Skip to main content

Effect of propionyl-L-carnitine on experimental models of peripheral arteriopathy in the rat

  • Chapter
The Carnitine System

Part of the book series: Developments in Cardiovascular Medicine ((DICM,volume 162))

  • 69 Accesses

Abstract

A common feature of several different vascular pathologies is an imbalance of energy metabolism in vascular and muscular tissues in the area affected, independent of the cause of the disease (vascular obstruction, diabetic status, sickle cell anemia, etc.). The energy production in these tissues is strongly dependent on the oxidation of fatty acids via the Krebs cycle. The crucial role of L-carnitine (L-C) in this process has been recognized for many years and explained by many authors [1–3]. L-C is a fundamental cofactor for fatty acid oxidation within the mitochondria; fatty acids, free or CoA-activated, cannot penetrate the mitochondrial membrane, while carnitine esters are transported through the membrane into the mitochondria by carnitine translocase. A beneficial effect of L-C in patients with peripheral vascular diseases (PVD) has already been shown in different clinical studies, as well as, secondary carnitine deficiency in the skeletal muscles of these subjects [4–6]. During the search for new derivatives with an improved activity in comparison to the original compound, an acyl-derivative of L-C, propionyl-L-carnitine (PLC), has been selected. Biochemical studies have indicated that PLC presents some advantages over L-C, mainly due to its better transport into the cells, related to the presence of the propionyl group [7–9]. PLC can be converted to propionyl-CoA by carnitine acetyltransferase (CAT) and further to methylmalonyl-CoA by mitochondrial propionyl-CoA carboxylase. Whereas methyl malonyl-CoA remains in the matrix for further conversion to succinyl-CoA and subsequently to succinate (thereby entering the Krebs cycle), PLC may move across the mitochondrial inner membrane by a reversible transport mechanism. As a consequence, PLC administration could result in an activation of the Krebs cycle through an “anaplerotic effect” with a subsequent increase of energy supply [10, 11]. This enhancement of energy production should be particularly useful in all those conditions in which a reduced oxygen availability is involved.

“Propionyl-L-carnitine and pentoxifylline both provided protection against vascular damages caused by ergotamine tartrate administration, as evidenced by lower frequencies of epithelial tissue damage, at the site of injection, and a lower degree of ulceration and tissue necrosis. Propionyl-L-carnitine treatment (120mg/kg per os) significantly restored the walking ability of the rats when the treatment started one week after streptozotocin injection, and lasted up to 9 weeks.”

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 39.99
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 54.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Preview

Unable to display preview. Download preview PDF.

Unable to display preview. Download preview PDF.

References

  1. Bremer J. Carnitine — metabolism and function. Physiol Rev 1980; 63: 1420–1480.

    Google Scholar 

  2. Bieber LL. Carnitine. Annu Rev Biochem 1988; 57: 261–283.

    Article  PubMed  CAS  Google Scholar 

  3. Fritz IB, Arrigoni Martelli E. Sites of action of carnitine and its derivatives on the cardiovascular system: interactions with membranes. Trends Pharmacol Sci 1993; 14: 355–360.

    Article  PubMed  CAS  Google Scholar 

  4. Hiatt WR, Narwaz D, Brass EP. Carnitine metabolism during exercise in patients with peripheral vascular diseases. J Appl Physiol 1987; 62: 2383–2387.

    PubMed  CAS  Google Scholar 

  5. Brevetti G, Angelini C, Rosa M et al. Muscle carnitine deficiency in patients with severe peripheral vascular disease. Circulation 1991; 84: 1490–1495.

    Article  PubMed  CAS  Google Scholar 

  6. Brevetti G, Perna S. Metabolic and clinical effects of L-carnitine in peripheral vascular disease. In: Ferrari R, Di Mauro S, Sherwood G, editors. L-Carnitine and its role in medicine: From function to therapy. London: Academic Press, 1992: 359–378.

