Int J Stem Cells.  2014 May;7(1):1-11.

Comparative Study between Intravenous and Intraperitoneal Stem Cell Therapy in Amiodarone Induced Lung Injury in Rat

Affiliations
  • 1Department of Histology, Faculty of Medicine, Cairo University, Cairo, Egypt. mahakaah@yahoo.com
  • 2Department of Clinical Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt.

Abstract

BACKGROUND AND OBJECTIVES
The fibrosing form of lung injury (occupational, environmental, infective or drug induced) is associated with significant morbidity and mortality. Amiodarone (AM), often prescribed for control of arrhythmias is considered a potential cause. No effective treatment was confirmed, except lung transplantation. Intravenous (IV) stem cell therapy may produce pulmonary emboli or infarctions. Despite being commonly used in clinical practice, the intraperitoneal (IP.) route has been rarely used for cell delivery. The present study aimed at investigating and comparing the possible effect of IP stem cell therapy (SCT) on pulmonary toxicity versus the intravenous route in a rat model of amiodarone induced lung damage.
METHODS AND RESULTS
36 adult male albino rats were divided into 4 groups. Rats of AM group were given 30 mg/kg daily orally for 4 weeks. Rats of IV SCT group were injected with stem cells in the tail vein. Rats of IP SCT group received IP cell therapy. Histological, histochemical, immunohistochemical and morphometric studies were performed. Obstructed bronchioles, overdistended alveoli, reduced type I pneumocytes, increased thickness of alveolar septa and vessels wall besides increased area% of collagen fibers regressed in response to IV and IP SCT. The improvement was more obvious in IV group. The area% of Prussion blue +ve and CD105 +ve cells was significantly higher in IV group.
CONCLUSIONS
Cord blood MSC therapy proved definite amelioration of lung injury ending in fibrosis. The effect of IP SCT was slightly inferior to that of IV SCT, which may be overwhelmed by repeated IP injection.

Keyword

Mesenchymal stem cells; Amiodarone; Cord blood; Lung injury

MeSH Terms

Adult
Amiodarone*
Animals
Arrhythmias, Cardiac
Bronchioles
Cell- and Tissue-Based Therapy
Collagen
Fetal Blood
Fibrosis
Humans
Infarction
Lung
Lung Injury*
Lung Transplantation
Male
Mesenchymal Stromal Cells
Models, Animal
Mortality
Pneumocytes
Rats*
Stem Cells*
Veins
Amiodarone
Collagen

Figure

  • Fig. 1. (A) Section in the lung of a control rat showing two bronchioles (b), alveoli (a), alveolar sacs (S) and a vessel (v) (H&E, ×100). (B) Section in the lung of a control rat showing alveoli (a) and alveolar sacs (S) lined by multiple cells exhibiting flat nuclei (arrows). Note alveolar septa (I) (H&E, ×400).

  • Fig. 2. (A) Section in the lung of a rat in amiodarone group showing a small bronchiole with partial obliteration of the lumen by shed epithelial cells (*), thickened wall of two vessels (v), cellular aggregates in alveolar septa and cellular infiltration in the adventitia of bronhiole (arrowheads) (H&E, ×100). (B) Section in the lung of a rat in amiodarone group showing thickening of the alveolar septa exhibiting dense cellular infiltration (arrowheads), irregular alveoli (a) and alveolar spaces (S) lined by few cells exhibiting flat nuclei (arrows). Note multiple extravasated RBCs (*) (H&E, ×100). (C) Section in the lung of a rat in amiodarone group showing over-distended alveoli (a) with disrupted alveolar walls (*). Note cellular infiltrates in the alveolar septa (arrowheads) (H&E, ×400).

  • Fig. 3. (A) Section in the lung of a rat in IVI stem cell therapy group showing two bronchioles (b), one of them demonstrates cellular infiltrate in the adventitia (arrowhead) and a congested vessel (c) (H&E, ×100). (B) Section in the lung of a rat in IVI stem cell therapy group showing a bronchiole (b), a vessel with partially thickened wall (v), congested vessels (c) and extravasated RBCs (*) (H&E, ×100). (C) Section in the lung of a rat in IVI stem cell therapy group showing normal alveoli (a) and alveolar sacs (S) lined by multiple cells exhibiting flat nuclei (arrows). Note cellular infiltrates (arrowheads) and extravasated RBCs in few alveolar septa (*) (H&E, ×400).

