Acessibilidade / Reportar erro

Taurine Chloramine decreases cell viability and cytokine production in blood and spleen lymphocytes from septic rats

Abstract

Taurine (Tau) is an abundant amino acid in polymorphonuclear leukocytes that react with hypochlorous acid to form taurine chloramine (TauCl) under inflammatory conditions. We investigated potential interactions between lymphocytes and TauCl in rats submitted to cecal ligation. Animals were divided into sham or CLP groups (24 or 120 h) to isolate lymphocytes from blood and spleen. Lymphocytes were cultured at a concentration of 1×106 cells/mL and activated by concanavalin A. Tau and TauCl were added at 1, 10, and 100 μM. Cells were incubated with MTT to evaluate cell viability and cytokine concentration in the supernatant was determined. TauCl decreased lymphocyte viability and altered the secretion pattern of important inflammatory mediators in non-specific-phenotype manner. The effort to a is elucidate mechanisms of immune cell (dys)function in sepsis is important to better understand the complex regulation of immune system during sepsis development, and further studies are necessary to confirm TauCl as potential target in this context.

Key words
lymphocyte; sepsis; taurine; taurine chloramine; viability

INTRODUCTION

The Third International Consensus Definitions Task Force defined sepsis as life-threatening organ dysfunction caused by dysregulated host response to infection (Seymour et al. 2016SEYMOUR CW ET AL. 2016. Assessment of Clinical Criteria for Sepsis: For the Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA 315(8): 762-774.). Sepsis remains the leading cause of death in intensive care units (ICUs), despite remarkable advances in treatment of critical illnesses and progress in general intensive care medicine (Perner et al. 2017PERNER A ET AL. 2017. The intensive care medicine research agenda on septic shock. Intensive Care Med 43: 1294-1305.).

Although polymorphonuclears (PMNs) are mostly related to innate immunity, they also co-localized in T cells at sites of persistent infections, chronic inflammations or tumors (Müller et al. 2009MÜLLER I, MUNDER M, KROPF P & HÄNSCH GM. 2009. Polymorphonuclear neutrophils and T lymphocytes: strange bedfellows or brothers in arms? Trends Immunol 30: 522-530.). There is increasing evidence for mutual interaction between PMNs and T cells (Müller et al. 2009MÜLLER I, MUNDER M, KROPF P & HÄNSCH GM. 2009. Polymorphonuclear neutrophils and T lymphocytes: strange bedfellows or brothers in arms? Trends Immunol 30: 522-530.); thus, we aimed to evaluate possible interactions among chloramine and lymphocytes. Although MPO and Tau are separated in mature PMNs, the formation of TauCl inside PMNs is possible after reactive oxygen species (ROS)-driven permeabilization of azurophilic granules to the cytosol. This may occur during bacteria-induced apoptosis in PMNs Blomgran et al. 2007BLOMGRAN R, ZHENG L & STENDAHL O. 2007. Cathepsin-cleaved Bid promotes apoptosis in human neutrophils via oxidative stress-induced lysosomal membrane permeabilization. J Leukoc Biol 81: 1213-1223.).

In recent years, great attention has been focused on the mechanisms of immune cell death during sepsis. The early phase of sepsis is dominated by a hyperinflammatory state, which is mediated by systemic production of inflammatory cytokines, including interleukin 1 (IL-1), IL-6, tumor necrosis factor alpha (TNF-α) and gamma interferon (IFN-γ). This hyperinflammatory response may lead to organ damage and death in a subset of septic patients (Döcke et al. 1997DÖCKE WD RANDOW F, SYRBE U, KRAUSCH D, ASADULLAH K REINKE P, OLK HD & KOX W 1997. Monocyte deactivation in septic patients: restoration by IFN-gamma treatment. Nat Med 3: 678-681., Heidecke et al. 1999HEIDECKE CD, HENSLER T, WEIGHARDT H, ZANTL N, WAGNER H, SIEWERT JR & HOLZMANN B. 1999. Selective defects of T lymphocyte function in patients with lethal intraabdominal infection. Am J Surg 178: 288-292., Rittirsch et al. 2008RITTIRSCH D, FLIERL MA & WARD PA. 2008. Harmful molecular mechanisms in sepsis. Nat Rev Immunol 8: 776-787.). To balance the pro-inflammatory response, the anti-inflammatory cytokines, including IL-10, are produced. As a result, the majority of patients that survive the initial phase of pro-inflammatory sepsis progress to a protracted period of immune suppression (Hotchkiss et al. 2001HOTCHKISS RS ET AL. 2001. Sepsis-induced apoptosis causes progressive profound depletion of B and CD4 T lymphocytes in humans. J Immunol 166: 6952-6963., Rittirsch et al. 2008RITTIRSCH D, FLIERL MA & WARD PA. 2008. Harmful molecular mechanisms in sepsis. Nat Rev Immunol 8: 776-787.).

More recently IL-17, a pro-inflammatory cytokine expressed by activated memory CD4+ T cells was demonstrated to be important in sepsis (Jekarl et al. 2015JEKARL DW, KIM JY, LEE S, KIM M, KIM Y, HAN K, WOO SH & LEE WJ. 2015. Diagnosis and evaluation of severity of sepsis via the use of biomarkers and profiles of 13 cytokines: a multiplex analysis. Clin Chem Lab Med 53(4): 575-581.). In particular, IL-17A is a mediator of neutrophil stimulation and mobilization by T lymphocytes (Kolls & Linden 2004KOLLS JK & LINDEN A. 2004. Interleukin-17 family members and inflammation. Immunity 21: 467-476., Kasten et al. 2010KASTEN KR ET AL. 2010. Interleukin-7 (IL-7) treatment accelerates neutrophil recruitment through gamma delta T-cell IL-17 production in a murine model of sepsis. Infect Immun 78: 4714-4722.). Later stages of TH17 cell differentiation rely on the expression of IL-23, which is a potent inducer of IL-17 (Murphy et al. 2003MURPHY CA, LANGRISH CL, CHEN Y, BLUMENSCHEIN W, MCCLANAHAN T, KASTELEIN RA, SEDGWICK JD & CUA DJ. 2003. Divergent pro- and antiinflammatory roles for IL-23 and IL-12 in joint autoimmune inflammation. J Exp Med 198: 1951-1957., Langrish et al. 2005LANGRISH CL, CHEN Y, BLUMENSCHEIN WM, MATTSON J, BASHAM B, SEDGWICK JD, MCCLANAHAN T, KASTELEIN RA & CUA DJ. 2005. IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. J Exp Med 201: 233-240.). Although innate IL17 responses might eradicate most pathogens, it can lead to hyperinflammatory responses; moreover, innate immune cells activated by IL17 could damage the surrounding tissue (Hirota et al. 2012HIROTA K, AHLFORS H, DUARTE JH & STOCKINGER B. 2012. Regulation and function of innate and adaptive interleukin-17-producing cells. EMBO Rep 13: 113-120.).

