Toll-like receptor 4 regulates platelet function and contributes to coagulation abnormality and organ injury in hemorrhagic shock and resuscitation

Ning Ding, Guoqiang Chen, Rosemary Hoffman, Patricia A Loughran, Chhinder P Sodhi, David J Hackam, Timothy R Billiar, Matthew D Neal, Ning Ding, Guoqiang Chen, Rosemary Hoffman, Patricia A Loughran, Chhinder P Sodhi, David J Hackam, Timothy R Billiar, Matthew D Neal

Abstract

Background: Growing evidence indicates that the presence of toll-like receptor 4 (TLR4) on platelets is a key regulator of platelet number and function. Platelets exposed to TLR4 agonists may serve to activate other cells such as neutrophils and endothelial cells in sepsis and other inflammatory conditions. The functional significance of platelet TLR4 in hemorrhagic shock (HS), however, remains unexplored.

Methods and results: Using thromboelastography and platelet aggregometry, we demonstrate that platelet function is impaired in a mouse model of HS with resuscitation. Further analysis using cellular-specific TLR4 deletion in mice revealed that platelet TLR4 is essential for platelet activation and function in HS with resuscitation and that platelet TLR4 regulates the development of coagulopathy after hemorrhage and resuscitation. Transfusion of TLR4-negative platelets into mice resulted in protection from coagulopathy and restored platelet function. Additionally, platelet-specific TLR4 knockout mice were protected from lung and liver injury and exhibited a marked reduction in systemic inflammation as measured by circulating interleukin-6 after HS with resuscitation.

Conclusions: We demonstrate for the first time that platelet TLR4 is an essential mediator of the inflammatory response as well as platelet activation and function in HS and resuscitation.

Keywords: blood platelets; inflammation; shock, hemorrhagic; toll-like receptor 4.

Conflict of interest statement

Conflict of Interest Disclosures: None.

© 2014 American Heart Association, Inc.

Figures

Figure 1
Figure 1
Changes of platelet function in hemorrhagic shock mouse models. Thromboelastography (TEG) was used to investigate platelet function via maximal amplitude (MA) after HS or HS-R. A: MA value significantly decreased in HS-R mice compared with control and HS alone. Data shown are means ± SEM, n = 8–10 mice/group. *p

Figure 2

TLR4 contributes to platelet function…

Figure 2

TLR4 contributes to platelet function impairment in HS-R. A: TEG from wide type…

Figure 2
TLR4 contributes to platelet function impairment in HS-R. A: TEG from wide type (WT) mice and TLR4−/− subjected to HS-R or unmanipulated control. A: MA values showing reduction WT but not TLR4−/− mice after HS-R. Data shown are means ± SEM, n= 8–10 mice/group. *p

Figure 3

Platelet TLR4 contributes to platelet…

Figure 3

Platelet TLR4 contributes to platelet function impairment in HS-R. A: Whole blood of…

Figure 3
Platelet TLR4 contributes to platelet function impairment in HS-R. A: Whole blood of cell specific TLR4−/− mice were subjected to HS-R or unmanipulated control and analyzed by TEG. Data shown are means ± SEM, n= 4–6 mice/group. *ploxP/loxP and TLR4loxP/Pf4-cre mice following HS-R or control. Data are means ± SEM, n= 4–6 mice/group. *p<0.05 vs. control.

Figure 4

Assessment of platelet activation by…

Figure 4

Assessment of platelet activation by flow cytometry. Platelet rich plasma from TLR4 loxP/Pf4-cre…

Figure 4
Assessment of platelet activation by flow cytometry. Platelet rich plasma from TLR4loxP/Pf4-cre and TLR4loxP/loxP mice was isolated and stained for CD41 and CD62p (100uM ADP). A: Percentage of CD62p positive. Data are means ± SEM, n = 8–10 mice/group. *p<0.05 vs. control. B: Representative dual parameter dot-plot figures obtained by flow cytometry for CD41 and CD62p. C: Treatment of PRP from both TLR4loxP/Pf4-cre and TLR4loxP/loxP mice with 0.1U/ml thrombin confirms equal CD62p upregulation. Data are means ± SEM, n = 5 mice/group. p=NS between thrombin groups.

