Internal Medicine and Reproduction 2015 Program Proceedings

Seventh Annual Symposium From Our Practice to Yours

September 23rd - September 26th, 2015

Northeast Association of Equine Practitioners

1 NEAEP Mission Statement

The mission of the NEAEP is to improve the health and welfare of byproviding state- of-the-art professional education and supporting the economic security of the equine industry by complementing established local associations and giving equine veterinarians, farriers, technicians, veterinary students and owners a unified voice at the state and regional levels.

2 2015 Internal Medicine/Reproduction

Table of Contents

New Strategies For Managing Neonatal Maladjustment Syndrome. Peter R. Morresey...... 4

Update on Anaplasmosis – Made No Better by a Name Change. Peter R. Morresey...... 10

Head Shakers – Is There Anything New Under The Sun? Peter R. Morresey...... 14

Managing Strangles Outbreaks Ashley G. Boyle...... 21

Strangles- Unusual Cases and Effective Treatments Ashley G. Boyle...... 25

Diagnosis and Management of the Sick Foal in the Field Ashley G. Boyle...... 28

Treating Colitis In The Field. Rose D. Nolen-Walston...... 30

Clenbuterol – The Good, The Bad, And The Ugly Rose D. Nolen-Walston...... 37

Equine Blood Transfusion - What You Need To Know To Get The Job Done. Rose D. Nolen-Walston...... 41

Guttural Pouch Disease- More Than Strangles Rachel Gardner...... 46

New Thinking on Gastric Ulcer Diagnosis and Treatment Rachel Gardner...... 50

Evaluation and Management of the Recumbent Horse Rachel Gardner...... 54

Nonexertional Rhabdomyolysis Stephanie J. Valberg ...... 56

Exertional Rhabdomyolysis - Causes of Poor Performance Stephanie J. Valberg...... 65

Equine Genetic Diseases - Genetic Testing: What Is Available And When To Use It Stephanie J. Valberg...... 71

The Pleurals and the Peritoneals: Body Cavity Cytology Tracy Stokol...... 78

A Case-Based Approach to Equine Blood Tracy Stokol...... 83

The ABCs of Acid-Base Analysis or the View from the Eye of a Clinical Pathologist Tracy Stokol...... 96

Non-traditional therapies for endometritis: science or voodoo? Charles F. Scoggin...... 102

The impact of aging on fertility, pregnancy, and offspring vigor in broodmares Charles F. Scoggin...... 108

The benefit of routine monitoring and supplementation of progesterone in early pregnancy Charles F. Scoggin...... 114

Management of mares for breeding with frozen semen: pre- and post-insemination considerations for maximum fertility Kristina Lu...... 118 How to successfully integrate assisted reproductive techniques in your busy practice Kristina Lu...... 124 Diagnostic methods and interpretation for endometritis Kristina Lu...... 129

3 New Strategies For Managing Neonatal Maladjustment Syndrome. Peter R. Morresey BVSc MACVSc DipACT DipACVIM (LA) Rood and Riddle Equine Hospital, PO Box 12070, Lexington, KY 40580

Introduction Neonatal Maladjustment Syndrome, Hypoxic-Ischemic Encephalopathy, or Perinatal Asphyxia Syndrome: foals poorly adapted to extrauterine life have been categorized in many ways. Clinical manifestation can be subtle or profound. This talk will summarize current literature explaining what is happening in these foals, provide insights into the things we can change and those we can’t, and walk through the author’s approach to these cases.

Pathophysiology of Neonatal Maladjustment Syndrome Cerebral ischemia may result from systemic hypoxemia depressing myocardial performance. Reduced cerebral blood flow results in decreased delivery of oxygen and energy substrates to the brain resulting in a combined hypoxic-ischemic insult. Anaerobic metabolism depletes the brain’s stores of glucose and high energy phosphates (ATP and phosphocreatine) with resulting accumu- lation of lactate and inorganic phosphate. (Yager, 1992) Energy failure impairs glutamate uptake resulting in extracellular accumulation leading to tonic overstimulation of postsynaptic excitotoxic amino acid receptors (excitotoxicity). Reperfusion and reoxygenation in the early post-ischemic recovery period following this insult is itself harmful. Maternal infection has been shown strongly associated with neonatal brain injury. (Willough- by, Jr., 2002) Intrauterine infection may indirectly injure the brain via the induction of sepsis and poor perfusion, or directly by the production of inflammatory mediators that devitalize neurons. (Eklind, 2001) The neonatal brain has been experimentally shown as highly susceptible to free radical-me- diated injury. (Edwards, 1997) Under normal conditions, mitochondrial function generates low concentrations of the superoxide anion and hydrogen peroxide. These are scavenged by multiple enzyme systems including superoxide dismutase, catalase, and glutathione peroxidase. Other nonenzymatic antioxidants include a-tocopherol (vitamin E) and ascorbic acid (vitamin C). During reperfusion following ischemia, oxygen free radicals react with membrane phospholipids to form oxidized lipids with pro-inflammatory actions. (Grow, 2002) Regardless of the inciting cause, neural injury results in increased release of neurotrans- mitters that in turn generates an excess of second messengers, apoptosis and increases in in- tracellular sodium and calcium. (Grow, 2002) Accumulation of sodium concurrent with failure of energy dependent cell membrane ion pumps (e.g. Na+-K+ ATPase) leads rapidly to cell swelling. Accumulation of calcium due to glutamate excess and mediated by NMDA receptors can lead to activation of calcium-dependent phospholipases, nitric oxide synthase, and proteases. Increased activation of phospholipase A2 (PLA2) in postsynaptic neurons results in increased free arachi- donic acid and PAF, enhancing glutamate-mediated neurotransmission and further potentiating NMDA receptor activity, while decreasing glutamate reuptake. Cerebral ischemia induces cyc- lo-oxygenase-2 (COX-2) expression facilitating metabolism of arachidonic acid to inflammatory lipid mediators. A self-reinforcing cycle is therefore initiated. (Arundine, 2004;Bazan, 1995)

A number of equine neonatal studies have shown the presence of intracranial hemorrhage associated with neurological compromise. Compared to a control group, convulsive foals showed hemorrhage in various locations of the brain. (Palmer, 1975) Also present was necrosis of the

4 cerebral cortex, diencephalon and brain stem. Minimal hemorrhage occurred in the brains of control foals. In another study, central nervous system hemorrhage was found more frequently in premature foals when compared to those born at full term, in foals born dead compared to live births, and in foals born following assisted delivery compared to foals born without intervention.

Clinical presentation of Neonatal Maladjustment Syndrome The most common neurological problems of foals include depression, seizure activity, ab- normal behavior, loss of affinity for the mare and loss of the suckle reflex. All these signs are referable to dysfunction of the brain. Expression of neurological lesions will be dependent on the stage of development which the inciting damage occurred. The result of cerebral hypoxic or ischemic insult to the neonate, NMS/HIE/PAS is arguably the most common seizure precipitating syndrome faced by the equine clinician. The diagnosis is dependent upon a compatible history and clinical examination findings while ruling out other con- ditions. (Green and Mayhew, I. G., 1990) Excitatory amino acids, calcium ions, free radicals, nitric oxide, pro-inflammatory cytokines and products of lipid peroxidation are all thought to contribute to the syndrome, with the neonatal brain having an increased susceptibility to damage by these agents compared to adults. (Anderson, 1988;Arango, 2006;Arundine, 2004) In addition to neurological manifestations respiratory compromise, renal insufficiency, and gastrointestinal ileus with or without mucosal compromise can occur. Aberrant pulmonary func- tion can result in abnormal gas exchange, in particular decreased ventilation leading to retention of carbon dioxide. As a result, central depression and abnormal blood acid base balance may result. Renal compromise leads to electrolyte and fluid balance derangements. With the loss of normal gastrointestinal function, diarrhea and sepsis can result from intestinal bacterial over- growth.

Diagnostics in neurological conditions Complete blood count (CBC) and serum chemistry A CBC aids in the diagnosis of systemic infection that may have spread to the CNS. Serum chemistry assays major organ system function, chiefly liver (removal of blood borne toxins) and kidney (assessment of electrolyte homeostasis). Immunoglobulin G (IgG) levels following colostrum are a direct measure of immune sufficiency in the previously hypogammaglobulinemic neonatal foal. Blood gas evaluation assesses pulmonary gas exchange and acid-base status of the foal.

Cerebrospinal fluid (CSF) analysis Cytology and microbiological culture of the CSF aids diagnosis, antimicrobial selection and prognosis in suspected cases of meningitis. Total protein concentration, nucleated cell count and RBC count should be routinely performed. Where increased intracranial pressure is suspected aspiration of CSF should be avoided as herniation of the brain through the foramen magnum may occur.

Radiography, computed tomography (CT) and Magnetic resonance imaging (MRI) Congenital malformations, disruption of bony structures and deviation of neural tissue by trauma can be visualized with radiography and CT. Both plain and contrast radiographic studies are possible. Soft tissue masses, foci of infection or vascular disruption are more readily imaged with MRI.

5 Treatment goals Once a diagnosis of a neurological disorder has been made, the treatment plan should seek to achieve the following clinical goals: • Manage seizure episodes and any neurologic dysfunction • Restore cerebral perfusion and oxygenation if compromised • Control cerebral edema • Control cerebral inflammation • Provide metabolic requirements of the debilitated patient • Address concurrent physical injuries and medical conditions if present

• Manage seizure episodes and any neurologic dysfunction Drugs for seizure control readily accessible to the practicing equine veterinarian are limited. Therapeutic targets include glutamate accumulation, aberrant calcium fluxes due to excessive activation of the N-methyl-D-aspartate (NMDA) glutamate receptors, free radical formation, lipid peroxidation, and generation of arachidonic acid metabolites. (Lipton, 1994;Nilsson, 1996)

• Restore cerebral perfusion and oxygenation if compromised Fluid therapy In addition to meeting the metabolic needs of the patient, appropriate fluid therapy is essential to ensure adequate cerebral perfusion which may have been compromised by a cerebral insult. The brain controls its own perfusion rate and pressure despite fluctuations in arterial blood delivery by autoregulation, the loss of which has been reported after cerebral ischemia and reperfusion or traumatic brain injury. Systemic dehydration has not been shown to decrease existing cerebral edema, and a negative fluid balance has been proven to be detrimental to overall patient out- come. (Clifton, 2002) Supplemental oxygen An appropriate fluid balance maintains cerebral perfusion and oxygenation while avoiding increased intracranial pressure. In situations where cerebral insult has occurred, judicious use of supplemental oxygen may of further benefit as the avoidance of hypoxemia improves neurologi- cal outcome following traumatic brain injury. (Chesnut, 1993)

•Control cerebral edema Edema of the brain decreases perfusion and oxygenation, along with causing cerebral com- pression against bony confinement and further trauma. Edema may be due to disruption of the blood brain barrier (BBB), cellular disruption causing intracellular water collection, or osmotic imbalances between blood and tissue. Iatrogenic over-hydration potentiates CNS edema follow- ing cranial trauma and exacerbates pulmonary edema in recumbent patients (affecting ventilation and oxygenation). Current information suggests elevation of the head during recumbency pre- vents cerebral edema and improves survival, although the mainstay of edema control has tradi- tionally been the use of hyperosmolar agents. (Feldman, 1992)

•Control cerebral inflammation Non-steroidal anti-inflammatory drugs (NSAIDs) attenuate arachidonic acid metabolites that promote platelet aggregation, and facilitate inflammatory and immune reactions. Experimentally, COX inhibitors improve cerebral blood flow, decrease edema, protect COX-2–expressing neu- rons, and attenuate microglial activation.

6 Glucocorticoids are immunosuppressive, diminish upregulation of pro-inflammatory cyto- kines and reduce monocyte infiltration following cerebral ischemia. Glucocorticoids downregulate cytokine-mediated COX-2 expression by monocytes and astrocytes and inhibit phospholipase A2, attenuating release of arachidonic acid from the cell membrane. The use of glucocorticoids in cases of cerebral insult is controversial however benefit in cases of acute bacterial meningitis has been shown. (van de Beek, 2007)

• Address concurrent physical injuries and medical conditions if present Wounds, contusions and decubital ulceration require topical treatments and dressings, and if sufficiently extensive concurrent systemic antimicrobials to counter bacterial dissemination in the compromised neonate. Leg wraps will avoid distal limb edema and secondary limb injury. Head protection ‘bumpers’ minimize secondary neurologic injury from repeated cranial trauma in recumbent or seizing foals. Corneal ulceration secondary to abrasion or exposure keratitis can be prevented by regular application of ophthalmic lubricants. If recumbent, the foal requires a supportive bed and a means to cleanly eliminate body waste. Elevation of the head (up to 30 degrees) minimizes cerebral edema. Sedation of the seizing patient may be required to minimize self-trauma. Options are limited however a2 adrenergic agonists and benzodiazepines possess favorable qualities including the added benefit of muscle relaxation which decreases overall energy requirements. Contraindi- cated drugs include butorphanol (may increase CSF pressure) and acepromazine (lowers the seizure threshold).

• Neurosteroids: their function and role in NMS Recent developments in perinatal endocrinology have indicated that neurosteroids are inte- gral to the transition to extrauterine life (Diesch, 2013). These progestagen compounds cross the blood-brain barrier and have neuromodulatory effects, inducing a syndrome of depression com- patible with NMS but in the absence of hypoxic-ischemic insult. This syndrome has been exper- imentally reproduced in healthy foals by the administration of allopregnanolone (Madigan, 2012). Treatment of the reversion to fetal consciousness has been reported by the use of a ‘squeeze technique’, whereby the foal is restrained by ropes applying pressure to the mid-thorax region for a time period sufficient to mimic vaginal delivery (Madigan and ALEMAN, M. R., 2014;Toth, 2012).

