Circulating Pneumolysin Is a Potent Inducer of Cardiac Injury during Pneumococcal Infection

Yasir Alhamdi, Daniel R Neill, Simon T Abrams, Hesham A Malak, Reham Yahya, Richard Barrett-Jolley, Guozheng Wang, Aras Kadioglu, Cheng-Hock Toh, Yasir Alhamdi, Daniel R Neill, Simon T Abrams, Hesham A Malak, Reham Yahya, Richard Barrett-Jolley, Guozheng Wang, Aras Kadioglu, Cheng-Hock Toh

Abstract

Streptococcus pneumoniae accounts for more deaths worldwide than any other single pathogen through diverse disease manifestations including pneumonia, sepsis and meningitis. Life-threatening acute cardiac complications are more common in pneumococcal infection compared to other bacterial infections. Distinctively, these arise despite effective antibiotic therapy. Here, we describe a novel mechanism of myocardial injury, which is triggered and sustained by circulating pneumolysin (PLY). Using a mouse model of invasive pneumococcal disease (IPD), we demonstrate that wild type PLY-expressing pneumococci but not PLY-deficient mutants induced elevation of circulating cardiac troponins (cTns), well-recognized biomarkers of cardiac injury. Furthermore, elevated cTn levels linearly correlated with pneumococcal blood counts (r=0.688, p=0.001) and levels were significantly higher in non-surviving than in surviving mice. These cTn levels were significantly reduced by administration of PLY-sequestering liposomes. Intravenous injection of purified PLY, but not a non-pore forming mutant (PdB), induced substantial increase in cardiac troponins to suggest that the pore-forming activity of circulating PLY is essential for myocardial injury in vivo. Purified PLY and PLY-expressing pneumococci also caused myocardial inflammatory changes but apoptosis was not detected. Exposure of cultured cardiomyocytes to PLY-expressing pneumococci caused dose-dependent cardiomyocyte contractile dysfunction and death, which was exacerbated by further PLY release following antibiotic treatment. We found that high PLY doses induced extensive cardiomyocyte lysis, but more interestingly, sub-lytic PLY concentrations triggered profound calcium influx and overload with subsequent membrane depolarization and progressive reduction in intracellular calcium transient amplitude, a key determinant of contractile force. This was coupled to activation of signalling pathways commonly associated with cardiac dysfunction in clinical and experimental sepsis and ultimately resulted in depressed cardiomyocyte contractile performance along with rhythm disturbance. Our study proposes a detailed molecular mechanism of pneumococcal toxin-induced cardiac injury and highlights the major translational potential of targeting circulating PLY to protect against cardiac complications during pneumococcal infections.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Fig 1. PLY-expressing but not PLY-deficient pneumococci…
Fig 1. PLY-expressing but not PLY-deficient pneumococci induce cardiac injury and inflammation.
(A-C) Representative Western blots showing circulating cTnI and cTnT in murine plasma following i.v. injection of D39/PLN-A (n = 4) (1x106 CFU) (A) serotype-1, sequence type 300 and 306 (n = 5) (1x106 CFU) (B), and serotype 6B and PLY-deletion pneumococci (ΔPLY) (1x106 CFU) (C). (D) Mean±SD of circulating cTnI from 4 mice (each group) that survived at 24 h after treatment with D39, PLN-A, ST300, ST306, ΔCbpA, 6B and ΔPLY (1x106 CFU each). *p<0.05 as compared to 0 h. (E) Linear correlation between pneumococcal CFU counts and circulating cTnI levels at various time points (n = 21). (F) Circulating levels of cTnI in plasma of mice following i.v. injection of D39 (1x106 CFU) with or without liposomes (lipo). (n = 3 each group). *p<0.05 as compared to 0 h, #p<0.05 as compared to D39 group. (G) Survival curves of mice infected (i.v.) with D39,PLN-A and ΔPLY (n = 10 for D39, n = 5 for PLN-A, n = 4 for ΔPLY). (H) Circulating levels of cTnI in plasma of mice that died and those that survived within the first 30 h post-D39 infection (n = 4 each group). *p<0.05 as compared to survival group. (I) Pathological examination of murine heart sections after infection with D39, serotype-1, serotype 6B and ΔCbpA pneumococci. H&E representative images (a-h) of murine heart sections under x4 magnification (a-c) and x60 magnification (d-h). Hearts from mice infected with D39 (30 h post infection, d), serotype-1 (e), 6B (f) and ΔCbpA (g) show inflammatory cell infiltration (arrows) at x60 magnification, these are absent in a normal heart section (h). (i-j) Immunohistochemistry images showing absence of pneumococcal capsule staining in heart from mice infected with D39 (i) and serotype-1 (j), despite presence of inflammatory cell infiltrations (arrows). (k) Fresh pneumococci (D39) (arrows) were stained in parallel, as a positive control for pneumococcal capsule staining. (l-n) Representative immunohistochemistry images showing absence of active caspase-3 staining in heart section from mice infected with D39 (l), serotype-1 (m) and 6B (n) despite presence of inflammatory cell infiltrations (arrows). (o) Gut microvilli of a septic mouse showing positive active caspase-3 signal (arrows), was used as a positive control for active caspase-3 staining.