Figure 5

TLR4 regulates platelet aggregation following…

Figure 5

TLR4 regulates platelet aggregation following HS-R. Aggregometry was performed on wild-type (TLR4 +)…

Figure 5
TLR4 regulates platelet aggregation following HS-R. Aggregometry was performed on wild-type (TLR4 +) or mice lacking TLR4 on their platelets (TLR4 −). Results are expressed as the area under the aggregometry curve, measured as arbitrary units/minute (AU*min). A: Ex vivo collagen (5ug/ml) treatment of platelets demonstrates no significant difference in aggregation between groups. N=8 mice/group. p=NS. B: Aggregometry from either control (pre-shock blood draw) vs HS-R (post-shock). Collagen (5ug/ml) was present as the agonist in all samples. N=5 mice/group. C: Representative aggregometry tracings from control and HS-R. Each y axis block represents 10% change and each x axis block is 1 minute.

Figure 6

Effect of HS-R on platelet…

Figure 6

Effect of HS-R on platelet sequestration into lung and liver. TLR4 loxP/Pf4-cre mice…

Figure 6
Effect of HS-R on platelet sequestration into lung and liver. TLR4loxP/Pf4-cre mice and TLR4loxP/loxP mice were subjected to HS-R or unmanipulated control. Platelet sequestration into lung and liver of the mice were assessed by measuring the expression of CD41 using western blotting and immunofluorescence. A: The expression of CD41 in liver (A) and lung (B), analyzed by Western blotting. Data are means ± SEM, n = 8–10 mice/group. *p<0.05 vs. control. Livers (C) and lungs (D) were stained with CD41 (green), F-actin (white) antibody and counterstained with Hoechst to detect nuclei (blue) and imaged by confocal microscopy (Olympus FV1000 confocal, magnification ×200). Representative images of five individual experiments.

Figure 7

Effect of HS-R on lung…

Figure 7

Effect of HS-R on lung and liver injury and cytokine release. TLR4 loxP/Pf4-cre…

Figure 7
Effect of HS-R on lung and liver injury and cytokine release. TLR4loxP/Pf4-cre mice and TLR4loxP/loxP mice were subjected to HS-R or unmanipulated control and liver and lung injury was assessed by histology, circulating AST and ALT concentrations, lung MPO activity and circulating IL-6. Liver (A) and lung (B) were sectioned and stained with H&E and tissue damage induced by HS-R was assessed by light microscopy (Nikon FX series). Black arrows show the regions of necrosis. (Original magnification ×100). Serum AST (C) and ALT (D) levels were assayed and are presented as means ± SEM (n = 8–10 mice/group). Aliquots of the mouse lung tissues were homogenized to prepare total protein samples and MPO levels were assayed by ELISA (E). The levels are presented as means ± SEM (n = 8–10 mice/group). Serum samples from mice treated as described above were used to assay IL-6 concentration by ELISA (F) and the levels are presented as means ± SEM (n = 8–10 mice/group). *p<0.05 vs. control. #p<0.05 vs. HS-R group of TLR4loxP/loxP mice.

Figure 8

Platelet TLR4 is necessary and…

Figure 8

Platelet TLR4 is necessary and sufficient for the induction of platelet dysfunction and…