Reference List Anderson DK, Waters TR and Means ED. Pretreatment with alpha tocopherol enhances neurologic recov- ery after experimental spinal cord compression injury. J Neurotrauma 1988;5(1):61-7. Arango MF and Mejia-Mantilla JH. Magnesium for acute traumatic brain injury. Cochrane Database Syst Rev 2006;(4):CD005400. Arundine M and Tymianski M. Molecular mechanisms of glutamate-dependent neurodegeneration in isch- emia and traumatic brain injury. Cell Mol Life Sci 2004;61(6):657-68. Bazan NG, Rodriguez de Turco EB and Allan G. Mediators of injury in neurotrauma: intracellular signal transduction and gene expression. J Neurotrauma 1995;12(5):791-814. Chesnut RM, Marshall LF, Klauber MR, et al. The role of secondary brain injury in determining outcome from severe head injury. J Trauma 1993;34(2):216-22. Clifton GL, Miller ER, Choi SC, et al. Fluid thresholds and outcome from severe brain injury. Crit Care Med 2002;30(4):739-45.

7 Diesch TJ and Mellor DJ. Birth transitions: Pathophysiology, the onset of consciousness and possible impli- cations for neonatal maladjustment syndrome in the foal. Equine Vet J 2013;45(6):656-60.

Edwards AD, Yue X, Cox P, et al. Apoptosis in the brains of infants suffering intrauterine cerebral injury. Pediatr Res 1997;42(5):684-9.

Eklind S, Mallard C, Leverin AL, et al. Bacterial endotoxin sensitizes the immature brain to hypoxic--isch- aemic injury. Eur J Neurosci 2001;13(6):1101-6.

Feldman Z, Kanter MJ, Robertson CS, et al. Effect of head elevation on intracranial pressure, cerebral perfusion pressure, and cerebral blood flow in head-injured patients. Journal of Neurosurgery 1992;76(2): 207-11.

Green S, Mayhew IG. Neurologic disorders. In: Koterba A, Drummond W, Kosch P, editors. Equine Clinical Neonatology. 1 ed. Philadelphia: Lea and Febiger; 1990. p. 496-530.

Grow J and Barks JD. Pathogenesis of hypoxic-ischemic cerebral injury in the term infant: current concepts. Clin Perinatol 2002;29(4):585-602.

Lipton SA and Rosenberg PA. Excitatory amino acids as a final common pathway for neurologic disorders. N Engl J Med 1994;330(9):613-22.

Madigan JE, Aleman MR, inventors. Methods of diagnosing and treating neonatal reversion to fetal con- sciousness. 2014; US 20140213563 A1.

Madigan JE, Haggett EF, Pickles KJ, et al. Allopregnanolone infusion induced neurobehavioural alterations in a neonatal foal: Is this a clue to the pathogenesis of neonatal maladjustment syndrome? Equine Vet J 2012;44:109-12.

Nilsson P, Laursen H, Hillered L, et al. Calcium movements in traumatic brain injury: the role of glutamate receptor-operated ion channels. J Cereb Blood Flow Metab 1996;16(2):262-70.

Palmer AC and Rossdale PD. Neuropathology of the convulsive foal syndrome. J Reprod Fertil Suppl 1975;(23):691-4.

Toth B, Aleman M, Brosnan RJ, et al. Evaluation of squeeze-induced somnolence in neonatal foals. American journal of veterinary research 2012;73(12):1881-9.

van de Beek D, de Gans J, McIntyre P, et al. Corticosteroids for acute bacterial meningitis. Cochrane Data- base Syst Rev 2007;(1):CD004405.

Willoughby RE, Jr. and Nelson KB. Chorioamnionitis and brain injury. Clin Perinatol 2002;29(4):603-21.

Yager JY, Brucklacher RM and Vannucci RC. Cerebral energy metabolism during hypoxia-ischemia and early recovery in immature rats. Am J Physiol 1992;262(3 Pt 2):H672-H677.

8 Sedation

Xylazine 0.02-1 mg/kg IV or IM Short acting, may promote seizures Detomidine 0.005-0.04 mg/kg IV or IM Long acting Anti-inflammatories Flunixin 0.5-1 mg/kg IV q12h Ketoprofen 1-2 mg/kg IV q12h Firocoxxib 0.1 mg/kg IV q24h Dimethyl sulfoxide 1 g/kg IV q12h Administer as 10% solution

Seizure control medications Diazepam 0.05-0.4 mg/kg IV Short-acting. Midazolam 0.2 mg/kg bolus IV Maintain with 0.1-0.2 mg/kg/h constant rate infusion. 4-10 mg/kg IV over 15 min Long-acting. Maintain long term 5-10 mg/kg PO bid Pentobarbital 3-5 mg/kg IV May induce severe respiratory depression. Use only when non-re sponsive to other agents. Potassium bromide 10 mg/kg PO q 8 h. Useful adjunct therapy or may be used as the sole agent for long- term management.

Control cerebral edema Hypertonic saline Up to 7 ml/kg IV q12h IV as 3% solution Mannitol 1 mg/kg iv q12h IV as 20% solution

Miscellaneous Magnesium sulfate 0.05 mg/kg IV over 30 min. Control excitotoxic glutamate neurotransmitter cascade. Vitamin C 100 mg/kg IV q12h Anti-oxidant Vitamin E 10-20u/kg PO q24h Lipid peroxidation Thiamine 10 mg/kg iv q24h 9 Update on Anaplasmosis – Made No Better by a Name Change. Peter R. Morresey BVSc MACVSc DipACT DipACVIM (LA) Rood and Riddle Equine Hospital, PO Box 12070, Lexington, KY 40580

Introduction Formerly called equine granulocytic ehrlichiosis, anaplasmosis (Anaplasma phagocytophi- lum) is an infectious disease with a seasonal occurrence. While predominantly seen in northern California, cases have been reported in numerous states and other countries. The disease is known to be tick-borne and not transmissible between horses and humans although the same bacterial strains are responsible for the infection. This talk will summarize manifestation, diagno- sis and treatment of this condition.

Epidemiology The first case of equine granulocytic anaplasmosis was described in California in 1969 (Grib- ble, 1969). Anaplasma phagocytophilum frequently infects horses in North America (Madi- gan, 2000) within the range of its tick vector (Ixodes sp). Areas include the northeast, midwest, northwest and southeast. Infection has also been documented in Canada, Scandinavia, Europe (Dzi-Ögiel, 2013), Great Britain, North Africa (M’ghirbi, 2012), and South America. A seasonal occurrence is noted: late fall through spring. As no effective transovarial transmission has been shown, a reservoir animal is necessary for the maintenance in nature (Woldehiwet, 2006). In California, knowledge of natural reservoirs of A. phagocytophilum is limited. Immature I. pacificus commonly feed on small reptiles and mammals, birds, deer and wild carnivore. Once adult, I. pacificus infests dogs, coyotes, horses, deer, wild mammals, several rodent species and cattle predominantly. Persistent bacteremia, particularly during the summer may facilitate maintenance reservoir of A. phagocytophilum in California. Detection of concurrent Bartonella henselae, Borrelia burgdorferi, and A. phagocytophilum infection of I. pacificus ticks has been demonstrated in California (Holden, 2006). In the eastern US, A. phagocytophilum has been documented in mice, chipmunks, and voles (Telford, 1996;Tyzzer, 1938;Walls, 1997), however the most significant reservoir forA. phagocy- tophilum is the white-footed mouse with a seroprevalence ranging between 1-50% (Nicholson, 1998;Telford, 1996). Severity of clinical disease of A. phagocytophilum varies widely between locations. In the United States, human disease manifestations are more severe when compared to disease in Eu- rope where it is relatively mild. However in the US cattle disease is rare yet cattle and sheep in Europe have significant signs of disease (Woldehiwet, 2006). Cross species reactivity occurs. Seroconversion in horses with the absence of disease has been documented following inoculation with the bovine variant of A. phagocytophilum (Pusterla, 1998). Experimental infection with the human adapted strain however caused clinical disease indistinguishable from natural infection in one study (Barlough, 1995), and was fatal to one horse in another study (Franzen, 2007). Persistence of A. phagocytophilum has been detected in the bloodstream of horses following experimental infection. This was unrelated to the occurrence of clinical disease, suggesting per- sistence may occur with natural infection during an acute outbreak (Franzen, 2009). Cyclic bac- teremia has also been documented following experimental infection of lambs (Granquist, 2010).

10 Pathogenesis In the horse, A. phagocytophilum infection has an incubation period of approximately 10 days (range 1-3 weeks) post introduction via a tick-bite (Dzi-Ögiel, 2013). The pathogenesis of disease is poorly understood. Once inoculated through the dermis by the bite of a tick, lymphatic spread or dissemination via the bloodstream is likely. Thereafter cells of the lymphoreticular and hemopoietic systems are invaded leading to the characteristic clinical signs. Replication occurs within vacuoles of professional phagocytes. It is thought that following invasion of the liver, spleen, lungs and other organs, localized inflammatory reactions are initiated and inflammatory cell ac- cumulation is promoted. Inflammatory mediators IL-1ß, TNF-a, and IL-8 are thought responsible (Kim, 2002). Pancytopenia is thought to result from peripheral sequestration, consumption and lysis of cellular blood constituents. Host immunological defense dysfunction or suppression appears to result from A. phagocy- tophilium infection. Affected horses can develop a wide range of opportunistic infections with bacteria, fungi and viruses. Both humoral and cell-mediated immunity are affected, with phago- cytic and migratory functional deficits present.

Clinical disease Clinical signs initially include fever (101.5-106°F) and mild depression. As disease progress- es, fever becomes undulating (101.5-106°F). Anorexia may occur. Petechiation and ecchymoses can occur. Edema develops in the subcutaneous tissue and fascia. Regional vasculitis is com- mon, with the limb subcutaneous tissue and fascia predominantly affected. This leads to reluc- tance to move. In many cases ataxia develops. Icterus is often seen. Cardiac arrhythmias (ven- tricular tachycardia and premature ventricular contractions) may be heard. A transient systolic heart murmur has been reported in experimentally infected horses, being ascribed to physiologic turbulence (Franzen, 2005;Madigan, 1993).

Subclinical infections are frequent, therefore many A. phagocytophilum infections are likely undetected, this being exacerbated by many of the symptoms appearing rather nonspecific for any particular disease (Madigan, 2000).

Severity of disease and duration of clinical signs appears age related. In adults, disease progresses with signs characteristic of disease: fever, anorexia, edema, reluctance to move and ataxia. Horses under 4 years of age develop milder disease with signs similar to older horses, while those under 1 year of age may present with only fever.

Neither laminitis nor abortion have been reported subsequent to A. phagocytophilium infec- tion, in contrast to many differential diagnoses.

Differential diagnosis As the clinical signs of anaplasmosis are many and varied in the horse, a number of differential diagnoses need to be considered as they can result in more serious pathology should they not be ruled out by physical or laboratory examination: • Borreliosis: Lyme disease can also present with vague signs of fever, depression and lame- ness. Vector range is similar to anaplasmosis.

11 • Encephalitis (Equine viral encephalitides): Acute onset of high fever, ataxia and severe depression. • Purpura hemorrhagica: High fever, vasculitis, limb edema, petechiation and laminitis potential. • Equine infectious anemia: High fever, depression, anemia, and petechiation. • Equine viral arteritis: High fever, limb edema and depression. • Potomac Horse Fever (Neorickettsia risticii): Similar vector range. High fever and malaise, with diarrhea and usually profound leukopenia. Rapid onset of laminitis. • Equine protozoal myeloencephalitis Insidious or sometimes rapid onset of ataxia. • Equine herpesvirus-1: High fever, depression and ataxia. • West Nile virus: High fever, depression, muscle fasciculations and ataxia. • Liver disease: Icterus, moderate to severe depression, anorexia, weakness, ataxia, edema, hemorrhage, diarrhea and anemia. Leptospirosis: Fever, depression and multiple organ dysfunction may result.

Diagnosis Anaplasmosis presents with a range of non-specific signs, however fever, anemia, icterus, a reluctance to move and swollen legs are generally seen, and highly suggestive in areas known to be prone to infection. Laboratory findings include thrombocytopenia, anemia and leucopenia. These abnormal- ities have been shown both in natural and experimental infections (Bermann, 2002;Franzen, 2009;Reubel, 1998). The diagnosis is made by detection of typical inclusions (morulae) in neutrophil granulo- cytes and from EDTA-blood by PCR or later by antibody seroconversion with an IFA test. It can be detected in buffy coat blood smears early in the course of clinical infection and illness as morula in neutrophils, with increasing numbers visible in a Giemsa-stained blood smear after the first few days of pyrexia (peak ehrlichemia days 3-5). Between 0.5-16% of neutrophils may be affected, appearing 2.6±1.5 days post onset of fever (Franzen, 2005). Detection of A. phagocytophilum DNA by PCR of EDTA whole blood samples (EDTA blood) is consistently positive 2-3 days before appearance of clinical signs, and persists 4-9 days following resolution of clinical signs. The PCR has been shown to be much more sensitive than microscopic investigation (Franzen, 2005). This is especially useful both early and late in the course of infection where the number of morulae are low and make cytological diagnosis diffi- cult. Serology is of marginal utility as in the acute phase of the infection horses are usually sero- negative (Franzen, 2005). Isolated serum samples do not differentiate between current or past infection. Experimentally, seroconversion has been demonstrated between 12-16 days post inoculation. Maximum IFA titers are found within 3-7 days subsequent to the onset of positivity (Franzen, 2005). A fourfold increase in titer confirms recent exposure. A positive IFAT persists for up to 300d post inoculation with the organism (Nyindo, 1978).

Treatment The administration of oxytetracycline (7mg/kg iv q 24h) is an effective treatment (Madigan, 1993), However, experimental infection has revealed some horses can make a full recovery without treatment (Franzen, 2009). This may account for the relatively high seropositivity re-

12 ported in European studies (Hansen, 2010;Passamonti, 2010). Seroprevalence between horses with or without likelihood of vector-borne diseases was similar, indicating subclinical infections were likely to be commonplace (Passamonti, 2010). Prognosis Complete recovery from clinical signs of disease, including neurological symptoms, occurs within 2 to 3 weeks of onset (Franzen, 2005;Madigan, 1993). Recovered horses are solidly immune for in excess of two years and are not believed to develop a carrier state. Persistence of infection has been suggested with some European strains, but further verification is required. Tick control measures are mandatory for control of disease. There is no vaccine.

Reference List

Barlough JE, Madigan JE, DeRock E, et al. Protection against Ehrlichia equi is conferred by prior infection with the human granulocytotropic Ehrlichia (HGE agent). J Clin Microbiol 1995;33(12):3333-4.

Bermann F, Davoust B, Fournier PE, et al. Ehrlichia equi (Anaplasma phagocytophila) infection in an adult horse in France. Vet Rec 2002;150(25):787-8.

Dzi-Ögiel B, Adaszek L, Kalinowski M, et al. Equine granulocytic anaplasmosis. Research in Veterinary Science 2013;95(2):316-20.

Franzen P, Aspan A, Egenvall A, et al. Acute clinical, hematologic, serologic, and polymerase chain reac- tion findings in horses experimentally infected with a European strain ofAnaplasma phagocytophilum. J Vet Intern Med 2005;19(2):232-9.

Franzen P, Aspan A, Egenvall A, et al. Molecular evidence for persistence of Anaplasma phagocytophilum in the absence of clinical abnormalities in horses after recovery from acute experimental infection. J Vet Intern Med 2009;23(3):636-42.

Franzen P, Berg AL, Aspan A, et al. Death of a horse infected experimentally with Anaplasma phagocy- tophilum. Vet Rec 2007;160(4):122-5.

Granquist EG, Bårdsen K, Bergström K, et al. Variant -and individual dependent nature of persistent Ana- plasma phagocytophilum infection. Acta Vet Scand 2010;52(1):25.

Gribble DH. Equine ehrlichiosis. J Am Vet Med Assoc 1969;155(2):462-9.

Hansen M, Christoffersen M, Thuesen L, et al. Seroprevalence of Borrelia burgdorferi sensu lato and Anaplasma phagocytophilum in Danish horses. Acta Vet Scand 2010;52(1):1-6.

Holden K, Boothby JT, Kasten RW, et al. Co-detection of Bartonella henselae, Borrelia burgdorferi, and Anaplasma phagocytophilum in Ixodes pacificus Ticks from California, USA. Vector-Borne and Zoonotic Diseases 2006;6(1):99-102.

Kim HY, Mott J, Zhi N, et al. Cytokine gene expression by peripheral blood leukocytes in horses ex- perimentally infected with Anaplasma phagocytophila. Clinical and diagnostic laboratory immunology 2002;9(5):1079-84.

Head Shakers – Is There Anything New Under The Sun?

13 Head Shakers – Is There Anything New Under The Sun? Peter R. Morresey BVSc MACVSc DipACT DipACVIM (LA) Rood and Riddle Equine Hospital, PO Box 12070, Lexington, KY 40580

Introduction Head shaking in horses is a syndrome of many alleged causes and numerous touted treat- ments. It is a source of frustration to owners, trainers and veterinarians alike. Often a diagnosis of exclusion, a systematic and comprehensive work-up is necessary to eliminate treatable organic disease that may manifest as shaking of the head. Contemporary theories on pathophysiology, diagnostic approaches, and current treatment of head shaking are discussed. Signalment The mean age at onset (or recognition) of headshaking has been reported in various studies, both clinical and owner observational, as 7.3, 7.5, and 9 years (Lane, 1987;Madigan, 2001;Mills, 2002). in these studies were over-represented (63–78%) however breed predilection was detected. In the populations studied, many affected horses were acquired by the current owner in late autumn or winter when headshaking was not present or apparent to the new owner (Lane, 1987;Madigan, 2001). Clinical syndrome Headshaking syndrome is commonly seen during exercise, particularly at a trot when the head is held relatively stationary. However, it can also occur at rest or during other gaits. Affect- ed horses display a characteristic headshaking or head-tossing in a vertical plane. Other signs include snorting, rapid movements of the upper lip, nasal discharge, lacrimation, and rubbing or striking the nares or side of the face with a foreleg or fixed object. The majority of affected horses have a seasonal worsening of signs. Onset occurs in spring with signs continuing until late summer or autumn. This suggests photoperiod associated neuro- humoral changes, changes in temperature and humidity, allergen exposure or unknown environ- mental factors contribute to the onset of the headshaking syndrome. Pathophysiology of head shaking Shaking of the equine head can be associated with frustration or anticipation of activity. It is also a response to noxious stimuli – insects, poorly fitting tack and restraint. While there is a long list of potential causes of headshaking, many are not amenable to treatment. Those amenable to medical or surgical treatment in include temporohyoid osteoarthropathy, and allergic reactions. Other causes include hormonal imbalances and vascular changes. Temporohyoid osteoarthropathy (THO) Headshaking with or without deficits of the facial and/or vestibular nerves has been attributed to bony proliferation of the temporohyoid joint and proximal shaft of the stylohyoid bone. While some reports indicate an infectious etiology (recovery of bacteria via tympanocentesis or nec- ropsy (Blythe, 1997;Power, 1983), others have not (Naylor, 2010). The predilection for Quarter horses, Paints and Appaloosas also speaks against an infectious etiology. Affected horses are most commonly presented when neurological deficits develop. Bony proliferation leads to impingement of either or both of the facial and vestibular nerves in the area of the acoustic meatus. The onset of neurologic signs may be acute (vestibular ataxia, facial

14 nerve deficits), chronic (persistent corneal ulceration resulting from diminished tear production or blinking), or initially subtle but progressive (as with the onset of a head tilt, trouble with the , or difficulty chewing) (Blythe, 1997;Walker, 2002). Trigeminal neuralgia/neuropathy (TN) In the horse, TN triggers include bright light or sunshine (photic headshaking), touch, auditory and olfactory stimuli (Madigan, 1995;Madigan, 2001;Mills, 2002). Also, exercise-induced increas- es in airflow through the nasal passages, blood flow to nasal and turbinate mucosa, and maxillary arterial pulse profile (Newton, 2000). As a cause of headshaking, TN was first considered over a century ago (1899) following suc- cessful treatment by infraorbital neurectomy (Williams, 1899). Further supporting evidence has been provided more recently, as perineural anesthesia of branches of the trigeminal nerve has led to transient improvement, with lasting improvement reported following neurectomy, chemical sclerosis, or coil compression of the infraorbital or maxillary nerves (Mair, 1999;Mair, 1992;New- ton, 2000;Roberts, 2009;Roberts, 2013). Paralleling TN as a cause of headshaking in horses, TN in humans can cause a shooting or burning sensation across the face. The maxillary branch is most often affected with right-sided disease more prevalent (considered due to a narrower right foramen rotundum and foramen ovale through which the nerve passes). In people with classical TN (parallels idiopathic headshaking in horses) is partial demyelination of the trigeminal nerve is the only pathological finding, and is thought to be associated with vascular compression. It is considered that the resulting demyelination disturbs trigeminal nerve electrophysiology, resulting in ectopic electrical impulses that precipitate facial pain (Zakrzewska, 2002). Histopathology of trigeminal nerve, the ganglion and several branches from headshaking horses has not revealed demyelination or other pathology (Newton, 2001). However, increased excitability of the nerve has been demonstrated following stimulation of the trigeminal nerve in four horses with headshak- ing syndrome (Pickles, 2011b). Trigeminal ganglion infection with latent EHV-1 has been investigated as a cause of altered trigeminal nerve function. However, latent EHV-1 as detected by real-time PCR, was recovered from only one of eight geldings with headshaking syndrome (Aleman, 2012). Luteinizing hormone (LH) Headshaking syndrome is observed most frequently in geldings and displays a seasonal oc- currence. Increased activity of gonadotropin releasing hormone (GnRH) leading to increased cir- culating concentrations of luteinizing hormone (LH) and follicle-stimulating hormone (FSH), could lead to seasonal changes in trigeminal nerve excitability in affected geldings (Pickles, 2011a). However, GnRH vaccine was administered to geldings with headshaking syndrome decreased LH and FSH concentrations but did not ameliorate signs of headshaking (Pickles, 2011a). Allergy As headshaking has a seasonal prevalence, it has been hypothesized that headshaking syn- drome may be initiated by allergic reactions (LANE, 1987). Cough (27% of cases evaluated) and excessive tracheal mucus (32% of horses examined by airway endoscopy) in keeping with recurrent airway obstruction (RAO or ‘heaves’). Allergic associated with these conditions in humans, affecting up to 30% of adults and 40% of children, may trigger headshaking syndrome. Serous nasal secretions, congestion, sneezing, and airflow obstruction present in humans with al- lergic rhinitis can be associated with facial pain. However investigation in horses (nasal mucosal biopsies, gross and microscopic examination of the head and nasal passages), of headshakers

15 has not supported this hypothesis. Vasomotor rhinitis Non-allergic rhinopathy (vasomotor rhinitis) has been documented to cause headshaking syn- drome (Lane, 1987;McGorum, 1990). Being non-seasonal and non-allergic in origin, it is consid- ered that altered autonomic control of nasal mucosal blood flow and secretions precipitates the condition. With increased airflow during exercise, clinical signs that are indistinguishable from allergic rhinitis may develop. Vasoconstrictive drugs (i.e. decongestants) and intranasal cortico- steroids can alleviate signs. Photic headshaking In humans, photic stimulation via the optic nerve leads to a tickling sensation in the nasal mucosa (Whitman, 1993). This may be the result of light stimulation of the optic nerves causing activation of the maxillary branch of the trigeminal nerve, or regional parasympathetic branches (Everett, 1964). It has been postulated that the photic trigger is present in some horses with head- shaking similar to this human photic sneeze (Madigan, 1995). Clinical signs of photic headshak- ers are indistinguishable from those of non-photic headshakers (Madigan, 2001;Newton, 2000). This suggests multiple triggers activate the same final effector trigeminal response. Such stimuli include sound (metal sound, clap), eating (hard carrots, fibrous hay), and nasal mucosal stimula- tion via diagnostic nasal swabbing (Aleman, 2014).

Diagnosis History and characteristics of headshaking A detailed history should be taken from the owner. This can include management procedures, diet, exercise and environmental changes. When investigating idiopathic headshaking following exclusion of other diseases, the characteristic head motion (rapid downward motion of the nose followed by upward flinging of the head) should be present. Physical examination In addition to a complete general examination which includes the oral cavity, ophthalmic and otoscopic examinations are needed. Endoscopy of the upper airway, including the guttural pouch- es, and radiography of the head and throatlatch region are indicated. Computed tomography (CT) and magnetic resonance imaging (MRI) improve visualization of both bone and soft tissue. Local anesthesia of the infraorbital nerve Used as an aid to diagnosis but reported success is low. Infraorbital anesthesia with 2% mepivacaine improved 3/19, had no effect on 8/19, and worsened 8/19 horses (Mair, 1999). In another study only 1/8 horses improved and only by 50% however the volume of mepivacaine was lower (Newton, 2000). A higher volume of local anesthetic infiltration may however affect adjacent nerves. Bilateral anesthesia of the posterior ethmoidal nerve (maxillary nerve) improved 13/17 horses (Newton, 2000) and 23/27 horses (Roberts, 2013) with headshaking. However the location of the nerve requires an extended period for diffusion of the anesthetic to achieve a full effect (Aleman, 2014;Newton, 2000).

16 Treatment Headshaking may be a response to a noxious stimulus, whether physiological or behavioral. Removal of the source of discomfort or treatment of the underlying organic disease is necessary in all cases. However, since the etiology of idiopathic headshaking is unknown, there are no spe- cific or demonstrably curative treatments currently. The basis of control is therefore management (where possible) and not cure. Currently reported treatments have a low success rate, and do not correct the underlying abnormal trigeminal neurophysiology. As some horses have been noted to go into spontaneous remission, it seems possible that the aberrant trigeminal nerve physiology and activity might be reversible. Ameliorating the disturbed nerve function appears to hold promise as the key to successful treatment. Environmental management to avoid established precipitating stimuli is central to control. This may involve riding at night or in low light situations, but this is impractical especially for com- petition horses. Nose nets and face masks have enjoyed some success, with up to 75% of own- ers reporting measurable improvement with appliances ranging from full coverage of the muzzle and lips, to coverage of only the nostrils and upper lip (Mills, 2003). Medication usage is commonplace, with a number of compounds reported to be at least par- tially effective in reducing severity of presentation (Table 1). As with all pharmaceutical usage, deleterious effects and tendency to overuse are of concern, as is violation of regulations relating to competition withdrawal times. Surgical approaches have been reported. Infraorbital neurectomy first appeared in the veter- inary literature in the late 1800s (Williams, 1899). Response is minimal and a high post-surgical complication rate suggests this procedure is contraindicated (Mair, 1999). Chemical sclerosis of the posterior ethmoidal nerve similarly suffers from a lack of efficacy with a high rate of reoccur- rence reported (Newton, 2000). The placement of platinum coils to compress the caudal infra- orbital nerve has been reported used in horses unresponsive to medical therapy, with up to 50% success achieved however multiple surgeries were required in some horses due to repeated re- occurrence of headshaking (Roberts, 2009;Roberts, 2013). Due to post-operative complications including increased severity of nose rubbing progressing to self-trauma, surgical interventions should be considered salvage procedures for horses non-responsive to other modalities and at risk of euthanasia.

17 Table 1

Treatment Dose/frequency Comments

Cyproheptadine 0.3mg/kg po q12h Moderate improvement. Lethargy

Carbamazepine 2-8mg/kg po q6h-q12h Moderate improvement. (may be used in combination Unpredictable efficacy with ).

Hydroxyzine 1mg/kg po q12h Moderate improvement.

Fluphenazine 50mg im. Repeat 1-4 monthly Neurological dysfunction.

Phenobarbital 3-6mg/kg po q12h Calmative. Sedation.

Gabapentin 5-20 mg/kg po q12h-q24h Anecdotal reports, variable.

Corticosteroids Standard dosage, also Variable success pulse therapy reported.

Magnesium Variable Calmative.

Melatonin 15-18mg po q24h Dose 5pm. Altered hair shedding.

Sodium cromoglycate eye drops Apply OU q6h Seasonal head shakers.

Adapted from: (Madigan, 2001) (Mair, 1999) (Madigan, 1995) (Newton, 2000) (Pickles, 2014) (Stalin, 2008) (Tomlinson, 2013)

Reference List

Aleman M, Pickles KJ, Simonek G, et al. Latent Equine Herpesvirus-1 in Trigeminal Ganglia and Equine Idiopathic Headshaking. J Vet Intern Med 2012;26(1):192-4.

18 Aleman M, Rhodes D, Williams DC, et al. Sensory Evoked Potentials of the Trigeminal Nerve for the Diag- nosis of Idiopathic Headshaking in a Horse. J Vet Intern Med 2014;28(1):250-3.

Blythe LL. Otitis media and interna and temporohyoid osteoarthropathy. The Veterinary clinics of North America Equine practice 1997;13(1):21-42.

Everett HC. Sneezing in response to light. Neurology 1964;14(5):483.

Lane JG and Mair TS. Observations on headshaking in the horse. Equine Vet J 1987;19(4):331-6.

Madigan JE, Kortz G, Murphy C, et al. Photic headshaking in the horse: 7 cases. Equine Vet J 1995;27(4):306- 11.

Madigan JE and Bell SA. Owner survey of headshaking in horses. Journal of the American Veterinary Medical Association 2001;219(3):334-7.

Mair TS. Assessment of bilateral infra-orbital nerve blockade and bilateral infra-orbital neurectomy in the investigation and treatment of idiopathic headshaking. Equine Vet J 1999;31(3):262-4.

Mair TS, Howarth S and Lane JG. Evaluation of some prophylactic therapies for the idiopathic headshaker syndrome. Equine Vet J 1992;24(S11):10-2.

McGorum BC and Dixon PM. Vasomotor rhinitis with headshaking in a pony. Equine Vet J 1990;22(3):220-2.

Mills DS, Cook S, Taylor K, et al. Analysis of the variations in clinical signs shown by 254 cases of equine headshaking. The Veterinary record 2002;150(8):236-40.

Mills DS and Taylor K. Field study of the efficacy of three types of nose net for the treatment of headshaking in horses. The Veterinary record 2003;152(2):41-4.

Naylor RJ, Perkins JD, Allen S, et al. Histopathology and computed tomography of age-associated degeneration of the equine temporohyoid joint. Equine Vet J 2010;42(5):425-30.

Newton SA. The fuctional anatomy of the trigeminal nerve of the horse University of Liverpool; 2001.

Newton SA, Knottenbelt DC and Eldridge PR. Headshaking in horses: possible aetiopathogenesis suggested by the results of diagnostic tests and several treatment regimes used in 20 cases. Equine Vet J 2000;32(3):208-16.

Pickles KJ, Berger J, Davies R, et al. Use of a gonadotrophin-releasing hormone vaccine in headshaking horses. Veterinary Record 2011a;168(1):19.

Pickles KJ, Gibson TJ, Johnson CB, et al. Preliminary investigation of somatosensory evoked potentials in equine headshaking. The Veterinary record 2011b;168(19):511.

Pickles K, Madigan J and Aleman M. Idiopathic headshaking: Is it still idiopathic? The Veterinary Journal 2014;201(1):21-30.

Power HT, Watrous BJ and de Lahunta A. Facial and vestibulocochlear nerve disease in six horses. Journal of the American Veterinary Medical Association 1983;183(10):1076-80.

Roberts VLH, Mckane SA, Williams A, et al. Caudal compression of the infraorbital nerve: A novel surgical technique for treatment of idiopathic headshaking and assessment of its efficacy in 24 horses. Equine Vet J 2009;41(2):165-70.

19 Roberts VLH, Perkins JD, Skärlina E, et al. Caudal anaesthesia of the infraorbital nerve for diagnosis of idiopathic headshaking and caudal compression of the infraorbital nerve for its treatment, in 58 horses. Equine Vet J 2013;45(1):107-10.

Stalin CE, Boydell IP and Pike RE. Treatment of seasonal headshaking in three horses with sodium cromoglycate eye drops. The Veterinary record 2008;163(10):305-6.

Tomlinson JE, Neff P, Boston RC, et al. Treatment of Idiopathic Headshaking in Horses with Pulsed High-Dose . J Vet Intern Med 2013;27(6):1551-4.

Walker AM, Sellon DC, Comelisse CJ, et al. Temporohyoid Osteoarthropathy in 33 Horses (1993-2000). J Vet Intern Med 2002;16(6):697-703.

Whitman BW and Packer RJ. The photic sneeze reflex Literature review and discussion. Neurology 1993;43(5):868.

Williams LW. Involuntary shaking of the head and its treatment by trifacial neurectomy. American Veterinary Review 1899;23:321-6.

Zakrzewska JM. Diagnosis and Differential Diagnosis of Trigeminal Neuralgia. The Clinical Journal of Pain 2002;18(1).

20 Managing Strangles Outbreaks Ashley G. Boyle, DVM, DACVIM Assistant Professor of Medicine, Section Field Service Department of Clinical Studies, New Bolton Center, University of Pennsylvania 382 West Street Rd. Kennett Square, PA [email protected] Pathophysiology

S. equi subsp equi is inhaled or ingested after direct contact with mucopurulent discharge from infected horses or contaminated equipment. The bacterium attaches to the crypts and epithelium of the lingual and palatine tonsils. The organism enters the mandibular and pharyngeal lymph nodes. Clinical signs develop 3 to 14 days after exposure. 1,2 Clinical Signs

The first clinical sign of strangles is acute-onset fever (often >103°F), followed by lethargy, depression, bilateral mucopurulent nasal discharge, lymphadenopathy, and abscessation of the retropharyngeal and mandibular lymph nodes. Occasionally, the parotid and cranial cervical lymph nodes are affected. Retropharyngeal lymph node enlargement can lead to narrowing of the pharynx, resulting in upper respiratory noise, dysphagia, and neck extension. Empyema results when the retropharyngeal lymph nodes drain pus into the guttural pouches. Horses may develop respiratory distress due to retropharyngeal abscesses that are not externally mature. Clinical signs are more severe in immunologically naïve (1 through 5 years of age), geriatric (older than 20 years), and immunocompromised horses.1,2,3 Some horses may develop complications such as metastatic abscessation, purpura hemorrhagica, and myositis. Transmission

Shedding of S. equi subsp equi begins 2 to 3 days after onset of fever. In most cases, shedding persists for a minimum of 2 to 3 weeks. Horses that have recovered from strangles have been shown to shed for an additional 6 weeks.1 If organisms are harbored in the guttural pouches, horses have been shown to shed S. equi subsp equi for up to 2.5 years. These outwardly healthy horses (i.e., carriers) that still shed organisms are a source of infection when introduced into a new population of horses.6,7,8 Transmission occurs through nose-to-nose contact; proximity; equipment (e.g., water buckets, feed buckets, tack, twitches); clothing; and equipment of owners, caretakers, farriers, and veterinarians.1 Under laboratory conditions, S. equi subsp equi has been shown to persist on wood for 63 days at 35.6°F and for 48 days on glass and wood at 68°F.9 One study modeling field conditions revealed that the organism was found to persist for less than 4 days, 23 but moist environments (e.g., water buckets) allow the organism to persist for extended periods.1,2 Seventy-five percent of horses that have been infected with S. equi subsp equi and have not been treated with antimicrobials develop lasting immunity for approximately 5 years or longer. 1,2,10,11 Diagnostic Testing

Early definitive diagnosis is essential for containing this highly infectious disease. Cytologic eval- uation reveals gram-positive extracellular cocci in long chains supports a diagnosis of a ß-hemo-

21 lytic streptococcus organism. Historically, the gold standard for diagnosis is bacterial culture of abscess aspirates, nasopharyngeal swabs, nasopharyngeal revealing S. equi subsp equi. This is the preferred method on aspirates of mature abscesses, but takes a minimum of 24 hours to ob- tain results. In our clinical practice, we now use polymerase chain reaction (PCR) testing to detect the DNA of the organism as the gold standard which can also be performed on nasopharyngeal swab, nasopharyngeal wash, and guttural pouch wash samples.1 Nasopharyngeal washes are preferable to nasopharyngeal swabs due to a larger sampling area, but guttural pouch sampling is the most reliable, although more expensive and requires specialized equipment. PCR testing is more sensitive than bacterial culture22 and should always be used in combination. Treatment

The goal of treating strangles is to control transmission and eliminate infection while providing future immunity to the disease. Uncomplicated cases of strangles are often left to run their course with supportive care, providing lasting immunity. Affected horses should be isolated in a clean, dry stall and fed moist, palatable food. NSAIDs should be used judiciously to decrease swelling and promote eating. Hot compresses or topical 20% ichthammol can be used to accelerate maturation of abscessation. Mature external abscesses should be lanced to allow drainage, followed by daily lavage of open abscesses using dilute povidone iodine solution. This speeds resolution of absces- sation as well as alleviation of compression of the pharynx. 2 Methods Of Outbreak Control

Most outbreaks are thought to originate from introduction of an infected horse to a naïve popula- tion. All new horses should be isolated for 3 weeks and monitored for any signs of disease, includ- ing fever. If cost is not prohibitive, horses should be screened for S. equi subsp equi infection us- ing nasopharyngeal washes. Many farms with repeated infections have resorted to screening for infection via endoscopic evaluation and PCR testing of guttural pouch lavages. The Animal Health Trust in the United Kingdom recently developed a new serologic test that detects antigens differ- ently than the SeM ELISA. This new test appears to be more sensitive for detecting animals with recent exposure (as little as 2 weeks) to S. equi subsp equi and has been used as a screening tool to determine who needs endoscopic examinations upon arrival to a farm during quarantine prior to introduction into the herd. However, this test is currently not available in the United States and there is no way to distinguish vaccinated animals from recently exposed animals.15

Once an outbreak has occurred, twice-daily monitoring of rectal temperatures of all horses on the farm is essential to contain the outbreak. Because febrile horses do not shed disease for the initial 2 days, immediate identification of febrile horses enables caretakers to isolate these horses before shedding occurs. All movement of horses to and from the farm should be stopped until they are determined to be noninfectious. All equipment (e.g., pitchforks, buckets, grooming tools) for an affected horse should be isolated and used only for that horse. Personnel handling infect- ed horses should wear barrier precautions (i.e., gowns, gloves, plastic boots that cover shoes) and, ideally, should not handle noninfected horses or should handle infected horses last. Water buckets should be disinfected daily. 1,8 Facilities and equipment should be cleaned first to remove all organic material and then disinfected with phenols, iodophors, or chlorhexidine compounds or steam cleaned. 1,9 Surfaces and equipment must be allowed to dry thoroughly. Paddocks that hold infected horses should be rested for 4 weeks. A minimum of two weeks after all cases of strangles have resolved, one guttural pouch lavage along with an endoscopic examination for empyema

22 or chondroids should be obtained from convalescing horses and their contacts at approximately weekly intervals and tested for S. equi subsp equi via PCR to detect carriers. This is preferable to the previously recommended three nasopharyngeal washes. Horses have been found positive in their guttural pouches despite three negative nasopharyngeal washes. 16 A series of 3 nasopha- ryngeal swabs, collected 1 week apart, will result in detection by a positive culture on at least one of the swabs in approximately 60% of carrier animals. Concurrent testing of these swabs by PCR increases the likelihood of detection to over 90% of carrier animals. 15 For the purpose of efficien- cy, I recommend treating each guttural pouch with penicillin at the time of endoscopic examination unless there is gross contamination of the guttural pouch which would require aggressive lavage. A minimum of three weeks should be waited prior to retesting a treated, previously positive guttur- al pouch via PCR . The percentage of carriers per outbreak could be as high as 10%.1,8, 12,17,18 It is important to remember that SeM ELISA does not detect carrier status.19

The use of vaccination during an outbreak is controversial. The 2005 ACVIM Consensus State- ment recommends that live vaccine should be administered only to healthy animals with no known exposure to infected horses during an outbreak, but no published data show that use during an outbreak is detrimental.1 The AAEP Infectious Disease Committee does not recommend vacci- nation during an outbreak.13 It is suggested that horses recovering from infection should not be vaccinated for 1-2 years. 20

Eradication of this disease will not be possible until the subpopulation of carriers is eliminated and the development of new vaccines that distinguish vaccinates from exposed are available. 21

REFERENCES

1. Sweeney CR, Timoney JF, Newton JR, Hines MT. Streptococcus equi infections in horses: guidelines for treatment, control, and prevention of strangles. J Vet Intern Med 2005:19:123-134.

2. Boyle AG. Streptococcus equi subspecies equi Infection (Strangles) in horses. Compendium Continuing Education for the Practicing Veterinarian 2011; 33:online.

3. Sweeney CR, Benson CE, Whitlock RH, et al. Description of an epizootic and persistence of Streptococ- cus equi infections in horses. JAVMA 1989;9:1281-1286.

4. Ford J, Lokai MD. Complications of Streptococcus equi infection. Equine Pract 1980;4:41-44.

5. Radostits OM, Gay CC, Blood DC, et al. Purpura hemorrhagica. In: Veterinary Medicine: A Textbook of the Diseases of Cattle, Sheep, Pigs, Goats and Horses. 8th ed. London: Balliere Tindall; 1999:1713-1714.

6. Carlson GP. Diseases of the hematopoietic and hemolymphatic systems. In: Smith BP, ed. Large Animal Internal Medicine. 3rd ed. St. Louis: CV Mosby; 2002:1043.

7. Galan JE, Timoney JF. Immune complexes in purpura hemorrhagica of the horse contain IgA and M an- tigen of Streptococcus equi. J Immunol 1985;135:3134-3137.

8. Newton JR, Verheyen K, Talbot NC, et al. Control of strangles outbreaks by isolation of guttural pouch carriers identified using PCR and culture of Streptococcus equi. Equine etV J 2000;32:515-526.

9. Jorm LR. Laboratory studies on the survival of Streptococcus equi on surfaces. In: Plowright W, Ross- dale PD, Wade JF, eds. Proceedings of Equine infectious Diseases VI. Newmarket, UK: R & W Publica-

23 tions;1992:39-43.

10. Pusterla N, Watson JL, Affolter VK, et al. Purpura haemorrhagica in 53 horses. Vet Rec 2003;153:118- 121.

11. Freeman DE. Diagnosis and treatment of diseases of the guttural pouch (part 2). Compend Contin Educ Pract Vet 1980;2:S25-S31.

12. Newton JR, Wood JN, Dunn KA, et al. Naturally occurring persistent and asymptomatic infection of the guttural pouches of horses with Streptococcus equi. Vet Rec 1997;140:84-90.

13. AAEP. Strangles (Streptococcus equi). Accessed June 2009 at www.aaep.org/strangles.htm.

14. Waller A, Robinson C, Newton JR. Further thoughts on the eradication of strangles in equids. JAVMA 2007;231:1335.

15. http://www.aht.org.uk/cms-display/bact_bloodadvice.html Accessed January 2012.

16. Getting a grip on Strangles, Havermeyer Workshop, October 2012.

17. Newton JR, Wood JN, Dunn KA, et al. Naturally occurring persistent and asymptomatic infection of the guttural

pouches of horses with Streptococcus equi. Vet Rec 1997;140:84-90.

18. Verheyen K, Newton JR, Talbot NC, et al. Elimination of guttural pouch infection and inflammation in asymptomatic carriers of Streptococcus equi. Equine Vet J 2000;32:527-532.

19. Davidson A, Traub-Dargatz JL, Magnuson R, et al. Lack of correlation between antibody titers to fibrinogen binding protein of Streptococcus equi and persistent carriers of strangles. JVet Diagn Invest 2008;20:457-462.

20. Wilson JH. Vaccine efficacy and controversies. Proc 51st Annu Meet AAEP 2005;409-420.

21. Waller A, Robinson C, Newton JR. Further thoughts on the eradication of strangles in equids. JAVMA 2007;231:1335.

22. Boyle, AG, Boston RC, O’Shea K, Young S, Rankin SC, Optimization of an in vitro assay to detect Strep- tococcus equi subsp. equi. Vet. Microbiol. 159: 406-410, 2012.

23. Weese JS, Jarlot C, Morley PS, Survival of Streptococcus equi on surfaces in an outdoor environment. Can Vet J. 2009 Sep;50(9):968-70.

24. Boyle AG et al. J Am Vet Med Assoc 2009;235: 973.

24 Strangles- Unusual Cases and Effective Treatments Ashley G. Boyle, DVM, DACVIM Assistant Professor of Medicine, Section Field Service Department of Clinical Studies, New Bolton Center, University of Pennsylvania 382 West Street Rd. Kennett Square, PA [email protected] Usual Abscess Locations

S. equi subsp equi can infect any lymph node including and limited to periocular resulting in S. equi positive ocular discharge, parotid lymph nodes, and suprascapular. Bastard Strangles (Metastatic Abscessation)

In as many as 20% of cases, S. equi subsp equi spreads via hematogenous or lymphatic spread. This results in metastatic abscessation which can affect any organ system. Aspiration of muco- purulent discharge or hematogenous or lymphatic spread to the lungs can cause pneumonia. Abscessation in the mesentery, liver, spleen, and kidneys is common, leading to peritonitis and clinical signs of colic. Abscessation of the cranial mediastinal lymph nodes can cause tracheal compression and respiratory distress. Neurologic signs are present when abscessation occurs in the brain.1 The mortality rate of horses with strangles is less than 2%, but the presence of bastard strangles increases the mortality rate to as high as 62%. 2,4 Purpura Hemorrhagica

Purpura hemorrhagica is an aseptic necrotizing vasculitis that can occur in mature horses after repeated natural exposure to infection or after vaccination of horses that have had strangles.1,5,6,7 The actual incidence of this type III hypersensitivity response secondary to strangles or vaccina- tion is unknown. 1,2 Clinical signs range from mild to life-threatening, including pitting edema of the head, trunk, and distal limbs as well as petechiae and ecchymoses. In some cases, antigen– antibody complexes affect other sites, including the gastrointestinal tract, muscles, lungs, and kidneys. 1,2,4,5 Guttural Pouch Empyema And Carriers

Carriers can appear outwardly healthy but harbor S. equi subsp equi in their guttural pouches. De- finitive determination of carrier status requires endoscopic examination of the guttural pouches as well as culture and PCR testing of guttural pouch lavage fluid. 1,2,7,8 Often, if there is no external abscessation, there will be internal abscessation of the retropharyngeal lymph nodes. This can recognized early by painful palpation of the throat latch region and squeezing the trachea. These horses are at high risk for rupture into the guttural pouch and asphyxiation and often require pre- sentative tracheostomies. Any horse with guttural pouch empyema is also at risk for aspiration pneumonia. Serologic Testing

Serologic testing involves the use of ELISA to detect the SeM protein. Serology is useful for de- tecting recent, but not current, infection; assessing the need for vaccination; identifying horses that may be predisposed to purpura hemorrhagica; and diagnosing S. equi subsp equi–associat-

25 ed purpura hemorrhagica and metastatic abscessation.1,2,6

Various authors have suggested that horses with high serum SeM-specific antibody titers may be predisposed to developing purpura hemorrhagica when vaccinated against S equi.1 A 2009 study24 identified factors associated with an increased likelihood that horses would have SeM-spe- cific antibody titers ≥ 1:1,600. Results indicated that older horses, horses other than Thorough- breds and , and horses that had been vaccinated with an attenuated-live intranasal S equi vaccine between 1 and 3 years previously had an increased likelihood of having a serum SeM-specific antibody titer ≥ 1:1,600. We also found that 26/35 (74%) horses that had been vac- cinated with IN strangles when their SeM titer was equal to 1:1,600 did not have any complications associated with purpura.24 Therefore, the recommendation is to not vaccinate when the titer is ≥ 1: 3,200. Treatment

While the use of antimicrobials for treating strangles is controversial, horses with complications such as metastatic disease or purpura definitely require the use of systemic antimicrobials for ex- tended periods. In addition, horses with severely enlarged lymph nodes and dyspnea often require an emergency tracheostomy 1,2,3,7 and intensive supportive care. The preferred antimicrobial is penicillin (procaine penicillin [22,000 to 44,000 IU/kg IM q12h] or aqueous potassium penicillin [22,000 to 44,000 IU/kg IV q6h]). The use of antimicrobials during the acute phase of fever and depression may prevent abscess formation and decrease shedding but also the development of lasting immunity. Cases of purpura hemorrhagica also require the use of systemic corticosteroids (dexamethasone, 0.1 to 0.2 mg/kg IV or IM q12–24h; prednisolone, 0.5 to 1 mg/kg PO q24h) for an average of 3 weeks to reduce systemic vasculitis.2,10

Elimination of guttural pouch empyema requires repeated lavage with a solution of 20% acetylcys- teine in buffered saline via polyethylene tubing through an endoscope or via a chambers catheter. Chondroids are particularly difficult to remove, possibly requiring manual removal with endoscopic equipment such as a memory helical polyp retrieval basket, repeated lavage via indwelling cath- eters, or surgical removal.1,2,8,12,13 Successful elimination of S. equi subsp equi in these carriers requires local treatment of the guttural pouch with a penicillin gel after removal of the material within the guttural pouch.1,12,13 Repeated local treatment and systemic treatment with procaine penicillin or potassium penicillin for 7 to 10 days are necessary for refractory cases.2,8

References

1. Sweeney CR, Timoney JF, Newton JR, Hines MT. Streptococcus equi infections in horses: guidelines for treatment, control, and prevention of strangles. J Vet Intern Med 2005:19:123-134.

2. Boyle AG. Streptococcus equi subspecies equi Infection (Strangles) in horses. Compendium Continuing Education for the Practicing Veterinarian 2011; 33:online.

3. Sweeney CR, Benson CE, Whitlock RH, et al. Description of an epizootic and persistence of Streptococ- cus equi infections in horses. JAVMA 1989;9:1281-1286.

4. Ford J, Lokai MD. Complications of Streptococcus equi infection. Equine Pract 1980;4:41-44.

5. Radostits OM, Gay CC, Blood DC, et al. Purpura hemorrhagica. In: Veterinary Medicine: A Textbook of the Diseases of Cattle, Sheep, Pigs, Goats and Horses. 8th ed. London: Balliere Tindall; 1999:1713-1714.

26 6. Carlson GP. Diseases of the hematopoietic and hemolymphatic systems. In: Smith BP, ed. Large Animal Internal Medicine. 3rd ed. St. Louis: CV Mosby; 2002:1043.

7. Galan JE, Timoney JF. Immune complexes in purpura hemorrhagica of the horse contain IgA and M an- tigen of Streptococcus equi. J Immunol 1985;135:3134-3137.

8. Newton JR, Verheyen K, Talbot NC, et al. Control of strangles outbreaks by isolation of guttural pouch carriers identified using PCR and culture of Streptococcus equi. Equine etV J 2000;32:515-526.

9. Jorm LR. Laboratory studies on the survival of Streptococcus equi on surfaces. In: Plowright W, Ross- dale PD, Wade JF, eds. Proceedings of Equine infectious Diseases VI. Newmarket, UK: R & W Publica- tions;1992:39-43.

10. Pusterla N, Watson JL, Affolter VK, et al. Purpura haemorrhagica in 53 horses. Vet Rec 2003;153:118- 121.

11. Freeman DE. Diagnosis and treatment of diseases of the guttural pouch (part 2). Compend Contin Educ Pract Vet 1980;2:S25-S31.

12. Newton JR, Wood JN, Dunn KA, et al. Naturally occurring persistent and asymptomatic infection of the guttural pouches of horses with Streptococcus equi. Vet Rec 1997;140:84-90.

13. AAEP. Strangles (Streptococcus equi). Accessed June 2009 at www.aaep.org/strangles.htm.

14. Waller A, Robinson C, Newton JR. Further thoughts on the eradication of strangles in equids. JAVMA 2007;231:1335.

15. http://www.aht.org.uk/cms-display/bact_bloodadvice.html Accessed January 2012.

16. Getting a grip on Strangles, Havermeyer Workshop, October 2012.

17. Newton JR, Wood JN, Dunn KA, et al. Naturally occurring persistent and asymptomatic infection of

the guttural pouches of horses with Streptococcus equi. Vet Rec 1997;140:84-90.

18. Verheyen K, Newton JR, Talbot NC, et al. Elimination of guttural pouch infection and inflammation in asymptomatic carriers of Streptococcus equi. Equine Vet J 2000;32:527-532.

19. Davidson A, Traub-Dargatz JL, Magnuson R, et al. Lack of correlation between antibody titers fibrinogen binding protein of Streptococcus equi and persistent carriers of strangles. J Vet Diagn Invest 2008;20:457- 462.

20. Wilson JH. Vaccine efficacy and controversies. Proc 51st Annu Meet AAEP 2005;409-420.

21. Waller A, Robinson C, Newton JR. Further thoughts on the eradication of strangles in equids. JAVMA

2007;231:1335.

22. Boyle, AG, Boston RC, O’Shea K, Young S, Rankin SC, Optimization of an in vitro assay to detect Strep- tococcus equi subsp. equi. Vet. Microbiol. 159: 406-410, 2012.

23. Weese JS, Jarlot C, Morley PS, Survival of Streptococcus equi on surfaces in an outdoor environment.

Can Vet J. 2009 Sep;50(9):968-70.

24. Boyle AG et al. J Am Vet Med Assoc 2009;235: 973.

27 Diagnosis and Management of the Sick Foal in the Field Ashley G. Boyle, DVM, DACVIM Assistant Professor of Medicine, Section Field Service Department of Clinical Studies, New Bolton Center, University of Pennsylvania 382 West Street Rd. Kennett Square, PA [email protected]

The following cases will be presented. Case 1:

A 2 hour foal was examined when the maiden mare presented for being colicky post-partum. The foal nursed with assistance and then on her own by 4 hours. The foal had bright red gums but was otherwise bright, alert and responsive. The foal was placed on 10 mg/ kg Ceftiofur (Naxcel) q 12 hours SQ. The placenta was examined and was within normal limits. IgG on radioimmunodiffusion was measured at 12 hours of age and was >1600 mg/dL. Fibrino- gen was increased at 465 mg/dL. Foal was continued to receive Ceftiofur for 1 week. Foal was doing well and very difficult to treat. Fibrinogen had decreased to 400 mg/dL. Antibiotics were discontinued at that time. Foal was seen to trot by two different veterinarians on day 6 and day 7 during an exam of the dam and was sound. At 11 days of age the foal was down, presenting with a septic physitis.

We will discuss recent work on failure of passive transfer, neonatal sepsis and intrauterine infec- tions, the use of SAA and fibrinogen to monitor treatment, and the use of blood cultures.

Case 2:

A 2 week old TB colt presents with a wet abdomen and enlarged umbilicus. We will discuss how this foal was managed on the farm, risk factors associated with development of umbilical prob- lems and when surgery is needed.

Case 3:

We will present an outbreak of fevers on a small breeding farm that was character- ized by contagious diarrhea. We will discuss differentials for foal diarrhea and how this outbreak was complicated by concurrent Rhodococcus equi infections.

Case 4:

A 4 year old maiden Arab mare is rejecting a her foal at birth. We will discuss management and chemical strategies to allow for acceptance of the foal and recommendations for foals in future years.

28 References

Cohen N. Rhodococcus equi foal pneumonia.Vet Clin North Am Equine Pract. 2014 Dec;30(3):609- 22.

Giguère S1, Cohen ND, Chaffin MK, Hines SA, Hondalus MK, Prescott JF, Slovis NM. Rhodo- coccus equi: clinical manifestations,virulence, and immunity. J Vet Intern Med. 2011 Nov- Dec;25(6):1221-30.

Liepman RS, Dembek KA, Slovis NM, Reed SM, Toribio RE. Validation of IgG cut-off values and their association with survival in neonatal foals. Equine Vet J. 2015 Feb 12. doi: 10.1111/evj.12428.

Palmer J. Update on the management of neonatal sepsis in horses. Vet Clin North Am Equine Pract. 2014 Aug;30(2):317-36.

29 Treating Colitis In The Field. Rose D. Nolen-Walston, DVM, DACVIM (LAIM) Kennett Square, PA

Introduction Colitis in horses is a scary disease on several levels. First, it results in huge fluid losses, sometimes up to 100 liters a day. These can be challenging to replace and keep up with. Second, regardless of etiology, a leaky colon wall frequently leads to endotoxemia, which wreaks havoc on the horse’s physiology. Systemic Inflammatory Response Syndrome (SIRS) is a rapid result, leading to organ failure, especially of the horse’s main shock organ, the foot. And thirdly, a few important differential diagnoses are either contagious or even zoonotic, leading to issues with biosecurity and barn management.

Whats Causes Colitis? Potomac Horse Fever •What it looks like: • Summer/fall • Big fever (~104° F) • Laminitis EARLY • Leukocytosis after a couple of days • How to diagnose: • Blood and fecal PCR – UCDavis or UKy • Treat with oxytet *before* tests come (feet fall off) s Grain Overload • What it looks like: • Usually history of access to grain is present • How to diagnose: • History • Grain in reflux? s C. Difficile and perfringens • What it looks like: • Usually foals or after antimicrobial treatment • Mild to very severe • Foals: bloody diarrhea • How to diagnose: • Fecal TOXIN ELISA • Not culture! • C. diff fecal PCR s Salmonella • What it looks like: • Big fever, sometimes reflux too • No appetite

30 • Usually leukopenic, sometimes leukocytosis • Usually after GI surgery (#1 risk factor) • How to diagnose: • Fecal PCR is best screening test 1. Pick the lab carefully 2. UPenn: very good • Culture? Use to confirm s

Coronavirus • What it looks like: • Big fever (~105° F) • Sometimes mild colic, mild diarrhea • Often profound neutopenia (<1000/ul) • But doesn’t look toxic! • Multiple animals (sometimes) • How to diagnose: • Fecal PCR UCDavis s

Cantharadin toxicosis (“Blister beetle”) • What it looks like: • Diarrhea, colic, hematuria • Severe hypocalcemia • How to diagnose: • Western alfalfa hay – find the beetles • Cantharidin levels in urine s

Larval cyathostomiasis • What it looks like: • Bad diarrhea • Profound hypoproteinemia • How to diagnose: • Fecal egg count NOT USEFUL 1. it’s the larvae that cause disease, and they do not lay eggs 2. no correlation between number of adults and larvae 3. can only get a definitive diagnosis on necropsy or biopsy • Thick colon on U/S (1cm!) • Red larvae in manure, on sleeve • Treat with moxidectin and steroids s

Small colon impaction • What it looks like • Masquerades as colitis: low volume diarrhea, colic (often bloated), fever • How to diagnose: • Rectal: “feels like an anaconda in the abdomen” s

“Colonic dysbiosis”

31 • What it looks like: • Any sort of colitis where the etiologic agent can’t be found • ~50% of cases • How to diagnose: • Diagnosis of exclusion

Commons Clinical Signs on PE: Think Inflammatory! • Fever • Inappetence, dullness, depression • Hyperemic mucous membranes • Injected sclera • Tachycardia • Hypovolemia s How sick is the horse? Gums • Have owner describe color: should be the color of the palm of their hand • Usually is they say “pale” that’s a good sign s

Heart rate • <44 bpm: no hurry • 48-60: evaluate soon • > 60: evaluate ASAP, consider referral • > 80: decompensating s

Lactate

THE SCORECARD OF THE BODY! Lactate meter, handheld, costs $375, $2/strip, uses a drop of blood. Tells you if you are winning, or if you are losing.

• Normal: <2 mmol/L (usually <1!) • 2-4: getting in some trouble • 4-8: very bad things are happening • 8-12: decompensating fast • >12: may not make it to the hospital

Hematologys PCV • Above 50%? Get worried • Splitting! • PCV goes up, TS goes down? Get really worried s

Total protein

32 • Don’t usually run into problems unless < 4/g/dL • Relative hypoproteinemia?

Serums Chemistry Sodium • Colitis cases: it’s always 126 mEq/L! (okay, not always, but oddly often) • If it is <128, monitor • If it is <125 mEq/L, start treating s • If it is <118 mEq/L, call a friendly internist! Potassium s • Don’t let it get too low – stay above 3 mEq/L Bicarb? • Stay on top of the sodium, it will fix itself • Sometimes the best way to stay on top of the sodium is to give more, in the form of sodium bicarb.

Ultrasonographys Look for fluid in the colon (ventrally), wall thickness, sand shadows.

Treatings Colitis In The Field Fluids Anti-endotoxics Anti-diarrheals Laminitis prophylaxis Definitive treatment Monitoring s

Fluids: IV • Minimum effective amount: 20 L (5%) • Can lose 5L/h • Maximum bolus speed 5-7 L/h • Hypertonic NaCl? Just as a bandaid • Colloids? Not in the field s

Fluids: how much? • 30 ml/kg boluses (15L per horse) until volume replete • Warm extremities • Better MM color • Lower HR • PEEING! • ***lactate coming down*** s

Fluids: enteral • Cheap and easy • Okay, not easy

33 • NG tube • Once a day (2x12L?) • Taped in, funnel • Bolus: up to 10 L every 30 minutes! • Ileus: they will spit them back at you • Realistically, 4-8L q 2-6 h • Taped in, coil set, CRI • Enteral feeding tube (thin) • Can still eat! • Use old fluid set (coil, 2 x 5Lbags with the corners cut off) • 1-5L/h s

Fluids: electrolytes Isotonic: per liter, add • 5.6 g of table salt • 0.6 g of Lite Salt (50% NaCl, 50% KCl), • 3.4 g of baking soda (NaHCO2) • 1 ml (by volume) ~ 1g (use a syringe!) s

I usually use ½ strength or ¼ strength • Isotonic is ~9ml powdered ‘lytes per liter • Half strength, 4-5 ml powder/liter s

Anti-endotoxic: Polymixin B • Binds endotoxin in the blood • 3,000-7,000 u/kg IV q 8-12h • 1.5 - 3 million units • 3 - 6 vials/horse • Dilute in saline, give slow s

Anti-endotoxic: Flunixin meglumine • Inhibition of inflammatory mediators • 0.25 mg/kg IV q 8h • 2.5-3 ml s

Toxin binder: Smectite Clay • Binds Clostridial toxins in the lumen of the colon • Go big or go home: • 3lb loading dose • 1 lb q6h, if you can! s

Intestinal Proctectants and Probiotics • Bismuth subsalicylate • Saccharomyces boulardii • Probiotic • Prospective clinical trial – better than any of the other probiotics

34 s

Fecal Transfaunation • Very effective in humans with C diff • No studies in horses • Healthy horse, make a slurry from fresh manure, give 2-6 L s

Cryoprophylaxis • Ice boots • On 24 hours a day • “Jacks boots” $65/pair • Bagged ice • *** If you wait until they are sore: too late!*** s

Definitive Treatment: Antibiotics • PHF: • oxytet if you even suspect it (summer time, geography) • C diff/perf: • metronidazole 15 mg/kg PO q 12 • Double the dose if per rectum • Salmonella • No! (caveat) • If neutrophils are <1000/ul, consider treating with broad spectrum parenteral antibiotics • 1/3 of severe colitis cases are bacteremic; consider antibiotics if signs of severe sepsis • Coronavirus • No! • Ignore the low neutrophils, you’ll be fine s

Monitoring • Creatinine • Oxytet + Banamine + dehydration + endotoxin = kidney damage • Measure creat every couple of days • PCV/lactate • Decrease or stop fluids if the horse can hold these steady • Try to maintain lactate at <2 mmol/L • Total solids • If dropping to 4, consider referral for colloids • Do not decrease fluids to hold TS up! Perfusion is KING. • Sodium • Vital to monitor with foals, can get dangerously low dangerouslky fast • Anything under 120 needs serious attention, very careful not to replace too fast (fatal neuro complications) • Triglycerides • Minis, ponies, donkeys will die of hepatic lipidosis if they are in anegative enrgy balance too long

35 • Measure triglycerides daily • Supplement calories (feed!) or IV glucose (CRI – hard in the field) if they get to 250-500 mg/dL • Add insulin if necessary (hard in the field, start at 0.1 u/kg SQ twice daily)

36 Clenbuterol – The Good, The Bad, And The Ugly Rose D. Nolen-Walston, DVM, DACVIM (LAIM) Kennett Square, PA

Introduction Clenbuterol is the only FDA-approved medication for horses with reversible bronchospasm, administered with a wide dosage range (0.8 to 3.2 ug/kg, q 12 h for up to 30 days). It is delivered as an oral syrup, and bioavailibility in horses is excellent (83.9%). This is in contrast to albuterol, which has poor oral absorption in the horse, and should therefore be avoided in favor of clen- buterol if medication per os is desirable. A long-acting ß2-adrenoceptor agonist, clenbuterol is administered (Ventipulmin®) to horses for a variety of purposes. Beta-2 adrenergic agonists are a mainstay in the treatment of bronchoconstrictive diseases of both humans and animals, including asthma and COPD in man, and in horses, Recurrent Airway Obstruction (RAO) and Inflammatory Airway Disease (IAD), a disease which affects up to a third of racehorses in training. ß2 agonists activate the sympathetic innervation of bronchial smooth muscle resulting in bronchodilation and mucokinesis in the lungs, and causes smooth muscle relaxation in the myometrium, and blood vessels of the liver and kidney. Clenbuterol has been shown to increase dynamic compliance and decrease pulmonary resistance in ponies bronchoconstricted with intravenous histamine. It has a dose-dependent effect, but is not effective in all horses. Normal horses treated with clen- buterol do not demonstrate bronchodilation in response to clenbuterol as there is minimal smooth muscle tone in the small airways. At the commonly administered dose of 0.8ug/kg, only 25% of horses with RAO show a clinical response, which increases up to 75% at the 3.2ug/kg dose. Like horses with RAO, horses with IAD also have a hyperreactivity to inhaled histamine leading to bronchoconstriction. In addition to bronchodilation, clenbuterol has also been shown to have anti-inflammatory effects, speed mucociliary clearance, re-partition fat to muscle, and decrease mucus production by goblet cells.

This lecture will focus largely on the effects clenbuterol, both beneficial and negative, the significance of tachyphylaxis, and make recommendations on its usage in horses.

Effects Of Beta-2 Agonists Bronchodilation and bronchoprotection In humans, ß2 agonists are also widely acknowledged to attenuate bronchoconstriction from both direct and indirect stimuli. Less evaluation of the bronchoprotective effect of clenbuterol has been performed in the horse, although studies in ponies showed no protective effect of intrave- nous clenbuterol against inhaled histamine, but some effect against intravenous histamine. Like- wise, Mazan et al. also found no significant effect on histamine reactivity of 10 days of aerosolized salbutamol in horses with IAD, although based on human data, it is possible that bronchodilator tolerance had already occurred and thus the bronchoprotective window may have been missed.

Anabolic effects When skeletal myocyte ß-2 receptors are stimulated by clenbuterol, there is a direct an- abolic effect that causes an increase in muscle mass in humans, food animals, and horses. In addition, clenbuterol induces lipolysis and inhibits adipogenesis associated with alterations in the adipokines leptin and adiponectin in adult horses. This increases muscle-directed protein depo-

37 sition and reduces total body fat, which is generally known as repartitioning. Clenbuterol causes a switching from type I, oxidative, slow-twitch fibers to a type II, glycolytic, fast-twitch fiber phe- notype, even though ß2 adrenergic receptors are expressed more on slow-twitch than fast-twitch fibers. This effect is lessened when combined with exercise. Though these changes in fiber type are noted in horses, the contractile properties of isolated muscle fiber remain unchanged when treated with clenbuterol. Clenbuterol is anabolically active in humans and cattle, and in horses although its leptin- and adiponectin-mediated repartitioning effects cause an increase in muscle mass, this potential benefit is offset by a negative ergonomic effect. We recently performed a pair of double-blinded, placebo-controlled clinical trials which examined the interaction of exercise and chronic low-dose clenbuterol (0.8μg/kg PO q12 for 21 days) administration in both resting (non-working) and working (polo) horses. Our results showed a significant reduction in percent body fat of 8% and 12% in non-working and working horses, re- spectively that received clenbuterol. There was no evidence of tachyphylaxis of the lipolytic effect by day 21, and horses in the working group returned to their baseline percent body fat within 2 weeks of cessation of clenbuterol treatment. Additionally, there was no decrease in body weight during either trial, suggesting a true repartitioning of fat to muscle rather than simply a loss of adiposity. These results indicate that at even the commonly administered low dose, clenbuterol reduces body fat when compared to a placebo and may have repartitioning effects that are both lipolytic and anabolic in nature.

Effects on performance In light of its anabolic effects, clenbuterol has been investigated as a therapeutic com- pound to treat muscle wasting in humans, but due to concerns of abuse, it has been banned in athletes by the International Olympic Committee and it is illegal as a growth promoter in food ani- mals. In horses, however, clenbuterol has long been a legal therapeutic medication in most racing jurisdictions, with published withdrawal times. In a number of treadmill studies it has not been it has not been shown to improve performance; in fact any changes noted have been negative ergogenic effects, and may also have deleterious effects on cardiac function in horses.

Anti-inflammatory effects Clenbuterol, like other cyclic AMP inducing drugs, has been shown to have anti-inflam- matory properties. In horses with RAO, it protects against the effects of aerosolized endotoxin, hay dust suspension, and Aspergillus fumigatus antigen, showing a significant beneficial effects on lung function, bronchoalveolar lavage neutrophil influx, and pro-inflammatory cytokines and chemokines expression in pulmonary alveolar macrophages. This effect held true even when endotoxin was administered intravenously, where pre-treatment with clenbuterol reduced peak rectal temperature and peak plasma TNFß concentration in adult horses.

Diagnosis of anhidrosis Unlike other mammalian species that sweat in response to cholinergic stimulation, sweating in horses is mediated by ß-2 adrenergic receptors. It is likely that anhidrosis, a naturally occurring syndrome of reduced sweat production, is due to gradual failure of the secretory process of the sweat glands apparently caused by desensitization and down-regulation of the ß-2 receptors in response to endogenous catecholamines. This process occurs most commonly in horses living in hot, humid climates where there is continual stimulation of sweating, although it is unclear why only some horses are affected. Diagnosis of anhidrosis is usually performed using intradermal

38 ß-agonists, either non-specific (epinephrine) or specific (terbutaline or salbutamol). Normal hors- es sweat at a 1:1,000,000 concentration of epinephrine, and that failure to sweat at this concen- tration is diagnostic for hypo- or anhidrosis.

Tocolytic effects Clenbuterol is used as a tocolytic in horses to slow the progression of labor in dystocias, allowing easier manual mutation of the fetus.

Negative side effects Negative side effects of the ß2 agonists are those expected by adrenergic activation. They include characterized sweating, tachycardia, agitation by decreases in aerobic capacity, time to fatigue, cardiac function, and VO2max. The use of ß2-agonists is nonetheless common in both human and equine athletes with lower airway disease, despite these significant side effects and the rapid occurrence of desensitization or tachyphylaxis to the effects of this class of drug. This is suggested by data from Kearns et alia, who found that at 3.2 µg/kg, side effects including extreme sweating were initially observed but subsided after 10 days, Clenbuterol has been hypothesized to affect thermoregulation, which may be a component to the reduced aerobic capacity of horses receiving high doses for extended periods.

Tachyphylaxis In asthmatic humans as well as other mammalian models of bronchoconstriction, chronic treatment with ß2 agonists results in tolerance or tachyphylaxis (desensitization) to the effects of the drug. This can be seen as a positive attribute, in that the negative side-effects of β2 agonist administration such as sweating, tachycardia, and agitation decrease over the first few weeks of use, but also is associated with a decreased efficacy of the bronchodilatory effects of the treat- ment and an actual worsening of asthma control. Minimal data are available regarding the effect of long-term ß2 agonist treatment on bronchodilation in horses, although small studies in horses with RAO and IAD showed conflicting results, with little-to-no effect seen. The cause of tolerance to ß2 agonists appears multifactorial. In the horse, Abraham et al. showed that 12 days of intravenous clenbuterol in adult horses resulted in a reduced density and responsiveness of ß2-adrenoceptor on lymphocytes, suggesting that these effects are conserved across species. In the treatment of human asthma, concurrent use of a corticosteroid attenuates the development of tachyphylaxis to bronchodilators and it is now widely recommended that daily ß2-agonists should be used only in conjunction with inhaled corticosteroids to prevent rapid de- sensitization. Although no studies have been performed to evaluate the effect of this drug com- bination on lung function in horses with IAD/RAO, it has been demonstrated that the decrease in lymphocyte ß2-receptor density and function in horses receiving clenbuterol is reversed and prevented by administration of dexamethasone. In contrast to other mammals, equine sweat glands contain ß2 receptors that show rapid desensitization toß2 agonists leading to sub-normal sweat production in vitro. In an experimental setting, there is a gradual decline in sweat production over the course of prolonged epinephrine infusion, and several studies have demonstrated similar findings of apparent desensitization of sweat gland ß-2 receptors both in vivo and ex vivo, where cultured equine sweat glands show a rapid and homologous desensitization to epinephrine. However, after 21 days of oral clenbuterol at 0.8 µg/kg twice daily, we found no reduction of sweating using an intradermal epinephrine sweat at concentrations of epinephrine as low as 1:1,000,000, thus suggesting that any reduction

39 in sweating caused by chronic clenbuterol is likely to be sub-clinical. Using a blinded, randomized cross-over designed trial, we showed clenbuterol at standard dosages (0.8 μg/kg per os every 12 hours) resulted in a decreased bronchoconstrictive response to inhaled histamine compared to horses receiving placebo treatment. Using flowmetric pleth- ysmography and inhaled histamine bronchoprovocation, 5 out of 8 horses (62%) had a ≥100% increase in PC35 (35% percent change in DELTAflow) peaking at day 14 of administration. This degree of efficacy is somewhat higher than previous studies that show 25% of RAOhorses demonstrating clinical evidence of bronchodilation at the same dose. This may be due to superior sensitivity of pulmonary function testing compared to clinical examination for detecting bronchoc- onstriction, the greater reversibility of bronchoconstriction in horses with IAD compared to those with RAO-induced airway remodeling, or differences in airway secretion volume or composition between the two diseases. Additionally, all horses showing a bronchoprotective effect demon- strated tachyphylaxis by day 21 of treatment; 7/8 horses had PC35 values that were actually low- er than baseline, indicating pulmonary function may have worsened during the treatment period. This is consistent with human data that show prolonged administration of ß-2 agonists results in worse than baseline lung function. Based on in vitro studies in the horse and extensive human clinical data, it is likely that extended use of clenbuterol should be combined with inhaled cortico- steroids, although the safety and efficacy of this approach has not been validated in the horse. However, our study suggests that clenbuterol alone is less effective as a bronchodilator after 14 contiguous days of treatment, and courses of administration should not exceed 2 weeks without interruption.

40 Equine Blood Transfusion - What You Need To Know To Get The Job Done. Rose D. Nolen-Walston, DVM, DACVIM (LAIM) Kennett Square, PA

1. Blood products a. Whole blood o No commercial blood banks for horses o Almost all blood is collected from donor immediately before use b. Plasma o Usually acquired frozen from a commercial source o Uses: o failure of passive transfer o colloid support in FOALS (need too much volume for adults, $$$) o replacement of coag factors (fresh-frozen (FFP) only) o “Specialty” plasmas (commercially available) come from animals hyper-immunized against specific pathogens: o Hi-Gamm: high IgM, for FPT in foals o J-5: E. coli cell wall (endotoxemia) o anti-botulism o anti-Rhodococcus o anti-West Nile Virus c. Washed RBC o Rarely used, produced at point-of-care o ideal for foals with Neonatal isoerythrolysis d. Packed RBC o Rarely used, produced at point-of-care o Ideal for animals with hemolysis (such as Red Maple Toxicity) that do not need extra volume (plasma) e. Platelets o Rarely used, produced at point-of-care o Can only be stored at room temperature (up to 3 days) as refrigeration causes platelets to become non-functional

2. Equine blood types a. seven blood groups: A, C, D, K, P, Q, and U b. each group can have multiple factors. For example, horses with blood group A can have factors a, b, c, d, e, f, g. These factors are surface markers on the RBC. o So each horse has a blood type such as Qa or Pd… o 400,000 combinations! c. Antibodies o Anti-RBC antibodies are typically acquired by exposure (usually pregnancy - see Neonatal isoerythrolysis) o anti-Ca antibodies may naturally occur, and cause mild agglutination reactions, not usually clinically significant in vivo

41 o As horses age, they seem to develop more allo-antibodies, possibly due to cross-reaction with ingested protein, exposed by diet? (Dr. Sean Owens, personal communica- tion)

3. Cross-matching Horses have minimal naturally occurring anti-RBC antibodies and often do not make RBC anti- bodies until they have been administered blood multiple times, transfusions can usually be per- formed without crossmatch if necessary, as long as you are careful to watch for transfusion reac- tions and get informed consent from the owner.

a. Major crossmatch - same as small animal, human o Donor erythrocytes are washed and incubated with recipient serum b. Minor crossmatch crossmatch - same as small animal, human o Donor serum is incubated with washed recipient erythrocytes c. Types of crossmatch reaction o Agglutination o caused by agglutinins o Hemolysis o caused by hemolysins o detected after adding complement to the reaction mixture o technically quite difficult, usually only performed in large hospitals/labs d. Storing blood samples for crossmatches. o As many large hospitals maintain a blood donor herd, it would make sense to store samples from these donors in the lab so that crossmatches can be performed without re-phlebotomizing the horses each time. o ***Unfortunately, a study that we did at New Bolton in 2011 demonstrates that crossmatch results are not stable on aliquots of blood stored for as little as a week.***

4. Blood donor selection, blood collection a. Ideally Qa and Aa antigen and antibody negative o Usually Standardbreds and Quarterhorses o “universal donor”? b. Usually large, quiet (or maiden mare), PCV >35% c. Needs to be UTD on vaccines, anthelmintics, and especially testing for blood borne disease (i.e. Coggin’s negative). o Aseptically place a 14g (or even 10g) catheter ideally going up the neck, and connect to blood collection device. o Remove up to 15-18 ml/kg (***so approx 8-10L for a 500kg horse***) o Replace with 10-20L of crystalloid. o Can usually collect every 3 weeks or so. d. Collection systems o ACD, sodium citrate, CPDA-1 (best for storage). o Bags (human), large glass bottles also available.

5. Equine blood product administration a. Normal blood volume for an adult horse is ~8% of bodyweight (80ml/kg)

42 b. Blood deficit can be calculated using an intuitive formula:

(normal PCV - current PCV)/normal PCV x 0.08 x bodyweight (kg)

o However, many horses who have had acute hemorrhage will still have a normal PCV; typically the plasma protein will drop before the PCV o Assuming donor blood is ~ PCV 40%, every 2.2 ml of whole blood/kg body weight will increase the PCV by 1% c. Indications for whole blood o Anemia with clinical signs o tachycardia (HR>60 bpm for an adult) o colic (low GI perfusion) o dullness, depression, anorexia o usually occur when PCV <12-15% if acute loss, may get as low as 9-10% with chronic anemia before signs are seen d. Transfusion technique o Same as small animals, humans o Replace transfer set every 4 liters o Monitor for signs of reaction (increased rectal temperature, piloerection, sweating, tachycardia, colic, diarrhea) o TPR every 2 minutes for first 30 minutes, then every 15 minutes until transfusion is complete o Treat reactions by stopping transfusion, giving crystalloid fluids and epinephrine if necessary (1-5ml of 1:1,000 epinephrine SQ or 1-2ml IV in an average 450kg horse) e. Transfused RBC lifespan o Normal RBC lifespan is 150 days o Half-life of 20-30 days donated RBCs o too many blood-types for “perfect” matching o takes 5 days to mount an immune response o Bone marrow response is essential o ~50 days for autologous re-transfused blood

6. Blood storage a. Equine blood is usually collected at the time of use and not banked. b. Can be stored for 28 days, but red cell half-life drops a little. o CPDA-1 best, glass bottles worst for storage o Use same storage techniques as small animal

Diseases requiring blood transfusion

A. Neonatal isoerythrolysis o neonatal foal develops severe intravascular hemolysis due to colostral antibodies o most common in *multiparous* Standardbreds and 1. Pathophysiology a. Mare is bred to stallion with different blood type

43 b. She becomes sensitized to foal antigens in utero c. She makes antibodies against foals’s red cells d. Foal ingests colostrum with anti-RBC antibodies e. Type II hypersensitivity reaction → severe hemolytic anemia o Qa antigen and Aa antigen are responsible for most cases a. Therefore mares that are negative for Qa and/or Aa antigens are at highest risk for having an NI foal. o ~19% of and ~17% of Standardbreds b. Occasionally other groups (Dc, Ua, Ab and Pa) c. Mules (horse dam, sire) have very high incidence of NI because of DONKEY RBC ANTIGEN; therefore 100% of matings are incompatible.

Breed Qa(-) Aa(-) Incidence of NI Thoroughbred 39% 15% 0.05% Standardbred 100% 44% 2% N/A N/A 8-10%

2. Clinical signs a. NORMAL AT BIRTH b. Hemolysis becomes clinically apparent from 12 hours to 5 days post-birth foal c. Dull, lethargic d. Tachycardic e. Icteric f. Variable hemoglobinemia, hemoglobinuria, anemia

3. Treatment and Prevention a. Transfuse if necessary (tachycardia, dullness, PCV <10-15%) o Whose blood? 1. WASHED dam’s RBC 2. whole blood from an unrelated gelding b. Prevent by: o Not re-breeding to stallion with incompatible blood type o Cross-matching mare and stallion during late pregnancy to see if mare is pro- ducing antibodies o Performing a jaundice foal agglutination test, which is a stall-side cross-match that looks for agglutination between mare colostrum or serum and foal blood. 1. If any of these are positive, withhold colostrum from foal (can be diffi- cult!)

