream or in the hematopoietic organs can be significantly diminished, other functions are either unchanged or even enhanced. This juxtaposition illustrates that there is no global defect. The mechanisms called reprogramming or trained innate immunity are probably aimed at preventing a generalized deleterious inflammatory reaction, and work to maintain the defense mechanisms at their due levels.
Collapse
|
6
|
Jędrzejewski T, Piotrowski J, Pawlikowska M, Wrotek S, Kozak W. Extract from Coriolus versicolor fungus partially prevents endotoxin tolerance development by maintaining febrile response and increasing IL-6 generation. J Therm Biol 2019; 83:69-79. [PMID: 31331527 DOI: 10.1016/j.jtherbio.2019.05.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/09/2019] [Accepted: 05/10/2019] [Indexed: 11/25/2022]
Abstract
Endotoxin tolerance is defined as a reduced endotoxin-induced fever following repeated injections of lipopolysaccharide (LPS). Clinical examples of endotoxin tolerance include sepsis or cystic fibrosis. This state is characterized by inhibition of pro-inflammatory cytokines production and decrease in nuclear factor-kappa B (NF-κB) activation. Extract from Coriolus versicolor (CV) fungus is classified as a biological response modifier, which exhibits various biological activities, including immunopotentiating properties. The aim of study was to examine the effect of CV extract injection on body core temperature of Wistar rats during LPS-induced endotoxin tolerance. Body temperature was measured using biotelemetry. CV extract was injected intraperitoneally (100 mg kg-1) 2 h prior to the first LPS peritoneal administration (50 μg/kg). Endotoxin tolerance was induced by three consecutive daily injections of LPS at the same dose. We also investigated the influence of CV extract pre-injection on the properties of peripheral blood mononuclear cells (PBMCs) isolated from LPS-treated rats in response to LPS stimulation ex vivo. PBMCs were isolated 2 h after the first LPS injection. After 24 h pre-incubation, the cells were stimulated with LPS (1 μg ml-1) for 4 h. Our results revealed that CV extract partially prevents endotoxin tolerance through maintaining febrile response in rats following consecutive exposure to LPS. This state was accompanied by the ability of PBMCs isolated from rats injected with CV extract and LPS to release larger amounts of interleukin 6 and greater NF-κB activation in response to LPS stimulation ex vivo compared with the cells derived from rats injected only with LPS. Data also showed that CV extract augmented mitogenic effect of LPS on PBMCs and caused increase in reactive oxygen species generation. We concluded that CV extract, by a modifying effect on body temperature during endotoxin tolerance, can be consider as the immunostimulating agent, which prevents the non-specific refractoriness described in patients with sepsis or ischemia.
Collapse
Affiliation(s)
- Tomasz Jędrzejewski
- Department of Immunology, Faculty of Biology and Environment Protection, Nicolaus Copernicus University, 1 Lwowska Street, Torun, 87-100, Poland.
| | - Jakub Piotrowski
- Department of Immunology, Faculty of Biology and Environment Protection, Nicolaus Copernicus University, 1 Lwowska Street, Torun, 87-100, Poland.
| | - Małgorzata Pawlikowska
- Department of Immunology, Faculty of Biology and Environment Protection, Nicolaus Copernicus University, 1 Lwowska Street, Torun, 87-100, Poland.
| | - Sylwia Wrotek
- Department of Immunology, Faculty of Biology and Environment Protection, Nicolaus Copernicus University, 1 Lwowska Street, Torun, 87-100, Poland.
| | - Wieslaw Kozak
- Department of Immunology, Faculty of Biology and Environment Protection, Nicolaus Copernicus University, 1 Lwowska Street, Torun, 87-100, Poland.
| |
Collapse
|
7
|
Salomão R, Ferreira B, Salomão M, Santos S, Azevedo L, Brunialti M. Sepsis: evolving concepts and challenges. Braz J Med Biol Res 2019; 52:e8595. [PMID: 30994733 PMCID: PMC6472937 DOI: 10.1590/1414-431x20198595] [Citation(s) in RCA: 184] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 02/11/2019] [Indexed: 12/15/2022] Open
Abstract
Sepsis remains a major cause of morbidity and mortality worldwide, with increased burden in low- and middle-resource settings. The role of the inflammatory response in the pathogenesis of the syndrome has supported the modern concept of sepsis. Nevertheless, a definition of sepsis and the criteria for its recognition is a continuous process, which reflects the growing knowledge of its mechanisms and the success and failure of diagnostic and therapeutic interventions. Here we review the evolving concepts of sepsis, from the "systemic inflammatory response syndrome triggered by infection" (Sepsis-1) to "a severe, potentially fatal, organic dysfunction caused by an inadequate or dysregulated host response to infection" (Sepsis-3). We focused in the pathophysiology behind the concept and the criteria for recognition and diagnosis of sepsis. A major challenge in evaluating the host response in sepsis is to characterize what is protective and what is harmful, and we discuss that, at least in part, the apparent dysregulated host response may be an effort to adapt to a hostile environment. The new criteria for recognition and diagnosis of sepsis were derived from robust databases, restricted, however, to developed countries. Since then, the criteria have been supported in different clinical settings and in different economic and epidemiological contexts, but still raise discussion regarding their use for the identification versus the prognostication of the septic patient. Clinicians should not be restricted to definition criteria when evaluating patients with infection and should wisely use the broad array of information obtained by rigorous clinical observation.
Collapse
Affiliation(s)
- R. Salomão
- Disciplina de Infectologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - B.L. Ferreira
- Disciplina de Infectologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - M.C. Salomão
- Departamento de Moléstias Infecciosas e Parasitárias Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - S.S. Santos
- Disciplina de Infectologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - L.C.P. Azevedo
- Unidade de Terapia Intensiva do Hospital Sírio Libanês, São Paulo, SP, Brasil
| | - M.K.C. Brunialti
- Disciplina de Infectologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| |
Collapse
|
8
|
Fensterheim BA, Young JD, Luan L, Kleinbard RR, Stothers CL, Patil NK, McAtee-Pereira AG, Guo Y, Trenary I, Hernandez A, Fults JB, Williams DL, Sherwood ER, Bohannon JK. The TLR4 Agonist Monophosphoryl Lipid A Drives Broad Resistance to Infection via Dynamic Reprogramming of Macrophage Metabolism. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:3777-3789. [PMID: 29686054 PMCID: PMC5964009 DOI: 10.4049/jimmunol.1800085] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 03/28/2018] [Indexed: 12/21/2022]
Abstract
Monophosphoryl lipid A (MPLA) is a clinically used TLR4 agonist that has been found to drive nonspecific resistance to infection for up to 2 wk. However, the molecular mechanisms conferring protection are not well understood. In this study, we found that MPLA prompts resistance to infection, in part, by inducing a sustained and dynamic metabolic program in macrophages that supports improved pathogen clearance. Mice treated with MPLA had enhanced resistance to infection with Staphylococcus aureus and Candida albicans that was associated with augmented microbial clearance and organ protection. Tissue macrophages, which exhibited augmented phagocytosis and respiratory burst after MPLA treatment, were required for the beneficial effects of MPLA. Further analysis of the macrophage phenotype revealed that early TLR4-driven aerobic glycolysis was later coupled with mitochondrial biogenesis, enhanced malate shuttling, and increased mitochondrial ATP production. This metabolic program was initiated by overlapping and redundant contributions of MyD88- and TRIF-dependent signaling pathways as well as downstream mTOR activation. Blockade of mTOR signaling inhibited the development of the metabolic and functional macrophage phenotype and ablated MPLA-induced resistance to infection in vivo. Our findings reveal that MPLA drives macrophage metabolic reprogramming that evolves over a period of days to support a macrophage phenotype highly effective at mediating microbe clearance and that this results in nonspecific resistance to infection.
Collapse
Affiliation(s)
- Benjamin A Fensterheim
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37212
| | - Jamey D Young
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37212
| | - Liming Luan
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232; and
| | - Ruby R Kleinbard
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232; and
| | - Cody L Stothers
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37212
| | - Naeem K Patil
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232; and
| | | | - Yin Guo
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232; and
| | - Irina Trenary
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235
| | - Antonio Hernandez
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232; and
| | - Jessica B Fults
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232; and
| | - David L Williams
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614
| | - Edward R Sherwood
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37212
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232; and
| | - Julia K Bohannon
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232; and
| |
Collapse
|
9
|
Sharma NK, Tashima AK, Brunialti MKC, Ferreira ER, Torquato RJS, Mortara RA, Machado FR, Assuncao M, Rigato O, Salomao R. Proteomic study revealed cellular assembly and lipid metabolism dysregulation in sepsis secondary to community-acquired pneumonia. Sci Rep 2017; 7:15606. [PMID: 29142235 PMCID: PMC5688086 DOI: 10.1038/s41598-017-15755-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 11/01/2017] [Indexed: 12/21/2022] Open
Abstract
Sepsis is a life-threatening disorder characterized by organ dysfunction and a major cause of mortality worldwide. The major challenge in studying sepsis is its diversity in such factors as age, source of infection and etiology. Recently, genomic and proteomic approaches have improved our understanding of its complex pathogenesis. In the present study, we use quantitative proteomics to evaluate the host proteome response in septic patients secondary to community-acquired pneumonia (CAP). Samples obtained at admission and after 7 days of follow-up were analyzed according to the outcomes of septic patients. The patients' proteome profiles were compared with age- and gender-matched healthy volunteers. Bioinformatic analyses of differentially expressed proteins showed alteration in the cytoskeleton, cellular assembly, movement, lipid metabolism and immune responses in septic patients. Actin and gelsolin changes were assessed in mononuclear cells using immunofluorescence, and a higher expression of gelsolin and depletion of actin were observed in survivor patients. Regarding lipid metabolism, changes in cholesterol, HDL and apolipoproteins were confirmed using enzymatic colorimetric methods in plasma. Transcriptomic studies revealed a massive change in gene expression in sepsis. Our proteomic results stressed important changes in cellular structure and metabolism, which are possible targets for future interventions of sepsis.
Collapse
Affiliation(s)
- Narendra Kumar Sharma
- Division of Infectious Diseases, Escola Paulista de Medicina, Hospital São Paulo, Universidade Federal de Sao Paulo, Sao Paulo, 04039-032, Brazil
| | - Alexandre Keiji Tashima
- Departamento de Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, São Paulo, 04023-900, Brazil
| | - Milena Karina Colo Brunialti
- Division of Infectious Diseases, Escola Paulista de Medicina, Hospital São Paulo, Universidade Federal de Sao Paulo, Sao Paulo, 04039-032, Brazil
| | - Eden Ramalho Ferreira
- Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina, Universidade Federal de Sao Paulo, Sao Paulo, 04023-062, Brazil
| | - Ricardo Jose Soares Torquato
- Departamento de Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, São Paulo, 04023-900, Brazil
| | - Renato Arruda Mortara
- Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina, Universidade Federal de Sao Paulo, Sao Paulo, 04023-062, Brazil
| | - Flavia Ribeiro Machado
- Intensive Care Unit, Hospital São Paulo, Escola Paulista de Medicina, Universidade Federal de Sao Paulo, Sao Paulo, 04024-002, Brazil
| | - Murillo Assuncao
- Intensive Care Unit, Hospital Israelita Albert Einstein, Sao Paulo, 05652- 900, Brazil
| | - Otelo Rigato
- Division of Infectious Diseases, Escola Paulista de Medicina, Hospital São Paulo, Universidade Federal de Sao Paulo, Sao Paulo, 04039-032, Brazil
- Intensive Care Unit, Hospital Sirio Libanes, Sao Paulo, 01409-001, Brazil
| | - Reinaldo Salomao
- Division of Infectious Diseases, Escola Paulista de Medicina, Hospital São Paulo, Universidade Federal de Sao Paulo, Sao Paulo, 04039-032, Brazil.
| |
Collapse
|
10
|
Dobbs KR, Embury P, Vulule J, Odada PS, Rosa BA, Mitreva M, Kazura JW, Dent AE. Monocyte dysregulation and systemic inflammation during pediatric falciparum malaria. JCI Insight 2017; 2:95352. [PMID: 28931756 DOI: 10.1172/jci.insight.95352] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 08/16/2017] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Inflammation and monocytes are thought to be important to human malaria pathogenesis. However, the relationship of inflammation and various monocyte functions to acute malaria, recovery from acute malaria, and asymptomatic parasitemia in endemic populations is poorly understood. METHODS We evaluated plasma cytokine levels, monocyte subsets, monocyte functional responses, and monocyte inflammatory transcriptional profiles of 1- to 10-year-old Kenyan children at the time of presentation with acute uncomplicated malaria and at recovery 6 weeks later; these results were compared with analogous data from asymptomatic children and adults in the same community. RESULTS Acute malaria was marked by elevated levels of proinflammatory and regulatory cytokines and expansion of the inflammatory "intermediate" monocyte subset that returned to levels of healthy asymptomatic children 6 weeks later. Monocytes displayed activated phenotypes during acute malaria, with changes in surface expression of markers important to innate and adaptive immunity. Functionally, acute malaria monocytes and monocytes from asymptomatic infected children had impaired phagocytosis of P. falciparum-infected erythrocytes relative to asymptomatic children with no blood-stage infection. Monocytes from both acute malaria and recovery time points displayed strong and equivalent cytokine responsiveness to innate immune agonists that were independent of infection status. Monocyte transcriptional profiles revealed regulated and balanced proinflammatory and antiinflammatory and altered phagocytosis gene expression patterns distinct from malaria-naive monocytes. CONCLUSION These observations provide insights into monocyte functions and the innate immune response during uncomplicated malaria and suggest that asymptomatic parasitemia in children is not clinically benign. FUNDING Support for this work was provided by NIH/National Institute of Allergy and Infectious Diseases (R01AI095192-05), the Burroughs Wellcome Fund/American Society of Tropical Medicine and Hygiene, and the Rainbow Babies & Children's Foundation.
Collapse
Affiliation(s)
- Katherine R Dobbs
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio, USA.,Division of Pediatric Infectious Diseases, University Hospitals Rainbow Babies and Children's Hospital, Cleveland, Ohio, USA
| | - Paula Embury
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio, USA
| | - John Vulule
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Peter S Odada
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Bruce A Rosa
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Makedonka Mitreva
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri, USA
| | - James W Kazura
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio, USA
| | - Arlene E Dent
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio, USA.,Division of Pediatric Infectious Diseases, University Hospitals Rainbow Babies and Children's Hospital, Cleveland, Ohio, USA
| |
Collapse
|
11
|
Esquerdo KF, Sharma NK, Brunialti MKC, Baggio-Zappia GL, Assunção M, Azevedo LCP, Bafi AT, Salomao R. Inflammasome gene profile is modulated in septic patients, with a greater magnitude in non-survivors. Clin Exp Immunol 2017; 189:232-240. [PMID: 28369745 DOI: 10.1111/cei.12971] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2017] [Indexed: 01/08/2023] Open
Abstract
Inflammasome signalling induces the processing and secretion of interleukin (IL)-1β and IL-18 which, coupled with pyroptosis, activate further the inflammatory response. In the present study we evaluated the expression of genes involved in inflammasome signalling pathways in septic patients, their interaction networks and the predicted functions modulated in survivors and non-survivors. Twenty-seven patients with sepsis secondary to community-acquired pneumonia admitted to intensive care units from three general hospitals in São Paulo were included into the study. We performed a polymerase chain reaction (PCR) array encompassing 35 genes related to the nucleotide-binding oligomerization domain and leucine-rich repeat-containing (NLR)-inflammasome in peripheral blood mononuclear cells obtained at admission and after 7 days of follow-up. Eleven healthy volunteers were used as the reference group. Increased NLRC4 and NLRP3 and decreased nucleotide-binding oligomerization domain (NOD1), and NLRP1 expression was observed in septic patients compared to healthy individuals; the IL-1β and IL-18 expression levels were also high in the patients. The gene expression changes followed the same patterns in surviving and non-surviving patients, with higher magnitudes observed in non-survivors. Functional analyses revealed, however, that activation and inhibition intensity for representing functions were different in survivors and non-survivors, as for production of reactive oxygen species, synthesis of nitric oxide and for the control of bacterial infections. Our results showed that the genes involved in the activation of the NLR-inflammasome cascades were altered substantially in septic patients, with a higher number of altered genes and a higher intensity in the disturbance of gene expression found among patients dying of sepsis.
Collapse
Affiliation(s)
- K F Esquerdo
- Escola Paulista de Medicina, Hospital São Paulo, Universidade Federal de Sao Paulo, São Paulo, Brazil
| | - N K Sharma
- Escola Paulista de Medicina, Hospital São Paulo, Universidade Federal de Sao Paulo, São Paulo, Brazil
| | - M K C Brunialti
- Escola Paulista de Medicina, Hospital São Paulo, Universidade Federal de Sao Paulo, São Paulo, Brazil
| | - G L Baggio-Zappia
- Escola Paulista de Medicina, Hospital São Paulo, Universidade Federal de Sao Paulo, São Paulo, Brazil
| | - M Assunção
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | | | - A T Bafi
- Escola Paulista de Medicina, Hospital São Paulo, Universidade Federal de Sao Paulo, São Paulo, Brazil
| | - R Salomao
- Escola Paulista de Medicina, Hospital São Paulo, Universidade Federal de Sao Paulo, São Paulo, Brazil
| |
Collapse
|
12
|
Fensterheim BA, Guo Y, Sherwood ER, Bohannon JK. The Cytokine Response to Lipopolysaccharide Does Not Predict the Host Response to Infection. THE JOURNAL OF IMMUNOLOGY 2017; 198:3264-3273. [PMID: 28275139 DOI: 10.4049/jimmunol.1602106] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 02/13/2017] [Indexed: 12/18/2022]
Abstract
The magnitude of the LPS-elicited cytokine response is commonly used to assess immune function in critically ill patients. A suppressed response, known as endotoxin tolerance, is associated with worse outcomes, yet endotoxin tolerance-inducing TLR4 ligands are known to protect animals from infection. Thus, it remains unknown whether the magnitude of the LPS-elicited cytokine response provides an accurate assessment of antimicrobial immunity. To address this, the ability of diverse TLR ligands to modify the LPS-elicited cytokine response and resistance to infection were assessed. Priming of mice with LPS, monophosphoryl lipid A (MPLA), or poly(I:C) significantly reduced plasma LPS-elicited proinflammatory cytokines, reflecting endotoxin tolerance, whereas CpG-ODN-primed mice showed augmented cytokine production. In contrast, LPS, MPLA, and CpG-ODN, but not poly(I:C), improved the host response to a Pseudomonas aeruginosa infection. Mice primed with protective TLR ligands, including CpG-ODN, showed reduced plasma cytokines during P. aeruginosa infection. The protection imparted by TLR ligands persisted for up to 15 d yet was independent of the adaptive immune system. In bone marrow-derived macrophages, protective TLR ligands induced a persistent metabolic phenotype characterized by elevated glycolysis and oxidative metabolism as well as augmented size, granularity, phagocytosis, and respiratory burst. Sustained augmentation of glycolysis in TLR-primed cells was dependent, in part, on hypoxia-inducible factor 1-α and was essential for increased phagocytosis. In conclusion, the magnitude of LPS-elicited cytokine production is not indicative of antimicrobial immunity after exposure to TLR ligands. Additionally, protective TLR ligands induce sustained augmentation of phagocyte metabolism and antimicrobial function.
