Skip to main content
Top
Published in: Journal of Translational Medicine 1/2021

Open Access 01-12-2021 | Septicemia | Research

Pulmonary midkine inhibition ameliorates sepsis induced lung injury

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

Published in: Journal of Translational Medicine | Issue 1/2021

Login to get access

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
Appendix
Available only for authorised users
Literature
1.
go back to reference 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):919–24.CrossRef 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):919–24.CrossRef
2.
go back to reference 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):259–72.CrossRef 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):259–72.CrossRef
3.
go back to reference Weng L, et al. Sepsis-related mortality in China: a descriptive analysis. Intensive Care Med. 2018;44(7):1071–80.CrossRef Weng L, et al. Sepsis-related mortality in China: a descriptive analysis. Intensive Care Med. 2018;44(7):1071–80.CrossRef
4.
go back to reference Singer M, et al. The third international consensus definitions for sepsis and septic shock (sepsis-3). JAMA. 2016;315(8):801–10.CrossRef Singer M, et al. The third international consensus definitions for sepsis and septic shock (sepsis-3). JAMA. 2016;315(8):801–10.CrossRef
5.
go back to reference Angus DC, van der Poll T. Severe sepsis and septic shock. N Engl J Med. 2013;369(9):840–51.CrossRef Angus DC, van der Poll T. Severe sepsis and septic shock. N Engl J Med. 2013;369(9):840–51.CrossRef
6.
go back to reference Schmidt EP, et al. The pulmonary endothelial glycocalyx regulates neutrophil adhesion and lung injury during experimental sepsis. Nat Med. 2012;18(8):1217–23.CrossRef Schmidt EP, et al. The pulmonary endothelial glycocalyx regulates neutrophil adhesion and lung injury during experimental sepsis. Nat Med. 2012;18(8):1217–23.CrossRef
7.
go back to reference 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.CrossRef 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.CrossRef
8.
9.
go back to reference Johansson PI, Stensballe J, Ostrowski SR. Shock induced endotheliopathy (SHINE) in acute critical illness—a unifying pathophysiologic mechanism. Crit Care. 2017;21(1):25.CrossRef Johansson PI, Stensballe J, Ostrowski SR. Shock induced endotheliopathy (SHINE) in acute critical illness—a unifying pathophysiologic mechanism. Crit Care. 2017;21(1):25.CrossRef
10.
go back to reference Muramatsu T. Structure and function of midkine as the basis of its pharmacological effects. Br J Pharmacol. 2014;171(4):814–26.CrossRef Muramatsu T. Structure and function of midkine as the basis of its pharmacological effects. Br J Pharmacol. 2014;171(4):814–26.CrossRef
11.
go back to reference Krzystek-Korpacka M, et al. Midkine, a multifunctional cytokine, in patients with severe sepsis and septic shock: a pilot study. Shock. 2011;35(5):471–7.CrossRef Krzystek-Korpacka M, et al. Midkine, a multifunctional cytokine, in patients with severe sepsis and septic shock: a pilot study. Shock. 2011;35(5):471–7.CrossRef
12.
go back to reference Chang W, et al. Plasma midkine is associated with 28-Day mortality and organ function in sepsis. J Intensive Care Med. 2019;35(11):1290–6.CrossRef Chang W, et al. Plasma midkine is associated with 28-Day mortality and organ function in sepsis. J Intensive Care Med. 2019;35(11):1290–6.CrossRef
13.
go back to reference Hobo A, et al. The growth factor midkine regulates the renin-angiotensin system in mice. J Clin Invest. 2009;119(6):1616–25.CrossRef Hobo A, et al. The growth factor midkine regulates the renin-angiotensin system in mice. J Clin Invest. 2009;119(6):1616–25.CrossRef
14.
go back to reference 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.CrossRef 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.CrossRef
15.
go back to reference Correa TD, Takala J, Jakob SM. Angiotensin II in septic shock. Crit Care. 2015;19:98.CrossRef Correa TD, Takala J, Jakob SM. Angiotensin II in septic shock. Crit Care. 2015;19:98.CrossRef
16.
go back to reference Klein N, et al. Angiotensin-(1–7) protects from experimental acute lung injury. Crit Care Med. 2013;41(11):e334-343.CrossRef Klein N, et al. Angiotensin-(1–7) protects from experimental acute lung injury. Crit Care Med. 2013;41(11):e334-343.CrossRef
17.
go back to reference Montezano AC, et al. Angiotensin II and vascular injury. Curr Hypertens Rep. 2014;16(6):431.CrossRef Montezano AC, et al. Angiotensin II and vascular injury. Curr Hypertens Rep. 2014;16(6):431.CrossRef
18.
go back to reference Nguyen Dinh Cat A, et al. Angiotensin II, NADPH oxidase, and redox signaling in the vasculature. Antioxid Redox Signal. 2013;19(10):1110–20.CrossRef Nguyen Dinh Cat A, et al. Angiotensin II, NADPH oxidase, and redox signaling in the vasculature. Antioxid Redox Signal. 2013;19(10):1110–20.CrossRef
19.
go back to reference 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):131–6.CrossRef 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):131–6.CrossRef