Figure 8
Platelet TLR4 is necessary and sufficient for the induction of platelet dysfunction and organ injury in HS-R. Wild-type (LOXP) and mice lacking TLR4 on platelets (PF4-cre) were depleted of their platelets using anti-CD41 antibody and subsequently transfused donor platelets isolated from either their littermates (LOXPtransLOXP, PF4-cretransPF4-cre) as controls or from donors with opposite TLR4 status with respect to their platelets (LOXPtransPF4-cre, PF4-cretransLOXP) to test the specific role of TLR4 on platelets in HS-R. (A) TEGs pre and post HS-R are shown and the MA are quantified in (B) as means ± SEM (n = 5 mice/group). (C) rSerum AST levels including both sham and HS-R (means ± SEM (n = 5 mice/group)). *p<0.01 vs. control. #p<0.05 vs. HS-R group of LOXPtransLOXP mice.
All figures (8)
Similar articles
Cited by
References
    1. Smith TL, Weyrich AS. Platelets as central mediators of systemic inflammatory responses. Thromb Res. 2011;127:391–394. - PMC - PubMed
    1. Tweardy DJ, Khoshnevis MR, Yu B, Mastrangelo MA, Hardison EG, López JA. Essential role for platelets in organ injury and inflammation in resuscitated hemorrhagic shock. Shock. 2006;26:386–390. - PubMed
    1. Bauer E, Chanthaphavong RS, Sodhi CP, Hackam DJ, Billiar TR, Bauer PM. Genetic deletion of TLR4 on platelets attenuates experimental pulmonary hypertension. Circ Res. 2014 Mar 17; [Epub ahead of print]. - PMC - PubMed
    1. Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell. 2010;140:805–820. - PubMed
    1. Blasius AL, Beutler B. Intracellular toll-like receptors. Immunity. 2010;32:305–315. - PubMed
Show all 50 references
Publication types
MeSH terms
Full text links [x]
[x]
Cite
Copy Download .nbib .nbib
Format: AMA APA MLA NLM
Figure 2
Figure 2
TLR4 contributes to platelet function impairment in HS-R. A: TEG from wide type (WT) mice and TLR4−/− subjected to HS-R or unmanipulated control. A: MA values showing reduction WT but not TLR4−/− mice after HS-R. Data shown are means ± SEM, n= 8–10 mice/group. *p

Figure 3

Platelet TLR4 contributes to platelet…

Figure 3

Platelet TLR4 contributes to platelet function impairment in HS-R. A: Whole blood of…

Figure 3
Platelet TLR4 contributes to platelet function impairment in HS-R. A: Whole blood of cell specific TLR4−/− mice were subjected to HS-R or unmanipulated control and analyzed by TEG. Data shown are means ± SEM, n= 4–6 mice/group. *ploxP/loxP and TLR4loxP/Pf4-cre mice following HS-R or control. Data are means ± SEM, n= 4–6 mice/group. *p<0.05 vs. control.

Figure 4

Assessment of platelet activation by…

Figure 4

Assessment of platelet activation by flow cytometry. Platelet rich plasma from TLR4 loxP/Pf4-cre…

Figure 4
Assessment of platelet activation by flow cytometry. Platelet rich plasma from TLR4loxP/Pf4-cre and TLR4loxP/loxP mice was isolated and stained for CD41 and CD62p (100uM ADP). A: Percentage of CD62p positive. Data are means ± SEM, n = 8–10 mice/group. *p<0.05 vs. control. B: Representative dual parameter dot-plot figures obtained by flow cytometry for CD41 and CD62p. C: Treatment of PRP from both TLR4loxP/Pf4-cre and TLR4loxP/loxP mice with 0.1U/ml thrombin confirms equal CD62p upregulation. Data are means ± SEM, n = 5 mice/group. p=NS between thrombin groups.

Figure 5

TLR4 regulates platelet aggregation following…

Figure 5

TLR4 regulates platelet aggregation following HS-R. Aggregometry was performed on wild-type (TLR4 +)…

Figure 5
TLR4 regulates platelet aggregation following HS-R. Aggregometry was performed on wild-type (TLR4 +) or mice lacking TLR4 on their platelets (TLR4 −). Results are expressed as the area under the aggregometry curve, measured as arbitrary units/minute (AU*min). A: Ex vivo collagen (5ug/ml) treatment of platelets demonstrates no significant difference in aggregation between groups. N=8 mice/group. p=NS. B: Aggregometry from either control (pre-shock blood draw) vs HS-R (post-shock). Collagen (5ug/ml) was present as the agonist in all samples. N=5 mice/group. C: Representative aggregometry tracings from control and HS-R. Each y axis block represents 10% change and each x axis block is 1 minute.