Fig 2. Circulating pore-forming PLY mediates cardiac…
Fig 2. Circulating pore-forming PLY mediates cardiac injury and inflammation in vivo.
(A) Representative Western blots showing circulating cTnI and cTnT in murine plasma following i.v. injection of PLY (200 ng/g) and PdB (400 ng/g). (B) Mean±SD of circulating cTnI from 4 mice (each group) that survived at 24 h after treatment with PLY and PdB. *p<0.05 as compared to PdB group. (C) Survival curves of mice injected (i.v.) with PLY (400 and 200 ng/g) or PdB (400 ng/g) (n = 8 each group). (D) Pathological examination of murine heart sections after infection with purified PLY (200 ng/g) (a, b and d) and PdB (400 ng/g) (c). H&E representative images (a-c) of murine heart sections under x60 magnification showing inflammatory cell infiltrations (arrows) in mice dying within 6 hours (a) and those surviving for 24 hours (b). (c) PdB injection causes no pathological abnormality in murine myocardium. (d) Absence of active caspase-3 staining in murine myocardium following PLY injection.
Fig 3. Wild type (D39) but not…
Fig 3. Wild type (D39) but not PLY-deficient (PLN-A) pneumococci cause cardiomyocyte dysfunction and death in vitro.
(A) Viability of HL-1 cells was assessed at 8 h after incubation with increasing CFUs of D39 and PLN-A. Viability of untreated cells (UT) was set at 100%. Data are presented as Mean±SD (n = 4). (B) Time course of HL-1 cell viability after incubation with 5x106 CFU/ml D39 or PLN-A in the presence or absence of antibiotics (AB). Data are presented as Mean±SD (n = 4). (C) and (D) Effects of D39 and PLN-A (5x106 CFU/ml) on the total number of spontaneously contracting HL-1 cells over time using antibiotic-free (C) and antibiotic-containing media (D). Data are presented as Mean±SD (n = 4). (E) Effects of D39 and PLN-A (5x106 CFU/ml) on the rhythm and rate of contraction of HL-1 cells over time. Typical traces of spontaneous contraction are presented. (F-J) Effects of D39 and PLN-A (5x106 CFU/ml) on Peak Shortening (F), maximum velocity of shortening (+dL/dt) (G), Time to Peak (TTP) (H), Time to 90%-re-lengthening tR90 (I) and maximum velocity of relaxation (-dL/dt) (J) of HL-1 cells after 4 h treatment are presented as Mean±SD (n = 9 from 3 independent experiments). *ANOVA test, p<0.05.
Fig 4. PLY at sub-lytic doses adversely…
Fig 4. PLY at sub-lytic doses adversely affect cardiomyocyte function in vitro.
(A) Viability of HL-1 cells was assessed 30 min after incubation with increasing concentrations of PLY and PdB using the WST-8 assay. Viability of untreated cells (UT) was set at 100%. Data are presented as Mean±SD. * p<0.05 (n = 4). (B) Time course of HL-1 cell viability after incubation with 1.5 μg/ml PLY or PdB. *p<0.05 (n = 3). (C) Effects of increasing concentrations of PLY on the total number of spontaneously contracting HL-1 cells over time. Data are presented as Mean±SD. *p<0.05 (n = 4). (D) Representative traces of cardiomyocyte contraction before and after PLY and PdB treatment (n = 4). (E-I) Effects of PLY and PdB (1.0 μg/ml) on Peak Shortening (E), +dL/dt (F), TTP (G), tR90 (H) and -dL/dt (I) of HL-1 cells after 30 min treatment are presented as Mean±SD. *p<0.05 (n = 9 from 3 independent experiments).
