Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Mechanisms of Disease: cytokine and adipokine signaling in uremic cachexia

Abstract

Clinical wasting is an important risk factor for mortality in uremic patients and is reported to have a prevalence of 30–60%. 'Malnutrition' is often inappropriately used to describe a group of nutritional abnormalities in uremic patients, which are characterized by anorexia, increased basal metabolic rate, loss of lean body mass, and declining levels of serum proteins. This syndrome—more accurately described as 'cachexia'—manifests as growth failure in children with uremia. Acidosis and inflammation are important causes of uremic cachexia but the underlying molecular mechanism is not well understood. Concentrations of circulating cytokines, such as leptin, tumor necrosis factor-α, interleukin-1, and interleukin-6, are elevated in patients with end-stage renal disease and correlate with the degree of cachexia in these individuals. Other energy-modulating hormones such as ghrelin, and adipokines such as adiponectin and resistin, are also perturbed in uremia and could contribute to nutritional abnormalities. We recently showed that elevated levels of circulating cytokines might be an important contributor to uremia-associated cachexia via signaling through the central melanocortin system. Small-molecule melanocortin antagonists, which are biologically active when administered orally or intraperitoneally, are now available and have been used successfully to ameliorate experimental cachexia. These findings could form the basis of a novel therapeutic strategy for uremic cachexia.

Key Points

  • The prevalence of clinical wasting—an important risk factor for mortality—in end-stage renal disease is 30–60%

  • Cachexia in uremic patients manifests as anorexia, increased basal metabolic rate, loss of lean body mass, and declining levels of serum proteins

  • The mechanisms through which acidosis and inflammation cause uremic cachexia are not well understood

  • New data indicate that circulating cytokines—levels of which are increased in end-stage renal disease—and other energy-modulating hormones might act on the hypothalamic melanocortin system in the pathogenesis of uremic cachexia

  • Antagonists of melanocortin receptors can ameliorate experimental cachexia

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Control of energy homeostasis by peripheral hormones and their central signaling pathways.

Similar content being viewed by others

References

  1. Gruppen MP et al. (2003) Cardiac disease in young adult patients with end-stage renal disease since childhood: a Dutch cohort study. Kidney Int 63: 1058–1065

    Article  Google Scholar 

  2. Sarnak MJ and Levey AS (2000) Cardiovascular disease and chronic renal disease: a new paradigm. Am J Kidney Dis 35: S117–S131

    Article  CAS  Google Scholar 

  3. Cianciaruso B et al. (1995) Cross-sectional comparison of malnutrition in continuous ambulatory peritoneal dialysis and hemodialysis patients. Am J Kidney Dis 26: 475–486

    Article  CAS  Google Scholar 

  4. Tisdale MJ (1997) Biology of cachexia. J Natl Cancer Inst 89: 1763–1773

    Article  CAS  Google Scholar 

  5. Kotler DP et al. (1989) Magnitude of body-cell-mass depletion and the timing of death from wasting in AIDS. Am J Clin Nutr 50: 444–447

    Article  CAS  Google Scholar 

  6. Sherry BA et al. (1989) Anticachectin/tumor necrosis factor-alpha antibodies attenuate development of cachexia in tumor models. FASEB J 3: 1956–1962

    Article  CAS  Google Scholar 

  7. Fong Y et al. (1989) Cachectin/TNF or IL-1 alpha induces cachexia with redistribution of body proteins. Am J Physiol 256: R659–665

    CAS  PubMed  Google Scholar 

  8. Plata-Salaman CR et al. (1996) Anorexia induced by chronic central administration of cytokines at estimated pathophysiological concentrations. Physiol Behav 60: 867–875

    Article  CAS  Google Scholar 

  9. Scribner BH et al. (1960) The treatment of chronic uremia by means of intermittent hemodialysis: a preliminary report. Trans Am Soc Artif Intern Organs 6: 114–122

    CAS  PubMed  Google Scholar 

  10. Anderstam B et al. (1996) Middle-sized molecule fractions isolated from uremic ultrafiltrate and normal urine inhibit ingestive behavior in the rat. J Am Soc Nephrol 7: 2453–2460

    CAS  PubMed  Google Scholar 

  11. Mamoun AH et al. (1999) Evidence of splanchnic-brain signaling in inhibition of ingestive behavior by middle molecules. J Am Soc Nephrol 10: 309–314

    CAS  PubMed  Google Scholar 

  12. Pecoits-Filho R et al. (2005) Chronic kidney disease and inflammation in pediatric patients: from bench to playground. Pediatr Nephrol 20: 714–720

    Article  Google Scholar 

  13. Kaizu Y et al. (2003) Association between inflammatory mediators and muscle mass in long-term hemodialysis patients. Am J Kidney Dis 42: 295–302

    Article  CAS  Google Scholar 

  14. Bologa RM et al. (1998) Interleukin-6 predicts hypoalbuminemia, hypocholesterolemia, and mortality in hemodialysis patients. Am J Kidney Dis 32: 107–114