Figure 6

Effect of HS-R on platelet…

Figure 6

Effect of HS-R on platelet sequestration into lung and liver. TLR4 loxP/Pf4-cre mice…

Figure 6
Effect of HS-R on platelet sequestration into lung and liver. TLR4loxP/Pf4-cre mice and TLR4loxP/loxP mice were subjected to HS-R or unmanipulated control. Platelet sequestration into lung and liver of the mice were assessed by measuring the expression of CD41 using western blotting and immunofluorescence. A: The expression of CD41 in liver (A) and lung (B), analyzed by Western blotting. Data are means ± SEM, n = 8–10 mice/group. *p<0.05 vs. control. Livers (C) and lungs (D) were stained with CD41 (green), F-actin (white) antibody and counterstained with Hoechst to detect nuclei (blue) and imaged by confocal microscopy (Olympus FV1000 confocal, magnification ×200). Representative images of five individual experiments.

Figure 7

Effect of HS-R on lung…

Figure 7

Effect of HS-R on lung and liver injury and cytokine release. TLR4 loxP/Pf4-cre…

Figure 7
Effect of HS-R on lung and liver injury and cytokine release. TLR4loxP/Pf4-cre mice and TLR4loxP/loxP mice were subjected to HS-R or unmanipulated control and liver and lung injury was assessed by histology, circulating AST and ALT concentrations, lung MPO activity and circulating IL-6. Liver (A) and lung (B) were sectioned and stained with H&E and tissue damage induced by HS-R was assessed by light microscopy (Nikon FX series). Black arrows show the regions of necrosis. (Original magnification ×100). Serum AST (C) and ALT (D) levels were assayed and are presented as means ± SEM (n = 8–10 mice/group). Aliquots of the mouse lung tissues were homogenized to prepare total protein samples and MPO levels were assayed by ELISA (E). The levels are presented as means ± SEM (n = 8–10 mice/group). Serum samples from mice treated as described above were used to assay IL-6 concentration by ELISA (F) and the levels are presented as means ± SEM (n = 8–10 mice/group). *p<0.05 vs. control. #p<0.05 vs. HS-R group of TLR4loxP/loxP mice.

Figure 8

Platelet TLR4 is necessary and…

Figure 8

Platelet TLR4 is necessary and sufficient for the induction of platelet dysfunction and…

