Skip to main content
Top
Published in: Molecular Imaging and Biology 4/2020

Open Access 01-08-2020 | Research Article

Multimodal Imaging Techniques Show Differences in Homing Capacity Between Mesenchymal Stromal Cells and Macrophages in Mouse Renal Injury Models

Authors: Arthur Taylor, Jack Sharkey, Rachel Harwood, Lauren Scarfe, Michael Barrow, Matthew J. Rosseinsky, Dave J. Adams, Bettina Wilm, Patricia Murray

Published in: Molecular Imaging and Biology | Issue 4/2020

Login to get access

Abstract

Purpose

The question of whether mesenchymal stromal cells (MSCs) home to injured kidneys remains a contested issue. To try and understand the basis for contradictory findings reported in the literature, our purpose here was to investigate whether MSC homing capacity is influenced by administration route, the type of injury model used, and/or the presence of exogenous macrophages.

Procedures

To assess the viability, whole-body biodistribution, and intra-renal biodistribution of MSCs, we used a multimodal imaging strategy comprising bioluminescence and magnetic resonance imaging. The effect of administration route (venous or arterial) on the ability of MSCs to home to injured renal tissue, and persist there, was assessed in a glomerular injury model (induced by the nephrotoxicant, Adriamycin) and a tubular injury model induced by ischaemia-reperfusion injury (IRI). Exogenous macrophages were used as a positive control because these cells are known to home to injured mouse kidneys. To assess whether the homing capacity of MSCs can be influenced by the presence of exogenous macrophages, we used a dual-bioluminescence strategy that allowed the whole-body biodistribution of the two cell types to be monitored simultaneously in individual animals.

Results

Following intravenous administration, no MSCs were detected in the kidneys, irrespective of whether the mice had been subjected to renal injury. After arterial administration via the left cardiac ventricle, MSCs transiently populated the kidneys, but no preferential homing or persistence was observed in injured renal tissue after unilateral IRI. An exception was when MSCs were co-administered with exogenous macrophages; here, we observed some homing of MSCs to the injured kidney.