Taurine chloramine (TauCl) is an oxidative product released during the inflammatory response. Taurine (Tau) is generated from cysteine and the most abundant free amino acid in mammalian monocytes and tissues (Vinton et al. 1986VINTON NE, LAIDLAW SA, AMENT ME & KOPPLE JD. 1986. Taurine concentrations in plasma and blood cells of patients undergoing long-term parenteral nutrition. Am J Clin Nutr 44: 398-404., Learn et al. 1990LEARN DB, FRIED VA & THOMAS EL. 1990. Taurine and hypotaurine content of human leukocytes. J Leukoc Biol 48: 174-182.), found in all animal cells at millimolar concentrations, in the plasma and extracellular fluids in much lower ranges (Shuller-Levis & Park 2003) and at the site of inflammation reaches micromolar concentrations (Jang et al. 2009JANG JS, PIAO S, CHA YN & KIM C. 2009. Taurine Chloramine Activates Nrf2, Increases HO-1 Expression and Protects Cells from Death Caused by Hydrogen Peroxide. J Clin Biochem Nutr 45(1): 37-43.).

Tau protects activated neutrophils from inflammatory injury by detoxifying the highly oxidizing hypochlorous acid (HOCl/OCl−) produced from hydrogen peroxide (H2O2) (Thomas et al. 1986aTHOMAS EL, GRISHAM MB & JEFFERSON MM. 1986a. Cytotoxicity of Chloramines. Methods Enzymol 132: 585-593.). Some evidences showed that Tau can act as an antioxidant by enhancing expression and activities of defense enzymes such catalase, superoxide dismutase and glutathione peroxidase (Jang et al. 2009JANG JS, PIAO S, CHA YN & KIM C. 2009. Taurine Chloramine Activates Nrf2, Increases HO-1 Expression and Protects Cells from Death Caused by Hydrogen Peroxide. J Clin Biochem Nutr 45(1): 37-43.). Neonates, aging process and some specific conditions like trauma and sepsis limit the biosynthetic capacity of humans to produce Tau (Marcinkiewicz & Kontny 2014MARCINKIEWICZ J & KONTNY E. 2014. Taurine and inflammatory diseases. Amino Acids 46(1): 7-20.). It has been reported that Tau may protect immune cells against attack by chlorinated oxidants, besides protect tissues and inflammation sites (Cantin 1994CANTIN AM. 1994. Taurine modulation of hypochlorous acid-induced lung epithelial cell injury in vitro. Role of anion transport. J Clin Invest 93(2): 606-614.). In contrast, a study in synoviocyte cultures showed that Tau does not affect the functions of these cells in vitro (Kontny et al. 2003)

Taurine removes excessive HOCl by reacting readily with HOCl/OCl− and forming TauCl, which is more stable and weaker oxidant (Park et al. 1995PARK E, SCHULLER-LEVIS G & QUINN MR. 1995. Taurine chloramine inhibits production of nitric oxide and TNF-alpha in activated RAW 264.7 cells by mechanisms that involve transcriptional and translational events. J Immunol 154: 4778-4784.). Despite its lower reactivity it was shown that TauCl could induce apoptosis in lymphocytes (Klamt & Shacter 2005KLAMT F & SHACTER E. 2005. Taurine chloramine, an oxidant derived from neutrophils, induces apoptosis in human B lymphoma cells through mitochondrial damage. J Biol Chem 280(22): 21346-21352.) and inhibits inducible nitric oxide synthase and TNF-α gene expression (Barua et al. 2001BARUA M, LIU Y & QUINN MR. 2001. Taurine chloramine inhibits inducible nitric oxide synthase and TNF-alpha gene expression in activated alveolar macrophages: decreased NF-kappaB activation and IkappaB kinase activity. J Immunol 167(4): 2275-2281.). TauCl has been described as an oxidant with anti-inflamatory effects, with capacity of suppress production of mediators like cytokines, chemokines, nitric oxide and prostaglandin E2 (Marcinkiewicz et al. 1998, Kontny et al. 2000KONTNY E, SZCZEPAŃSKA K, KOWALCZEWSKI J, KUROWSKA M, JANICKA I, MARCINKIEWICZ J & MAŚLIŃSKI W. 2000. The mechanism of taurine chloramine inhibition of cytokine (interleukin-6, interleukin-8) production by rheumatoid arthritis fibroblast-like synoviocytes. Arthritis Rheum 43(10): 2169-2177.). Thus, we hypothesized that lymphocytes from septic animals are susceptible to the effects of TauCl and this could be one of the mechanisms of immunosuppression during sepsis.

The objective of this study was to assess the effect of Tau and TauCl on the viability of blood and splenic lymphocytes. Considering that Tau has a redundant role, we hypothesized that these compounds, mainly TauCl, could be act on the pathways that explain, at least in part, a pathophysiologic mechanism on the complex balance of stimulatory and inhibitory pathways in sepsis.

MATERIALS AND METHODS

Animals

Animal experiments were conducted according to protocols approved by the Animal Experimental Ethics Committee of Universidade do Extremo Sul Catarinense, Criciúma, Brazil (protocol number: 89/2012). Eight-week-old male Wistar rats (250-300g) were housed at 18°C-22°C, in 12 h/12 h light/dark conditions and 55-65% relative humidity. Wistar rats were divided randomly into two groups: sham (n=12) and sepsis (n =16).

Cecal and ligation puncture (CLP) model of sepsis

Wistar rats were subjected to CLP as previously described (Fink & Heard 1990FINK MP & HEARD SO. 1990. Laboratory models of sepsis and septic shock. J Surg Res 49(2): 186-196.). Briefly, the animals were randomized and anesthetized using a mixture of intraperitoneal ketamine (80 mg/kg) and xylazine (10 mg/kg). Under aseptic conditions, a 3-cm midline laparotomy was performed to expose the cecum and adjoining intestine. The cecum was ligated with a 3.0 silk suture at its base, below the ileocecal valve and punctured once with a 14- gauge needle. The cecum was then squeezed gently to extrude a small amount of feces through the perforation site. The cecum was then returned to the peritoneal cavity, and the laparotomy was closed with 4.0 silk sutures. Animals were resuscitated with regular saline (50mL/kg) subcutaneously (s.c.) immediately and 12h after CLP. All animals received antibiotics (ceftriaxone at 30 mg/kg) every 6h s.c. for 3 days and dipyrone sodium (80 mg/kg) (s.c.) immediately and 12h after CLP. To minimize variability between different experiments, the CLP procedure was always performed by the same investigator.