Fig 5. PLY binds to cardiomyocyte membrane…
Fig 5. PLY binds to cardiomyocyte membrane and disrupts Ca2+ homeostasis and membrane potential at sub-lytic concentrations.
(A) HL-1 cells were incubated with 5 μg/ml FITC-PLY and 1.0 μg/ml propidium iodide (PI) to monitor disruption of membrane integrity over time. The localisation of FITC-PLY (green) and PI (red) were recorded using time lapse confocal microscopy (LSM 710, Zeiss) in a maintained environment of 5% CO2 at 37°C. Arrows indicate membrane binding of FITC-PLY. Bar = 20 μm (n = 3). (B) HL-1 cells were treated with 5, 2.5, or 1 μg/ml FITC-PLY along with 1.0 μg/ml PI for 30 min. Following washing in PBS and fixation in 4% PFA, localisation of FITC-PLY and PI were visualized under the same conditions for comparison. Typical images of each dose are presented (n = 3). Arrows indicate membrane binding of FITC-PLY. Bar = 10 μm. (C) HL-1 cells were incubated with 5 μg/ml FITC-PdB and 1.0 μg/ml propidium iodide (PI) to monitor disruption of membrane integrity over time. The localisation of FITC-PdB (green) and PI (red) were recorded as described in (A). Arrows indicate membrane binding of FITC-PdB. Bar = 20 μm (n = 3). (D) Membrane potential changes detected in HL-1 cells following exposure to PLY (1 μg/ml). Top panel: histogram showing typical resting membrane potential under untreated (UT) conditions and following exposure to PLY presented as Mean±SD (n = 3).* p = 0.01. Bottom panel: Typical action potential of HL-1 cells treated without (UT) or with PLY. (E) Changes of [Ca2+]i of HL-1 cardiomyocytes with fura-2am as an indicator were recorded using the IonOptix. Typical traces before and after PLY or PdB treatment in presence and absence of extracellular Ca2+ are presented. Caffeine (10 mM) was used to induce Ca2+ release from sarcoplasmic reticulum stores within cardiomyocytes.
Fig 6. Profound calcium influx induces cardiomyocyte…
Fig 6. Profound calcium influx induces cardiomyocyte dysfunction and injury in response to sub-lytic PLY.
(A) Mean±SD of both systolic and diastolic [Ca2+]i of HL-cells within 1 min of treatment with PLY/PdB (1.0 μg/ml) from 4 independent experiments are presented. *p<0.05. (B) Change in intracellular Ca2+ transient amplitude over 30 min in response to different concentrations of PLY. Ca2+ transient amplitude at 0 min was set at 100%. Data are presented as Mean±SD (n = 4). (C) and (D) The effects of changing extracellular Ca2+ concentration (in media) of HL-1 cells from 2 to 3 mM on +dL/dt (C) and cell viability (D) in the absence (UT) and presence of 1.0 μg/ml PLY. Mean±SD from 3 independent experiments are presented. * p<0.01 when compared to 2 mM extracellular Ca2+.
Fig 7. PLY activates the PKCα-cTnI axis…
Fig 7. PLY activates the PKCα-cTnI axis in HL-1 cardiomyocytes to depress contractility.
(A) Representative Western blots and (B) a band-quantification histogram showing the distribution of PKCα and PKCβII in the cytosol “C” and membrane “M” fractions of HL-1 cells before (UT) and 30 min after PLY-treatment. GAPDH and Pan-Cadherin were used as markers for “C” and “M” fractions, respectively. Data are presented as Mean±SD (n = 3). * p<0.05 compared to UT. (C-E) Typical Western blots showing PLY effects on the association of PKCα and PKCβII with the myofilament fraction (C), the phosphorylation of cTnI at the S43 and T144 PKC-dependent phosphorylation sites (D) and the effects of PKCα inhibitor Go6976 (5 nM) and the PKCβII inhibitor LY333531 (10 nM) on PLY (1.0 μg/ml)-induced phosphorylation of cTnI at S43 and T144 phosphorylation sites (E) in HL-1 cells after 30 min of PLY treatment. cTnI was used as endogenous control (n = 3). (F) and (G) Effects of PLY (1.0 μg/ml) ± Go6976 (5 nM) or LY333531 (10 nM) on Peak Shortening (F) and +dL/dt (G) after 30 min of PLY treatment. Data are presented as Mean±SD (n = 9). *p<0.05 compared to UT.
Fig 8. PLY induces endoplasmic reticulum (ER)…
Fig 8. PLY induces endoplasmic reticulum (ER) stress pathway without progressing to apoptosis in HL-1 cardiomyocytes.
(A) Typical Western blots showing the activation of ER stress markers, (p-elF2α, p-IRE, BiP), JNK and ERK in HL-1 cells 30 min after PLY treatment. (B) Representative western blots showing the effect of PLY on the induction of apoptotic markers CHOP and active caspase-3 after 8 hour of treatment. Thapsigargin (Tg 5 μM) and Staurosporin (Stau 10 μM) were used as positive inducers of CHOP and active caspase-3 respectively. (C-G) Effects of PLY (1.0 μg/ml) and PLY+ 4-Phenylbutyric acid (4-PBA, an ER stress inhibitor, 10 mM) on Peak Shortening (C), (+dL/dt) (D), TTP (E), tR90 (F) and (-dL/dt) (G) of HL-1 cells after 30 min treatment are presented as Mean±SD (n = 9 from 3 independent experiments). * p<0.05.
Fig 9. Activation of PKCα-cTnI pathway and…
Fig 9. Activation of PKCα-cTnI pathway and ER stress in murine cardiomyocytes exposed to D39 or PLY.
(A) Representative Western blots and (B) a band-quantification histogram showing the distribution of PKCα and PKCβII in the cytosol “C” and membrane “M” fractions of murine cardiomyocytes under untreated (UT), D39- and PLN-A-infection (1x106 CFU) conditions (24 h post-infection). GAPDH and Pan-Cadherin were used as markers for “C” and “M” fractions, respectively. Data are presented as Mean±SD (n = 3). * p<0.05. (C) and (D) Typical Western blots showing the effects of D39/PLN-A (1x106 CFU) infection (C) and PLY/PdB (200 ng/g) i.v. injection (D) on the association of PKCα and PKCβII with the myofilament fraction of murine cardiomyocytes 24 h post injection. cTnI was used as an endogenous control (n = 3). (E) and (F) Typical Western blots illustrating the phosphorylation of cTnI at the S43 and T144 phosphorylation sites in murine cardiomyocytes following D39/PLN-A infection (E) and PLY/PdB injection (F) (24 h post injection). cTnI was used as an endogenous control (n = 3). (G) and (H) Typical Western blots showing activation of ER stress markers in murine cardiomyocytes at 24 h post-infection with D39/PLN-A (G) and PLY/PdB (H) (n = 3).
Fig 10. The mechanisms and effects of…
Fig 10. The mechanisms and effects of PLY on cardiomyocytes.
High lytic concentrations of PLY induce large pore formation to lyse cells. However, sub-lytic concentrations of PLY bind cellular membrane to induce smaller pores thereby triggering profound Ca2+ influx into cardiomyocytes. The resulting abnormal increment in intracellular Ca2+ concentration [Ca2+]i causes significant membrane depolarization, activation of detrimental signalling pathways (e.g. PKCα-cTnI axis, ER stress) and reductions in the Ca2+ transient amplitude to cause rhythm disturbance and depression in contractile force. Ultimately, Ca2+ overload causes cellular injury which may account for cardiac troponin leakage from cardiomyocytes into the circulation. Toxin-sequestering liposomes offer a potential novel therapeutic intervention against the toxic effects of circulating PLY.

References

    1. Ortqvist A, Hedlund J, Kalin M (2005) Streptococcus pneumoniae: epidemiology, risk factors, and clinical features. Semin Respir Crit Care Med 26: 563–574.
    1. Viasus D, Garcia-Vidal C, Manresa F, Dorca J, Gudiol F, et al. (2013) Risk stratification and prognosis of acute cardiac events in hospitalized adults with community-acquired pneumonia. J Infect 66: 27–33. 10.1016/j.jinf.2012.09.003
    1. Corrales-Medina VF, Suh KN, Rose G, Chirinos JA, Doucette S, et al. (2011) Cardiac complications in patients with community-acquired pneumonia: a systematic review and meta-analysis of observational studies. PLoS Med 8: e1001048 10.1371/journal.pmed.1001048
    1. Ramirez J, Aliberti S, Mirsaeidi M, Peyrani P, Filardo G, et al. (2008) Acute myocardial infarction in hospitalized patients with community-acquired pneumonia. Clin Infect Dis 47: 182–187. 10.1086/589246
    1. Corrales-Medina VF, Musher DM, Wells GA, Chirinos JA, Chen L, et al. (2012) Cardiac complications in patients with community-acquired pneumonia: incidence, timing, risk factors, and association with short-term mortality. Circulation 125: 773–781. 10.1161/CIRCULATIONAHA.111.040766
    1. Mandal P, Chalmers JD, Choudhury G, Akram AR, Hill AT (2011) Vascular complications are associated with poor outcome in community-acquired pneumonia. QJM 104: 489–495. 10.1093/qjmed/hcq247
    1. Moammar MQ, Ali MI, Mahmood NA, DeBari VA, Khan MA (2010) Cardiac troponin I levels and alveolar-arterial oxygen gradient in patients with community-acquired pneumonia. Heart Lung Circ 19: 90–92. 10.1016/j.hlc.2009.08.009
    1. Ahl J, Littorin N, Forsgren A, Odenholt I, Resman F, et al. (2013) High incidence of septic shock caused by Streptococcus pneumoniae serotype 3—a retrospective epidemiological study. BMC Infect Dis 13: 492 10.1186/1471-2334-13-492
    1. Orsini J, Mainardi C, Muzylo E, Karki N, Cohen N, et al. (2012) Microbiological profile of organisms causing bloodstream infection in critically ill patients. J Clin Med Res 4: 371–377. 10.4021/jocmr1099w
    1. Parrillo JE, Parker MM, Natanson C, Suffredini AF, Danner RL, et al. (1990) Septic shock in humans. Advances in the understanding of pathogenesis, cardiovascular dysfunction, and therapy. Ann Intern Med 113: 227–242.
    1. Zanotti-Cavazzoni SL, Hollenberg SM (2009) Cardiac dysfunction in severe sepsis and septic shock. Curr Opin Crit Care 15: 392–397. 10.1097/MCC.0b013e3283307a4e
    1. Bessiere F, Khenifer S, Dubourg J, Durieu I, Lega JC (2013) Prognostic value of troponins in sepsis: a meta-analysis. Intensive Care Med 39: 1181–1189. 10.1007/s00134-013-2902-3
    1. Singanayagam A, Singanayagam A, Elder DH, Chalmers JD (2012) Is community-acquired pneumonia an independent risk factor for cardiovascular disease? Eur Respir J 39: 187–196. 10.1183/09031936.00049111
    1. Musher DM, Rueda AM, Kaka AS, Mapara SM (2007) The association between pneumococcal pneumonia and acute cardiac events. Clin Infect Dis 45: 158–165.
    1. Paton JC, Andrew PW, Boulnois GJ, Mitchell TJ (1993) Molecular analysis of the pathogenicity of Streptococcus pneumoniae: the role of pneumococcal proteins. Annu Rev Microbiol 47: 89–115.
    1. Brown AO, Mann B, Gao G, Hankins JS, Humann J, et al. (2014) Streptococcus pneumoniae Translocates into the Myocardium and Forms Unique Microlesions That Disrupt Cardiac Function. PLoS Pathog 10: e1004383 10.1371/journal.ppat.1004383
    1. Kadioglu A, Weiser JN, Paton JC, Andrew PW (2008) The role of Streptococcus pneumoniae virulence factors in host respiratory colonization and disease. Nat Rev Microbiol 6: 288–301. 10.1038/nrmicro1871
    1. Kadioglu A, Gingles NA, Grattan K, Kerr A, Mitchell TJ, et al. (2000) Host cellular immune response to pneumococcal lung infection in mice. Infect Immun 68: 492–501.
    1. Stringaris AK, Geisenhainer J, Bergmann F, Balshusemann C, Lee U, et al. (2002) Neurotoxicity of pneumolysin, a major pneumococcal virulence factor, involves calcium influx and depends on activation of p38 mitogen-activated protein kinase. Neurobiol Dis 11: 355–368.
    1. Dogan S, Zhang Q, Pridmore AC, Mitchell TJ, Finn A, et al. (2011) Pneumolysin-induced CXCL8 production by nasopharyngeal epithelial cells is dependent on calcium flux and MAPK activation via Toll-like receptor 4. Microbes Infect 13: 65–75. 10.1016/j.micinf.2010.10.003
    1. Bermpohl D, Halle A, Freyer D, Dagand E, Braun JS, et al. (2005) Bacterial programmed cell death of cerebral endothelial cells involves dual death pathways. J Clin Invest 115: 1607–1615.
    1. McNeela EA, Burke A, Neill DR, Baxter C, Fernandes VE, et al. (2010) Pneumolysin activates the NLRP3 inflammasome and promotes proinflammatory cytokines independently of TLR4. PLoS Pathog 6: e1001191 10.1371/journal.ppat.1001191
    1. Bers DM (2000) Calcium fluxes involved in control of cardiac myocyte contraction. Circ Res 87: 275–281.
    1. Reuter H, Scholz H (1977) The regulation of the calcium conductance of cardiac muscle by adrenaline. J Physiol 264: 49–62.
    1. Vassalle M, Lin CI (2004) Calcium overload and cardiac function. J Biomed Sci 11: 542–565.
    1. Pyrko P, Kardosh A, Liu YT, Soriano N, Xiong W, et al. (2007) Calcium-activated endoplasmic reticulum stress as a major component of tumor cell death induced by 2,5-dimethyl-celecoxib, a non-coxib analogue of celecoxib. Mol Cancer Ther 6: 1262–1275.
    1. Layland J, Solaro RJ, Shah AM (2005) Regulation of cardiac contractile function by troponin I phosphorylation. Cardiovasc Res 66: 12–21.
    1. Henry BD, Neill DR, Becker KA, Gore S, Bricio-Moreno L, et al. (2015) Engineered liposomes sequester bacterial exotoxins and protect from severe invasive infections in mice. Nat Biotechnol 33: 81–88. 10.1038/nbt.3037
    1. Berry AM, Yother J, Briles DE, Hansman D, Paton JC (1989) Reduced virulence of a defined pneumolysin-negative mutant of Streptococcus pneumoniae. Infect Immun 57: 2037–2042.
    1. Berry AM, Ogunniyi AD, Miller DC, Paton JC (1999) Comparative virulence of Streptococcus pneumoniae strains with insertion-duplication, point, and deletion mutations in the pneumolysin gene. Infect Immun 67: 981–985.
    1. Wells SM, Sleeper M (2008) Cardiac troponins. Journal of Veterinary Emergency and Critical Care 18: 235–245.
    1. Mahajan VS, Jarolim P (2011) How to interpret elevated cardiac troponin levels. Circulation 124: 2350–2354. 10.1161/CIRCULATIONAHA.111.023697
    1. Claycomb WC, Lanson NA Jr., Stallworth BS, Egeland DB, Delcarpio JB, et al. (1998) HL-1 cells: a cardiac muscle cell line that contracts and retains phenotypic characteristics of the adult cardiomyocyte. Proc Natl Acad Sci U S A 95: 2979–2984.
    1. White SM, Constantin PE, Claycomb WC (2004) Cardiac physiology at the cellular level: use of cultured HL-1 cardiomyocytes for studies of cardiac muscle cell structure and function. Am J Physiol Heart Circ Physiol 286: H823–829.
    1. Boyett MR, Moore M, Jewell BR, Montgomery RA, Kirby MS, et al. (1988) An improved apparatus for the optical recording of contraction of single heart cells. Pflugers Arch 413: 197–205.
    1. Fillon S, Soulis K, Rajasekaran S, Benedict-Hamilton H, Radin JN, et al. (2006) Platelet-activating factor receptor and innate immunity: uptake of gram-positive bacterial cell wall into host cells and cell-specific pathophysiology. J Immunol 177: 6182–6191.
    1. Paton JC, Lock RA, Lee CJ, Li JP, Berry AM, et al. (1991) Purification and immunogenicity of genetically obtained pneumolysin toxoids and their conjugation to Streptococcus pneumoniae type 19F polysaccharide. Infect Immun 59: 2297–2304.
    1. Shewell LK, Harvey RM, Higgins MA, Day CJ, Hartley-Tassell LE, et al. (2014) The cholesterol-dependent cytolysins pneumolysin and streptolysin O require binding to red blood cell glycans for hemolytic activity. Proc Natl Acad Sci U S A 111: E5312–5320. 10.1073/pnas.1412703111
    1. Sahoo SK, Kim T, Kang GB, Lee JG, Eom SH, et al. (2009) Characterization of calumenin-SERCA2 interaction in mouse cardiac sarcoplasmic reticulum. J Biol Chem 284: 31109–31121. 10.1074/jbc.M109.031989
    1. Wheeler-Jones CP (2005) Cell signalling in the cardiovascular system: an overview. Heart 91: 1366–1374.
    1. Powers FM, Farias S, Minami H, Martin AF, Solaro RJ, et al. (1998) Cardiac myofilament protein function is altered during sepsis. J Mol Cell Cardiol 30: 967–978.
    1. Wu LL, Tang C, Liu MS (2001) Altered phosphorylation and calcium sensitivity of cardiac myofibrillar proteins during sepsis. Am J Physiol Regul Integr Comp Physiol 281: R408–416.
    1. Steinberg SF (2008) Structural basis of protein kinase C isoform function. Physiol Rev 88: 1341–1378. 10.1152/physrev.00034.2007
    1. Pyle WG, Chen Y, Hofmann PA (2003) Cardioprotection through a PKC-dependent decrease in myofilament ATPase. Am J Physiol Heart Circ Physiol 285: H1220–1228.
    1. Kooij V, Zhang P, Piersma SR, Sequeira V, Boontje NM, et al. (2013) PKCalpha-specific phosphorylation of the troponin complex in human myocardium: a functional and proteomics analysis. PLoS One 8: e74847 10.1371/journal.pone.0074847
    1. Way KJ, Chou E, King GL (2000) Identification of PKC-isoform-specific biological actions using pharmacological approaches. Trends Pharmacol Sci 21: 181–187.
    1. Groenendyk J, Agellon LB, Michalak M (2013) Coping with endoplasmic reticulum stress in the cardiovascular system. Annu Rev Physiol 75: 49–67. 10.1146/annurev-physiol-030212-183707
    1. Wang J, Hu X, Jiang H (2014) ER stress-induced apoptosis: A novel therapeutic target in heart failure. Int J Cardiol.
    1. Zhang Y, Xia Z, La Cour KH, Ren J (2011) Activation of Akt rescues endoplasmic reticulum stress-impaired murine cardiac contractile function via glycogen synthase kinase-3beta-mediated suppression of mitochondrial permeation pore opening. Antioxid Redox Signal 15: 2407–2424. 10.1089/ars.2010.3751
    1. Zhang B, Zhang Y, La Cour KH, Richmond KL, Wang XM, et al. (2013) Mitochondrial aldehyde dehydrogenase obliterates endoplasmic reticulum stress-induced cardiac contractile dysfunction via correction of autophagy. Biochim Biophys Acta 1832: 574–584. 10.1016/j.bbadis.2013.01.013
    1. Urano F, Wang X, Bertolotti A, Zhang Y, Chung P, et al. (2000) Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1. Science 287: 664–666.
    1. Zinszner H, Kuroda M, Wang X, Batchvarova N, Lightfoot RT, et al. (1998) CHOP is implicated in programmed cell death in response to impaired function of the endoplasmic reticulum. Genes Dev 12: 982–995.
    1. Braun JS, Sublett JE, Freyer D, Mitchell TJ, Cleveland JL, et al. (2002) Pneumococcal pneumolysin and H(2)O(2) mediate brain cell apoptosis during meningitis. J Clin Invest 109: 19–27.
    1. Park CS, Cha H, Kwon EJ, Sreenivasaiah PK, Kim do H (2012) The chemical chaperone 4-phenylbutyric acid attenuates pressure-overload cardiac hypertrophy by alleviating endoplasmic reticulum stress. Biochem Biophys Res Commun 421: 578–584. 10.1016/j.bbrc.2012.04.048
    1. Ayala P, Montenegro J, Vivar R, Letelier A, Urroz PA, et al. (2012) Attenuation of endoplasmic reticulum stress using the chemical chaperone 4-phenylbutyric acid prevents cardiac fibrosis induced by isoproterenol. Exp Mol Pathol 92: 97–104. 10.1016/j.yexmp.2011.10.012
    1. Chironna M, Sallustio A, De Robertis A, Quarto M, Germinario C (2010) Case report: fulminant pneumococcal sepsis in an unvaccinated asplenic patient in Italy. Euro Surveill 15.
    1. Dalla Bona E, Beltrame V, Liessi F, Sperti C (2012) Fatal pneumococcal sepsis eleven years after distal pancreatectomy with splenectomy for pancreatic cancer. JOP 13: 693–695. 10.6092/1590-8577/1074
    1. Iinuma Y, Hirose Y, Tanaka T, Kumagai K, Miyajima M, et al. (2007) Rapidly progressive fatal pneumococcal sepsis in adults: a report of two cases. J Infect Chemother 13: 346–349.
    1. Mehta NJ, Khan IA, Gupta V, Jani K, Gowda RM, et al. (2004) Cardiac troponin I predicts myocardial dysfunction and adverse outcome in septic shock. International journal of cardiology 95: 13–17.
    1. ver Elst KM, Spapen HD, Nguyen DN, Garbar C, Huyghens LP, et al. (2000) Cardiac troponins I and T are biological markers of left ventricular dysfunction in septic shock. Clin Chem 46: 650–657.
    1. Ammann P, Fehr T, Minder EI, Gunter C, Bertel O (2001) Elevation of troponin I in sepsis and septic shock. Intensive Care Med 27: 965–969.
    1. Spreer A, Kerstan H, Bottcher T, Gerber J, Siemer A, et al. (2003) Reduced release of pneumolysin by Streptococcus pneumoniae in vitro and in vivo after treatment with nonbacteriolytic antibiotics in comparison to ceftriaxone. Antimicrob Agents Chemother 47: 2649–2654.
    1. Jedrzejas MJ (2001) Pneumococcal virulence factors: structure and function. Microbiol Mol Biol Rev 65: 187–207; first page, table of contents.
    1. El-Rachkidy RG, Davies NW, Andrew PW (2008) Pneumolysin generates multiple conductance pores in the membrane of nucleated cells. Biochem Biophys Res Commun 368: 786–792. 10.1016/j.bbrc.2008.01.151
    1. Sonnen AF, Plitzko JM, Gilbert RJ (2014) Incomplete pneumolysin oligomers form membrane pores. Open Biol 4: 140044 10.1098/rsob.140044
    1. Miragoli M, Gaudesius G, Rohr S (2006) Electrotonic modulation of cardiac impulse conduction by myofibroblasts. Circ Res 98: 801–810.
    1. Beurg M, Hafidi A, Skinner L, Cowan G, Hondarrague Y, et al. (2005) The mechanism of pneumolysin-induced cochlear hair cell death in the rat. J Physiol 568: 211–227.
    1. Tang J, Wang G, Liu Y, Fu Y, Chi J, et al. (2011) Cyclosporin A induces cardiomyocyte injury through calcium-sensing receptor-mediated calcium overload. Pharmazie 66: 52–57.
    1. Wang Y, Goldhaber JI (2004) Return of calcium: manipulating intracellular calcium to prevent cardiac pathologies. Proc Natl Acad Sci U S A 101: 5697–5698.
    1. Thandroyen FT, Morris AC, Hagler HK, Ziman B, Pai L, et al. (1991) Intracellular calcium transients and arrhythmia in isolated heart cells. Circ Res 69: 810–819.
    1. Braz JC, Gregory K, Pathak A, Zhao W, Sahin B, et al. (2004) PKC-alpha regulates cardiac contractility and propensity toward heart failure. Nat Med 10: 248–254.
    1. Minamino T, Komuro I, Kitakaze M (2010) Endoplasmic reticulum stress as a therapeutic target in cardiovascular disease. Circ Res 107: 1071–1082. 10.1161/CIRCRESAHA.110.227819
    1. Palaniyandi SS, Sun L, Ferreira JC, Mochly-Rosen D (2009) Protein kinase C in heart failure: a therapeutic target? Cardiovasc Res 82: 229–239. 10.1093/cvr/cvp001
    1. Belmonte SL, Blaxall BC (2011) PKC-ing is believing: targeting protein kinase C in heart failure. Circ Res 109: 1320–1322. 10.1161/CIRCRESAHA.111.259358
    1. Neill DR, Fernandes VE, Wisby L, Haynes AR, Ferreira DM, et al. (2012) T regulatory cells control susceptibility to invasive pneumococcal pneumonia in mice. PLoS Pathog 8: e1002660 10.1371/journal.ppat.1002660
    1. Morton DB (1985) Pain and laboratory animals. Nature 317: 106
    1. Abrams ST, Zhang N, Manson J, Liu T, Dart C, et al. (2013) Circulating histones are mediators of trauma-associated lung injury. Am J Respir Crit Care Med 187: 160–169. 10.1164/rccm.201206-1037OC
    1. Lewis R, Asplin KE, Bruce G, Dart C, Mobasheri A, et al. (2011) The role of the membrane potential in chondrocyte volume regulation. J Cell Physiol 226: 2979–2986. 10.1002/jcp.22646
    1. Ke L, Qi XY, Dijkhuis AJ, Chartier D, Nattel S, et al. (2008) Calpain mediates cardiac troponin degradation and contractile dysfunction in atrial fibrillation. J Mol Cell Cardiol 45: 685–693. 10.1016/j.yjmcc.2008.08.012

Source: PubMed

3
購読する