scholarly journals Genetic and pharmacologic inhibition of Tpl2 kinase is protective in a mouse model of ventilator-induced lung injury

2014 ◽  
Vol 2 (1) ◽  
pp. 15 ◽  
Author(s):  
Evangelos Kaniaris ◽  
Katerina Vaporidi ◽  
Eleni Vergadi ◽  
Emmanuel E Theodorakis ◽  
Eumorfia Kondili ◽  
...  
Shock ◽  
2012 ◽  
Vol 38 (4) ◽  
pp. 375-380 ◽  
Author(s):  
Arie J. Hoogendijk ◽  
Maria T. Kuipers ◽  
Tom van der Poll ◽  
Marcus J. Schultz ◽  
Catharina W. Wieland

2008 ◽  
Vol 108 (2) ◽  
pp. 261-268 ◽  
Author(s):  
Rosanna Vaschetto ◽  
Jan W. Kuiper ◽  
Shyh Ren Chiang ◽  
Jack J. Haitsma ◽  
Jonathan W. Juco ◽  
...  

Background Mechanical ventilation can induce organ injury associated with overwhelming inflammatory responses. Excessive activation of poly(adenosine diphosphate-ribose) polymerase enzyme after massive DNA damage may aggravate inflammatory responses. Therefore, the authors hypothesized that the pharmacologic inhibition of poly(adenosine diphosphate-ribose) polymerase by PJ-34 would attenuate ventilator-induced lung injury. Methods Anesthetized rats were subjected to intratracheal instillation of lipopolysaccharide at a dose of 6 mg/kg. The animals were then randomly assigned to receive mechanical ventilation at either low tidal volume (6 ml/kg) with 5 cm H2O positive end-expiratory pressure or high tidal volume (15 ml/kg) with zero positive end-expiratory pressure, in the presence and absence of intravenous administration of PJ-34. Results The high-tidal-volume ventilation resulted in an increase in poly(adenosine diphosphate-ribose) polymerase activity in the lung. The treatment with PJ-34 maintained a greater oxygenation and a lower airway plateau pressure than the vehicle control group. This was associated with a decreased level of interleukin 6, active plasminogen activator inhibitor 1 in the lung, attenuated leukocyte lung transmigration, and reduced pulmonary edema and apoptosis. The administration of PJ-34 also decreased the systemic levels of tumor necrosis factor alpha and interleukin 6, and attenuated the degree of apoptosis in the kidney. Conclusion The pharmacologic inhibition of poly(adenosine diphosphate-ribose) polymerase reduces ventilator-induced lung injury and protects kidney function.


Author(s):  
Adrian S. Seah ◽  
Kara A. Grant ◽  
Gilman B. Allen ◽  
Minara Aliyeva ◽  
Jason H.T. Bates

2016 ◽  
Vol 123 (1) ◽  
pp. 143-151 ◽  
Author(s):  
Karl Michael Strosing ◽  
Simone Faller ◽  
Veronica Gyllenram ◽  
Helen Engelstaedter ◽  
Hartmut Buerkle ◽  
...  

2011 ◽  
Vol 301 (2) ◽  
pp. L161-L170 ◽  
Author(s):  
Otgonchimeg Rentsendorj ◽  
Mahendra Damarla ◽  
Neil R. Aggarwal ◽  
Ji-Young Choi ◽  
Laura Johnston ◽  
...  

Phosphodiesterase 2A (PDE2A) is stimulated by cGMP to hydrolyze cAMP, a potent endothelial barrier-protective molecule. We previously found that lung PDE2A contributed to a mouse model of ventilator-induced lung injury (VILI). The purpose of the present study was to determine the contribution of PDE2A in a two-hit mouse model of 1-day intratracheal (IT) LPS followed by 4 h of 20 ml/kg tidal volume ventilation. Compared with IT water controls, LPS alone (3.75 μg/g body wt) increased lung PDE2A mRNA and protein expression by 6 h with a persistent increase in protein through day 4 before decreasing to control levels on days 6 and 10. Similar to the PDE2A time course, the peak in bronchoalveolar lavage (BAL) neutrophils, lactate dehydrogenase (LDH), and protein concentration also occurred on day 4 post-LPS. IT LPS (1 day) and VILI caused a threefold increase in lung PDE2A and inducible nitric oxide synthase (iNOS) and a 24-fold increase in BAL neutrophilia. Compared with a control adenovirus, PDE2A knockdown with an adenovirus expressing a short hairpin RNA administered IT 3 days before LPS/VILI effectively decreased lung PDE2A expression and significantly attenuated BAL neutrophilia, LDH, protein, and chemokine levels. PDE2A knockdown also reduced lung iNOS expression by 53%, increased lung cAMP by nearly twofold, and improved survival from 47 to 100%. We conclude that in a mouse model of LPS/VILI, a synergistic increase in lung PDE2A expression increased lung iNOS and alveolar inflammation and contributed significantly to the ensuing acute lung injury.


2018 ◽  
Vol 19 (1) ◽  
Author(s):  
Junbo Zheng ◽  
Yongbo Huang ◽  
Diana Islam ◽  
Xiao-Yan Wen ◽  
Sulong Wu ◽  
...  

Author(s):  
Joshua Englert ◽  
Po-Shun Lee ◽  
Rebecca M. Baron ◽  
Augustine M.K. Choi

Sign in / Sign up

Export Citation Format

Share Document