  • Fig. 4. (A) Section in the lung of a rat in IPI stem cell therapy group showing three normal bronchioles (b) surrounded by infiltrating cells (arrowheads) and two vessels (v) with partial thickening of their walls. Note some congested vessels (c) (H&E, ×100). (B) Section in the lung of a rat in IPI stem cell therapy group showing a normal bronchiole (b) surrounded by infiltrating cells (arrowheads) and two vessels (v) with marked thickening of their walls (v) (H&E, ×100). (C) Section in the lung of a rat in IPI stem cell therapy group showing normal alveoli (a) and alveolar sacs (S) lined by multiple cells exhibiting flat nuclei (arrows). Note cellular infiltrates (arrowheads) and extravasated RBCs in multiple alveolar septa (*) (H&E, ×400).

  • Fig. 5. (A) Section in the lung of a control rat showing fine collagen fibers (arrowheads) in the alveolar septa, denser collagen fibers in the wall of a bronchiole (arrows) and in the adventitia (*) of a blood vessel (Masson’s trichrome, ×400). (B) Section in the lung of a rat in amiodarone group showing dense collagen fibers (arrows) and infiltrating cells (arrowheads) in the alveolar septa (Masson’s trichrome, ×400). (C) Section in the lung of a rat in IVI stem cell therapy group showing dense collagen fibers (*) in a septum and infiltrating cells (arrowhead) in another septum around normal alveoli (a) and alveolar sacs (S) (Masson’s trichrome, ×400). (D) Section in the lung of a rat in IPI stem cell therapy group showing dense collagen fibers (arrows) in some septa and infiltrating cells (arrowheads) in other septa (Masson’s trichrome, ×400).

  • Fig. 6. (A) Section in the lung of a control rat showing negative staining in the alveoli (a), alveolar sacs (S) and alveolar septa (I) (Prussian blue, ×400). (B) Section in the lung of a rat in IVI stem cell therapy group showing multiple Prussian blue positive cells (arrows) inside and around blood vessels (v), in the alveolar septa (I) and among the epithelial lining of a bronchiole (b) (Prussian blue, ×400). (C) Section in the lung of a rat in IPI stem cell therapy group showing some Prussian blue positive cells (arrows) inside blood vessels (v), in the alveolar septa (I) and in the adventitia of a bronchiole (b) (Prussian blue, ×400).

  • Fig. 7. (A) Section in the lung of a control rat showing negative immunostaining in the alveoli (a), alveolar sacs (S) and alveolar septa (I) (CD105 immunostaining, ×400). (B) Section in the lung of a rat in IVI stem cell therapy group showing multiple CD105 positive cells (arrows) in the alveolar septa and among the lining epithelium of the alveoli and alveolar sacs (CD105 immunostaining, ×400). (C) Section in the lung of a rat in IPI stem cell therapy group showing some CD105 positive cells (arrows) in the alveolar septa and among the lining epithelium of the alveoli and alveolar sacs (CD105 immunostaining, ×400).


