Pulmonary midkine inhibition ameliorates sepsis induced lung injury

Jing-Yuan Xu, Wei Chang, Qin Sun, Fei Peng, Yi Yang, Jing-Yuan Xu, Wei Chang, Qin Sun, Fei Peng, Yi Yang

Abstract

Background: Midkine is a multi-functional molecule participating in a various key pathological process. We aimed to evaluate the change of midkine in sepsis and its association with angiotensin-converting enzyme (ACE) system, as well as the mechanism by which midkine induced in sepsis and lung injury.

Methods: The peripheral blood sample of septic patients on admission was obtained and measured for midkine, ACE and angiotensin II. Cecal ligation and puncture (CLP) mouse model was used, and adeno-associated virus (AAV) was stilled trans-trachea for regional targeting midkine expression, comparing the severity of lung injury. Furthermore, we studied the in vitro mechanism of midkine activates ACE system by using inhibitors targeting candidate receptors of midkine, and its effects on the vascular endothelial cells.

Results: Plasma midkine was significantly elevated in sepsis, and was closely associated with ACE system. Both circulating and lung midkine was increased in CLP mouse, and was related to severe lung injury. Regional interfering midkine expression in lung tissue by AAV could alleviate acute lung injury in CLP model. In vitro study elucidated that Notch 2 participated in the activation of ACE system and angiotensin II release, induced by midkine and triggered vascular endothelial injury by angiotensin II induced reactive oxygen species production.

Conclusions: Midkine inhibition ameliorates sepsis induced lung injury, which might via ACE/Ang II pathway and the participation of Notch 2 in the stimulation of ACE. Trial registration Clinicaltrials.gov NCT02605681. Registered 12 November 2015.

Keywords: Angiotensin II; Angiotensin converting enzyme; Midkine; Sepsis.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
Clinical Profiles. a Plasma midkine remarkedly elevated in septic patients (n = 26) vs. healthy volunteers (n = 5). b The correlation between plasma midkine and ACE (Spearmen’s rho = 0.3793, P = 0.0676). c The correlation between plasma midkine and angiotensin II (Spearmen’s rho = 0.38, P = 0.07)
Fig. 2
Fig. 2
Midkine and ACE level in plasma and lung tissue. a Plasma midkine in CLP (n = 6) vs. sham (n = 6) model [ng/L, 747.3 (703.6–811.1) vs. 87 (64.5–199.5), P = 0.0095]. b Plasma ACE in CLP (n = 6) vs. sham (n = 6) [ng/mL, 454.1 (378.1–500.3) vs. 53.4 (46.3–63.8), P = 0.0022]. c Plasma angiotensin II in CLP (n = 6) vs. sham (n = 6) [ng/L, 7.2 (6.8–9.6) vs. 1.9 (1.2–3.3), P = 0.002]. d Midkine and ACE mRNA level in lung tissue in CLP (n = 6) vs. sham (n = 6), mRNA amplification was normalized to β-actin. e Midkine and ACE protein level was determined by Western blots, and the representative result was shown. Quantitative analysis of midkine and ACE protein using densitometry, normalized to β-actin
Fig. 3
Fig. 3
Targeted abrogation of midkine in the lung tissue ameliorated acute lung injury in CLP model. a Immunofluorescence staining of GFP of lung tissue in control mouse (WT), mouse transduced with vehicle AAV (AAV. Veh) and mouse transduced with AAV carrying midkine RNAi sequence (AAV.RNAi.MK, n = 6 in each group). Lower panel showed immunohistochemistry staining of midkine in lung tissue in WT, AAV.Veh and AAV.RNAi.MK group (n = 6 in each group). b Midkine and ACE protein levels in lung tissue determined by Western blots in control mouse (WT), mouse transduced with vehicle AAV (AAV.Veh) and mouse transduced with AAV carrying midkine RNAi sequence (AAV.RNAi.MK) (n = 6 in each group). c Immunohistochemistry staining of midkine in lung tissue of control mouse (WT) and mouse transduced with AAV midkine RNAi (AAV.RNAi.MK) with sham operation (upper panel), and the respective group mouse with CLP (lower panel) (n = 6 in each group). d H&E staining of control mouse (WT), mouse transduced with vehicle AAV (AAV.Veh) and mouse transduced with AAV carrying midkine RNAi sequence (AAV.RNAi.MK) in CLP vs. sham mouse were shown. Quantitative assessment by lung histopathological score was presented (n = 6). e BALF analysis from control mouse (WT), mouse transduced with vehicle AAV (AAV.Veh) and mouse transduced with AAV carrying midkine RNAi (AAV.RNAi.MK) in CLP vs sham mouse were shown (n = 6). Cell counts (upper left), TNF-α in BALF (upper right), protein contents in BALF (lower left) and lung wet/dry ratio (lower right) were shown (n = 6 in each group). f ACE hydrolytic activity determined by FAPGG in control mouse (WT), mouse transduced with vehicle AAV (AAV.Veh) and mouse transduced with AAV with midkine RNAi (AAV.RNAi.MK) in CLP vs. sham mouse was shown (left). ACE activity was measured at indicated time point. Angiotensin I was added to lung homogenization and the converted angiotensin II was determined (right) (n = 6 in each group)
Fig. 4
Fig. 4
Pulmonary microvascular endothelial cell injury was mediated by ACE system via Notch 2 receptor. a ACE mRNA level in pulmonary microvascular endothelial cell (PMVEC) in control group, cells stimulated by midkine (20 ng/mL and 100 ng/mL), and normalized to β-actin (upper left). ACE protein level in PMVEC lysates in control group, cells stimulated by midkine (20 ng/mL and 100 ng/mL). Quantitative analysis using densitometry normalized to β-actin was shown (lower left). PMVEC ACE activity determined by FAPGG in control group, cells stimulated by midkine, and cell stimulated by midkine and blocked with inhibitors targeting Notch 2 (DAPT), ALK (LDK378) and EGFR (Erlotinib), respectively. ACE activity was measured at indicated time point (upper right). Angiotensin II in the supernatant was determined after angiotensin I was added to the culture medium, in control group, cells stimulated with midkine, and cells stimulated with midkine blocked by DAPT (lower right). b Notch 2 and ACE protein level in PMVEC in control group, cells stimulated with midkine (100 ng/mL), and cells stimulated by midkine and blocked by DAPT. Quantitative analysis of full length (FL) and intra-cellular domain (NICD) of Notch 2 and ACE, using densitometry, normalized to β-actin. c Cell viability determined by CCK-8 assay of PMVEC in blank, control group, cells stimulated by midkine, cells stimulated by midkine and blocked by DAPT and ACEI respectively. Reactive oxygen species (ROS) production in PMVEC in blank, control group, cells stimulated by midkine, cells stimulated by midkine and blocked by DAPT and ACEI respectively. d The expression of inter-cellular adhesive molecules including VCAM-1 and VE-cadherin were determined by Western blots. The expression of VCAM-1 and VE-cadherin in PMVEC were shown in control group, cells stimulated by midkine, cells stimulated by midkine and blocked by DAPT and ACEI, respectively. e Immunofluorescence staining of VE-Cadherin and ZO-1 in PMVEC in control group, cells stimulated by midkine and cells stimulated by midkine but prohibited by DAPT