    Google Scholar 

  7. Siliprandi N, Di Lisa F, Pivetta A, Miotto G, Siliprandi D. Transport and function of L-carnitine and L-propionylcarnitine: relevance to some cardiomyopathies and cardiac ischemia. Z Kardiol 1987; 76(Suppl 5): 34–40.

    PubMed  CAS  Google Scholar 

  8. Paulson DJ, Traxler JS, Schmidt MJ, Noonan JJ, Shug AL. Protection of the ischaemic myocardium by L-propionylcarnitine: effects on the recovery of cardiac output after ischaemia and reperfusion, carnitine transport, and fatty acid oxidation. Cardiovasc Res 1986; 20: 536–541.

    Article  PubMed  CAS  Google Scholar 

  9. Siliprandi N, Di Lisa F, Menabö R. Propionyl-L-carnitine: biochemical significance and possible role in cardiac metabolism. Cardiovasc Drugs Ther 1991; 5(Suppl 1): 11–15.

    Article  PubMed  Google Scholar 

  10. Hülsmann WC. Biochemical profile of propionyl-L-carnitine. Cardiovasc Drugs Ther 1991; 5(Suppl 1): 7–9.

    Article  PubMed  Google Scholar 

  11. Corsico N, Casantini C, Arrigoni Martelli E. Study of the effect of propionyl-Lcarnitine on experimental models of peripheral arteriopathies. Pharm Res 1993; Suppl 27: 79–80.

    Article  CAS  Google Scholar 

  12. Murai T, Muraoka K, Saga K et al. Effect of beraprost sodium on peripheral circulation insufficiency in rats and rabbits. Arzneimittelforsch 1989; 39: 856–859.

    PubMed  CAS  Google Scholar 

  13. Bertelli A, Conte A, Ronca G, Segnini D, Yu G. Protective effect of propionylcarnitine against peroxidative damage to arterial endothelium membranes. Int J Tissue React 1991; 13: 41–43.

    PubMed  CAS  Google Scholar 

  14. Mathien GM, Terjung RL. Influence of training following bilateral stenosis of the femoral artery in rats. Am J Physiol 1986; 250: H1050–H1059.

    PubMed  CAS  Google Scholar 

  15. Ashida S, Ishihara M, Ogawa H, Abiko Y. Protective effect of ticlopidine on experimentally induced peripheral arterial occlusive disease in rats. Thromb Res 1980; 18: 55–67.

    Article  PubMed  CAS  Google Scholar 

  16. Hara H, Shimada H, Kitajima M, Tamao Y. Effect of (±)-2-(dimethylamino)-l-[[o-(m-methoxyphenethyl)phenoxy]methyl] ethyl hydrogen succinate on experimental models of peripheral obstructive disease. Arzneimittelforsch 1991; 41: 616–620.

    PubMed  CAS  Google Scholar 

  17. Corsico N, Nardone A, Lucreziotti MR et al. Effect of propionyl-L-carnitine of peripheral arteriopathy: a functional, histologic, and NMR spectroscopic study. Cardiovasc Drugs Ther 1993; 7: 241–251.

    Article  PubMed  CAS  Google Scholar 

  18. Corsico N, Aureli T, Di Cocco ME, Nardone A, Arrigoni Martelli E, Conti F. Propionyl-L-carnitine restored impaired muscle energy metabolism in an experimental model of peripheral vascular insufficiency. A study by 31P-NMR spectroscopy. FASEB J 1992; 6: A1369 (Abstr).

    Google Scholar 

  19. Capuani G, Aureli T, Miccheli A et al. An investigation of experimental arteriopathy in rat hind limb by “in vivo” 31P-NMR spectroscopy. Effect of propionyl-L-carnitine. Br J Pharmacol. In press.

    Google Scholar 

  20. Miccheli A, Aureli T, Delfini M et al. Study on influence of inactivation enzymes techniques and extraction procedures on cerebral phosphorylate metabolite level by 31P NMR spectroscopy. Cell Mol Biol 1988; 34: 591–603.

    PubMed  CAS  Google Scholar 

  21. Challiss RAJ, Blackledge MJ, Radda GK. Spatially resolved changes in diabetic rat skeletal muscle metabolism in vivo studied by 31P-n.m.r. spectroscopy. Biochem J 1990; 268: 111–115.

    PubMed  CAS  Google Scholar 

  22. Pugliese G, Tilton RG, Williamson JR. Glucose-induced metabolic imbalances in the pathogenesis of diabetic vascular disease. Diabetes 1990; 39: 312–323.

    Article  PubMed  CAS  Google Scholar 

  23. Williamson JR, Tilton RG, Kilo C. The polyol pathway and diabetic vascular complications. In: Conti F, Andreani D, Guerignian JL, Striker LGE, editors. Diabetic complications: epidemiology and pathogenic mechanism. New York: Raven Press, 1991: 45–58.

    Google Scholar 

  24. Corsico N, Cantagallo A, Nardone A, Morabito E, Serafini S, Arrigoni Martelli E. Vascular and cardiac alterations in diabetic rats: prevention by propionyl-L-carnitine. Proc Symp Vascular Complications of Diabetes Mellitus; Boston, 1993: 21 (Abstr).

    Google Scholar 

  25. Lassila R, Lepantalo M, Lindfors O. Peripheral arterial disease — natural outcome. Acta Med Scand 1986; 220: 295–301.

    Article  PubMed  CAS  Google Scholar 

  26. Regensteiner JG, Wolfel EE, Brass EP et al. Chronic changes in skeletal muscle histology and function in peripheral arterial disease. Circulation 1993; 87: 413–421.

    Article  PubMed  CAS  Google Scholar 

  27. Hiatt WR, Nawaz D, Brass EP. Carnitine metabolism during exercise in patients with peripheral vascular disease. J Appl Physiol 1987; 62: 2383–2387.

    PubMed  CAS  Google Scholar 

  28. Hiatt WR, Wolfel EE, Regensteiner JG, Brass EP. Skeletal muscle carnitine metabolism in patients with unilateral peripheral arterial disease. J Appl Phyiol 1992; 73: 346–353.

    CAS  Google Scholar 

  29. Arduini A, Molajoni F, Dottori S et al. Effect of oral propionyl-L-carnitine treatment on membrane phospholipid fatty acid turnover in diabetic rat erythrocytes. Diabetes. Accepted pending revision.

    Google Scholar 

  30. Hülsmann WC, Dubelaar ML. Carnitine requirement of vascular endothelial and smooth muscle cells in imminent ischemia. Mol Cell Biochem 1992; 116: 125–129.

    Article  PubMed  Google Scholar 

  31. Arduini A, Gorbunov N, Arrigoni Martelli E et al. Effect of L-carnitine and its acetate and propionate esters on the molecular dynamics of human erythrocyte membrane. Biochim Biophys Acta 1993; 1146: 229–235.

    Article  PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 1995 Springer Science+Business Media Dordrecht

About this chapter

Cite this chapter

Corsico, N., Arrigoni-Martelli, E. (1995). Effect of propionyl-L-carnitine on experimental models of peripheral arteriopathy in the rat. In: De Jong, J.W., Ferrari, R. (eds) The Carnitine System. Developments in Cardiovascular Medicine, vol 162. Springer, Dordrecht. https://doi.org/10.1007/978-94-011-0275-9_26

Download citation

  • DOI: https://doi.org/10.1007/978-94-011-0275-9_26

  • Publisher Name: Springer, Dordrecht

  • Print ISBN: 978-94-010-4122-5

  • Online ISBN: 978-94-011-0275-9

  • eBook Packages: Springer Book Archive

Publish with us

Policies and ethics