Reference

References

1. Kinder BW, Shariat C, Collard HR, Koth LL, Wolters PJ, Golden JA, Panos RJ, King TE Jr. Undifferentiated connective tissue disease-associated interstitial lung disease: changes in lung function. Lung. 2010. 188:143–149.
Article
2. Vanfleteren LE, Linssen CF. Role of microorganisms in interstitial lung disease. Curr Opin Pulm Med. 2010. 16:489–495.
Article
3. Van Cott TE, Yehle KS, DeCrane SK, Thorlton JR. Amiodarone-induced pulmonary toxicity: case study with syndrome analysis. Heart Lung. 2013. 42:262–266.
Article
4. O’Brien K, Troendle J, Gochuico BR, Markello TC, Salas J, Cardona H, Yao J, Bernardini I, Hess R, Gahl WA. Pirfenidone for the treatment of Hermansky-Pudlak syndrome pulmonary fibrosis. Mol Genet Metab. 2011. 103:128–134.
Article
5. Range FT, Hilker E, Breithardt G, Buerke B, Lebiedz P. Amiodarone-induced pulmonary toxicity--a fatal case report and literature review. Cardiovasc Drugs Ther. 2013. 27:247–254.
Article
6. Ding DC, Shyu WC, Lin SZ. Mesenchymal Stem Cells. Cell Transplant. 2011. 20:5–14.
Article
7. Bashir Q, Robinson SN, de Lima MJ, Parmar S, Shpall E. Umbilical cord blood transplantation. Clin Adv Hematol Oncol. 2010. 8:786–801.
8. Maurer MH. Proteomic definitions of mesenchymal stem cells. Stem Cells Int. 2011. 2011:704256–704264.
Article
9. Prockop DJ, Olson SD. Clinical trials with adult stem/progenitor cells for tissue repair: let’s not overlook some essential precautions. Blood. 2007. 109:3147–3151.
Article
10. van Laar JM, Sullivan K. Stem cell transplantation in systemic sclerosis. Curr Opin Rheumatol. 2013. 25:719–725.
Article
11. Park M, Lee SH, Lee YH, Yoo KH, Sung KW, Koo HH, Kang HJ, Park KD, Shin HY, Ahn HS, Chung NG, Cho B, Kim HK, Koh KN, Im HJ, Seo JJ, Han DK, Baek HJ, Kook H, Hwang TJ, Lee EK, Hah JO, Lim YJ, Jung HJ, Park JE, Jang MJ, Chong SY, Oh D; Korean Cord Blood Transplantation Working Party. Pre-engraftment syndrome after unrelated cord blood transplantation: a predictor of engraftment and acute graft-versus-host disease. Biol Blood Marrow Transplant. 2013. 19:640–646.
Article
12. Ghionzoli M, Cananzi M, Zani A, Rossi CA, Leon FF, Pierro A, Eaton S, De Coppi P. Amniotic fluid stem cell migration after intraperitoneal injection in pup rats: implication for therapy. Pediatr Surg Int. 2010. 26:79–84.
Article
13. Kolettis TM, Agelaki MG, Baltogiannis GG, Vlahos AP, Mourouzis I, Fotopoulos A, Pantos C. Comparative effects of acute vs. chronic oral amiodarone treatment during acute myocardial infarction in rats. Europace. 2007. 9:1099–1104.
Article
14. Osman FRO, Aboul Fotouh GI, Zickri MB, El Deeb D. Histological and ultrastructural study on the effect of some antiarrhythmic drugs on the thyroid gland of albino rat. MD Thesis. 2004.
15. Lee YG, Hwang JW, Park SB, Shin IS, Kang SK, Seo KW, Lee YS, Kang KS. Reduction of liver fibrosis by xenogeneic human umbilical cord blood and adipose tissue-derived multi-potent stem cells without treatment of an immunosuppressant. TERM. 2008. 5:613–621.
16. Castelo-Branco MT, Soares ID, Lopes DV, Buongusto F, Martinusso CA, do Rosario A Jr, Souza SA, Gutfilen B, Fonseca LM, Elia C, Madi K, Schanaider A, Rossi MI, Souza HS. Intraperitoneal but not intravenous cryopreserved mesenchymal stromal cells home to the inflamed colon and ameliorate experimental colitis. PLoS One. 2012. 7:e33360.
Article
17. Stocum DL, Zupanc GK. Stretching the limits: stem cells in regeneration science. Dev Dyn. 2008. 237:3648–3671.
Article
18. Dua HS, Joseph A, Shanmuganathan VA, Jones RE. Stem cell differentiation and the effects of deficiency. Eye (Lond). 2003. 17:877–885.
Article
19. Koch TG, Heerkens T, Thomsen PD, Betts DH. Isolation of mesenchymal stem cells from equine umbilical cord blood. BMC Biotechnol. 2007. 7:26.
Article
20. Kraitchman DL, Heldman AW, Atalar E, Amado LC, Martin BJ, Pittenger MF, Hare JM, Bulte JW. In vivo magnetic resonance imaging of mesenchymal stem cells in myocardial infarction. Circulation. 2003. 107:2290–2293.
Article