References

    1. Levy MM, 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(12):919–924. doi: 10.1016/S1473-3099(12)70239-6.
    1. Fleischmann C, et al. Assessment of global incidence and mortality of hospital-treated sepsis. Current estimates and limitations. Am J Respir Crit Care Med. 2016;193(3):259–272. doi: 10.1164/rccm.201504-0781OC.
    1. Weng L, et al. Sepsis-related mortality in China: a descriptive analysis. Intensive Care Med. 2018;44(7):1071–1080. doi: 10.1007/s00134-018-5203-z.
    1. Singer M, et al. The third international consensus definitions for sepsis and septic shock (sepsis-3) JAMA. 2016;315(8):801–810. doi: 10.1001/jama.2016.0287.
    1. Angus DC, van der Poll T. Severe sepsis and septic shock. N Engl J Med. 2013;369(9):840–851. doi: 10.1056/NEJMra1208623.
    1. Schmidt EP, et al. The pulmonary endothelial glycocalyx regulates neutrophil adhesion and lung injury during experimental sepsis. Nat Med. 2012;18(8):1217–1223. doi: 10.1038/nm.2843.
    1. Yu WK, et al. Vascular endothelial cadherin shedding is more severe in sepsis patients with severe acute kidney injury. Crit Care. 2019;23(1):18. doi: 10.1186/s13054-019-2315-y.
    1. Ince C, et al. The endothelium in sepsis. Shock. 2016;45(3):259–270. doi: 10.1097/SHK.0000000000000473.
    1. Johansson PI, Stensballe J, Ostrowski SR. Shock induced endotheliopathy (SHINE) in acute critical illness—a unifying pathophysiologic mechanism. Crit Care. 2017;21(1):25. doi: 10.1186/s13054-017-1605-5.
    1. Muramatsu T. Structure and function of midkine as the basis of its pharmacological effects. Br J Pharmacol. 2014;171(4):814–826. doi: 10.1111/bph.12353.
    1. Krzystek-Korpacka M, et al. Midkine, a multifunctional cytokine, in patients with severe sepsis and septic shock: a pilot study. Shock. 2011;35(5):471–477. doi: 10.1097/SHK.0b013e3182086001.
    1. Chang W, et al. Plasma midkine is associated with 28-Day mortality and organ function in sepsis. J Intensive Care Med. 2019;35(11):1290–1296. doi: 10.1177/0885066619861580.
    1. Hobo A, et al. The growth factor midkine regulates the renin-angiotensin system in mice. J Clin Invest. 2009;119(6):1616–1625. doi: 10.1172/JCI37249.
    1. Doerschug KC, et al. Renin-angiotensin system activation correlates with microvascular dysfunction in a prospective cohort study of clinical sepsis. Crit Care. 2010;14(1):R24. doi: 10.1186/cc8887.
    1. Correa TD, Takala J, Jakob SM. Angiotensin II in septic shock. Crit Care. 2015;19:98. doi: 10.1186/s13054-015-0802-3.
    1. Klein N, et al. Angiotensin-(1–7) protects from experimental acute lung injury. Crit Care Med. 2013;41(11):e334–343. doi: 10.1097/CCM.0b013e31828a6688.
    1. Montezano AC, et al. Angiotensin II and vascular injury. Curr Hypertens Rep. 2014;16(6):431. doi: 10.1007/s11906-014-0431-2.
    1. Nguyen Dinh Cat A, et al. Angiotensin II, NADPH oxidase, and redox signaling in the vasculature. Antioxid Redox Signal. 2013;19(10):1110–1120. doi: 10.1089/ars.2012.4641.
    1. Liu L, et al. Losartan, an antagonist of AT1 receptor for angiotensin II, attenuates lipopolysaccharide-induced acute lung injury in rat. Arch Biochem Biophys. 2009;481(1):131–136. doi: 10.1016/j.abb.2008.09.019.
    1. Honda Y, et al. Midkine deteriorates cardiac remodeling via epidermal growth factor receptor signaling in chronic kidney disease. Hypertension. 2016;67(5):857–865. doi: 10.1161/HYPERTENSIONAHA.115.06922.