Collection, isolation and stimulation of blood and splenic lymphocytes

Blood from both sham and sepsis groups was collected through cardiac puncture and stored in EDTA tubes in 24 and 120 hours after CLP surgery. Spleens were simultaneously collected. Blood and spleen lymphocytes were isolated by Ficoll-Hypaque solution gradient separation (Sigma-Aldrich Co, St. Louis, MO, USA), using standard protocols (Boyum 1977BOYUM A. 1977. Separation of lymphocytes, lymphocyte subgroups and monocytes: a review. Lymphology 10(2): 71-76., Mishell & Dutton 1967MISHELL RI & DUTTON RW. 1967. Immunization of dissociated spleen cell cultures from normal mice. J Exp Med 126: 423-442.). Cells were suspended in RPMI 1640 media (Sigma-Aldrich) supplemented with 20% heat-inactivated fetal bovine serum (FBS) (Life Technologies, Grand Island, NY, USA), penicillin (100 UI/mL, Sigma-Aldrich), streptomycin (100 µg/mL, Sigma-Aldrich) and processed for further staining with Giemsa stain (Sigma-Aldrich) and Trypan blue (Sigma-Aldrich). Cells were counted in a Neubauer chamber, and the viability was determined using 94% by Trypan blue exclusion dye. T cells were suspended in 96-well plates in the presence of concanavalin A (Con A, Sigma-Aldrich) at 2 µg/mL (Fazzino et al. 2010FAZZINO F, OBREGÓN F & LIMA L. 2010. Taurine and proliferation of lymphocytes in physically restrained rats. J Biomed Sci 17: S24.). The final volume was 200μl with density of 1 × 106 cells/ml. The plates were incubated at 37°C in 5% CO2, for 24 hours.

Preparation of Tau and TauCl

TauCl was frechly prepared according standard protocols (Thomas et al. 1986aTHOMAS EL, GRISHAM MB & JEFFERSON MM. 1986a. Cytotoxicity of Chloramines. Methods Enzymol 132: 585-593., bTHOMAS EL, GRISHAM MB & JEFFERSON MM. 1986b. Preparation and Characterization of Chloramines. Methods Enzymol 132: 569-585.) with modifications. Briefly, 25 mM sodium hypochlorite (Sigma-Aldrich) was added to 100 mM Tau (Sigma-Aldrich) in phosphate buffered saline (PBS) (pH 7,8-8,0). Each preparation of TauCl was monitored by ultraviolet absorption (252nm, 429 M-1cm-1) to confirm the presence of the chloramine. Tau was prepared in PBS (pH 7,4). Stock solutions of Tau (1; 10 and 100 µM TauCl) were used.

Citotoxicity assay and measurement of cell viability

Twenty-four hours after culture, lymphocytes were incubated with Tau or TauCl (at 1; 10; 100 µM concentrations) for 1 hour (Ogino et al. 2009OGINO T, THAN TA, HOSAKO M, OZAKI M, OMORI M & OKADA S. 2009. Taurine chloramine: a possible oxidant reservoir. Adv Exp Med Biol 643: 451-461.). After this period, supernatants were collected and frozen at –80°C. Cell viability was assessed by the mitochondria-dependent reduction of thiazolyl blue tetrazolium bromide (MTT) (Sigma-Aldrich) to formazan crystals (Mosmann 1983MOSMANN T. 1983. Rapid colorimetric assay for cellular growth and survival: applications to proliferation and cytotoxicity assays. J Immunol Methods 65: 55-63.). After 4 hours incubation with MTT (500 µg/mL) 100 µL of dimethyl sulfoxide (DMSO) (Sigma-Aldrich) was added. The absorbance of DMSO-dissolved MTT crystals was measured at 560-630 nm with a SpectraMax M3 microplate reader (Molecular Devices, Sunnyvale, California, USA).

Cytokine measurements

To measure IL-17A, IL-10, TNF-α, IL-6, IFN-γ and IL-23, sandwich ELISAs were performed (Peprotech Inc. Connecticut, USA). The concentration for each cytokine was calculated using a recombinant protein as the standard. The linear ranges were: 15,6 to 1000 pg/mL (IL-23); 16 to 1000 pg/mL (IL-17A); 39 to 2500 pg/mL (IL-10); 17 to 3000 pg/mL (TNF-α); 32-8000 pg/mL (IL-6); 25-4000 pg/mL (IFN-γ).

Statistical analysis

All experiments were performed in triplicate and analyzed with the SPSS statistical package (SPSS 20.0 software). Values were represented as means ± SD. ANOVA with post-hoc Tukey test were performed for ELISAs and Multi-factorial ANOVA tests were used for MTT assays. P values < 0.05 were considered significant.

RESULTS

Cell viability by MTT assay

In general, cell viability alteration was more often seen at 24 hours than 120 hours both in blood and spleen in non-dependent dose manner and did not follow any specific pattern. Tau and Tau-Cl treatments (1, 10 and 100 µM) reduced blood cell viability after 24 hours when compared to the corresponding sham group (Figure 1a). A different pattern was seen in blood lymphocytes at 120 hours (Figure 1b), when Tau did not reduce viability. However, cell viability decreased upon exposure to 10 and 100 µM TauCl.

Figure 1
Cell viability measurement by MTT assay. Viability of lymphocytes treated with Tau and TauCl in different molar concentrations. Rats were submitted to CLP or Sham, and 24 or 120 hours after the surgery their blood and spleens were collected, the lymphocytes were isolated, cultured and the cell viability was measured by MTT assay. (a) blood, 24 hours; (b) blood, 120 hours; (c) spleen, 24 hours; (d) spleen, 120 hours. * = P < 0.05 for CLP compared with sham group (Tau-treated). # = P < 0.05 for CLP compared with Sham group (TauCl-treated). (n = 5).

Splenic lymphocytes were less sensitive than blood cells 24 hours after treatment with both Tau and Tau-Cl. Only 1 µM TauCl decreased splenic cell viability after 24 hours when compared to the relative sham group (Figure 1c). After 120 hours, no sensitivity to Tau and TauCl in CLP group was observed.

