Thromb Haemost 2012; 108(05): 981-991
DOI: 10.1160/TH12-03-0196
Animal Models
Schattauer GmbH

Infrared fluorescence for vascular barrier breach in vivo – A novel method for quantitation of albumin efflux

Annette von Drygalski
1   University of California San Diego, Department of Medicine, San Diego, California, USA
2   The Scripps Research Institute, Department of Molecular and Experimental Medicine, La Jolla, California, USA
,
Christian Furlan-Freguia
3   The Scripps Research Institute, Department of Immunology and Microbial Science, La Jolla, California, USA
,
Laurent O. Mosnier
2   The Scripps Research Institute, Department of Molecular and Experimental Medicine, La Jolla, California, USA
,
Subramanian Yegneswaran
2   The Scripps Research Institute, Department of Molecular and Experimental Medicine, La Jolla, California, USA
,
Wolfram Ruf
3   The Scripps Research Institute, Department of Immunology and Microbial Science, La Jolla, California, USA
,
John H. Griffin
1   University of California San Diego, Department of Medicine, San Diego, California, USA
2   The Scripps Research Institute, Department of Molecular and Experimental Medicine, La Jolla, California, USA
› Author Affiliations
Further Information

Publication History

Received: 26 March 2012

Accepted after major revision: 28 August 2012

Publication Date:
29 November 2017 (online)

Summary

Vascular hyperpermeability contributes to morbidity in inflammation. Current methodologies for in vivo assessment of permeability based on extravasation of Evans Blue (EB)-bound albumin are cumbersome and often lack sensitivity. We developed a novel infrared fluorescence (IRF) methodology for measurement of EB-albumin extravasation to quantify vascular permeability in murine models. Vascular permeability induced by endotoxaemia was examined for all solid organs, brain, skin and peritoneum by IRF and the traditional absorbance-based measurement of EB in tissue extracts. Organ IRF increased linearly with increasing concentrations of intravenous EB (2.5–25 mg/kg). Tissue IRF was more sensitive for EB accumulation compared to the absorbance-based method. Accordingly, differences in vascular permeability and organ EB accumulation between lipopolysaccharide-treated and saline-treated mice were often significant when analysed by IRF-based detection but not by absorbance-based detection. EB was detected in all 353 organs analysed with IRF but only in 67% (239/353) of organs analysed by absorbance-based methodology, demonstrating improved sensitivity of EB detection in organs with IRF. In contrast, EB in plasma after EB administration was readily measured by both methods with high correlation between the two methods (n=116, r2=0.86). Quantitation of organ-specific EB-IRF differences due to endotoxin was optimal when IRF was compared between mice matched for weight, gender, and age, and with appropriate corrections for organ weight and EB plasma concentrations. Notably, EB-IRF methodology leaves organs intact for subsequent histopathology. In summary, EB-IRF is a novel, highly sensitive, rapid, and convenient method for the relative quantification of EB in intact organs of treatment versus control mice.

 
  • References

  • 1 Goldenberg NM, Steinberg BE, Slutsky AS. et al. Broken barriers: a new take on sepsis pathogenesis. Sci Transl Med. 2011 3. 88ps25.
  • 2 Lee WL, Slutsky AS. Sepsis and endothelial permeability. N Engl J Med 2010; 363: 689-691.
  • 3 Russel JA. The current management of septic shock. Minerva Med 2008; 99: 431-458.
  • 4 Caster WO, Simon AB, Armstrong WD. An Evans blue method for the determination of plasma volume in the soft tissues of the rat. J Appl Physiol 1954; 6: 724-726.
  • 5 Caster WO, Simon AB, Armstrong WD. Evans blue space in tissues of the rat. Am J Physiol 1955; 183: 317-321.
  • 6 Caster WO, Garamella JJ, Veleso A. et al. Effect of acute coronary occlusion upon the movement of Evans blue dye into different areas of pig heart muscle. Am Heart J 1958; 56: 658-661.
  • 7 Darrow AL, Fung-Leung WP, Ye RD. et al. Biological consequences of thrombin receptor deficiency in mice. Thromb Haemost 1996; 76: 860-866.
  • 8 Castellino FJ, Liang Z, Volkir SP. et al. Mice with a severe deficiency of the endothelial protein C receptor gene develop, survive, and reproduce normally, and do not present with enhanced arterial thrombosis after challenge. Thromb Haemost 2002; 88: 462-472.
  • 9 Xu Z, Castellino FJ, Ploplis VA. Plasminogen activator inhibitor-1 (PAI-1) is cardioprotective in mice by maintaining microvascular integrity and cardiac architecture. Blood 2010; 115: 2038-2047.
  • 10 Linderkamp O, Mader T, Butenandt O. et al. Plasma volume estimation in severely ill infants and children using a simplified Evans blue method. Eur J Pediatr 1977; 125: 135-141.
  • 11 Finigan JH, Boueiz A, Wilkinson E. et al. Activated protein C protects against ventilator-induced pulmonary capillary leak. Am J Physiol Lung Cell Mol Physiol 2009; 296: L1002-L1011.
  • 12 Ryan AJ, McCoy DM, McGowan SE. et al. Alveolar sphingolipids generated in response to TNF-alpha modifies surfactant biophysical activity. J Appl Physiol 2003; 94: 253-258.
  • 13 Verbrugge SJ, Vazquez de AG, Gommers D. et al. Exogenous surfactant preserves lung function and reduces alveolar Evans blue dye influx in a rat model of ventilation-induced lung injury. Anesthesiology 1998; 89: 467-474.
  • 14 Zhuang J, Xu J, Zhang C. et al. IL-1beta acutely increases pulmonary SP and permeability without associated changes in airway resistance and ventilation in anesthetized rats. Respir Physiol Neurobiol 2011; 175: 12-19.
  • 15 Knezevic N, Tauseef M, Thennes T. et al. The G protein betagamma subunit mediates reannealing of adherens junctions to reverse endothelial permeability increase by thrombin. J Exp Med 2009; 206: 2761-2777.
  • 16 Rice BW, Contag CH. The importance of being red. Nat Biotechnol 2009; 27: 624-625.
  • 17 Kerschen EJ, Fernandez JA, Cooley BC. et al. Endotoxemia and sepsis mortality reduction by non-anticoagulant activated protein C. J Exp Med 2007; 204: 2439-2448.
  • 18 Dallal MM, Chang SW. Evans blue dye in the assessment of permeability-surface are product in perfused rat lungs. J Appl Physiol 1994; 77: 1030-1035.
  • 19 Green TP, Johnson DE, Marchessault RP. et al. Transvascular flux and tissue accrual of Evans blue: effects of endotoxin and histamine. J Lab Clin Med 1988; 111: 173-183.
  • 20 Rogers DF, Boschetto P, Barnes PJ. Plasma exudation Correlation between Evans blue dye and radiolabeled albumin in guinea pig airways in vivo. J Pharmacol Methods 1989; 21: 309-315.
  • 21 Zuccarello M, Anderson DK. Protective effect of a 21-aminosteroid on the blood-brain barrier following subarachnoid hemorrhage in rats. Stroke 1989; 20: 367-371.
  • 22 Liu W, Hendren J, Qin XJ. et al. Normobaric hyperoxia attenuates early blood-brain barrier disruption by inhibiting MMP-9-mediated occludin degradation in focal cerebral ischemia. J Neurochem 2009; 108: 811-820.
  • 23 Muzumdar RH, Huffman DM, Calvert JW. et al. Acute humanin therapy attenuates myocardial ischemia and reperfusion injury in mice. Arterioscler Thromb Vasc Biol 2010; 30: 1940-1948.