Proton pump inhibitors protect mice from acute systemic inflammation and induce long-term cross-tolerance

E Balza, P Piccioli, S Carta, R Lavieri, M Gattorno, C Semino, P Castellani, A Rubartelli, E Balza, P Piccioli, S Carta, R Lavieri, M Gattorno, C Semino, P Castellani, A Rubartelli

Abstract

Incidence of sepsis is increasing, representing a tremendous burden for health-care systems. Death in acute sepsis is attributed to hyperinflammatory responses, but the underlying mechanisms are still unclear. We report here that proton pump inhibitors (PPIs), which block gastric acid secretion, selectively inhibited tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) secretion by Toll-like receptor (TLR)-activated human monocytes in vitro, in the absence of toxic effects. Remarkably, the oversecretion of IL-1β that represents a hallmark of monocytes from patients affected by cryopyrin-associated periodic syndrome is also blocked. Based on these propaedeutic experiments, we tested the effects of high doses of PPIs in vivo in the mouse model of endotoxic shock. Our data show that a single administration of PPI protected mice from death (60% survival versus 5% of untreated mice) and decreased TNF-α and IL-1β systemic production. PPIs were efficacious even when administered after lipopolysaccharide (LPS) injection. PPI-treated mice that survived developed a long-term cross-tolerance, becoming resistant to LPS- and zymosan-induced sepsis. In vitro, their macrophages displayed impaired TNF-α and IL-1β to different TLR ligands. PPIs also prevented sodium thioglycollate-induced peritoneal inflammation, indicating their efficacy also in a non-infectious setting independent of TLR stimulation. Lack of toxicity and therapeutic effectiveness make PPIs promising new drugs against sepsis and other severe inflammatory conditions.