Figure 8
Platelet TLR4 is necessary and sufficient for the induction of platelet dysfunction and organ injury in HS-R. Wild-type (LOXP) and mice lacking TLR4 on platelets (PF4-cre) were depleted of their platelets using anti-CD41 antibody and subsequently transfused donor platelets isolated from either their littermates (LOXPtransLOXP, PF4-cretransPF4-cre) as controls or from donors with opposite TLR4 status with respect to their platelets (LOXPtransPF4-cre, PF4-cretransLOXP) to test the specific role of TLR4 on platelets in HS-R. (A) TEGs pre and post HS-R are shown and the MA are quantified in (B) as means ± SEM (n = 5 mice/group). (C) rSerum AST levels including both sham and HS-R (means ± SEM (n = 5 mice/group)). *p<0.01 vs. control. #p<0.05 vs. HS-R group of LOXPtransLOXP mice.
All figures (8)
Figure 3
Figure 3
Platelet TLR4 contributes to platelet function impairment in HS-R. A: Whole blood of cell specific TLR4−/− mice were subjected to HS-R or unmanipulated control and analyzed by TEG. Data shown are means ± SEM, n= 4–6 mice/group. *ploxP/loxP and TLR4loxP/Pf4-cre mice following HS-R or control. Data are means ± SEM, n= 4–6 mice/group. *p<0.05 vs. control.
Figure 4
Figure 4
Assessment of platelet activation by flow cytometry. Platelet rich plasma from TLR4loxP/Pf4-cre and TLR4loxP/loxP mice was isolated and stained for CD41 and CD62p (100uM ADP). A: Percentage of CD62p positive. Data are means ± SEM, n = 8–10 mice/group. *p<0.05 vs. control. B: Representative dual parameter dot-plot figures obtained by flow cytometry for CD41 and CD62p. C: Treatment of PRP from both TLR4loxP/Pf4-cre and TLR4loxP/loxP mice with 0.1U/ml thrombin confirms equal CD62p upregulation. Data are means ± SEM, n = 5 mice/group. p=NS between thrombin groups.
Figure 5
Figure 5
TLR4 regulates platelet aggregation following HS-R. Aggregometry was performed on wild-type (TLR4 +) or mice lacking TLR4 on their platelets (TLR4 −). Results are expressed as the area under the aggregometry curve, measured as arbitrary units/minute (AU*min). A: Ex vivo collagen (5ug/ml) treatment of platelets demonstrates no significant difference in aggregation between groups. N=8 mice/group. p=NS. B: Aggregometry from either control (pre-shock blood draw) vs HS-R (post-shock). Collagen (5ug/ml) was present as the agonist in all samples. N=5 mice/group. C: Representative aggregometry tracings from control and HS-R. Each y axis block represents 10% change and each x axis block is 1 minute.
Figure 6
Figure 6
Effect of HS-R on platelet sequestration into lung and liver. TLR4loxP/Pf4-cre mice and TLR4loxP/loxP mice were subjected to HS-R or unmanipulated control. Platelet sequestration into lung and liver of the mice were assessed by measuring the expression of CD41 using western blotting and immunofluorescence. A: The expression of CD41 in liver (A) and lung (B), analyzed by Western blotting. Data are means ± SEM, n = 8–10 mice/group. *p<0.05 vs. control. Livers (C) and lungs (D) were stained with CD41 (green), F-actin (white) antibody and counterstained with Hoechst to detect nuclei (blue) and imaged by confocal microscopy (Olympus FV1000 confocal, magnification ×200). Representative images of five individual experiments.
Figure 7
Figure 7
Effect of HS-R on lung and liver injury and cytokine release. TLR4loxP/Pf4-cre mice and TLR4loxP/loxP mice were subjected to HS-R or unmanipulated control and liver and lung injury was assessed by histology, circulating AST and ALT concentrations, lung MPO activity and circulating IL-6. Liver (A) and lung (B) were sectioned and stained with H&E and tissue damage induced by HS-R was assessed by light microscopy (Nikon FX series). Black arrows show the regions of necrosis. (Original magnification ×100). Serum AST (C) and ALT (D) levels were assayed and are presented as means ± SEM (n = 8–10 mice/group). Aliquots of the mouse lung tissues were homogenized to prepare total protein samples and MPO levels were assayed by ELISA (E). The levels are presented as means ± SEM (n = 8–10 mice/group). Serum samples from mice treated as described above were used to assay IL-6 concentration by ELISA (F) and the levels are presented as means ± SEM (n = 8–10 mice/group). *p<0.05 vs. control. #p<0.05 vs. HS-R group of TLR4loxP/loxP mice.
Figure 8
Figure 8
Platelet TLR4 is necessary and sufficient for the induction of platelet dysfunction and organ injury in HS-R. Wild-type (LOXP) and mice lacking TLR4 on platelets (PF4-cre) were depleted of their platelets using anti-CD41 antibody and subsequently transfused donor platelets isolated from either their littermates (LOXPtransLOXP, PF4-cretransPF4-cre) as controls or from donors with opposite TLR4 status with respect to their platelets (LOXPtransPF4-cre, PF4-cretransLOXP) to test the specific role of TLR4 on platelets in HS-R. (A) TEGs pre and post HS-R are shown and the MA are quantified in (B) as means ± SEM (n = 5 mice/group). (C) rSerum AST levels including both sham and HS-R (means ± SEM (n = 5 mice/group)). *p<0.01 vs. control. #p<0.05 vs. HS-R group of LOXPtransLOXP mice.

References

    1. Smith TL, Weyrich AS. Platelets as central mediators of systemic inflammatory responses. Thromb Res. 2011;127:391–394.
    1. Tweardy DJ, Khoshnevis MR, Yu B, Mastrangelo MA, Hardison EG, López JA. Essential role for platelets in organ injury and inflammation in resuscitated hemorrhagic shock. Shock. 2006;26:386–390.
    1. Bauer E, Chanthaphavong RS, Sodhi CP, Hackam DJ, Billiar TR, Bauer PM. Genetic deletion of TLR4 on platelets attenuates experimental pulmonary hypertension. Circ Res. 2014 Mar 17; [Epub ahead of print].
    1. Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell. 2010;140:805–820.
    1. Blasius AL, Beutler B. Intracellular toll-like receptors. Immunity. 2010;32:305–315.
    1. Moresco EM, LaVine D, Beutler B. Toll-like receptors. Curr Biol. 2011;21:R488–R493.
    1. Weyrich AS, Lindemann S, Zimmerman GA. The evolving role of platelets in inflammation. J Thromb Haemost. 2003;1:1897–1905.
    1. Weyrich AS, Zimmerman GA. Platelets: signaling cells in the immune continuum. Trends Immunol. 2004;25:489–495.
    1. Brown GT, McIntyre TM. Lipopolysaccharide signaling without a nucleus: kinase cascades stimulate platelet shedding of proinflammatory IL-1β-rich microparticles. J Immunol. 2011;186:5489–5496.
    1. Zhang G, Han J, Welch EJ, Ye RD, Voyno-Yasenetskaya TA, Malik AB, et al. Lipopolysaccharide stimulates platelet secretion and potentiates platelet aggregation via TLR4/MyD88 and the cGMP-dependent protein kinase pathway. J Immunol. 2009;182:7997–8004.
    1. Cognasse F, Hamzeh-Cognasse H, Lafarge S, Delezay O, Pozzetto B, McNicol A, Garraud O. Toll-like receptor 4 ligand can differentially modulate the release of cytokines by human platelets. Br J Haematol. 2008;141:84–91.
    1. Scott T, Owens MD. Thrombocytes respond to lipopolysaccharide through Toll-like receptor-4, and MAP kinase and NF-kappaB pathways leading to expression of interleukin-6 and cyclooxygenase-2 with production of prostaglandin E2. Mol Immunol. 2008;45:1001–1008.
    1. Ståhl AL, Svensson M, Mörgelin M, et al. Lipopolysaccharide from enterohemorrhagic Escherichia coli binds to platelets through TLR4 and CD62 and is detected on circulating platelets in patients with hemolytic uremic syndrome. Blood. 2006;108:167–176.
    1. Aslam R, Speck ER, Kim M, Crow AR, Bang KW, Nestel FP, et al. Platelet Toll-like receptor expression modulates lipopolysaccharide-induced thrombocytopenia and tumor necrosis factor-alpha production in vivo. Blood. 2006;107:637–641.
    1. Clark SR, Ma AC, Tavener SA, McDonald B, Goodarzi Z, Kelly MM, et al. Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat Med. 2007;13:463–469.
    1. Andonegui G, Kerfoot SM, McNagny K, Ebbert KV, Patel KD, Kubes P. Platelets express functional Toll-like receptor-4. Blood. 2005;106:2417–2423.
    1. Mollen KP, Anand RJ, Tsung A, Prince JM, Levy RM, Billiar TR. Emerging paradigm: toll-like receptor 4-sentinel for the detection of tissue damage. Shock. 2006;26:430–437.
    1. Benhamou Y, Favre J, Musette P, Renet S, Thuillez C, Richard V, et al. Toll-like receptors 4 contribute to endothelial injury and inflammation in hemorrhagic shock in mice. Crit Care Med. 2009;37:1724–1728.