Conclusions

Our findings strongly suggest that MSCs do not home to injured kidneys.
Appendix
Available only for authorised users
Literature
1.
go back to reference Torres Crigna A, Daniele C, Gamez C et al (2018) Stem/stromal cells for treatment of kidney injuries with focus on preclinical models. Front Med 5 Torres Crigna A, Daniele C, Gamez C et al (2018) Stem/stromal cells for treatment of kidney injuries with focus on preclinical models. Front Med 5
2.
go back to reference Burks SR, Nguyen BA, Tebebi PA, Kim SJ, Bresler MN, Ziadloo A, Street JM, Yuen PS, Star RA, Frank JA (2015) Pulsed focused ultrasound pretreatment improves mesenchymal stromal cell efficacy in preventing and rescuing established acute kidney injury in mice. Stem Cells 33:1241–1253CrossRef Burks SR, Nguyen BA, Tebebi PA, Kim SJ, Bresler MN, Ziadloo A, Street JM, Yuen PS, Star RA, Frank JA (2015) Pulsed focused ultrasound pretreatment improves mesenchymal stromal cell efficacy in preventing and rescuing established acute kidney injury in mice. Stem Cells 33:1241–1253CrossRef
3.
go back to reference Morigi M, Imberti B, Zoja C, Corna D, Tomasoni S, Abbate M, Rottoli D, Angioletti S, Benigni A, Perico N, Alison M, Remuzzi G (2004) Mesenchymal stem cells are renotropic, helping to repair the kidney and improve function in acute renal failure. J Am Soc Nephrol 15:1794–1804CrossRef Morigi M, Imberti B, Zoja C, Corna D, Tomasoni S, Abbate M, Rottoli D, Angioletti S, Benigni A, Perico N, Alison M, Remuzzi G (2004) Mesenchymal stem cells are renotropic, helping to repair the kidney and improve function in acute renal failure. J Am Soc Nephrol 15:1794–1804CrossRef
4.
go back to reference Bi B, Schmitt R, Israilova M, Nishio H, Cantley LG (2007) Stromal cells protect against acute tubular injury via an endocrine effect. J Am Soc Nephrol 18:2486–2496CrossRef Bi B, Schmitt R, Israilova M, Nishio H, Cantley LG (2007) Stromal cells protect against acute tubular injury via an endocrine effect. J Am Soc Nephrol 18:2486–2496CrossRef
5.
go back to reference Geng Y, Zhang L, Fu B et al (2014) Mesenchymal stem cells ameliorate rhabdomyolysis-induced acute kidney injury via the activation of M2 macrophages. Stem Cell Res Ther 5:80–80CrossRef Geng Y, Zhang L, Fu B et al (2014) Mesenchymal stem cells ameliorate rhabdomyolysis-induced acute kidney injury via the activation of M2 macrophages. Stem Cell Res Ther 5:80–80CrossRef
6.
go back to reference Schrepfer S, Deuse T, Reichenspurner H et al (2007) Stem cell transplantation: the lung barrier. Transplantation P 39:573–576CrossRef Schrepfer S, Deuse T, Reichenspurner H et al (2007) Stem cell transplantation: the lung barrier. Transplantation P 39:573–576CrossRef
7.
go back to reference Eggenhofer E, Benseler V, Kroemer A et al (2012) Mesenchymal stem cells are short-lived and do not migrate beyond the lungs after intravenous infusion. Front Immunol 3:297–297CrossRef Eggenhofer E, Benseler V, Kroemer A et al (2012) Mesenchymal stem cells are short-lived and do not migrate beyond the lungs after intravenous infusion. Front Immunol 3:297–297CrossRef
8.
go back to reference Fischer UM, Harting MT, Jimenez F, Monzon-Posadas WO, Xue H, Savitz SI, Laine GA, Cox CS Jr (2009) Pulmonary passage is a major obstacle for intravenous stem cell delivery: the pulmonary first-pass effect. Stem Cells Dev 18:683–692CrossRef Fischer UM, Harting MT, Jimenez F, Monzon-Posadas WO, Xue H, Savitz SI, Laine GA, Cox CS Jr (2009) Pulmonary passage is a major obstacle for intravenous stem cell delivery: the pulmonary first-pass effect. Stem Cells Dev 18:683–692CrossRef
9.
go back to reference Scarfe L, Taylor A, Sharkey J et al (2018) Non-invasive imaging reveals conditions that impact distribution and persistence of cells after in vivo administration. Stem Cell Res Ther 9:332CrossRef Scarfe L, Taylor A, Sharkey J et al (2018) Non-invasive imaging reveals conditions that impact distribution and persistence of cells after in vivo administration. Stem Cell Res Ther 9:332CrossRef
10.
go back to reference Wang Y, He J, Pei X, Zhao W (2013) Systematic review and meta-analysis of mesenchymal stem/stromal cells therapy for impaired renal function in small animal models. Nephrology 18:201–208CrossRef Wang Y, He J, Pei X, Zhao W (2013) Systematic review and meta-analysis of mesenchymal stem/stromal cells therapy for impaired renal function in small animal models. Nephrology 18:201–208CrossRef
11.