Figures

Figure 1
Figure 1
OME inhibits IL-1β and TNF-α secretion induced by different PAMPs in human healthy monocytes. (a and b) Healthy monocytes were incubated in the medium at neutral pH (pH 7.4) or acidic pH (pH 6.5) with LPS (100 ng/ml) in the absence or presence of OME (300 μM). Secreted IL-1β (a) and TNF-α (b) were quantified after 18 and 6 h, respectively. Data are expressed as ng/ml (N=5, mean±S.E.M.). (cg) Dose–response experiments with 10–300 μM of OME (c), ESO (d), lansoprazole (e), pantoprazole (f) and rabeprazole (g) were performed. Supernatants were collected after 18 or 6 h to quantify IL-1β (left panels) and TNF-α (right panels). Data are expressed as the percentage of secretion of PPI versus PPI-untreated cells; mean±S.E.M. of four experiments. (h and i) Monocytes were stimulated for 18 and 6 h with LPS (100 ng/ml), R848 (5 μg/ml) and zymosan (ZYM, 20 μg/ml), alone or in combination (LRZ), in the presence or absence of OME. Secreted IL-1β (h) and TNF-α (i) were quantified as above. Data are expressed as ng/ml (N=5, mean±S.E.M.). *P<0.05; **P<0.01; ***P<0.001
Figure 2
Figure 2
OME prevents secretion by monocytes from patients affected by CAPS. Monocytes from CAPS patients (N=4) and healthy donors (N=4) were stimulated with 100 ng/ml of LPS alone or in combination with OME (300 μM). Secreted IL-1β was quantified by enzyme-linked immunosorbent assay (ELISA) in 18 h supernatants. Data are expressed as ng/ml. **P<0.01; ***P<0.001
Figure 3
Figure 3
OME downmodulates IL-1β and TNF-α secretion with different mechanisms. (a) Real-time PCR of IL-1β and TNF-α mRNA levels, 3 h from exposure to LPS or LPS+OME. Data are expressed as fold change of mRNA levels in cells stimulated with LPS or LPS+OME versus untreated cells (mean of normalized expression±S.E.M.; N=4). (b) IL-1β (18 h) and TNF-α (6 h) secreted by monocytes from the same subjects analyzed in (a). Data are expressed as the percent of secretion by LPS+OME versus LPS (mean±S.E.M.). (c) Western blot analysis of intracellular pro-IL-1β in monocytes unstimulated or at different time points from LPS stimulation, with or without OME. α-Tubulin is used as the loading control. One representative experiment out of five is shown. (d and e) IL-1β secreted by human monocytes (d) or murine peritoneal macrophages (e) primed 3 h (d) or 18 h (e) with LPS and then exposed 30 min to ATP, at 1 mM (d) or 5 mM (e) or to 20 μM nigericin (Nig, 20 min) with or without OME (mean±S.E.M.; N=4). (f) Western blot of p10 caspase-1 in cell lysates from murine macrophages. α-Tubulin is shown as the loading control (one representative experiment out of three). (g) Monocytes stimulated 3 h with LPS were loaded with PBFI and incubated in medium alone (LPS) or with 20 μM nigericin without (LPS+Nig) or with 300 μM OME (LPS+Nig+OME). Data are expressed as 340/380 ratio of the PBFI fluorescence intensity measured every 60 s for 20 min (mean±S.E.M.; N=3). (h) PBFI fluorescence intensity after 20 min in the different culture conditions depicted in (g) is expressed as percent versus time 0. *P<0.05, **P<0.01; ***P<0.001
Figure 4
Figure 4
Macrophages do not express the gastric H+/K+ ATPase, but display surface v-ATPases. (a) Reverse transcription-polymerase chain reaction (RT-PCR) analysis of mRNA coding for the β-subunit of the gastric H+/K+ proton pump on peritoneal mouse macrophages (Mφ) untreated or exposed 3 h to LPS (Mφ+LPS). Data are expressed as fold changes of normalized expression versus murine stomach (mean±S.E.M. of three experiments). (b and c) PBMCs from healthy donors were double stained with anti-v-ATPase and anti-CD14 antibody (Ab) or anti-CD3 Ab time 0 or 3 or 6 h after exposure to LPS and analyzed by FACS. In (b), data are expressed as the relative fluorescence intensity (RFI) of v-ATPase in CD14+ (monocytes) and CD3+ (T-lymphocytes) cells (mean±S.E.M. of three independent experiments). Statistical analysis is referred to monocytes and evaluated versust0. *P<0.05; **P<0.01 versus (c) a representative experiment of costaining (out of 3) is shown: 41% of CD14+ cells (upper plot) and 1.88% of CD3+ cells are positive for surface v-ATPases at 3 h from LPS exposure. *P<0.05; **P<0.01
Figure 5
Figure 5
ESO protects mice from LPS shock, suppresses the systemic production of TNF-α and IL-1β and induces resistance to LPS rechallenge. (a) Mice were injected intravenously with LPS (12.5 mg/kg) alone (N=40), or received intraperitoneally ESO (12.5 mg/kg) 30 min before (N=40), 30 min after (N=10) or 90 min after (N=7) the LPS injection. Mice were monitored for survival. (b) TNF-α (left panel) and IL-1β (right panel) in sera from LPS- and LPS+ESO-treated mice were quantified (ng/ml) 90 min (TNF-α) or 4 h (IL-1β) after LPS injection, respectively (mean±S.E.M., N=11). (c) Fifteen mice ESO treated and that survived the first LPS shock (survived LPS+ESO) were rechallenged with LPS, without any treatment, 3 weeks after the first LPS injection. As control, 10 naive mice were injected with LPS. Mice were monitored for survival. (d) TNF-α (left) and IL-1β (right) levels (ng/ml) were detected in the serum of naive and rechallenged mice by enzyme-linked immunosorbent assay (ELISA) (mean±S.E.M.; N=8 for TNF-α, N=7 for IL-1β). (e) Twelve mice receive a single injection of ESO 15 days before LPS challenge (ESO pre-treated). A control group of 10 naive mice received LPS only. Mice were monitored for 21 days for survival. (f) Serum levels of TNF-α (left) or IL-1β (right) from ESO pre-treated and naive mice were quantified as above. Data are expressed as ng/ml (mean±S.E.M.; n=6). *P<0.05, **P<0.01 and ***P<0.001
Figure 6
Figure 6
Macrophages from mice survived LPS rechallenge secrete less TNF-α and IL-1β in response to different TLR agonists. (a and b) Peritoneal macrophages from ESO-treated mice survived the first LPS shock (N=4) and from naive mice (N=6) were stimulated with LPS (a) or primed 18 h with LPS and then exposed 30 min to ATP (b), and TNF-α (a) and IL-1β (a and b) were quantified in supernatants after 4 or 18 h. (c) Real-time PCR analysis of P2X7 receptor mRNA from macrophages from naive mice or from mice survived the first LPS shock (survived LPS+ESO) after 3 h exposure to LPS. (d–f) Peritoneal macrophages from ESO-treated mice survived the second LPS shock (N=4) and from naive mice (N=6) were stimulated with LPS, zymosan (ZYM), R848, Pam(3)CSK(4) (PAM3), poly(I:C) and flagellin (Flag) for 4 (d) or 18 h (e) or with LPS, Pam(3)CSK(4) or poly(I:C) for 18 h followed by 30 min with 5 mM ATP (f). Secreted TNF-α (d) and IL-1β (e and f) were quantified (ng/ml; mean±S.E.M.; N=4). *P<0.05; **P<0.01; ***P<0.001
Figure 7
Figure 7
ESO-treated mice that survived LPS shock display resistance to zymosan-induced generalized inflammation. ESO-treated mice that recovered from the first LPS challenge (survived LPS+ESO, N=5) and naive mice (N=7) were injected with zymosan (Zym, 1 g/kg). Mice were monitored for survival (a) and for loss of body weight (b; N=4). (c) Serum levels of TNF-α (left; N=4 naive+Zym and N=5 survived Zym-treated mice) and of IL-1β (right; N=3 naive+Zym and N=4 survived Zym-treated mice) were quantified (ng/ml; mean±S.E.M.). *P<0.05
Figure 8
Figure 8
ESO prevents thioglycollate-induced peritonitis. (a and c) Peritoneal cells isolated 4 h (a) or 72 h (c) from untreated (Unt) mice or from mice injected intraperitoneally with thioglycollate alone (Thio) or thioglycollate 30 min after intraperitoneal injection with ESO (Thio+ESO) were counted. Data are expressed as the total number of infiltrating inflammatory cells (mean±S.E.M.; N=3). (b and d) The relative percent of macrophages, lymphocytes and granulocytes in peritoneum lavage from the same mice was calculated at 4 h (b) and 72 h (d). (e, f and g) MIP-2 (e), KC (f) and MCP-1 (g) levels were determined in serum and peritoneal lavage 4 h after thioglicollate treatment in untreated or ESO-treated mice (pg/ml; N=3, mean±S.E.M.) *P<0.05; **P<0.01; ***P<0.001