20.
go back to reference Honda Y, et al. Midkine deteriorates cardiac remodeling via epidermal growth factor receptor signaling in chronic kidney disease. Hypertension. 2016;67(5):857–65.CrossRef Honda Y, et al. Midkine deteriorates cardiac remodeling via epidermal growth factor receptor signaling in chronic kidney disease. Hypertension. 2016;67(5):857–65.CrossRef
21.
go back to reference 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):859–69.CrossRef 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):859–69.CrossRef
22.
go back to reference Huang Y, et al. Midkine induces epithelial-mesenchymal transition through Notch2/Jak2-Stat3 signaling in human keratinocytes. Cell Cycle. 2008;7(11):1613–22.CrossRef Huang Y, et al. Midkine induces epithelial-mesenchymal transition through Notch2/Jak2-Stat3 signaling in human keratinocytes. Cell Cycle. 2008;7(11):1613–22.CrossRef
23.
go back to reference Rhodes A, et al. Surviving sepsis campaign: international guidelines for management of sepsis and septic shock: 2016. Intensive Care Med. 2017;43(3):304–77.CrossRef Rhodes A, et al. Surviving sepsis campaign: international guidelines for management of sepsis and septic shock: 2016. Intensive Care Med. 2017;43(3):304–77.CrossRef
24.
go back to reference Rittirsch D, et al. Immunodesign of experimental sepsis by cecal ligation and puncture. Nat Protoc. 2009;4(1):31–6.CrossRef Rittirsch D, et al. Immunodesign of experimental sepsis by cecal ligation and puncture. Nat Protoc. 2009;4(1):31–6.CrossRef
25.
go back to reference Sue RD, et al. CXCR2 is critical to hyperoxia-induced lung injury. J Immunol. 2004;172(6):3860–8.CrossRef Sue RD, et al. CXCR2 is critical to hyperoxia-induced lung injury. J Immunol. 2004;172(6):3860–8.CrossRef
26.
go back to reference Zhang H, et al. IL-6 trans-signaling promotes pancreatitis-associated lung injury and lethality. J Clin Invest. 2013;123(3):1019–31.CrossRef Zhang H, et al. IL-6 trans-signaling promotes pancreatitis-associated lung injury and lethality. J Clin Invest. 2013;123(3):1019–31.CrossRef
27.
go back to reference 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):719–27.CrossRef 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):719–27.CrossRef
28.
go back to reference 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.CrossRef 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.CrossRef
29.
go back to reference Weckbach LT, et al. Midkine acts as proangiogenic cytokine in hypoxia-induced angiogenesis. Am J Physiol Heart Circ Physiol. 2012;303(4):H429-438.CrossRef Weckbach LT, et al. Midkine acts as proangiogenic cytokine in hypoxia-induced angiogenesis. Am J Physiol Heart Circ Physiol. 2012;303(4):H429-438.CrossRef
30.
go back to reference 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;762:464–71.CrossRef 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;762:464–71.CrossRef
31.
go back to reference 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):1781–7.CrossRef 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):1781–7.CrossRef
32.
go back to reference Misa K, et al. Involvement of midkine in the development of pulmonary fibrosis. Physiol Rep. 2017;5(16):e13383.CrossRef Misa K, et al. Involvement of midkine in the development of pulmonary fibrosis. Physiol Rep. 2017;5(16):e13383.CrossRef
33.
go back to reference 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):315–23.CrossRef 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):315–23.CrossRef
34.
go back to reference Shindo E, et al. The growth factor midkine may play a pathophysiological role in rheumatoid arthritis. Mod Rheumatol. 2017;27(1):54–9.CrossRef Shindo E, et al. The growth factor midkine may play a pathophysiological role in rheumatoid arthritis. Mod Rheumatol. 2017;27(1):54–9.CrossRef
35.
go back to reference Paul M, Poyan Mehr A, Kreutz R. Physiology of local renin-angiotensin systems. Physiol Rev. 2006;86(3):747–803.CrossRef Paul M, Poyan Mehr A, Kreutz R. Physiology of local renin-angiotensin systems. Physiol Rev. 2006;86(3):747–803.CrossRef
36.
go back to reference 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):525–33.CrossRef 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):525–33.CrossRef
37.
go back to reference 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):5009–19.CrossRef 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):5009–19.CrossRef
38.
go back to reference 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):740–51.CrossRef 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):740–51.CrossRef
Metadata
Title
Pulmonary midkine inhibition ameliorates sepsis induced lung injury
Authors
Jing-Yuan Xu
Wei Chang
Qin Sun
Fei Peng
Yi Yang
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Journal of Translational Medicine / Issue 1/2021
Electronic ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-021-02755-z

Other articles of this Issue 1/2021

Journal of Translational Medicine 1/2021 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.