Cytokine secretion

As cytokine production by adherent and non-adherent mononuclear leukocytes plays a critical role in inflammation, a lymphocyte cytokine production profile in the presence of TauCl was analyzed. We did not investigate the effect of Tau in cytokine production because it did not influence the production of these mediators, as described previously (Marcinkiewicz et al. 1995MARCINKIEWICZ J, GRABOWSKA A, BERETA J & STELMASZYNSKA. 1995. Taurine chloramine, product of activated neutrophils, inhibits in vitro the generation of nitric oxide and other macrophage inflammatory mediators. J Leukoc Biol 58: 667-674., 1999MARCINKIEWICZ J, NOWAK B, GRABOWSKA A, BOBEK M, PETROVSKA L & CHAIN B. 1999. Regulation of murine dendritic cell functions in vitro by taurine chloramine, a major product of the neutrophil myeloperoxidase-halide system. Immunology 98: 371-378.). TauCl did not modify IL-17 secretion was decreased in splenic lymphocytes treated with 10 µM TauCl 120 hours after sepsis induction (Figure 2a and b). The secretion of IL-23, which is a potent inducer of IL-17 (Murphy et al. 2003MURPHY CA, LANGRISH CL, CHEN Y, BLUMENSCHEIN W, MCCLANAHAN T, KASTELEIN RA, SEDGWICK JD & CUA DJ. 2003. Divergent pro- and antiinflammatory roles for IL-23 and IL-12 in joint autoimmune inflammation. J Exp Med 198: 1951-1957., Langrish et al. 2005LANGRISH CL, CHEN Y, BLUMENSCHEIN WM, MATTSON J, BASHAM B, SEDGWICK JD, MCCLANAHAN T, KASTELEIN RA & CUA DJ. 2005. IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. J Exp Med 201: 233-240.), was altered only at 1 µM TauCl, as compared to untreated cells (Figure 3a and b). Both blood and splenic lymphocytes increased the secretion of IFN-γ when treated with TauCl (Figure 4a and b). However, 10 µM TauCl reduced the production of IFN-γ secretion in splenic lymphocytes.

Figure 2
The effect of TauCl on the production of the pro-inflammatory mediator, IL-17A. Lymphocytes (1x106 cells/mL) were preincubated with TauCl (1, 10 or 100 uM) for 1 hour. After this, supernatants were collected and IL-17A was measured by ELISA for (a) blood and (b) spleen. Results are expressed as means ± SD. # = P < 0.05, compared with sham rats at 120 hours (n=5).
Figure 3
The effect of TauCl on the production of the pro-inflammatory mediator, IL-23. Lymphocytes (1x106 cells/mL) were preincubated with TauCl (1, 10 or 100 uM) for 1 hour. After this, supernatants were collected and IL-23 was measured by ELISA for (a) blood and (b) spleen. Results are expressed as means ± SD. * = P < 0.05 compared with sham rats at 24 hours. # = P < 0.05 compared with sham rats at 120 hours (n=5).
Figure 4
The effect of TauCl on the production of the pro-inflammatory mediator, IFN-gamma. Lymphocytes (1x106 cells/mL) were preincubated with TauCl (1, 10 or 100 uM) for 1 hour. After this, supernatants were collected and IFN-gamma was measured by ELISA for (a) blood and (b) spleen. Results are expressed as means ± SD. * = P < 0.05 compared with sham rats at 24 hours. # = P < 0.05 compared with sham rats at 120 hours (n=5).

TNF-α secretion by spleen decreased significantly after 24 hours, as compared to sham (Figure 5a and b). The last pro-inflammatory cytokine evaluated was IL-6, an important mediator released during sepsis, however, the effect of TauCl in IL-6 secretion was not consistent (Figure 6a and b).

Figure 5
The effect of TauCl on the production of the pro-inflammatory mediator, TNF-α. Lymphocytes (1x106 cells/mL) were preincubated with TauCl (1, 10 or 100 uM) for 1 hour. After this, supernatants were collected and TNF-α was measured by ELISA for (a) blood and (b) spleen. Results are expressed as means ± SD. * = P < 0.05 compared with sham rats at 24 hours (n=5).
Figure 6
The effect of TauCl on the production of the pro-inflammatory mediator, IL-6. Lymphocytes (1x106 cells/mL) were preincubated with TauCl (1, 10 or 100 uM) for 1 hour. After this, supernatants were collected and IL-6 was measured by ELISA for (a) blood and (b) spleen. Results are expressed as means ± SD. * = P <0.05 compared with sham rats at 24 hours. # = P < 0.05 compared with sham rats at 120 hours (n=5).

It is also important to analyze an anti-inflammatory mediator that acts as an immune regulator in sepsis. TauCl decreased IL-10 secretion primarily in spleen cells (Figure 7) in the doses of 1 and 10 µM.

Figure 7
The effect of TauCl on the production of the pro-inflammatory mediator, IL-10. Lymphocytes (1x106 cells/mL) were preincubated with TauCl (1, 10 or 100 uM) for 1 hour. After this, supernatants were collected and IL-10 was measured by ELISA for (a) blood and (b) spleen. Results are expressed as means ± SD. * = P < 0.05 compared with sham rats 24 hours. # = P < 0.05 compared with sham rats 120 hours (n=5).

DISCUSSION

Determine the effects to TauCl on production of important inflammatory mediators such as cytokines by lymphocytes seems to be fundamental to understanding its role in immune dysfunction associated with sepsis.