go back to reference Zhuo W, Liao L, Fu Y et al (2013) Efficiency of endovenous versus arterial administration of mesenchymal stem cells for ischemia-reperfusion–induced renal dysfunction in rats. Transplantation P 45:503–510CrossRef Zhuo W, Liao L, Fu Y et al (2013) Efficiency of endovenous versus arterial administration of mesenchymal stem cells for ischemia-reperfusion–induced renal dysfunction in rats. Transplantation P 45:503–510CrossRef
12.
go back to reference Tögel F, Yang Y, Zhang P et al (2008) Bioluminescence imaging to monitor the in vivo distribution of administered mesenchymal stem cells in acute kidney injury. Am J Physiol-Renal 295:F315–F321CrossRef Tögel F, Yang Y, Zhang P et al (2008) Bioluminescence imaging to monitor the in vivo distribution of administered mesenchymal stem cells in acute kidney injury. Am J Physiol-Renal 295:F315–F321CrossRef
13.
go back to reference Sun Y, Yu H, Zheng D, Cao Q, Wang Y, Harris D, Wang Y (2011) Sudan Black B reduces autofluorescence in murine renal tissue. Arch Pathol Lab Med 135:1335–1342CrossRef Sun Y, Yu H, Zheng D, Cao Q, Wang Y, Harris D, Wang Y (2011) Sudan Black B reduces autofluorescence in murine renal tissue. Arch Pathol Lab Med 135:1335–1342CrossRef
14.
go back to reference Scarfe L, Brillant N, Kumar JD, et al. (2017) Preclinical imaging methods for assessing the safety and efficacy of regenerative medicine therapies. npj Regen Med 2:28 Scarfe L, Brillant N, Kumar JD, et al. (2017) Preclinical imaging methods for assessing the safety and efficacy of regenerative medicine therapies. npj Regen Med 2:28
15.
go back to reference Guiteras R, Sola A, Flaquer M, Hotter G, Torras J, Grinyó JM, Cruzado JM (2017) Macrophage overexpressing NGAL ameliorated kidney fibrosis in the UUO mice model. Cell Physiol Biochem 42:1945–1960CrossRef Guiteras R, Sola A, Flaquer M, Hotter G, Torras J, Grinyó JM, Cruzado JM (2017) Macrophage overexpressing NGAL ameliorated kidney fibrosis in the UUO mice model. Cell Physiol Biochem 42:1945–1960CrossRef
16.
go back to reference Wang Y, Wang YP, Zheng G, Lee VW, Ouyang L, Chang DH, Mahajan D, Coombs J, Wang YM, Alexander SI, Harris DC (2007) Ex vivo programmed macrophages ameliorate experimental chronic inflammatory renal disease. Kidney Int 72:290–299CrossRef Wang Y, Wang YP, Zheng G, Lee VW, Ouyang L, Chang DH, Mahajan D, Coombs J, Wang YM, Alexander SI, Harris DC (2007) Ex vivo programmed macrophages ameliorate experimental chronic inflammatory renal disease. Kidney Int 72:290–299CrossRef
17.
go back to reference Taylor A, Sharkey J, Plagge A et al (2018) Multicolour in vivo bioluminescence imaging using a NanoLuc-based BRET reporter in combination with firefly luciferase. Contrast Media Mol I 2018:10CrossRef Taylor A, Sharkey J, Plagge A et al (2018) Multicolour in vivo bioluminescence imaging using a NanoLuc-based BRET reporter in combination with firefly luciferase. Contrast Media Mol I 2018:10CrossRef
18.
go back to reference Diduch DR, Coe MR, Joyner C et al (1993) Two cell lines from bone marrow that differ in terms of collagen synthesis, osteogenic characteristics, and matrix mineralization. J Bone Joint Surg 75:92–105CrossRef Diduch DR, Coe MR, Joyner C et al (1993) Two cell lines from bone marrow that differ in terms of collagen synthesis, osteogenic characteristics, and matrix mineralization. J Bone Joint Surg 75:92–105CrossRef
19.
go back to reference Raschke WC, Baird S, Ralph P, Nakoinz I (1978) Functional macrophage cell lines transformed by abelson leukemia virus. Cell 15:261–267CrossRef Raschke WC, Baird S, Ralph P, Nakoinz I (1978) Functional macrophage cell lines transformed by abelson leukemia virus. Cell 15:261–267CrossRef
20.
go back to reference Kutner RH, Zhang XY, Reiser J (2009) Production, concentration and titration of pseudotyped HIV-1-based lentiviral vectors. Nat Protoc 4:495–505CrossRef Kutner RH, Zhang XY, Reiser J (2009) Production, concentration and titration of pseudotyped HIV-1-based lentiviral vectors. Nat Protoc 4:495–505CrossRef
21.
go back to reference Barrow M, Taylor A, Nieves DJ et al (2015) Tailoring the surface charge of dextran-based polymer coated SPIONs for modulated stem cell uptake and MRI contrast. Biomater Sci-UK 3:608–616CrossRef Barrow M, Taylor A, Nieves DJ et al (2015) Tailoring the surface charge of dextran-based polymer coated SPIONs for modulated stem cell uptake and MRI contrast. Biomater Sci-UK 3:608–616CrossRef
22.