    Article  CAS  Google Scholar 

  15. Goodman MN (1994) Interleukin-6 induces skeletal muscle protein breakdown in rats. Proc Soc Exp Biol Med 205: 182–185

    Article  CAS  Google Scholar 

  16. Tsujinaka T et al. (1996) Interleukin 6 receptor antibody inhibits muscle atrophy and modulates proteolytic systems in interleukin 6 transgenic mice. J Clin Invest 97: 244–249

    Article  CAS  Google Scholar 

  17. Aguilera A et al. (1998) Anorexigen (TNF alpha, cholecystokinin) and orexigen (neuropeptide Y) in peritoneal dialysis patients. Their relationship with nutritional parameters. Nephrol Dial Transplant 13: 1476–1483

    Article  CAS  Google Scholar 

  18. Guttridge DC et al. (2000) NF-κB induced loss of MyoD messenger RNA: Possible role in muscle decay and cachexia. Science 289: 2363–2365

    Article  CAS  Google Scholar 

  19. Plata-Salaman CR (1998) Cytokines and anorexia: a brief overview. Semin Oncol 25: 4–11

    Google Scholar 

  20. Schwartz MW and Morton GJ (2002) Obesity: Keeping hunger at bay. Nature 418: 595–597

    Article  CAS  Google Scholar 

  21. Ahima RS and Staels B (2000) Leptin. Ann Rev Physiol 62: 413–437

    Article  CAS  Google Scholar 

  22. Halaas J et al. (1995) Weight-reducing effects of plasma protein encoded by the obese gene. Science 269: 543–549

    Article  CAS  Google Scholar 

  23. Sharma K et al. (1997) Plasma leptin is partly cleared by the kidney and is elevated in hemodialysis patients. Kidney Int 51: 1980–1985

    Article  CAS  Google Scholar 

  24. Stenvinkel P et al. (2000) Increases in serum leptin during peritoneal dialysis are associated with inflammation and a decrease in lean body mass. J Am Soc Nephrol 11: 1303–1309

    CAS  PubMed  Google Scholar 

  25. Daschner M et al. (1998) Inappropriate elevation of serum leptin in children with chronic renal failure. European Study Group for Nutritional Treatment of Chronic Renal Failure in Chilhood. J Am Soc Nephrol 9: 1074–1079

    CAS  PubMed  Google Scholar 

  26. Wolf G et al. (2002) Leptin and renal disease. Am J Kidney Dis 39: 1–11

    Article  CAS  Google Scholar 

  27. Heimburger O and Stenvinkel P (2005) Adipokines in chronic kidney disease—fat tissue gives nephrologists a message. Perit Dial Int 25: 340–342

    PubMed  Google Scholar 

  28. Qi Y et al. (2004) Adiponectin acts in the brain to decrease body weight. Nature Med 10: 524–529

    Article  CAS  Google Scholar 

  29. Axelsson J et al. (2006) Elevated resistin levels in chronic kidney disease are associated with decreased glomerular filtration rate and inflammation, but not with insulin resistance. Kidney Int 69: 596–604

    Article  CAS  Google Scholar 

  30. Gomez-Ambrosi J et al. (2002) Weight loss in tumour-bearing mice is not associated with changes in resistin gene expression in white adipose tissue. Horm Metab Res 34: 674–677

    Article  CAS  Google Scholar 

  31. Palanivel R and Sweeney G (2005) Regulation of fatty acid uptake and metabolism in L6 skeletal muscle cells by resistin. FEBS Lett 579: 5049–5054

    Article  CAS  Google Scholar 

  32. Yoshimoto A et al. (2002) Plasma ghrelin and desacyl ghrelin concentrations in renal failure. J Am Soc Nephrol 13: 2748–2752

    Article  CAS  Google Scholar 

  33. Rodriguez Ayala E et al. (2004) Associations between plasma ghrelin levels and body composition in end-stage renal disease: a longitudinal study. Nephrol Dial Transplant 19: 421–426

    Article  CAS  Google Scholar 

  34. Wynne K et al. (2005) Subcutaneous ghrelin enhances acute food intake in malnourished patients who receive maintenance peritoneal dialysis: a randomized, placebo-controlled trial. J Am Soc Nephrol 6: 2111–2118

    Article  Google Scholar 

  35. Cheung W et al. (2005) Role of leptin and melanocortin signaling in uremia-associated cachexia. J Clin Invest 115: 1659–1665

    Article  CAS  Google Scholar 

  36. Madiehe AM et al. (2003) Hyperleptinemia and reduced TNF-alpha secretion cause resistance of db/db mice to endotoxin. Am J Physiol Regul Integr Comp Physiol 284: R763–R770

    Article  CAS  Google Scholar 

  37. Marks DL et al. (2001) Role of central melanocortin system in cachexia. Cancer Res 61: 1432–1438

    CAS  PubMed  Google Scholar 

  38. Mitch WE and Goldberg AL (1996) Mechanisms of muscle wasting: the role of the ubiquitin-proteasome pathway. N Engl J Med 335: 1897–1905

    Article  CAS  Google Scholar 

  39. Mitch WE et al. (1994) Metabolic acidosis stimulates muscle protein degradation by activating the adenosine triphosphate-dependent pathway involving ubiquitin and proteasomes. J Clin Invest 93: 2127–2133

    Article  CAS  Google Scholar 

  40. Isozaki U et al. (1996) Protein degradation and increased mRNAs encoding proteins of the ubiquitin-proteasome proteolytic pathway in BC3H1 myocytes require an interaction between glucocorticoids and acidification. Proc Natl Acad Sci USA 93: 1967–1971

    Article  CAS  Google Scholar 

  41. Price SR et al. (1996) Muscle wasting in insulinopenic rats results from activation of the ATP-dependent, ubiquitin-proteasome proteolytic pathway by a mechanism including gene transcription. J Clin Invest 98: 1703–1708

    Article  CAS  Google Scholar 

  42. Motil KJ et al. (1981) Whole-body leucine and lysine metabolism: response to dietary protein intake in young men. Am J Physiol 240: E712–E721

    CAS  PubMed  Google Scholar 

  43. Tom K et al. (1995) Long-term adaptive responses to dietary protein restriction in chronic renal failure. Am J Physiol 268: E668–E677

    CAS  PubMed  Google Scholar 

  44. Maroni BJ et al. (1997) Mechanisms permitting nephrotic patients to achieve nitrogen equilibrium with a protein-restricted diet. J Clin Invest 99: 2479–2487

    Article  CAS  Google Scholar 

  45. Mitch WE (2002) Requirements for protein, calories, and fat in the predialysis patient. In Handbook of Nutrition and the Kidney, 144–165 (Eds Mitch WE and Klahr S) Philadelphia: Lippincott, Williams and Wilkins

    Google Scholar 

  46. Simons JP et al. (1999) Weight loss and low body cell mass in males with lung cancer: relationship with systemic inflammation, acute-phase response, resting energy expenditure, and catabolic and anabolic hormones. Clin Sci 97: 215–223

    Article  CAS  Google Scholar 

  47. Munro R and Capell H (1997) Prevalence of low body mass in rheumatoid arthritis: associations with the acute phase response. Ann Rheum Dis 56: 326–329

    Article  CAS  Google Scholar 

  48. Stenvinkel P et al. (2000) Are there two types of malnutrition in chronic renal failure? Evidence for relationships between malnutrition, inflammation and atherosclerosis (MIA syndrome). Nephrol Dial Transplant 15: 953–960

    Article  CAS  Google Scholar 

  49. Pupim LB et al. (2002) Intradialytic parenteral nutrition improves protein and energy homeostasis in chronic hemodialysis patients. J Clin Invest 110: 483–492

    Article  CAS  Google Scholar 

  50. Betts PR and McGrath G (1974) Growth patterns and dietary intake of children with chronic renal insufficiency. Br Med J 2: 189–193

    Article  CAS  Google Scholar 

  51. Simmons JM et al. (1971) Relation of calorie deficiency to growth failure in children on hemodialysis and the growth response to calorie supplementation. N Engl J Med 285: 653–656

    Article  CAS  Google Scholar 

  52. Arnold WC et al. (1983) Effect of calorie supplementation on growth in children with uremia. Kidney Int 24: 205–209

    Article  CAS  Google Scholar 

  53. Walker S et al. (2003) Catch-up growth with normal parathyroid hormone levels in chronic renal failure. Pediatr Nephrol 18: 1236–1241

    Article  Google Scholar 

  54. Jones RW et al. (1982) The effects of chronic renal failure in infancy on growth, nutritional status and body compositioin. Pediatr Res 16: 784–791

    Article  CAS  Google Scholar 

  55. Markison S et al. (2005) The regulation of feeding and metabolic rate and the prevention of murine cancer cachexia with a small-molecule melanocortin-4 receptor antagonist. Endocrinology 146: 2766–2773

    Article  CAS  Google Scholar 

  56. Horvath TL (2005) The hardship of obesity: a soft-wired hypothalamus. Nature Neurosci 8: 561–565

    Article  CAS  Google Scholar 

  57. Cone RD (2005) Anatomy and regulation of the central melanocortin system. Nature Neurosci 8: 571–578

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by grants, R01 DK 50780 and K24 DK 59475 to RHM and K08 DK 62207 to DLM, from the NIH.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Robert H Mak.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Mak, R., Cheung, W., Cone, R. et al. Mechanisms of Disease: cytokine and adipokine signaling in uremic cachexia. Nat Rev Nephrol 2, 527–534 (2006). https://doi.org/10.1038/ncpneph0273

Download citation

  • Received:

  • Accepted:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncpneph0273

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing