<<

IN-DEPTH: GERIATRIC MEDICINE/METABOLICS

Review of Current Understanding of Pituitary Pars Intermedia Dysfunction

Dianne McFarlane, DVM, PhD, Diplomate ACVIM

Equine pituitary pars intermedia dysfunction (PPID) is a commonly diagnosed disease in the aged equine population. Recognition of PPID has increased considerably over the past two decades, in part because of an increase in client awareness of the condition as well as an increase in the number of aged horses receiving veterinary equine PPID care. This review focuses on the most current understanding of the development and progression of equine PPID. Author’s address: Oklahoma State University, Department of Physiological Sciences, Center of Veterinary Health Sciences, Still- water, OK 74078; e-mail: [email protected]. © 2013 AAEP.

1. Introduction is produced in the pars intermedia of the healthy horse.1 Anatomy and Function of the Equine Pituitary Equine pars intermedia activity has been shown The equine lies ventral to the optic to be inhibited by and stimulated by chiasm, separated from the brain by a fold of dura thyrotopin-releasing .2,3 The pars inter- mater known as the diaphragma sellae. It is sus- media receives direct innervation from the dopami- pended from the by the infundibular nergic of the periventricular nucleus of stalk. The equine pituitary gland has four lobes; the hypothalamus. These project through the pars distalis, pars intermedia, the infundibular stalk along the periphery of the (collectively known as the adenohypophysis), and pars nervosa, then travel into the pars intermedia, pars nervosa (neurohypophysis). where they terminate on the endocrine cells of the Melanotropes of the pars intermedia produce a pars intermedia, the melanotropes. Dopamine is hormone precursor , pro-opiomelancortin released at the pars intermedia from the nerve ter- (POMC), which undergoes extensive tissue-specific minals of the hypothalamic periventricular neurons cleavage to yield adrenocorticotropic hormone (ACTH), that synapse directly on the melanotropes.4 In the melanocyte-stimulating (MSH), ␤-endor- presence of dopamine secretion of pars intermedia, phin, corticotrophin-like intermediate lope POMC-derived peptide hormones are decreased. (CLIP), , and several other small pep- If dopamine is removed either by surgically cutting tides. In the pars intermedia of the healthy ani- the hypothalamic pituitary connection or by geneti- mal, the primary hormones produced are ␣-MSH, cally deleting the dopamine , melanotropes ␤-endorphin, and CLIP. Nearly all plasma ACTH will proliferate, hypertrophy, and increase produc- originates from the pars distalis, and minimal ACTH tion of POMC-derived .5–7

NOTES

294 2013 ր Vol. 59 ր AAEP PROCEEDINGS IN-DEPTH: GERIATRIC MEDICINE/METABOLICS The products of POMC are diverse and highly curs to the neurons, although at this pleiotrophic in function. ␣-MSH has a role in metab- time it is unknown whether oxidative stress is a olism, obesity, stress, and inflammation. Because of cause or consequence of PPID.25–27 these critical functions, this hormone is currently the Neuronal accumulation and aggregation of mis- focus of significant research attention, with more than folded is a mechanism that contributes 1250 PubMed citations in the past 5 years alone. to the pathogenesis of most neurodegenerative dis- ␣-MSH induces an anorexic response and feeling of eases, including Parkinson’s disease, a dopamin- satiety8 and has broad anti-inflammatory effects that ergic neurodegenerative disease of aged people. include decreased production of a wide array of cyto- In Parkinson disease, the protein that accumulates kines and other molecules, factors that contribute in the dopaminergic neurons is ␣-synuclein. ␣- to inflammation.9,10 ␣-MSH also impairs neutro- Synuclein is natively unfolded; however, under cer- phil function, including oxidative burst, , tain cellular conditions, ␣-synuclein can aggregate and adhesion.11,12 CLIP is the cleavage product in dopaminergic nerve terminals, disrupting cellular generated from the c-terminal portion of ACTH. function and triggering death.28 Conditions Little is known about the function of CLIP; how- that promote accumulation of ␣-synuclein include ever, both CLIP and its cleavage product, ␤-cell excessive concentration caused by increased produc- tropin, have been shown to stimulate the release of tion or decreased clearance, oxidation or nitration, from rodent beta cells.13 ␤-Endorphin is a and synuclein .29,30 ␣-Synuclein potent endogenous ␮-receptor that protein and was found to be in- functions in analgesia and reduction of pain- creased in the pars intermedia of horses with associated inflammation. PPID.21 In addition to being more abundant, pars Similar to several other species such as hamsters intermedia ␣-synuclein appears to be excessively and sheep, activity of the pars intermedia in horses nitrated in horses with PPID, a modification that has a robust seasonal rhythm, with increased output promotes aggregation.21 It is unknown if failure occurring as day length shortens.14–19 As a result, of protein clearance also contributes to ␣-synuclein the plasma concentration of the pars intermedia accumulation in horses with PPID. Misfolded pro- hormones, including ␣-MSH, are greater in the au- teins are removed primarily through autophagy, tumn (August through October) than in the winter the process by which damaged proteins or organelles or spring.18,19 It has been suggested that this ad- are recycled by the .31 Assessment of au- aptation helps to prepare the animal for the meta- tophagy in the periventricular neurons of horses bolic and nutritional pressures of the approaching with PPID is ongoing. winter. 3. Clinical Signs of PPID and Diagnostic Testing 2. Pituitary Pars Intermedia Dysfunction Clinical signs of PPID probably are the result of Equine PPID is associated with increased size and overexpression of the pars intermedia hormones. activity of the pars intermedia. At necropsy, horses Late in the disease, it is also possible that loss of with PPID have enlarged pituitary glands caused hormones from the adjacent, compressed lobes of the by hyperplasia, hypertrophy, and a single large or pituitary may also contribute to the clinical syn- multiple small adenomas. Enlargement of the pars drome. Weight loss caused by muscle atrophy, be- intermedia is often accompanied by compression of havioral changes, secondary infections, and changes adjacent . PPID was previously charac- in haircoat are some of the most common signs of terized as a benign neoplasia of the equine pituitary PPID. Laminitis occurs with PPID but less fre- gland; however, clinical, pharmacological, biochem- quently than was originally suggested. It is likely ical, and histological data all indicate that PPID is that laminitis occurs only in horses with PPID and a neurodegenerative disease with loss of inhibitory concurrent insulin dysregulation. Further work is dopaminergic input to the pars intermedia. Typi- ongoing to identify the mechanism of development of cal of any neurodegenerative disease, age is the endocrinopathic lamintis. primary risk factor for PPID, and progression of Testing for PPID involves measurement of en- clinical signs occur slowly, making early diagnosis dogenous hormone concentrations, including ACTH problematic. or ␣-MSH or dynamic testing. Dynamic testing, In horses with PPID, there is a marked reduc- which measures the response of the pars intermedia tion of dopamine in pars intermedia tissue20 as well to stimulation or inhibition, may be a more discrim- as a profound loss of dopaminergic periventricular inating approach to disease diagnosis. Because of nerve terminals and cell bodies.21 Further evi- the increase in PI activity in the fall, false-positive dence that PPID results from loss of dopamine is the diagnostic test results for PPID are common when improvement in clinical signs and plasma hormone testing is performed during the autumn if refer- concentration that is observed when horses with ence intervals are not adjusted for season.18–20 PPID are treated with a such as In addition, clinical signs of PPID are often more pergolide.22–24 Although the precise cause of neu- pronounced in the autumn, most notably an in- rodegeneration in PPID is unknown, several studies creased incidence of laminitis.32 Because pasture have provided evidence that oxidative damage oc- composition also changes significantly with season,

AAEP PROCEEDINGS ր Vol. 59 ր 2013 295 IN-DEPTH: GERIATRIC MEDICINE/METABOLICS studies are needed to determine the role of hormone weight, and metabolic hormones in rams. Am J Physiol increase in seasonal development of laminitis.33 Regul Integr Comp Physiol 2001;281:R76–R90. 17. Lincoln GA, Richardson M. Photo-neuroendocrine control of Diagnostic testing for PPID will be discussed in more seasonal cycles in body weight, pelage growth and reproduc- detail in another session. tion: lessons from the HPD sheep model. Comp Biochem Physiol C Pharmacol Toxicol Endocrinol 1998;119:283–294. 18. McFarlane D, Donaldson MT, McDonnell SM, et al. Effects References of season and sample handling on measurement of plasma 1. Wilson MG, Nicholson WE, Holscher MA, et al. Proopioli- alpha-melanocyte-stimulating hormone concentrations in pomelanocortin peptides in normal pituitary, pituitary tumor horses and ponies. Am J Vet Res 2004;65:1463–1468. and plasma of normal and Cushing’s horses. 19. Donaldson MT, McDonnell SM, Schanbacher BJ, et al. 1982;110:941–954. Variation in plasma adrenocorticotropic hormone concentra- 2. McFarlane D, Beech J, Cribb A. Alpha-melanocyte stimu- tion and dexamethasone suppression test results with sea- lating hormone release in response to thyrotropin releasing son, age, and sex in healthy ponies and horses. J Vet Intern hormone in healthy horses, horses with pituitary pars inter- Med 2005;19:217–222. media dysfunction and equine pars intermedia explants. 20. Millington WR, Dybdal NO, Dawson R Jr, et al. Equine Domest Anim Endocrinol 2006;30:276–288. Cushing’s disease: differential regulation of beta-endorphin 3. Orth DN, Holscher MA, Wilson MG, et al. Equine Cushing’s processing in tumors of the intermediate pituitary. Endo- disease: plasma immunoreactive proopiolipomelanocortin crinology 1988;123:1598–1604. peptide and levels basally and in response to diag- 21. McFarlane D, Dybdal N, Donaldson MT, et al. Nitration nostic tests. Endocrinology 1982;110:1430–1441. and increased alpha-synuclein expression associated with do- 4. Saland LC. The mammalian pituitary intermediate lobe: paminergic in equine pituitary pars inter- an update on innervation and regulation. Brain Res Bull media dysfunction. J Neuroendocrinol 2005;17:73–80. 2001;54:587–593. 22. Donaldson MT, LaMonte BH, Morresey P, et al. Treatment 5. Saiardi A, Borrelli E. Absence of dopaminergic control on with pergolide or cyproheptadine of pituitary pars intermedia melanotrophs leads to Cushing’s-like syndrome in mice. dysfunction (equine Cushing’s disease). J Vet Intern Med Mol Endocrinol 1998;12:1133–1139. 2002;16:742–746. 6. Goudreau JL, Lindley SE, Lookingland KJ, et al. Evidence 23. Perkins GA, Lamb S, Erb HN, et al. Plasma adrenocortico- that hypothalamic periventricular dopamine neurons inner- tropin (ACTH) concentrations and clinical response in horses vate the intermediate lobe of the rat pituitary. Neuroendo- treated for equine Cushing’s disease with cyproheptadine or crinology 1992;56:100–105. pergolide. Equine Vet J 2002;34:679–685. 7. Engler D, Pham T, Liu JP, et al. Studies of the regulation of 24. Schott HC, Coursen CL, Eberhart SW, et al. The Michigan the hypothalamic-pituitary-adrenal axis in sheep with hypo- Cushing’s Project, in Proceedings. Am Assoc Equine Pract thalamic-pituitary disconnection, II: evidence for in vivo 2001;47:22–24. ultradian hypersecretion of peptides by 25. Glover CM, Miller LM, Dybdal NO, et al. Extrapituitary the isolated anterior and intermediate pituitary. Endocri- and pituitary pathological findings in horses with pitui- nology 1990;127:1956–1966. tary pars intermedia dysfunction: a retrospective study. 8. Schuhler S, Horan TL, Hastings MH, et al. Feeding and J Equine Vet Sci 2009;29:146–153. behavioural effects of central administration of the melano- 26. Keen JA, McLaren M, Chandler KJ, et al. Biochemical in- cortin 3/4-R antagonist SHU9119 in obese and lean Siberian dices of vascular function, glucose metabolism and oxidative hamsters. Behav Brain Res 2004;152:177–185. stress in horses with equine Cushing’s disease. Equine Vet 9. Murphy MT, Richards DB, Lipton JM. Antipyretic potency J 2004;36:226–229. of centrally administered alpha-melanocyte stimulating hor- 27. McFarlane D, Cribb AE. Systemic and pituitary pars inter- mone. Science 1983;221:192–193. media capacity associated with pars intermedia 10. Lipton JM, Catania A. Anti-inflammatory actions of the oxidative stress and dysfunction in horses. Am J Vet Res neuroimmunomodulator alpha-MSH. Immunol Today 1997; 2005;66:2065–2072. 18:140–145. 28. Conway KA, Lee SJ, Rochet JC, et al. Acceleration of oli- 11. Catania A, Rajora N, Capsoni F, et al. The gomerization, not fibrillization, is a shared property of both alpha-MSH has specific receptors on neutrophils and reduced alpha-synuclein mutations linked to early-onset Parkinson’s chemotaxis in vitro. Peptides 1996;17:675–679. disease: implications for pathogenesis and therapy. Proc 12. Oktar BK, Yuksel M, Alican I. The role of cyclooxygenase Natl Acad SciUSA2000;97:571–576. inhibition in the effect of alpha-melanocyte stimulating hor- 29. Betarbet R, Canet-Aviles RM, Sherer TB, et al. Intersecting mone on reactive oxygen species production by rat peritoneal pathways to neurodegeneration in Parkinson’s disease: ef- neutrophils. 2004;71:1–5. fects of the pesticide rotenone on DJ-1, alpha-synuclein, and 13. Marshall JB, Kapcala LP, Manning LD, et al. Effect of the - system. Neurobiol Dis 2006;22: corticotropin-like intermediate lobe peptide on pancreatic 404–420. exocrine function in isolated rat pancreatic lobules. J Clin 30. Kirik D, Rosenblad C, Burger C, et al. Parkinson-like neu- Invest 1984;74:1886–1889. rodegeneration induced by targeted overexpression of alpha- 14. Logan A, Weatherhead B. Photoperiodic dependence of sea- synuclein in the nigrostriatal system. J Neurosci 2002;22: sonal changes in pituitary content of melanocyte-stimulating 2780–2791. hormone. Neuroendocrinol 1980;30:309–312. 31. Cuervo AM, Stefanis L, Fredenburg R, et al. Impaired deg- 15. Lincoln GA, Baker BI. Seasonal and photoperiod-induced radation of mutant alpha-synuclein by -mediated changes in the secretion of alpha-melanocyte-stimulating autophagy. Science 2004;305:1292–1295. hormone in Soay sheep: temporal relationships with changes 32. Donaldson MT, Jorgensen AJ, Beech J. Evaluation of sus- in beta-endorphin, , follicle-stimulating hormone, ac- pected pituitary pars intermedia dysfunction in horses with tivity of the and growth of wool and horns. J Endocri- laminitis. J Am Vet Med Assoc 2004;224:1123–1127. nol 1995;144:471–481. 33. Hoffman RM, Wilson JA, Kronfeld DS, et al. Hydrolyzable 16. Lincoln GA, Rhind SM, Pompolo S, et al. Hypothalamic carbohydrates in pasture, hay, and horse feeds: direct control of photoperiod-induced cycles in food intake, body and seasonal variation. J Anim Sci 2001;79:500–506.

296 2013 ր Vol. 59 ր AAEP PROCEEDINGS