go back to reference Barrow M, Taylor A, García Carrión J et al (2016) Co-precipitation of DEAE-dextran coated SPIONs: how synthesis conditions affect particle properties, stem cell labelling and MR contrast. Contrast Media Mol I 11:362–370CrossRef Barrow M, Taylor A, García Carrión J et al (2016) Co-precipitation of DEAE-dextran coated SPIONs: how synthesis conditions affect particle properties, stem cell labelling and MR contrast. Contrast Media Mol I 11:362–370CrossRef
23.
go back to reference Thu MS, Bryant LH, Coppola T, Jordan EK, Budde MD, Lewis BK, Chaudhry A, Ren J, Varma NR, Arbab AS, Frank JA (2012) Self-assembling nanocomplexes by combining ferumoxytol, heparin and protamine for cell tracking by magnetic resonance imaging. Nat Med 18:463–467CrossRef Thu MS, Bryant LH, Coppola T, Jordan EK, Budde MD, Lewis BK, Chaudhry A, Ren J, Varma NR, Arbab AS, Frank JA (2012) Self-assembling nanocomplexes by combining ferumoxytol, heparin and protamine for cell tracking by magnetic resonance imaging. Nat Med 18:463–467CrossRef
24.
go back to reference Ariza de Schellenberger A, Poller WC, Stangl V, Landmesser U, Schellenberger E (2018) Macrophage uptake switches on OCT contrast of superparamagnetic nanoparticles for imaging of atherosclerotic plaques. Int J Nanomedicine 13:7905–7913CrossRef Ariza de Schellenberger A, Poller WC, Stangl V, Landmesser U, Schellenberger E (2018) Macrophage uptake switches on OCT contrast of superparamagnetic nanoparticles for imaging of atherosclerotic plaques. Int J Nanomedicine 13:7905–7913CrossRef
25.
go back to reference Scarfe L, Rak-Raszewska A, Geraci S et al (2015) Measures of kidney function by minimally invasive techniques correlate with histological glomerular damage in SCID mice with adriamycin-induced nephropathy. Sci Rep-UK 5:13601CrossRef Scarfe L, Rak-Raszewska A, Geraci S et al (2015) Measures of kidney function by minimally invasive techniques correlate with histological glomerular damage in SCID mice with adriamycin-induced nephropathy. Sci Rep-UK 5:13601CrossRef
26.
go back to reference Hesketh EE, Czopek A, Clay M, et al. (2014) Renal ischaemia reperfusion injury: a mouse model of injury and regeneration. JOVE-J Vis Exp:51816 Hesketh EE, Czopek A, Clay M, et al. (2014) Renal ischaemia reperfusion injury: a mouse model of injury and regeneration. JOVE-J Vis Exp:51816
27.
go back to reference Furuichi K, Kaneko S, Wada T (2009) Chemokine/chemokine receptor-mediated inflammation regulates pathologic changes from acute kidney injury to chronic kidney disease. Clin Exp Nephrol 13:9–14CrossRef Furuichi K, Kaneko S, Wada T (2009) Chemokine/chemokine receptor-mediated inflammation regulates pathologic changes from acute kidney injury to chronic kidney disease. Clin Exp Nephrol 13:9–14CrossRef
28.
go back to reference Morigi M, Rota C, Montemurro T, Montelatici E, Lo Cicero V, Imberti B, Abbate M, Zoja C, Cassis P, Longaretti L, Rebulla P, Introna M, Capelli C, Benigni A, Remuzzi G, Lazzari L (2010) Life-sparing effect of human cord blood-mesenchymal stem cells in experimental acute kidney injury. Stem Cells 28:513–522PubMed Morigi M, Rota C, Montemurro T, Montelatici E, Lo Cicero V, Imberti B, Abbate M, Zoja C, Cassis P, Longaretti L, Rebulla P, Introna M, Capelli C, Benigni A, Remuzzi G, Lazzari L (2010) Life-sparing effect of human cord blood-mesenchymal stem cells in experimental acute kidney injury. Stem Cells 28:513–522PubMed
29.
go back to reference Santeramo I, Herrera Perez Z, Illera A et al (2017) Human kidney-derived cells ameliorate acute kidney injury without engrafting into renal tissue. Stem Cell Transl Med 6:1373–1384CrossRef Santeramo I, Herrera Perez Z, Illera A et al (2017) Human kidney-derived cells ameliorate acute kidney injury without engrafting into renal tissue. Stem Cell Transl Med 6:1373–1384CrossRef
Metadata
Title
Multimodal Imaging Techniques Show Differences in Homing Capacity Between Mesenchymal Stromal Cells and Macrophages in Mouse Renal Injury Models
Authors
Arthur Taylor
Jack Sharkey
Rachel Harwood
Lauren Scarfe
Michael Barrow
Matthew J. Rosseinsky
Dave J. Adams
Bettina Wilm
Patricia Murray
Publication date
01-08-2020
Publisher
Springer International Publishing
Published in
Molecular Imaging and Biology / Issue 4/2020
Print ISSN: 1536-1632
Electronic ISSN: 1860-2002
DOI
https://doi.org/10.1007/s11307-019-01458-8

Other articles of this Issue 4/2020

Molecular Imaging and Biology 4/2020 Go to the issue