References

    1. Hotchkiss RS, Karl IE. The pathophysiology and treatment of sepsis. N Engl J Med 2003; 348: 138–150.
    1. Angus DC, van der Poll T. Severe sepsis and septic shock. N Engl J Med 2013; 369: 840–851.
    1. Lagu T, Rothberg MB, Shieh MS, Pekow PS, Steingrub JS, Lindenauer PK. Hospitalizations, costs, and outcomes of severe sepsis in the United States 2003 to 2007. Crit Care Med 2007; 40: 754–761.
    1. Levy MM, Artigas A, Phillips GS, Rhodes A, Beale R, Osborn T et al. Outcomes of the surviving sepsis campaign in intensive care units in the USA and Europe: a prospective cohort study. Lancet Infect Dis 2012; 12: 919–924.
    1. Yende S, Linde-Zwirble W, Mayr F, Weissfeld LA, Reis S, Angus DC. Risk of cardiovascular events in survivors of severe sepsis. Am J Respir Crit Care Med 2014; 189: 1065–1074.
    1. Angus DC. The search for effective therapy for sepsis: back to the drawing board? JAMA 2011; 306: 2614–2615.
    1. Lavieri R, Piccioli P, Carta S, Delfino L, Castellani P, Rubartelli A. TLR costimulation causes oxidative stress with unbalance of proinflammatory and anti-inflammatory cytokine production. J Immunol 2014; 192: 5373–5381.
    1. Cerami A, Beutler B. The role of cachectin/TNF in endotoxic shock and cachexia. Immunol Today 1988; 9: 28–31.
    1. Li P, Allen H, Banerjee S, Franklin S, Herzog L, Johnston C et al. Mice deficient in IL-1 beta-converting enzyme are defective in production of mature IL-1 beta and resistant to endotoxic shock. Cell 1995; 80: 401–411.
    1. Wang H, Bloom O, Zhang M, Vishnubhakat JM, Ombrellino M, Che J et al. HMG-1 as a late mediator of endotoxin lethality in mice. Science 1999; 285: 248–251.
    1. Williams SC. After Xigris, researchers look to new targets to combat sepsis. Nat Med 2012; 18: 1001.
    1. Noritomi DT, Soriano FG, Kellum JA, Cappi SB, Biselli PJ, Libório Ab et al. Metabolic acidosis in patients with severe sepsis and septic shock: a longitudinal quantitative study. Crit Care Med 2009; 37: 2733–2739.
    1. Suetrong B, Walley KR. Lactic acidosis in sepsis: it's not all anaerobic: implications for diagnosis and management. Chest 2016; 149: 252–261.
    1. Riemann A, Ihling A, Thomas J, Schneider B, Thews O, Gekle M. Acidic environment activates inflammatory programs in fibroblasts via a cAMP-MAPK pathway. Biochim Biophys Acta 2015; 1853: 299–307.
    1. Okajima F. Regulation of inflammation by extracellular acidification and proton-sensing GPCRs. Cell Signal 2013; 25: 2263–2271.
    1. Kelly B, O'Neill LA. Metabolic reprogramming in macrophages and dendritic cells in innate immunity. Cell Res 2015; 25: 771–784.
    1. Rubartelli A, Bajetto A, Allavena G, Cozzolino F, Sitia R. Post-translational regulation of interleukin 1 beta secretion. Cytokine 1993; 5: 117–124.
    1. Jancic CC, Cabrini M, Gabelloni ML, Rodríguez Rodrigues C, Salamone G, Trevani AS et al. Low extracellular pH stimulates the production of IL-1 beta by human monocytes. Cytokine 2012; 57: 258–268.
    1. Rajamäki K, Nordström T, Nurmi K, Åkerman KE, Kovanen PT, Öörni K et al. Extracellular acidosis is a novel danger signal alerting innate immunity via the NLRP3 inflammasome. J Biol Chem 2013; 288: 3410–3419.
    1. Edye ME, Lopez-Castejon G, Allan SM, Brough D. Acidosis drives damage-associated molecular pattern (DAMP)-induced interleukin-1 secretion via a caspase-1-independent pathway. J Biol Chem 2013; 288: 30485–30494.
    1. Shin JM, Sachs G. Pharmacology of proton pump inhibitors. Curr Gastroenterol Rep 2008; 10: 528–534.
    1. Robinson M, Horn J. Clinical pharmacology of proton pump inhibitors: what the practising physician needs to know. Drugs 2003; 63: 2739–2754.
    1. Peppicelli S, Bianchini F, Calorini F. Extracellular acidity, a ‘reappreciated' trait of tumor environment driving malignancy: perspectives in diagnosis and therapy. Cancer Metast Rev 2014; 33: 823–832.
    1. De Milito A, Canese R, Marino ML, Borghi M, Iero M, Villa A et al. pH-dependent antitumor activity of proton pump inhibitors against human melanoma is mediated by inhibition of tumor acidity. Int J Cancer 2010; 127: 207–219.
    1. Kedika RR, Souza RF, Spechler SJ. Potential anti-inflammatory effects of proton pump inhibitors: a review and discussion of the clinical implications. Dig Dis Sci 2009; 54: 2312–2317.
    1. Poli-de-Figueiredo LF, Garrido AG, Nakagawa N, Sannomiya P. Experimental models of sepsis and their clinical relevance. Shock 2008; 30: 53–59.
    1. Rubartelli A. Redox control of NLRP3 inflammasome activation in health and disease. J Leukocyte Biol 2012; 92: 951–958.
    1. Mariathasan S, Weiss DS, Newton K, McBride J, O'Rourke K, Roose-Girma M et al. Cryopyrin activates the inflammasome in response to toxins and ATP. Nature 2006; 440: 228–232.
    1. Muñoz-Planillo R, Kuffa P, Martínez-Colón G, Smith BL, Rajendiran TM, Núñez G. K+ efflux is the common trigger of NLRP3 inflammasome activation by bacterial toxins and particulate matter. Immunity 2013; 38: 1142–1153.
    1. Arlehamn CS, Pétrilli V, Gross O, Tschopp J, Evans TJ. The role of potassium in inflammasome activation by bacteria. J Biol Chem 2010; 285: 10508–10518.
    1. Nishi T, Forgac M. The vacuolar (H+)-ATPases – nature's most versatile proton pumps. Nat Rev Mol Cell Biol 2002; 3: 94–103.
    1. Fais S, De Milito A, You H, Qin W. Targeting vacuolar H+-ATPases as a new strategy against cancer. Cancer Res 2007; 67: 10627–10630.
    1. Röhss K, Wilder-Smith C, Kilhamn J, Fjellman M, Lind T. Suppression of gastric acid with intravenous esomeprazole and omeprazole: results of 3 studies in healthy subjects. Int J Clin Pharmacol Ther 2007; 45: 345–354.
    1. Ferrari D, Pizzirani C, Adinolfi E. The P2X7 receptor: a key player in IL-1 processing and release. J Immunol 2006; 176: 3877–3883.
    1. Genovese T, Di Paola R, Catalano P, Li JH, Xu W, Massuda E et al. Treatment with a novel poly(ADP-ribose) glycohydrolase inhibitor reduces development of septic shock-like syndrome induced by zymosan in mice. Crit Care Med 2004; 32: 1365–1374.
    1. Li YM, Baviello G, Vlassara H, Mitsuhashi T. Glycation products in aged thioglycolate medium enhance the elicitation of peritoneal macrophages. J Immunol Methods 1997; 201: 183–188.
    1. Henderson RB, Hobbs JAR, Mathies M, Hogg N. Rapid recruitment of inflammatory monocytes is independent of neutrophil migration. Blood 2013; 102: 328–325.
    1. Garlanda C, Dinarello CA. The interleukin-1 family: back to the future. Immunity 2013; 39: 1003–1018.
    1. Tracey KJ, Cerami A. Tumor necrosis factor: a pleiotropic cytokine and therapeutic target. Annu Rev Med 1994; 45: 491–503.
    1. Lv S, Han M, Yi R, Kwon S, Dai C, Wang R. Anti-TNF-alpha therapy for patients with sepsis: a systematic meta-analysis. Int J Clin Pract 2014; 68: 520–528.
    1. Dellinger RP, Levy MM, Rhodes A, Annane D, Gerlach H, Opal SM et al. Surviving Sepsis Campaign Guidelines Committee including the Pediatric Subgroup. Surviving sepsis campaign: international guidelines for management of severe sepsis and septic shock: 2012. Crit Care Med 2013; 41: 580–637.
    1. Kawai T, Akira S. The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat Immunol 2010; 11: 373–384.
    1. Coll RC, Robertson AA, Chae JJ, Higgins SC, Muñoz-Planillo R, Inserra MC et al. A small-molecule inhibitor of the NLRP3 inflammasome for the treatment of inflammatory diseases. Nat Med 2015; 21: 248–255.
    1. Wolosin JM. Ion transport studies with H+-K+-ATPase-rich vesicles: implications for HCl secretion and parietal cell physiology. Am J Physiol 1985; 248: G595–G607.
    1. Biswas SK, Lopez-Collazo E. Endotoxin tolerance: new mechanisms, molecules and clinical significance. Trends Immunol 2009; 30: 475–487.
    1. Deutschman CS, Tracey KJ. Sepsis: current dogma and new perspectives. Immunity 2014; 40: 463–475.
    1. Parikh N, Howden CW. The safety of drugs used in acid-related disorders and functional gastrointestinal disorders. Gastroenterol Clin N Am 2010; 39: 529–542.
    1. Bardou M, Martin J, Barkun A. Intravenous proton pump inhibitors: an evidence-based review of their use in gastrointestinal disorders. Drugs 2009; 69: 435–448.
    1. Dellinger RP, Levy MM, Carlet JM, Bion J, Parker MM, Jaeschke R et al. Surviving Sepsis Campaign: international guidelines for management of severe sepsis and septic shock Crit Care Med 2008; 36: 296–327.
    1. Carta S, Penco F, Lavieri R, Martini A, Dinarello CA, Gattorno M et al. Cell stress increases ATP release in NLRP3 inflammasome-mediated autoinflammatory diseases, resulting in cytokine imbalance. Proc Natl Acad Sci USA 2015; 112: 283528–283540.
    1. Simon P. Q-Gene: processing quantitative real-time RT-PCR data. Bioinformatics 2003; 19: 1439–1440.

Source: PubMed

3
S'abonner