Here it was demonstrated that cell viability did not show a consistent pattern of alteration. Tau decreased cell viability at 24 hours, similarly to TauCl, and this is the opposite from what was expected, since Tau could exert a physiological scavenging potential against reactive species generated by oxidative metabolism (Oliveira et al. 2010OLIVEIRA MW, MINOTTO JB, DE OLIVEIRA MR, ZANOTTO-FILHO A, BEHR GA, ROCHA RF, MOREIRA JC & KLAMT F. 2010. Scavenging and antioxidant potential of physiological taurine concentrations against different reactive oxygen/nitrogen species. Pharmacol Rep 62(1): 185-193.). In addition, sepsis has been associated with increased oxidant levels, depleted antioxidants, and accumulated markers of oxidative stress (Zhang et al. 2000ZHANG H, SLUTSKY AS & VINCENT JL. 2000. Oxygen free radicals in ARDS, septic shock and organ dysfunction. Intensive Care Med 26: 474-476.). Ritter et al. (2003)RITTER C ET AL. 2003. Oxidative parameters and mortality in sepsis induced by cecal ligation and perforation. Intensive Care Med 29(10): 1782-1789. demonstrated that superoxide dismutase (SOD) is predictive of mortality. The SOD/Catalase (CAT) imbalance seems to be an important factor in the oxidative stress during the letal sepsis (Andrades et al. 2005ANDRADES M, RITTER C, MOREIRA JC & DAL-PIZZOL F. 2005. Oxidative parameters diferences during non-lethal and lethal sepsis development. J Surg Res 125(1): 68-72.) and plasma SOD activity can be used as a biomarker in human sepsis (Guerreiro et al. 2010GUERREIRO MO, PETRONILHO F, ANDRADES M, CONSTANTINO L, MINA FG, MOREIRA JC, DAL-PIZZOL F & RITTER C. 2010. Plasma superoxide dismutase activity and mortality in septic patients [corrected]. J Trauma 69(6): E102-E106.). This Tau potencial could be related to the functional impairment of taurine transporter (taut) caused by peroxynitrite. This is likely due to the nitrosylation and/or nitration of amino acids residues of taut, leading to less intracellular transport of Tau (Koo et al. 2012KOO TS, KWON HJ, KIM MH, KIM DD, SHIM CK, CHUNG SJ & CHONG S. 2012. Functional impairment of rat taurine transporter by activation of nitrogen oxide through superoxide. Drug Metab Pharmacokinet 27: 286-293.). Ther is a considerable body of evidence supporting the modification and functional alteration of proteins by nitrogen oxides (NOs) (Dimmeler et al. 1994DIMMELER S, MESSMER UK, TIEGES G & BRUNE B. 1994. Modulation of glyceraldehyde-3-phospahet dehydrogenase in Salmonella abortus equi lipopolysaccharide-treated mice. Eur J Pharmacol 267: 105-112., Kuhn & Arthur 1996KUHN DM & ARTHUR RE JR. 1996. Inactivation of brain tryptophan hydroxylase by nitric oxide. J Neurochem 67: 1072-1077., Mohr et al. 1996MOHR S, STAMLER JS & BRUNE B. 1996. Posttranslational modification of glyceraldehyde3phosphate dehydrogenase by S-nitrosylation and subsequent NADH attachment. J Biol Chem 271: 4209-4214.). In a previous study, the functional activities of secondary active transport systems, including the Tau uptake system, were reduced at the barrier between systemic circulation and the cerebrospinal fluid after an induction of inflammation. In addition, in vitro Tau uptake activity was reduced, suggesting that this process is sensitive to NOs (Han et al. 2002HAN H, KIM SG, LEE MG, SHIM CK & CHUN SJ. 2002. Mechanism of the reduced elimination clearance of benzylpeninicilin from cerebrospinal fluid in rats with intracisternal administration of lipopolysaccharide. Drug Metab Dispos 30: 1214-1220.). In contrast, Park et al. (2002)PARK E, JIA J, QUINN MR & SCHULLER-LEVIS G. 2002. Taurine chloramine inhibits lymphocyte proliferation and decreases cytokine production in activated human leukocytes. Clin Immunol 102(2): 179-184. conducted a study that cell viability was unaffected by 0.1mM and 0.4 mM TauCl.

It was demonstrated that taurine haloamines, such as TauCl, inhibit the production of pro-inflammatory mediators like TNF-α, IL-1B and IL-6 (Marcinkiewicz et al. 1995MARCINKIEWICZ J, GRABOWSKA A, BERETA J & STELMASZYNSKA. 1995. Taurine chloramine, product of activated neutrophils, inhibits in vitro the generation of nitric oxide and other macrophage inflammatory mediators. J Leukoc Biol 58: 667-674., Park et al. 1995PARK E, SCHULLER-LEVIS G & QUINN MR. 1995. Taurine chloramine inhibits production of nitric oxide and TNF-alpha in activated RAW 264.7 cells by mechanisms that involve transcriptional and translational events. J Immunol 154: 4778-4784., Barua et al. 2001BARUA M, LIU Y & QUINN MR. 2001. Taurine chloramine inhibits inducible nitric oxide synthase and TNF-alpha gene expression in activated alveolar macrophages: decreased NF-kappaB activation and IkappaB kinase activity. J Immunol 167(4): 2275-2281.). Klamt & Shacter (2005)KLAMT F & SHACTER E. 2005. Taurine chloramine, an oxidant derived from neutrophils, induces apoptosis in human B lymphoma cells through mitochondrial damage. J Biol Chem 280(22): 21346-21352. suggest that TauCl may be involved in the resolution and termination of acute inflammation. Besides that, Kontny et al. (2003) proposed that TauCl may represent negative regulatory loop to damp inflammation during inflammatory response and low TauCl concentration can lead to a pathological chronic status and affirmed that TauCl mediated downregulatory response is not effective enough to prevent transition from the acute to the chronic phase in an experimental of cells stimulated with lipopolysaccharide (LPS) (Chorazy et al. 2002CHORAZY M, KONTNY E, MARCINKIEWICZ J & MAŚLIŃSKI W. 2002. Taurine chloramine modulates cytokine production by human peripheral blood mononuclear cells. Amino Acids 23(4): 407-413.).

Lymphocytes at 120 hours were more sensitive to TauCl treatment. Increase in spleen lymphocyte apoptosis has been shown to reduce survival in experimental animals with sepsis and they are the immune cells most affected by deregulated apoptotic cell death during sepsis (Wagner et al. 2007WAGNER TH, DREWRY AM, MACMILLAN S, DUNNE WM, CHANG KC, KARL IE, HOTCHKISS RS & COBB JP. 2007. Surviving sepsis: bcl-2 overexpression modulates splenocyte transcriptional responses in vivo. Am J Physiol 292: 1751-1759., Wesche-Soldato et al. 2007WESCHE-SOLDATO DE, CHUNG CS, GREGORY SH, SALAZAR-MATHER TP, AYALA CA & AYALA A. 2007. CD8 T cells promote inflammation and apoptosis in the liver after sepsis: role of Fas-FasL. Am J Pathol 171: 87-96., Tinsley et al. 2003TINSLEY KW, GRAYSON MH, SWANSON PE, DREWRY AM, CHANG KC, KARL IE & HOTCHKISS RS. 2003. Sepsis induces apoptosis and profound depletion of splenic interdigitating and follicular dendritic cells. J Immunol 171: 909-914.). Both extrinsic negative regulatory pathways and cell-intrinsic negative regulatory pathways have key roles in T cells exhaustion (Wherry 2011WHERRY EJ. 2011. T cell exhaustion. Nat Immunol 12: 492-499.). TauCl could be implicated in this context. Although functional effector T cells can transiently express inhibitory receptors during activation, prolonged and/or high expression of multiple inhibitory receptors is a key feature of the exhaustion of CD8+ and CD4+ T cells both in animal models and in humans (Virgin et al. 2009VIRGIN HW, WHERRY EJ & AHMED R. 2009. Redefining chronic viral infection. Cell 138: 30-50.). Thus, TauCl can act as a molecule that signal to down-regulation of lymphocytes after sepsis.