21. Haasters F, Prall WC, Anz D, Bourquin C, Pautke C, Endres S, Mutschler W, Docheva D, Schieker M. Morphological and immunocytochemical characteristics indicate the yield of early progenitors and represent a quality control for human mesenchymal stem cell culturing. J Anat. 2009. 214:759–767.
Article
22. Kiernan JA. Histological and histochemical methods: theory and practice. 3rd ed. London, New York, New Delhi: Arnold publisher;2001. 111–162.
23. Bancroft JD, Gamble M. Connective tissue stains. Gamble M, Bancroft JD, editors. Theory and practice of histological techniques. 6th ed. Edinburgh, London, Oxford, New York, Philadelphia, St Louis, Sydney and Toronto: Elsevier Health Sciences, Churchill Livingstone;2008. 150.
24. Ellis R. Perls Prussian blue Staining Protocol. South Australia: Pathology Division, Queen Elizabeth Hospital;2007.
25. Yagi H, Soto-Gutierrez A, Navarro-Alvarez N, Nahmias Y, Goldwasser Y, Kitagawa Y, Tilles AW, Tompkins RG, Parekkadan B, Yarmush ML. Reactive bone marrow stromal cells attenuate systemic inflammation via sTNFR1. Mol Ther. 2010. 18:1857–1864.
Article
26. Emsley R, Dunn G, White IR. Mediation and moderation of treatment effects in randomised controlled trials of complex interventions. Stat Methods Med Res. 2010. 19:237–270.
Article
27. Larsen BT, Vaszar LT, Colby TV, Tazelaar HD. Lymphoid hyperplasia and eosinophilic pneumonia as histologic manifestations of amiodarone-induced lung toxicity. Am J Surg Pathol. 2012. 36:509–516.
Article
28. Sood BG, Wykes S, Landa M, De Jesus L, Rabah R. Expression of eNOS in the lungs of neonates with pulmonary hypertension. Exp Mol Pathol. 2011. 90:9–12.
Article
29. Borders CW 3rd, Bennett S, Mount C, Claassen SL. A rare case of acute diffuse alveolar hemorrhage following initiation of amiodarone: a case report. Mil Med. 2012. 177:118–120.
Article
30. Gumuser G, Vural K, Varol T, Parlak Y, Tuglu I, Topal G, Sayit E. Assessment of lung toxicity caused by bleomycin and amiodarone by Tc-99m HMPAO lung scintigraphy in rats. Ann Nucl Med. 2013. 27:592–599.
Article
31. Roth FC, Mulder JE, Brien JF, Takahashi T, Massey TE. Cytotoxic interaction between amiodarone and desethylamiodarone in human peripheral lung epithelial cells. Chem Biol Interact. 2013. 204:135–139.
Article
32. Sato N, Kojima K, Horio Y, Goto E, Masunaga A, Ichiyasu H, Kohrogi H. Successful treatment of severe amiodarone pulmonary toxicity with polymyxin B-immobilized fiber column direct hemoperfusion. Chest. 2013. 143:1146–1150.
Article
33. Cheng K, Rai P, Plagov A, Lan X, Kumar D, Salhan D, Rehman S, Malhotra A, Bhargava K, Palestro CJ, Gupta S, Singhal PC. Transplantation of bone marrow-derived MSCs improves cisplatinum-induced renal injury through paracrine mechanisms. Exp Mol Pathol. 2013. 94:466–473.
Article
34. Hao H, Liu J, Shen J, Zhao Y, Liu H, Hou Q, Tong C, Ti D, Dong L, Cheng Y, Mu Y, Liu J, Fu X, Han W. Multiple intravenous infusions of bone marrow mesenchymal stem cells reverse hyperglycemia in experimental type 2 diabetes rats. Biochem Biophys Res Commun. 2013. 436:418–423.
Article
35. Gordon D, Pavlovska G, Glover CP, Uney JB, Wraith D, Scolding NJ. Human mesenchymal stem cells abrogate experimental allergic encephalomyelitis after intraperitoneal injection, and with sparse CNS infiltration. Neurosci Lett. 2008. 448:71–73.
Article
36. Roddy GW, Oh JY, Lee RH, Bartosh TJ, Ylostalo J, Coble K, Rosa RH Jr, Prockop DJ. Action at a distance: systemically administered adult stem/progenitor cells (MSCs) reduce inflammatory damage to the cornea without engraftment and primarily by secretion of TNF-α stimulated gene/protein 6. Stem Cells. 2011. 29:1572–1579.
Article
37. Yousefi F, Ebtekar M, Soleimani M, Soudi S, Hashemi SM. Comparison of in vivo immunomodulatory effects of intravenous and intraperitoneal administration of adipose-tissue mesenchymal stem cells in experimental autoimmune encephalomyelitis (EAE). Int Immunopharmacol. 2013. 17:608–616.
Article
Full Text Links
  • IJSC
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr