SAEVA Proceedings 2015 | Page 59

South African Equine Veterinary Association Congress 2015  Protea Hotel  Stellenbosch Pituitary pars intermedia dysfunction - diagnosis and treatment Schott HC* Professor, Equine Internal Medicine Department of Large Animal Clinical Sciences D-202 Veterinary Medical Center Michigan State University, East Lansing, MI 48824-1314 (517)-353-9710 [email protected] Although the frequency of diagnosis and treatment of pituitary pars intermedia dysfunction (PPID) in horses has clearly increased over the past decade, there is no evidence that the prevalence of PPID is actually increasing. Increased recognition of the disease is likely a consequence of clients maintaining their horses to more advanced ages as well as improved health care (e.g., diet and dentistry) being provided to older horses. Recent surveys of horse owners in Queensland, Australia and the United Kingdom revealed prevalences of 15-20% of PPID in horses and ponies 15 years of age and older, increasing to nearly 30% in horses over 30 years of age. There is no gender predilection and average age of affected horses is around 20 years. All breeds and types of equids can be affected with PPID but ponies appear to be at greater risk. Pathophysiology: In humans and dogs, Cushing‟s disease is most commonly attributed to a corticotroph adenoma in the pars distalis of the pituitary gland. These adenomas are thought to arise spontaneously. In contrast, Cushing‟s disease in horses is almost exclusively attributed to melanotrope hyperplasia or adenoma formation in the pars intermedia (PI) that appears to be due to loss of hypothalamic innervation. Abnormal PI tissue in horses contains markedly reduced amounts of dopamine, about 10% that of normal pars intermedia tissue, consistent with a specific loss of hypothalamic dopaminergic innervation. Further, tyrosine hydroxylase activity, the rate limiting enzyme for production of dopamine, is also markedly reduced in equids with PPID. Recent evidence suggests that this loss of dopaminergic innervation is due to oxidant-induced injury to hypothalamic tissue. Thus, a risk factor for affected horses may be reduced anti-oxidant defense mechanisms in neural tissue. Further, insoluble aggregates of the neural protein αsynuclein have been found in dopaminergic nerve terminals of PPID-affected horses. These protein aggregates are also found in humans with Parkinson‟s disease suggesting that the two neurodegenerative disorders may share a similar pathogenesis. However, the population of neurons affected in horses, as compared to humans, appears to be somewhat different leading to the difference in clinical signs observed in each species. In normal equids, hypothalamic dopaminergic neurons exert a tonic inhibitory action on PI melanotropes. Progressive loss of dopaminergic innervation over many years leads to unregulated enlargement of the PI in equids with PPID, initially by melanotrope hyperplasia and progressing to micro- and macroadenoma formation within the PI. A consequence of loss of PI dopaminergic innervation is excess production of the prohormone pro-opiomelanocortin (POMC, 241 amino acids). Processing of POMC by prohormone convertase 1 and prohormone convertase 2 leads to increased amounts of many POMC-derived peptides, including ACTH1-39 (POMC138-176) and αMSH (ACTH1-13) in jugular venous plasma. Excess amounts of POMC peptides are thought to be responsible for development of the variable clinical signs of PPID, although actual pathophysiology remains poorly understood. 59