    1. Fan J, Li Y, Levy RM, Fan JJ, Hackam DJ, Vodovotz Y, et al. Hemorrhagic shock induces NAD(P)H oxidase activation in neutrophils: role of HMGB1-TLR4 signaling. J Immunol. 2007;178:6573–6580.
    1. Prince JM, Levy RM, Yang R, Mollen KP, Fink MP, Vodovotz Y, et al. Toll-like receptor-4 signaling mediates hepatic injury and systemic inflammation in hemorrhagic shock. J Am Coll Surg. 2006;202:407–417.
    1. McGhan LJ, Jaroszewski DE. The role of toll-like receptor-4 in the development of multi-organ failure following traumatic haemorrhagic shock and resuscitation. Injury. 2012;43:129–136.
    1. Sodhi CP, Neal MD, Siggers R, Sho S, Ma C, Branca M, et al. Intestinal epithelial TLR4 regulates goblet cell development and is required for necrotizing enterocolitis in mice. Gastroenterology. 2012;143:708–718. e1–e5.
    1. Kohut LK, Darwiche SS, Brumfield JM, Frank AM, Billiar TR. Fixed volume or fixed pressure: a murine model of hemorrhagic shock. J Vis Exp. 2011;52:pii 2068.
    1. Nieswandt B, Echtenacher B, Wachs FP, Schroder J, Gessner JE, Schmidt RE, et al. Acute systemic reaction and lung alterations induced by an antiplatelet integrin gpIIb/IIIa antibody in mice. Blood. 1999;94:684–693.
    1. Rumbaut RE, Bellera RV, Randhawa JK, Shrimpton CN, Dasgupta SK, Dong JF, et al. Endotoxin enhances microvascular thrombosis in mouse cremaster venules via a TLR4-dependent, neutrophil-independent mechanism. Am J Physiol Heart Circ Physiol. 2006;290:H1671–H1679.
    1. Stark RJ, Aghakasiri N, Rumbaut RE. Platelet-derived Toll-like receptor 4 (Tlr-4) is sufficient to promote microvascular thrombosis in endotoxemia. PLoS One. 2012;7:e41254.
    1. Martini WZ, Cortez DS, Dubick MA, Park MS, Holcomb JB. Thrombelastography is better than PT, aPTT, and activated clotting time in detecting clinically relevant clotting abnormalities after hypothermia, hemorrhagic shock and resuscitation in pigs. J Trauma. 2008;65:535–543.
    1. Zarbock A, Ley K. The role of platelets in acute lung injury (ALI) Front Biosci. 2009;14:150–158.
    1. Zhao L, Ohtaki Y, Yamaguchi K, Matsushita M, Fujita T, Yokochi T, et al. LPS-induced platelet response and rapid shock in mice: contribution of O-antigen region of LPS and involvement of the lectin pathway of the complement system. Blood. 2002;100:3233–3239.
    1. Modlin RL, Brightbill HD, Godowski PJ. The toll of innate immunity on microbial pathogens. N Engl J Med. 1999;340:1834–1835.
    1. Cognasse F, Lafarge S, Chavarin P, Acquart S, Garraud O. Lipopolysaccharide induces sCD40L release through human platelets TLR4, but not TLR2 and TLR9. Intensive Care Med. 2007;33:382–384.
    1. Subeq YM, Hsu BG, Lin NT, Yang FL, Chao YF, Peng TC, et al. Hypothermia caused by slow and limited-volume fluid resuscitation decreases organ damage by hemorrhagic shock. Cytokine. 2012;60:68–75.
    1. Lv KY, Yu XY, Bai YS, Zhu SH, Tang HT, Ben DF, et al. Role of inhibition of p38 mitogen-activated protein kinase in liver dysfunction after hemorrhagic shock and resuscitation. J Surg Res. 2012;178:827–832.
    1. Nachman RL, Weksler B. The platelet as an inflammatory cell. Ann N Y Acad Sci. 1972;201:131–137.