Collapse
Affiliation(s)
- Benjamin A Fensterheim
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37235; and
| | - Yin Guo
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37235; and
| | - Edward R Sherwood
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37235; and.,Anesthesiology Research Division, Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37212
| | - Julia K Bohannon
- Anesthesiology Research Division, Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37212
| |
Collapse
|
13
|
Guzmán-Beltrán S, Torres M, Arellano M, Juárez E. Human macrophages chronically exposed to LPS can be reactivated by stimulation with MDP to acquire an antimicrobial phenotype. Cell Immunol 2017; 315:45-55. [PMID: 28258715 DOI: 10.1016/j.cellimm.2017.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 02/04/2017] [Accepted: 02/18/2017] [Indexed: 01/01/2023]
Abstract
Macrophages are important in host defense and can differentiate into functionally distinct subsets named classically (M1) or alternatively (M2) activated. In several inflammatory disorders, macrophages become tolerized to prevent deleterious consequences. This tolerization reduces the ability of macrophages to respond to bacterial components (e.g., LPS) maintaining a low level of inflammation but compromising the ability of macrophages to mount an effective immune response during subsequent pathogen encounters. In this study, we aimed to reactivate human monocyte-derived macrophages chronically exposed to LPS by re-stimulation with muramyl dipeptide (MDP). We observed an undefined profile of cell surface marker expression during endotoxin tolerance and absence of TNFα production. Stimulating macrophages chronically exposed to LPS with LPS+MDP restored TNFα, production together with an increased production of IL1, IL6, IFNγ, IL4, IL5 and IL10. These results suggest that macrophages chronically exposed to LPS possess a mixed M1-M2 phenotype with sufficient antimicrobial and homeostatic potential.
Collapse
Affiliation(s)
- Silvia Guzmán-Beltrán
- Departamento de Investigación en Microbiología, Instituto Nacional de Enfermedades Respiratorias, México City, Mexico
| | - Martha Torres
- Departamento de Investigación en Microbiología, Instituto Nacional de Enfermedades Respiratorias, México City, Mexico
| | - Monserrat Arellano
- Departamento de Investigación en Microbiología, Instituto Nacional de Enfermedades Respiratorias, México City, Mexico
| | - Esmeralda Juárez
- Departamento de Investigación en Microbiología, Instituto Nacional de Enfermedades Respiratorias, México City, Mexico.
| |
Collapse
|
14
|
Rios ECS, Soriano FG, Olah G, Gerö D, Szczesny B, Szabo C. Hydrogen sulfide modulates chromatin remodeling and inflammatory mediator production in response to endotoxin, but does not play a role in the development of endotoxin tolerance. JOURNAL OF INFLAMMATION-LONDON 2016; 13:10. [PMID: 27042162 PMCID: PMC4818437 DOI: 10.1186/s12950-016-0119-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 03/28/2016] [Indexed: 02/01/2023]
Abstract
Background Pretreatment with low doses of LPS (lipopolysaccharide, bacterial endotoxin) reduces the pro-inflammatory response to a subsequent higher LPS dose, a phenomenon known as endotoxin tolerance. Moreover, hydrogen sulfide (H2S), an endogenous gaseous mediator (gasotransmitter) can exert anti-inflammatory effects. Here we investigated the potential role of H2S in the development of LPS tolerance. THP1 differentiated macrophages were pretreated with the H2S donor NaHS (1 mM) or the H2S biosynthesis inhibitor aminooxyacetic acid (AOAA, 1 mM). Methods To induce tolerance, cells were treated with a low concentration of LPS (0.5 μg/ml) for 4 or 24 h, and then treated with a high concentration of LPS (1 μg/ml) for 4 h or 24 h. In in vivo studies, male wild-type and CSE-/- mice were randomized to the following groups: Control (vehicle); Endotoxemic saline for 3 days before the induction of endotoxemia with 10 mg/kg LPS) mg/kg; Tolerant (LPS at 1 mg/kg for 3 days, followed LPS at 10 mg/kg). Animals were sacrificed after 4 or 12 h; plasma IL-6 and TNF-α levels were measured. Changes in histone H3 and H4 acetylation were analyzed by Western blotting. Results LPS tolerance decreased pro-inflammatory cytokine production. AOAA did not affect the effect of tolerance on reducing cytokine production. Treatment of the cells with the H2S donor reduced cytokine production. Induction of the tolerance increased the acetylation of H3; AOAA reduced histone acetylation. H2S donation increased histone acetylation. Tolerance did not affect the responses to H2S with respect to histone acetylation. Conclusions In conclusion, both LPS tolerance and H2S donation decrease LPS-induced cytokine production in vitro and modulate histone acetylation. However, endogenous, CSE-derived H2S does not appear to play a significant role in the development of LPS tolerance.
Collapse
Affiliation(s)
- Ester C S Rios
- Department of Emergency Medicine, Universidade de São Paulo Medical School, São Paulo, Brazil
| | - Francisco G Soriano
- Department of Emergency Medicine, Universidade de São Paulo Medical School, São Paulo, Brazil
| | - Gabor Olah
- Department of Anesthesiology, University of Texas Medical Branch, 601 Harborside Drive, Building 21, Room 4.202D, Galveston, TX 77555-1102 USA
| | - Domokos Gerö
- Department of Anesthesiology, University of Texas Medical Branch, 601 Harborside Drive, Building 21, Room 4.202D, Galveston, TX 77555-1102 USA
| | - Bartosz Szczesny
- Department of Anesthesiology, University of Texas Medical Branch, 601 Harborside Drive, Building 21, Room 4.202D, Galveston, TX 77555-1102 USA
| | - Csaba Szabo
- Department of Anesthesiology, University of Texas Medical Branch, 601 Harborside Drive, Building 21, Room 4.202D, Galveston, TX 77555-1102 USA
| |
Collapse
|
15
|
Santos SS, Carmo AM, Brunialti MKC, Machado FR, Azevedo LC, Assunção M, Trevelin SC, Cunha FQ, Salomao R. Modulation of monocytes in septic patients: preserved phagocytic activity, increased ROS and NO generation, and decreased production of inflammatory cytokines. Intensive Care Med Exp 2016; 4:5. [PMID: 26879814 PMCID: PMC4754229 DOI: 10.1186/s40635-016-0078-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 02/09/2016] [Indexed: 01/22/2023] Open
Abstract
Background The nature of the inflammatory response underscoring the pathophysiology of sepsis has been extensively studied. We hypothesized that different cell functions would be differentially regulated in a patient with sepsis. We evaluated the modulation of monocyte functions during sepsis by simultaneously assessing their phagocytic activity, the generation of reactive oxygen species (ROS) and nitric oxide (NO), and the production of inflammatory cytokines (IL-6 and TNF-α). Methods Whole blood was obtained from patients with severe sepsis and septic shock both at admission (D0, n = 34) and after seven days of therapy (D7, n = 15); 19 healthy volunteers were included as a control group. The cells were stimulated with LPS, Pseudomonas aeruginosa, and Staphylococcus aureus. The ROS and NO levels were quantified in monocytes in whole blood by measuring the oxidation of 2,7-dichlorofluorescein diacetate and 4-amino-5-methylamino-2,7-difluorofluorescein diacetate, respectively. Intracellular IL-6 and TNF-α were detected using fluorochrome-conjugated specific antibodies. Monocyte functions were also evaluated in CD163+ and CD163− monocyte subsets. Results The monocytes from septic patients presented with preserved phagocytosis, enhanced ROS and NO generation, and decreased production of inflammatory cytokines compared with the monocytes from healthy volunteers. TNF-α and IL-6 increased and ROS generation decreased in D7 compared with D0 samples. In general, CD163+ monocytes produced higher amounts of IL-6 and TNF-α and lower amounts of ROS and NO than did CD163− monocytes. Conclusions We demonstrated that monocytes from septic patients, which are impaired to produce inflammatory cytokines, display potent phagocytic activity and increased ROS and NO generation.
Collapse
Affiliation(s)
- Sidnéia S Santos
- Escola Paulista de Medicina, Hospital São Paulo, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
| | - Amanda M Carmo
- Escola Paulista de Medicina, Hospital São Paulo, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
| | - Milena K C Brunialti
- Escola Paulista de Medicina, Hospital São Paulo, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
| | - Flavia R Machado
- Escola Paulista de Medicina, Hospital São Paulo, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
| | - Luciano C Azevedo
- Hospital Sírio Libanes, Universidade de Sao Paulo, Ribeirao Preto, Brazil
| | - Murillo Assunção
- Hospital Israelita Albert Einstein, Universidade de Sao Paulo, Ribeirao Preto, Brazil
| | - Sílvia C Trevelin
- Faculdade de Medicina, Universidade de Sao Paulo, Ribeirao Preto, Brazil
| | - Fernando Q Cunha
- Faculdade de Medicina, Universidade de Sao Paulo, Ribeirao Preto, Brazil
| | - Reinaldo Salomao
- Escola Paulista de Medicina, Hospital São Paulo, Universidade Federal de Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|
16
|
Alves-Januzzi AB, Brunialti MKC, Salomao R. CD163 and CD206 expression does not correlate with tolerance and cytokine production in LPS-tolerant human monocytes. CYTOMETRY PART B-CLINICAL CYTOMETRY 2016; 92:192-199. [PMID: 26352275 DOI: 10.1002/cyto.b.21321] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 08/14/2015] [Accepted: 09/01/2015] [Indexed: 01/23/2023]
Abstract
BACKGROUND Lipopolysaccharide (LPS)-tolerant monocytes produce small amounts of inflammatory cytokines, which is one of the characteristics of the alternative activated macrophages (AAM). These cells exhibited an increased expression of CD206 and CD163. Given the functional similarities of AAMs with the modulation of monocytes' functions observed during sepsis and LPS-tolerance, we evaluated whether the inhibition of inflammatory cytokine production by LPS-tolerant monocytes is associated with the phenotype of cells expressing CD206 and CD163. METHODS We investigated whether tolerant human monocytes would modulate their expression of CD206 and CD163, markers of alternative activation, and whether the level of their expression would be related to cytokines detection. Tolerance to LPS was induced in peripheral blood mononuclear cell by pre-incubating the cells with increasing concentrations of LPS. The expression of CD206 and CD163 and intracellular TNF-α and IL-6 was determined 24 h after LPS challenge by flow cytometry. RESULTS No differences in CD163 expression were observed between tolerant and non-tolerant cells, while the expression of CD206, which was decreased following LPS stimulation in non-tolerized cells, was further reduced in tolerant cells. Decreased production of inflammatory cytokines was observed in the tolerized cells, regardless of the expression of CD163 and CD206, with the exception of IL-6 in CD206+ monocytes, which was similarly expressed in both tolerized and non-tolerized cells. CONCLUSIONS The effect of LPS in the expression of CD163 and CD206 on monocytes is not reverted in LPS tolerant cells, and the inhibition of inflammatory cytokines in tolerant cells is not related with modulation of these receptors. © 2016 International Clinical Cytometry Society.