Our experiments analyzed two stages of sepsis development – 24 hours representing the acute inflammatory response and 120 hours representing a hypo-inflammatory response. In an attempt to explain these stages, Marcinkiewicz et al. (1998)MARCINKIEWICZ J, GRABOWSKA A, BERETA J, BRYNIARSKI K & NOWAK B. 1998. Taurine chloramine down-regulates the generation of murine neutrophil inflammatory mediators. Immunopharmacology 40(1): 27-38. suggested that in acute phase of inflammation TauCl contributes to the inflammatory function of cells like neutrophils, and the increase in its levels is also associated with a secondary down-regulation of the inflammatory process. Peripheral blood mononuclear cells (PBMCs) isolated from patients that succumbed to sepsis exhibit a reduction in cytokine secretion when stimulated in vitro, supporting the hypothesis that functional immune cell impairment may predispose a patient to a poorer outcome (Boomer et al. 2011BOOMER JS ET AL. 2011. Immunosuppression in patients who die of sepsis and multiple organ failure. JAMA 306: 2594-2605.). However, in our study it was not possible to observe a reduction in all of cytokines when CLP cells were TauCl-treated, at least in lymphocytes.

The proliferative response of peripheral blood mononuclear cells in septic patients after stimulation with mitogens was also markedly decreased in comparison with that in healthy individuals (Venet et al. 2009VENET F ET AL. 2009. Increased circulating regulatory T cells (CD4+CD25+CD127-) contribute to lymphocyte anergy in septic shock patients. Intensive Care Med 35: 678-686.). The fact that TauCl decreases these cytokines production in lymphocytes from septic animals, particularly at 120 hours, could compromise important host defenses, in a lymphocyte population non-specific manner. Studies have indicated that a shift from a TH1 to a TH2 response occurs that contributes to the late immunosuppression seen during sepsis and this study contributes to show that TH17 also can be involved in sepsis. Multiple inhibitory mechanisms were identified in immunosuppressive phase of sepsis, which encompasses the 120 hours timepoint, including the dominance of inhibitory over activating receptors, expansion of suppressive cell types and induction of inhibitory ligands on both APCs and tissue parenchymal cells. The presence of apoptotic cells inducing CD8+ TREGS mediates suppression by producing TNF-related apoptosis-inducing ligand (TRAIL) (Unsinger et al. 2010UNSINGER J, KAZAMA H, MCDONOUGH JS, GRIFFITH TS, HOTCHKISS RS & FERGUSON TA. 2010. Sepsis-induced apoptosis leads to active suppression of delayed-type hypersensitivity by CD8+ regulatory T cells through a TRAIL-dependent mechanism. J Immunol 184: 6766-6772.).

As a product of the chlorinating system of neutrophils, TauCl can act as an immune modulator of other cells, including lymphocytes, trying to promote changes in the hyper/hypo inflammatory axis. TauCl may therefore play a role in maintaining the delicate balance between mounting an effective immune response on one hand, and minimizing the destrictive effect of the inflammatory cells on the other (Marcinkiewicz et al. 1998MARCINKIEWICZ J, GRABOWSKA A, BERETA J, BRYNIARSKI K & NOWAK B. 1998. Taurine chloramine down-regulates the generation of murine neutrophil inflammatory mediators. Immunopharmacology 40(1): 27-38.). In a study with rheumatoid arthritis cells, due to prolonged activation, neutrophils accumulating in rheumatoid synovial fluid exhibit features indicative of partial functional “exhaustion” and these cells generate less TauCl in vitro than their peripheral blood counterparts, suggesting that the local concentration of TauCl in the local of inflammation is probably too low to exert anti-inflammatory effects (Kontny et al. 2002KONTNY E, WOJTECKA-ŁUKASIK E, RELL-BAKALARSKA K, DZIEWCZOPOLSKI W, MAŚLIŃSKI W & MAŚLINSKI S. 2002. Impaired generation of taurine chloramine by synovial fluid neutrophils of rheumatoid arthritis patients. Amino Acids 23(4): 415-418.). When activated adherent and non-adherent monocuclear leukocytes were treated with TauCl, production of cytokines tested – IL-6, IL-8 and IL-12 – was significantly down-regulated, except for TNF-α (Park et al. 2002PARK E, JIA J, QUINN MR & SCHULLER-LEVIS G. 2002. Taurine chloramine inhibits lymphocyte proliferation and decreases cytokine production in activated human leukocytes. Clin Immunol 102(2): 179-184.). In contrast, our results showed significantly decrease of TNF-α at 24 hours, and this could be a feedback inhibition of TH1 responses during sepsis (Biron 2001BIRON CA. 2001. Interferons alpha and beta as immune regulators--a new look. Immunity 14(6): 661-664.). In the initial phase of immune response TauCl (present at low concentration) may favors the resolution of protective inflammation by supporting systhesis of TNF-α (Chorazy et al. 2002CHORAZY M, KONTNY E, MARCINKIEWICZ J & MAŚLIŃSKI W. 2002. Taurine chloramine modulates cytokine production by human peripheral blood mononuclear cells. Amino Acids 23(4): 407-413.). As the inflammatory process progress a local accumulation of TauCl occurs, and this compound could prevent perpetuation of chronic inflammation by a gradual down-regulation of IL-6 (Tilg et al. 1994TILG H, TREHU E, ATKINS MB, DINARELLO CA & MIER JW. 1994. Interleukin-6 (IL-6) as an anti-inflammatory cytokine: induction of circulating IL-1 receptor antagonist and soluble tumor necrosis factor receptor p55. Blood 83(1): 113-118.). Another possible mechanism to control inflammation could be an increase in IL-10, but we and others (Marcinkiewicz & Kontny 2014MARCINKIEWICZ J & KONTNY E. 2014. Taurine and inflammatory diseases. Amino Acids 46(1): 7-20.) could not observe any effect of TauCl on IL-10 secretion.

CONCLUSION

Our data suggest that TauCl could contribute to immune modulation during sepsis by decreasing lymphocyte viability, especially at early stages (24 hours) and by altering secretion pattern of important inflammatory mediators. The effort to elucidate mechanisms of immune cell (dys)function in sepsis is important to better understand the complex regulation of immune system during sepsis development, and futher studies are necessary to confirm TauCl as a potential target in this context.

ACKNOWLEGMENTS

This work was supported by grants from the Universidade do Extremo Sul Catarinense (UNESC) and CAPES (Coordenação de Aperfeiçoamento de Pessoal de Nível Superior), both from Brazil.