    1. Kuroda T, Shiohara E. Leukocyte and platelet depletion protects the liver from damage induced by cholestasis and ischemia-reperfusion in the dog. Scand J Gastroenterol. 1996;31:182–190.
    1. Kuroda T, Shiohara E, Homma T, Furukawa Y, Chiba S. Effects of leukocyte and platelet depletion on ischemia-reperfusion injury to dog pancreas. Gastroenterology. 1994;107:1125–1134.
    1. Shimada Y, Kutsumi Y, Nishio H, Asazuma K, Tada H, Hayashi T, et al. Role of platelets in myocardial ischemia-reperfusion injury in dogs. Jpn Circ J. 1997;61:241–248.
    1. Lefer AM, Campbell B, Scalia R, Lefer DJ. Synergism between platelets and neutrophils in provoking cardiac dysfunction after ischemia and reperfusion: role of selectins. Circulation. 1998;98:1322–1328.
    1. Alves-Filho JC. Toll-like receptors on platelets: the key for disseminated intravascular coagulation in sepsis? Thromb Res. 2005;115:537–538.
    1. Funayama H, Huang L, Sato T, Ohtaki Y, Asada Y, Yokochi T, et al. Pharmacological characterization of anaphylaxis-like shock responses induced in mice by mannan and lipopolysaccharide. Int Immunopharmacol. 2009;9:1518–1524.
    1. Montrucchio G, Bosco O, Del Sorbo L, Fascio Pecetto P, Lupia E, Goffi A, et al. Mechanisms of the priming effect of low doses of lipopolysaccharides on leukocyte-dependent platelet aggregation in whole blood. Thromb Haemost. 2003;90:872–881.
    1. Kawabata Y, Yang TS, Yokochi TT, Matsushita M, Fujita T, Shibazaki M, et al. Complement system is involved in anaphylactoid reaction induced by lipopolysaccharides in muramyldipeptide treated mice. Shock. 2000;14:572–577.
    1. Ohba M, Shibazaki M, Sasano T, Inoue M, Takada H, Endo Y. Platelet responses and anaphylaxis- like shock induced in mice by intravenous injection of whole cells of oral streptococci. Oral Micro Immunol. 2004;19:26–30.
    1. Tsan MF, Gao B. Endogenous ligands of Toll-like receptors. J Leukoc Biol. 2004;76:514–519.
    1. Miyake K. Innate immune sensing of pathogens and danger signals by cell surface Toll-like receptors. Semin Immunol. 2007;19:3–10.
    1. Arumugam TV, Okun E, Tang SC, Thundyil J, Taylor SM, Woodruff TM. Toll-like receptors in ischemia-reperfusion injury. Shock. 2009;32:4–16.
    1. Chao W. Toll-like receptor signaling: a critical modulator of cell survival and ischemic injury in the heart. Am J Physiol Heart Circ Physiol. 2009;296:H1–H12.
    1. Yang R, Harada T, Mollen KP, Prince JM, Levy RM, Englert JA, et al. Anti-HMGB1 neutralizing antibody ameliorates gut barrier dysfunction and improves survival after hemorrhagic shock. Mol Med. 2006;12:105–114.
    1. Neal MD, Leaphart C, Levy R, Prince J, Billiar TR, Watkins S, et al. Enterocyte TLR4 mediates phagocytosis and translocation of bacteria across the intestinal barrier. J Immunol. 2006;176:3070–3079.
    1. Marcus AJ, Safier LB, Ullman HL, Broekman MJ, Islam N, Oglesby TD, et al. 12S,20-dihydroxyicosatetraenoic acid: a new icosanoid synthesized by neutrophils from 12S-hydroxyicosatetraenoic acid produced by thrombin- or collagen- stimulated platelets. Proc Natl Acad Sci U S A. 1984;81:903–907.

Source: PubMed

3
구독하다