Collapse
Affiliation(s)
- Amanda Barba Alves-Januzzi
- Division of Infectious Diseases, Department of Medicine, Escola Paulista De Medicina, Hospital Sao Paulo, Universidade Federal De Sao Paulo, Sao Paulo, Brazil
| | - Milena Karina Colo Brunialti
- Division of Infectious Diseases, Department of Medicine, Escola Paulista De Medicina, Hospital Sao Paulo, Universidade Federal De Sao Paulo, Sao Paulo, Brazil
| | - Reinaldo Salomao
- Division of Infectious Diseases, Department of Medicine, Escola Paulista De Medicina, Hospital Sao Paulo, Universidade Federal De Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
17
|
Kox M, van Eijk LT, Verhaak T, Frenzel T, Kiers HD, Gerretsen J, van der Hoeven JG, Kornet L, Scheiner A, Pickkers P. Transvenous vagus nerve stimulation does not modulate the innate immune response during experimental human endotoxemia: a randomized controlled study. Arthritis Res Ther 2015; 17:150. [PMID: 26049730 PMCID: PMC4480894 DOI: 10.1186/s13075-015-0667-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 05/29/2015] [Indexed: 01/12/2023] Open
Abstract
INTRODUCTION Vagus nerve stimulation (VNS) exerts beneficial anti-inflammatory effects in various animal models of inflammation, including collagen-induced arthritis, and is implicated in representing a novel therapy for rheumatoid arthritis. However, evidence of anti-inflammatory effects of VNS in humans is very scarce. Transvenous VNS (tVNS) is a newly developed and less invasive method to stimulate the vagus nerve. In the present study, we determined whether tVNS is a feasible and safe procedure and investigated its putative anti-inflammatory effects during experimental human endotoxemia. METHODS We performed a randomized double-blind sham-controlled study in healthy male volunteers. A stimulation catheter was inserted in the left internal jugular vein at spinal level C5-C7, adjacent to the vagus nerve. In the tVNS group (n = 10), stimulation was continuously performed for 30 minutes (0-10 V, 1 ms, 20 Hz), starting 10 minutes before intravenous administration of 2 ng kg(-1) Escherichia coli lipopolysaccharide (LPS). Sham-instrumented subjects (n = 10) received no electrical stimulation. RESULTS No serious adverse events occurred throughout the study. In the tVNS group, stimulation of the vagus nerve was achieved as indicated by laryngeal vibration. Endotoxemia resulted in fever, flu-like symptoms, and hemodynamic changes that were unaffected by tVNS. Furthermore, plasma levels of inflammatory cytokines increased sharply during endotoxemia, but responses were similar between groups. Finally, cytokine production by leukocytes stimulated with LPS ex vivo, as well as neutrophil phagocytosis capacity, were not influenced by tVNS. CONCLUSIONS tVNS is feasible and safe, but does not modulate the innate immune response in humans in vivo during experimental human endotoxemia. TRIAL REGISTRATION Clinicaltrials.gov NCT01944228. Registered 12 September 2013.
Collapse
Affiliation(s)
- Matthijs Kox
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), Geert Grooteplein 10, 6500, HB, Nijmegen, The Netherlands.
| | - Lucas T van Eijk
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), Geert Grooteplein 10, 6500, HB, Nijmegen, The Netherlands.
| | - Tim Verhaak
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), Geert Grooteplein 10, 6500, HB, Nijmegen, The Netherlands.
| | - Tim Frenzel
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), Geert Grooteplein 10, 6500, HB, Nijmegen, The Netherlands.
| | - Harmke D Kiers
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), Geert Grooteplein 10, 6500, HB, Nijmegen, The Netherlands.
| | - Jelle Gerretsen
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), Geert Grooteplein 10, 6500, HB, Nijmegen, The Netherlands.
| | - Johannes G van der Hoeven
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), Geert Grooteplein 10, 6500, HB, Nijmegen, The Netherlands.
| | - Lilian Kornet
- Medtronic Inc., Endepolsdomein 5, 6229, GW, Maastricht, The Netherlands.
| | - Avram Scheiner
- Medtronic Inc., 8200 Coral Sea St NE, Mounds View, MN, 55112, USA.
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), Geert Grooteplein 10, 6500, HB, Nijmegen, The Netherlands.
| |
Collapse
|
18
|
Generation of nitric oxide and reactive oxygen species by neutrophils and monocytes from septic patients and association with outcomes. Shock 2012; 38:18-23. [PMID: 22575991 DOI: 10.1097/shk.0b013e318257114e] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
In this study, our aims were to evaluate the reactive oxygen species (ROS) and nitric oxide (NO) generation by monocytes and neutrophils from septic patients and to correlate their levels with clinical outcomes. Forty-nine septic patients and 19 healthy volunteers were enrolled in the study. The ROS and NO production was quantified in monocytes and neutrophils in whole blood by flow cytometry, constitutively, and after stimulation with Staphylococcus aureus and Pseudomonas aeruginosa. Nitric oxide production by monocytes was higher in septic patients compared with healthy volunteers for all conditions and by neutrophils at baseline, and ROS generation in monocytes and neutrophils was higher in septic patients than in healthy volunteers for all conditions. Nitric oxide production by monocytes and neutrophils was decreased at day 7 compared with that at admission (day 0) in survivors at baseline and after stimulation with S. aureus. Reactive oxygen species production by the monocytes and neutrophils was decreased in survivors at day 7 compared with day 0 under all conditions, except by neutrophils at baseline. No difference was found in NO and ROS generation by monocytes and neutrophils between day 7 and day 0 in nonsurvivors. Generation of NO and ROS by neutrophils and monocytes is increased in septic patients, and their persistence is associated with poor outcome.