    1. Kuo AH, et al. Recruitment of insulin receptor substrate-1 and activation of NF-kappaB essential for midkine growth signaling through anaplastic lymphoma kinase. Oncogene. 2007;26(6):859–869. doi: 10.1038/sj.onc.1209840.
    1. Huang Y, et al. Midkine induces epithelial-mesenchymal transition through Notch2/Jak2-Stat3 signaling in human keratinocytes. Cell Cycle. 2008;7(11):1613–1622. doi: 10.4161/cc.7.11.5952.
    1. Rhodes A, et al. Surviving sepsis campaign: international guidelines for management of sepsis and septic shock: 2016. Intensive Care Med. 2017;43(3):304–377. doi: 10.1007/s00134-017-4683-6.
    1. Rittirsch D, et al. Immunodesign of experimental sepsis by cecal ligation and puncture. Nat Protoc. 2009;4(1):31–36. doi: 10.1038/nprot.2008.214.
    1. Sue RD, et al. CXCR2 is critical to hyperoxia-induced lung injury. J Immunol. 2004;172(6):3860–3868. doi: 10.4049/jimmunol.172.6.3860.
    1. Zhang H, et al. IL-6 trans-signaling promotes pancreatitis-associated lung injury and lethality. J Clin Invest. 2013;123(3):1019–1031. doi: 10.1172/JCI64931.
    1. Rui T, et al. Erythropoietin prevents the acute myocardial inflammatory response induced by ischemia/reperfusion via induction of AP-1. Cardiovasc Res. 2005;65(3):719–727. doi: 10.1016/j.cardiores.2004.11.019.
    1. Takenaka H, et al. Midkine prevents ventricular remodeling and improves long-term survival after myocardial infarction. Am J Physiol Heart Circ Physiol. 2009;296(2):H462–469. doi: 10.1152/ajpheart.00733.2008.
    1. Weckbach LT, et al. Midkine acts as proangiogenic cytokine in hypoxia-induced angiogenesis. Am J Physiol Heart Circ Physiol. 2012;303(4):H429–438. doi: 10.1152/ajpheart.00934.2011.
    1. Badila E, et al. Midkine proteins in cardio-vascular disease. Where do we come from and where are we heading to? Eur J Pharmacol. 2015;762:464–471. doi: 10.1016/j.ejphar.2015.06.040.
    1. Salaru DL, et al. Midkine, a heparin-binding growth factor, and its roles in atherogenesis and inflammatory kidney diseases. Nephrol Dial Transplant. 2016;31(11):1781–1787. doi: 10.1093/ndt/gfw083.
    1. Misa K, et al. Involvement of midkine in the development of pulmonary fibrosis. Physiol Rep. 2017;5(16):e13383. doi: 10.14814/phy2.13383.
    1. Zhang R, et al. Mechanical stress and the induction of lung fibrosis via the midkine signaling Pathway. Am J Respir Crit Care Med. 2015;192(3):315–323. doi: 10.1164/rccm.201412-2326OC.
    1. Shindo E, et al. The growth factor midkine may play a pathophysiological role in rheumatoid arthritis. Mod Rheumatol. 2017;27(1):54–59. doi: 10.1080/14397595.2016.1179860.
    1. Paul M, Poyan Mehr A, Kreutz R. Physiology of local renin-angiotensin systems. Physiol Rev. 2006;86(3):747–803. doi: 10.1152/physrev.00036.2005.
    1. Garcia V, et al. 20-HETE activates the transcription of Angiotensin-Converting Enzyme via Nuclear Factor-kappa B translocation and promoter binding. J Pharmacol Exp Ther. 2016;356(3):525–533. doi: 10.1124/jpet.115.229377.
    1. Gungor C, et al. Notch signaling activated by replication stress-induced expression of midkine drives epithelial-mesenchymal transition and chemoresistance in pancreatic cancer. Cancer Res. 2011;71(14):5009–5019. doi: 10.1158/0008-5472.CAN-11-0036.
    1. Masuda T, et al. Growth factor midkine promotes T-Cell activation through Nuclear Factor of activated T Cells signaling and Th1 cell differentiation in lupus nephritis. Am J Pathol. 2017;187(4):740–751. doi: 10.1016/j.ajpath.2016.12.006.

Source: PubMed

3
Se inscrever