REFERENCES

  • ANDRADES M, RITTER C, MOREIRA JC & DAL-PIZZOL F. 2005. Oxidative parameters diferences during non-lethal and lethal sepsis development. J Surg Res 125(1): 68-72.
  • BARUA M, LIU Y & QUINN MR. 2001. Taurine chloramine inhibits inducible nitric oxide synthase and TNF-alpha gene expression in activated alveolar macrophages: decreased NF-kappaB activation and IkappaB kinase activity. J Immunol 167(4): 2275-2281.
  • BIRON CA. 2001. Interferons alpha and beta as immune regulators--a new look. Immunity 14(6): 661-664.
  • BLOMGRAN R, ZHENG L & STENDAHL O. 2007. Cathepsin-cleaved Bid promotes apoptosis in human neutrophils via oxidative stress-induced lysosomal membrane permeabilization. J Leukoc Biol 81: 1213-1223.
  • BOOMER JS ET AL. 2011. Immunosuppression in patients who die of sepsis and multiple organ failure. JAMA 306: 2594-2605.
  • BOYUM A. 1977. Separation of lymphocytes, lymphocyte subgroups and monocytes: a review. Lymphology 10(2): 71-76.
  • CANTIN AM. 1994. Taurine modulation of hypochlorous acid-induced lung epithelial cell injury in vitro. Role of anion transport. J Clin Invest 93(2): 606-614.
  • CHORAZY M, KONTNY E, MARCINKIEWICZ J & MAŚLIŃSKI W. 2002. Taurine chloramine modulates cytokine production by human peripheral blood mononuclear cells. Amino Acids 23(4): 407-413.
  • CHORAZY-MASSALSKA M, KONTNY E, KORNATKA A, RELL-BAKALARSKA M, MARCINKIEWICZ J & MAŚLIŃSKI W. 2004. The effect of taurine chloramine on pro-inflammatory cytokine production by peripheral blood mononuclear cells isolated from rheumatoid arthritis and osteoarthritis patients. Clin Exp Rheumatol 22(6): 692-698.
  • DIMMELER S, MESSMER UK, TIEGES G & BRUNE B. 1994. Modulation of glyceraldehyde-3-phospahet dehydrogenase in Salmonella abortus equi lipopolysaccharide-treated mice. Eur J Pharmacol 267: 105-112.
  • DÖCKE WD RANDOW F, SYRBE U, KRAUSCH D, ASADULLAH K REINKE P, OLK HD & KOX W 1997. Monocyte deactivation in septic patients: restoration by IFN-gamma treatment. Nat Med 3: 678-681.
  • FAZZINO F, OBREGÓN F & LIMA L. 2010. Taurine and proliferation of lymphocytes in physically restrained rats. J Biomed Sci 17: S24.
  • FINK MP & HEARD SO. 1990. Laboratory models of sepsis and septic shock. J Surg Res 49(2): 186-196.
  • GUERREIRO MO, PETRONILHO F, ANDRADES M, CONSTANTINO L, MINA FG, MOREIRA JC, DAL-PIZZOL F & RITTER C. 2010. Plasma superoxide dismutase activity and mortality in septic patients [corrected]. J Trauma 69(6): E102-E106.
  • HAN H, KIM SG, LEE MG, SHIM CK & CHUN SJ. 2002. Mechanism of the reduced elimination clearance of benzylpeninicilin from cerebrospinal fluid in rats with intracisternal administration of lipopolysaccharide. Drug Metab Dispos 30: 1214-1220.
  • HEIDECKE CD, HENSLER T, WEIGHARDT H, ZANTL N, WAGNER H, SIEWERT JR & HOLZMANN B. 1999. Selective defects of T lymphocyte function in patients with lethal intraabdominal infection. Am J Surg 178: 288-292.
  • HIROTA K, AHLFORS H, DUARTE JH & STOCKINGER B. 2012. Regulation and function of innate and adaptive interleukin-17-producing cells. EMBO Rep 13: 113-120.
  • HOTCHKISS RS ET AL. 2001. Sepsis-induced apoptosis causes progressive profound depletion of B and CD4 T lymphocytes in humans. J Immunol 166: 6952-6963.
  • JANG JS, PIAO S, CHA YN & KIM C. 2009. Taurine Chloramine Activates Nrf2, Increases HO-1 Expression and Protects Cells from Death Caused by Hydrogen Peroxide. J Clin Biochem Nutr 45(1): 37-43.
  • JEKARL DW, KIM JY, LEE S, KIM M, KIM Y, HAN K, WOO SH & LEE WJ. 2015. Diagnosis and evaluation of severity of sepsis via the use of biomarkers and profiles of 13 cytokines: a multiplex analysis. Clin Chem Lab Med 53(4): 575-581.
  • KASTEN KR ET AL. 2010. Interleukin-7 (IL-7) treatment accelerates neutrophil recruitment through gamma delta T-cell IL-17 production in a murine model of sepsis. Infect Immun 78: 4714-4722.
  • KLAMT F & SHACTER E. 2005. Taurine chloramine, an oxidant derived from neutrophils, induces apoptosis in human B lymphoma cells through mitochondrial damage. J Biol Chem 280(22): 21346-21352.
  • KOLLS JK & LINDEN A. 2004. Interleukin-17 family members and inflammation. Immunity 21: 467-476.
  • KONTNY E, SZCZEPAŃSKA K, KOWALCZEWSKI J, KUROWSKA M, JANICKA I, MARCINKIEWICZ J & MAŚLIŃSKI W. 2000. The mechanism of taurine chloramine inhibition of cytokine (interleukin-6, interleukin-8) production by rheumatoid arthritis fibroblast-like synoviocytes. Arthritis Rheum 43(10): 2169-2177.
  • KONTNY E, WOJTECKA-ŁUKASIK E, RELL-BAKALARSKA K, DZIEWCZOPOLSKI W, MAŚLIŃSKI W & MAŚLINSKI S. 2002. Impaired generation of taurine chloramine by synovial fluid neutrophils of rheumatoid arthritis patients. Amino Acids 23(4): 415-418.
  • KOO TS, KWON HJ, KIM MH, KIM DD, SHIM CK, CHUNG SJ & CHONG S. 2012. Functional impairment of rat taurine transporter by activation of nitrogen oxide through superoxide. Drug Metab Pharmacokinet 27: 286-293.
  • KUHN DM & ARTHUR RE JR. 1996. Inactivation of brain tryptophan hydroxylase by nitric oxide. J Neurochem 67: 1072-1077.
  • LANGRISH CL, CHEN Y, BLUMENSCHEIN WM, MATTSON J, BASHAM B, SEDGWICK JD, MCCLANAHAN T, KASTELEIN RA & CUA DJ. 2005. IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. J Exp Med 201: 233-240.
  • LEARN DB, FRIED VA & THOMAS EL. 1990. Taurine and hypotaurine content of human leukocytes. J Leukoc Biol 48: 174-182.
  • MARCINKIEWICZ J, GRABOWSKA A, BERETA J, BRYNIARSKI K & NOWAK B. 1998. Taurine chloramine down-regulates the generation of murine neutrophil inflammatory mediators. Immunopharmacology 40(1): 27-38.
  • MARCINKIEWICZ J, GRABOWSKA A, BERETA J & STELMASZYNSKA. 1995. Taurine chloramine, product of activated neutrophils, inhibits in vitro the generation of nitric oxide and other macrophage inflammatory mediators. J Leukoc Biol 58: 667-674.
  • MARCINKIEWICZ J & KONTNY E. 2014. Taurine and inflammatory diseases. Amino Acids 46(1): 7-20.
  • MARCINKIEWICZ J, NOWAK B, GRABOWSKA A, BOBEK M, PETROVSKA L & CHAIN B. 1999. Regulation of murine dendritic cell functions in vitro by taurine chloramine, a major product of the neutrophil myeloperoxidase-halide system. Immunology 98: 371-378.
  • MISHELL RI & DUTTON RW. 1967. Immunization of dissociated spleen cell cultures from normal mice. J Exp Med 126: 423-442.
  • MOHR S, STAMLER JS & BRUNE B. 1996. Posttranslational modification of glyceraldehyde3phosphate dehydrogenase by S-nitrosylation and subsequent NADH attachment. J Biol Chem 271: 4209-4214.
  • MOSMANN T. 1983. Rapid colorimetric assay for cellular growth and survival: applications to proliferation and cytotoxicity assays. J Immunol Methods 65: 55-63.
  • MÜLLER I, MUNDER M, KROPF P & HÄNSCH GM. 2009. Polymorphonuclear neutrophils and T lymphocytes: strange bedfellows or brothers in arms? Trends Immunol 30: 522-530.
  • MURPHY CA, LANGRISH CL, CHEN Y, BLUMENSCHEIN W, MCCLANAHAN T, KASTELEIN RA, SEDGWICK JD & CUA DJ. 2003. Divergent pro- and antiinflammatory roles for IL-23 and IL-12 in joint autoimmune inflammation. J Exp Med 198: 1951-1957.
  • OGINO T, THAN TA, HOSAKO M, OZAKI M, OMORI M & OKADA S. 2009. Taurine chloramine: a possible oxidant reservoir. Adv Exp Med Biol 643: 451-461.
  • OLIVEIRA MW, MINOTTO JB, DE OLIVEIRA MR, ZANOTTO-FILHO A, BEHR GA, ROCHA RF, MOREIRA JC & KLAMT F. 2010. Scavenging and antioxidant potential of physiological taurine concentrations against different reactive oxygen/nitrogen species. Pharmacol Rep 62(1): 185-193.
  • PARK E, JIA J, QUINN MR & SCHULLER-LEVIS G. 2002. Taurine chloramine inhibits lymphocyte proliferation and decreases cytokine production in activated human leukocytes. Clin Immunol 102(2): 179-184.
  • PARK E, SCHULLER-LEVIS G & QUINN MR. 1995. Taurine chloramine inhibits production of nitric oxide and TNF-alpha in activated RAW 264.7 cells by mechanisms that involve transcriptional and translational events. J Immunol 154: 4778-4784.
  • PERNER A ET AL. 2017. The intensive care medicine research agenda on septic shock. Intensive Care Med 43: 1294-1305.
  • RITTER C ET AL. 2003. Oxidative parameters and mortality in sepsis induced by cecal ligation and perforation. Intensive Care Med 29(10): 1782-1789.
  • RITTIRSCH D, FLIERL MA & WARD PA. 2008. Harmful molecular mechanisms in sepsis. Nat Rev Immunol 8: 776-787.
  • SCHULLER-LEVIS GB & PARK E. 2003. Taurine: new implications for an old amino acid. FEMS Microbiol Lett 226(2): 195-202.
  • SEYMOUR CW ET AL. 2016. Assessment of Clinical Criteria for Sepsis: For the Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA 315(8): 762-774.
  • THOMAS EL, GRISHAM MB & JEFFERSON MM. 1986a. Cytotoxicity of Chloramines. Methods Enzymol 132: 585-593.
  • THOMAS EL, GRISHAM MB & JEFFERSON MM. 1986b. Preparation and Characterization of Chloramines. Methods Enzymol 132: 569-585.
  • TILG H, TREHU E, ATKINS MB, DINARELLO CA & MIER JW. 1994. Interleukin-6 (IL-6) as an anti-inflammatory cytokine: induction of circulating IL-1 receptor antagonist and soluble tumor necrosis factor receptor p55. Blood 83(1): 113-118.
  • TINSLEY KW, GRAYSON MH, SWANSON PE, DREWRY AM, CHANG KC, KARL IE & HOTCHKISS RS. 2003. Sepsis induces apoptosis and profound depletion of splenic interdigitating and follicular dendritic cells. J Immunol 171: 909-914.
  • UNSINGER J, KAZAMA H, MCDONOUGH JS, GRIFFITH TS, HOTCHKISS RS & FERGUSON TA. 2010. Sepsis-induced apoptosis leads to active suppression of delayed-type hypersensitivity by CD8+ regulatory T cells through a TRAIL-dependent mechanism. J Immunol 184: 6766-6772.
  • VENET F ET AL. 2009. Increased circulating regulatory T cells (CD4+CD25+CD127-) contribute to lymphocyte anergy in septic shock patients. Intensive Care Med 35: 678-686.
  • VINTON NE, LAIDLAW SA, AMENT ME & KOPPLE JD. 1986. Taurine concentrations in plasma and blood cells of patients undergoing long-term parenteral nutrition. Am J Clin Nutr 44: 398-404.
  • VIRGIN HW, WHERRY EJ & AHMED R. 2009. Redefining chronic viral infection. Cell 138: 30-50.
  • WAGNER TH, DREWRY AM, MACMILLAN S, DUNNE WM, CHANG KC, KARL IE, HOTCHKISS RS & COBB JP. 2007. Surviving sepsis: bcl-2 overexpression modulates splenocyte transcriptional responses in vivo. Am J Physiol 292: 1751-1759.
  • WESCHE-SOLDATO DE, CHUNG CS, GREGORY SH, SALAZAR-MATHER TP, AYALA CA & AYALA A. 2007. CD8 T cells promote inflammation and apoptosis in the liver after sepsis: role of Fas-FasL. Am J Pathol 171: 87-96.
  • WHERRY EJ. 2011. T cell exhaustion. Nat Immunol 12: 492-499.
  • ZHANG H, SLUTSKY AS & VINCENT JL. 2000. Oxygen free radicals in ARDS, septic shock and organ dysfunction. Intensive Care Med 26: 474-476.

Publication Dates

  • Publication in this collection
    20 Nov 2020
  • Date of issue
    2020

History

  • Received
    14 Oct 2019
  • Accepted
    23 Mar 2020
Academia Brasileira de Ciências Rua Anfilófio de Carvalho, 29, 3º andar, 20030-060 Rio de Janeiro RJ Brasil, Tel: +55 21 3907-8100 - Rio de Janeiro - RJ - Brazil
E-mail: aabc@abc.org.br