Collapse
|
19
|
|
20
|
Colo Brunialti MK, Santos MC, Rigato O, Machado FR, Silva E, Salomao R. Increased percentages of T helper cells producing IL-17 and monocytes expressing markers of alternative activation in patients with sepsis. PLoS One 2012; 7:e37393. [PMID: 22693573 PMCID: PMC3365066 DOI: 10.1371/journal.pone.0037393] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 04/22/2012] [Indexed: 12/24/2022] Open
Abstract
Background A shift from Th1 to Th2 as well as an increase in Treg CD4+T cell subsets has been reported in septic patients (SP). Furthermore, these patients display modulation of monocyte function, with reduced production of pro-inflammatory cytokines upon LPS stimulus, which resembles the phenotype of alternatively activated macrophages. In this study, we evaluated the percentages of T cells differentiated into Th1, Th17 and Treg subsets, as well as the percentage of monocytes expressing markers of alternatively activated monocytes/macrophages (AAM) in SP. Methodology/Principal Findings Peripheral blood mononuclear cells (PBMC) were obtained from 32 healthy volunteers (HV) and from SP at admission (D0, n = 67) and after 7 days of therapy (D7, n = 33). Th1 and Th17 (CD3+CD8−) lymphocytes were identified by the intracellular detection of IFN-γ and IL-17, respectively, spontaneously and after PMA/Io stimulation, and Treg cells were identified by Foxp3+CD127- expression. Monocytes were evaluated for CD206 and CD163 expression. Absolute numbers of CD4+T lymphocytes were measured in whole blood samples by flow cytometry. The Mann-Whitney or Wilcoxon test was applied, as appropriate. The percentage of Th1 cells was lower in SP than in HV at admission after PMA/Io stimulation, whereas the percentage of Th17 cells was higher. In patients’ follow-up samples, a higher percentage of Th1 cells and a lower percentage of Th17 cells were observed on D7 compared with the D0 samples. Treg cells remained unchanged. Septic patients showed a markedly increased proportion of monocytes expressing CD163 and CD206. Conclusions/Significance Upon in vitro stimulus, the percentage of T helper lymphocytes producing IL-17 was higher in SP than in HV at admission, and the percentage producing IFN-γ was lower, a pattern that was reversed during follow-up. The increased expression of CD163 and CD206 indicates that monocytes may acquire the AAM phenotype during sepsis.
Collapse
MESH Headings
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/metabolism
- Biomarkers/metabolism
- Cell Count
- Cell Differentiation/immunology
- Female
- Follow-Up Studies
- Gene Expression Regulation/immunology
- Humans
- Interleukin-17/biosynthesis
- Lectins, C-Type/metabolism
- Macrophages/cytology
- Macrophages/immunology
- Macrophages/metabolism
- Male
- Mannose Receptor
- Mannose-Binding Lectins/metabolism
- Middle Aged
- Monocytes/cytology
- Monocytes/immunology
- Monocytes/metabolism
- Prognosis
- Receptors, Cell Surface/metabolism
- Sepsis/diagnosis
- Sepsis/immunology
- Sepsis/metabolism
- T-Lymphocytes, Helper-Inducer/cytology
- T-Lymphocytes, Helper-Inducer/immunology
- T-Lymphocytes, Helper-Inducer/metabolism
- T-Lymphocytes, Regulatory/cytology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Th1 Cells/cytology
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Th17 Cells/cytology
- Th17 Cells/immunology
- Th17 Cells/metabolism
Collapse
Affiliation(s)
- Milena Karina Colo Brunialti
- Division of Infectious Diseases, Department of Medicine, Hospital Sao Paulo, Escola Paulista de Medicina, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
| | - Michelle Carolina Santos
- Division of Infectious Diseases, Department of Medicine, Hospital Sao Paulo, Escola Paulista de Medicina, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
| | - Otelo Rigato
- Division of Infectious Diseases, Department of Medicine, Hospital Sao Paulo, Escola Paulista de Medicina, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
- Critical Care Unit, Hospital Sirio-Libanes, Sao Paulo, Brazil
| | - Flavia Ribeiro Machado
- Division of Anesthesiology, Department of Surgery, Hospital Sao Paulo, Escola Paulista de Medicina, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
| | - Eliezer Silva
- Critical Care Unit, Hospital Albert Einstein, Sao Paulo, Brazil
| | - Reinaldo Salomao
- Division of Infectious Diseases, Department of Medicine, Hospital Sao Paulo, Escola Paulista de Medicina, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
- * E-mail:
| |
Collapse
|