B. Red maple toxicosis o Ingestion of wilted leaves and branches from Acer rubrum (Red maple) a. Aka swamp of soft maple b. Eastern USA i. Typically seen in summer and autumn ii. After winds or storms (blow downs)

Red Maple Toxic 44 iii. After trees have been felled iv. yard mulch and trimmings have been dumped in the field.

1. Pathophysiology o Toxic principle gallic acid? o Toxic dose1.5-3 gm/kg body weight (0.7-1.5 kg for 450-kg horse) o Causes severe intra- and extravascular hemolysis and methemoglobinemia

2. Clinical signs o Dull, lethargic o Tachycardia o Pale or muddy brown (methemoglobin) mucous membranes o Massive anemia, hemoglobinemia, hemoglobinuria o Death (~60% mortality) Red Maple 3. Treatment NonToxic o Transfuse if necessary (tachycardia, dullness, PCV <10-15%) • any matched donor, unmatched if first transfusion and crossmatching is unavailable o Crystalloids (cautiously) for diuresis (hemoglobin=nephrotoxin) o Activated charcoal to early cases? Usually too late for affected horses.

C. Hemorrhage i. During nasal/sinus surgery o because this is anticipated, some people bank autologous blood 1-4 weeks before surgery ii. Post-castration iii. Uterine artery rupture, other obstetric iv .Guttural pouch mycosis v. Thrombocytopenia vi. Trauma

D. Immune-mediated hemolytic anemia i. Usually hapten-mediated o penicillin, other drugs o neoplasia o Streptococcal infection o anything else! ii Diagnose using Coomb’s test, direct immunofluorescence (DIF) flow cytometry iii. Treat with immunosuppression (corticosteroids) and ideally remove the inciting cause.

45 Guttural Pouch Disease- More Than Strangles Rachel Gardner, DVM, DACVIM (LAIM) BW Furlong & Associates Oldwick, NJ

Diseases of the guttural pouch can be challenging to diagnose and treat due to the complexity and vital nature of the structures in and surrounding the area. The guttural pouches are paired diverticula of the auditory tube connecting the nasopharynx and the middle ear. They are air-filled sacs that are each divided into a medial and lateral compartment. The guttural pouches are sepa- rated from each other by a thin septum, and each pouch has a 300-500ml capacity. The pouches communicate with the nasopharynx through paired ostia. Cranial nerves IX (glossopharyngeal), X (vagus), XI (accessory) and XII (hypoglossal) are located adjacent to the guttural pouch. The internal carotid artery and external carotid artery, with its branches, are also located adjacent to the pouch. Cranial nerves V (trigeminal) and VII (facial) are also in close proximity. The function of the guttural pouches still requires elucidation, however warming of inspired air is a prevailing theory.

Clinical exam begins with physical examination and external palpation of the guttural pouch- es. Further diagnostic evaluation may utilize radiography, ultrasonography or endoscopy, how- ever endoscopy remains the gold standard for examination of the guttural pouches. Entrance of the endoscope into the guttural pouch is facilitated by lifting of the fibrocartilaginous flap on the lateral aspect of the pharynx using a probe fed through the biopsy channel of the endoscope, and twisting the scope to move the flap allowing access to the pouch. Endoscopy enables direct visu- alization of the pouches which may reveal blood, exudate, masses, lymphadenopathy, masses or direct evidence of infection. Additionally, the anatomy and flexibility of the temporohyoid articula- tion can be observed. Radiographs are useful for detecting fluid or chondroids within the guttural pouches. Computed tomography (CT) and magnetic resonance imaging (MRI) have more recent- ly been used for more detailed examination of the stylohyoid bone and its articulations in cases of temporohyoid osteoarthropathy, as well as for evaluation of masses or tumors within the guttural pouches.

The most common disease involving the guttural pouches is empyema, an accumulation of purulent material caused by bacterial infection. Pus may become inspissated and form chon- droids. Upper respiratory infection caused by Streptococcus equi equi is the most common cause of guttural pouch empyema, although infections with other organisms such as Streptococcus equi zooepidemicus may also result in empyema. Less common causes include trauma to the guttural pouch or pharynx, or abnormalities of the pharyngeal orifice. Common clinical signs include vari- able fever, nasal discharge, submandibular and retropharyngeal lymphadenopathy, respiratory stridor, dysphagia or extended head/neck carriage. Diagnosis is most easily made by endoscopic exam and observation of purulent discharge from the pharyngeal orifice on the affected side(s), pharyngeal collapse, and fluid, pus or chondroids within the affected guttural pouch. A fluid line or round opacities within the guttural pouch may be visible on standing radiographs. ulture and PCR may be done on fluid or chondroids from the guttural pouch, or from saline washes of the guttural pouch.

46 Guttural pouch empyema is typically responsive to a combination of saline lavage and sys- temic antibiotic therapy. Lavage may be performed through the biopsy channel of the endoscope, or by placement of an indwelling foley catheter. Chondroids may be removed manually using trans-endoscopic tools such as snares, forceps and baskets. When chondroids are numerous or difficult to remove, consideration for surgical treatment should be given. Response to treatment is typically good, although resolution and duration of treatment before negative cultures are obtained may be protracted and nerve injury may occasionally occur.

Guttural pouch mycosis is fungal infection of the dorsal surface of one or both guttural pouches. A variety of different fungi have been isolated, however Aspergillus (Emericella) fumigatus is most commonly isolated.

The most common clinical sign associated with guttural pouch mycosis is unilateral epistaxis due to erosion of the affected artery. Intermittent episodes of mild to moderate epistaxis are typi- cally observed, frequently followed by an episode of severe and potentially fatal hemorrhage. The internal carotid and maxillary arteries are most commonly affected, though the external carotid or any arterial branch within the guttural pouch may be involved. Dysphagia is also common with guttural pouch mycosis due to injury of the pharyngeal branches of the vagus and the glossopha- ryngeal nerves. Respiratory noise, Horner syndrome, mucopurulent nasal discharge and facial nerve or tongue paralysis, may also be present.

Guttural pouch mycosis may be suspected based on clinical signs, but diagnosis is reached by endoscopy. Endoscopy may reveal blood draining from the pharyngeal orifice of the affected guttural pouch, pharyngeal collapse or signs of dysphagia. When the endoscope is advanced into the guttural pouch, a classic finding of a diphtheritic membrane is observed on the roof of the guttural pouch and/or over the stylohyoid bone. In some cases the bone may be thickened. Care should be taken not to disrupt fungal plaques or blood clots upon advancement of the endoscope into the guttural pouch due to risk of severe hemorrhage. Both guttural pouches should be eval- uated, as bilateral involvement occurs in some cases.

Medical treatment is not commonly recommended due to slow response to treatment and the risk of fatal hemorrhage during the treatment period, but several oral and topical anti-fungal agents have been used to treat guttural pouch mycosis in horses with variable response. Spon- taneous resolution has been reported to occur, however if infection is in close proximity to arteries or nerves there is risk of hemorrhage or neuropathy, respectively, prior to resolution of infection. Supportive care secondary to hemorrhage and dysphagia may be necessary, and if nerve involve- ment is present non-steroidal anti-inflammatories should be administered. Surgical occlusion of the affected artery is recommended to prevent hemorrhage secondary to infection. In most cases, slow regression of the fungal plaques is observed following occlusion although progression follow- ing surgical occlusion has been reported. The prognosis for survival is good with surgical arterial occlusion. Neuropathies, including laryngeal hemiplegia or dysphagia, may be slow to resolve and may be permanent.

Guttural pouch tympany is distension of one, or less commonly both, guttural pouches with air in foals up to one year of age. There is a significantly increased prevalence of the disorder in fillies compared to colts, and Arabian and Paint horse foals are predisposed. A sex-specific quantitative

47 trait locus has been described.

Foals with guttural pouch tympany develop an elastic swelling in the parotid region which is non-painful to palpation. When severe, tympany can result in respiratory distress and rarely, dysphagia and secondary aspiration pneumonia. Endoscopy of the guttural pouches is used to establish a diagnosis, although there is typically no apparent abnormality in the opening of the guttural pouch.

Surgical fenestration of the median septum, removal of obstructive tissue at the guttural pouch opening or creation of a salpingopharyngeal fistula can be performed to allow movement of the air out of the guttural pouch. Repeat surgeries may be necessary for permanent resolution, however once surgical resolution is achieved the prognosis for complete recovery and athletic success is good. Catheterization of the affected guttural pouch was previously used for temporary relief of tympany, however by leaving the catheter(s) in place for 6-8 weeks, pressure necrosis of the gut- tural pouch opening occurs and has been shown to result in permanent resolution. Regardless of method of treatment, the prognosis is more guarded if aspiration pneumonia or nerve involvement is present.

Temporohyoid osteoarthropathy (THO) is a thickening of the proximal portion of the stylohyoid bone, the tympanohyoid and the squamous portion of the temporal bone. Eventual fusion of the temporohyoid joint occurs. It is a progressive disease which occurs, in most cases, secondary to hematogenously-spread bacteria from an inner or middle ear infection. Although infection is most commonly bacterial, fungal infection may also occur. The changes that occur with THO can be exacerbated by upward head and neck movement such as occurs with teeth floating, dental examination and nasogastric intubation, as well as through common motions such as swallowing and vocalizing. When THO is present, the petrous portion of the temporal bone may fracture due to excessive forces secondary to fusion of the temporohyoid joint. Inflammation surrounding the area of petrous temporal bone fracture results in neuropathy of the adjacent facial (CN VII) and vestibulocochlear (VIII) nerves. Fracture less commonly results in neuropathy of the glossopha- ryngeal and vagus nerves. Extension of the the middle or inner ear infection may occur following fracture resulting in meningitis or other cranial nerve involvement.

THO is most common in middle aged horses, with a median reported age of 10.8 years, though cases have been reported over a large variety of age ranges. A predisposition has recent- ly been described in horses that crib, and in this study nearly ⅓ of horses reported with THO were cribbers. Horses early in the course of THO are reported to show signs such as unusual chewing behaviors, head tossing, ear rubbing, reluctance during bitting and abnormal head/neck carriage. Acute vestibular signs, including asymetrical ataxia, head tilt, nystagmus and balance loss are common. Pain on palpation of the base of the ear is variably present but may be supportive of the diagnosis. The majority of horses with THO will show signs of facial paralysis including ipsi- lateral ear droop and contralateral nose pull. Decreased tear production and loss of the palpebral reflex are also common, which may result in corneal ulceration and exposure keratitis. Dysphagia is also reported secondary to pain with tongue movement or glossopharyngeal or vagus nerve dysfunction.

48 Endoscopy of used for diagnosis of THO and reveals thickening and irregularity of the proximal stylohyoid bone. Despite unilateral signs, both guttural pouches should be examined, as bilateral involvement is common. In bilateral involvement, the stylohyoid bone is usually thicker on the side with clinical signs. Thickening and osteitis of the stylohyoid and petrous temporal bones is evident on dorsoventral radiographs. Computed tomography (CT) is useful for determining the extent of articular and soft tissue involvement.

Treatment with broad spectrum antibiotics for likely bacterial infection is indicated. Antimicro- bial drugs such as ampicillin, trimethoprim-sulfa, enrofloxacin and chloramphenicol which are ef- fective against Staphylococcus aureus are recommended. Tympanocentesis may yield samples for cytology and bacterial culture and sensitivity. NSAIDs are recommended for control of pain and inflammation, and may be used for supplemental pain control. Temporary tar- sorrhaphy is recommended in cases with an ineffective palpebral response or corneal ulceration. Ceratohyoidectomy, in which a portion of the hyoid apparatus is surgically removed, decreases forces on the temporohyoid joint and is performed to reduce fracture recurrence. The prognosis for medical and/or surgical treatment for horses with THO is variable. Neurologic signs can persist despite appropriate treatment, and resolution may take up to a year to occur in responsive cases. Treatment is more effective in cases with more mild neurologic involvement and those that are treated more promptly following development of clinical signs.

Nasopharyngeal obstruction caused by trapping of air in the guttural pouches, similar to gut- tural pouch tympany in foals, has been described in adult horses due to head/neck flexion. A de- fect in the salpingopharyngeal fold is suspected. Affected horses demonstrate a respiratory noise and exercise intolerance associated with head/neck flexion. Laser fenestration of the median septum of the guttural pouch and resection of the salpingopharyngeal fold has been associated with a good prognosis for return to function.

Suggested reading:

Freeman DE. Update on Disorders and Treatment of the Guttural Pouch. Vet Clin Equine. 2015. 31: 63-89.

Freeman DE. Complications of Surgery for Diseases of the Guttural Pouch. Vet Clin Equine. 2009. 24: 485- 497.

Sparks HD, Stick JA, Brakenhoff JE, et al. Partial resection of the plica salpingopharyngeus for the treat- ment of three foals with bilateral tympany of the the auditory tube diverticulum (guttural pouch). JAVMA. 2009. 235(6): 731-733.

Walker AM, Sellon DC, Cornelisse CJ, et al. Temporohyoid osteoarthropathy in 33 horses (1993-2000). JVIM. 2002 Nov-Dec. 16(6): 697-703.

49 New Thinking on Gastric Ulcer Diagnosis and Treatment Rachel Gardner, DVM, DACVIM (LAIM) BW Furlong & Associates Oldwick, NJ

Equine gastric ulcer syndrome (EGUS) is a common and relatively easily diagnosed disease of the horse. Ulceration may involve the distal esophagus, squamous mucosa, glandular mucosa or proximal duodenum. Many differences in the risk factors, pathophysiology and healing have been highlighted between ulceration in the squamous and glandular portions of the stomach, and thus more recent literature has suggested that they no longer be considered as a single disease entity.

The equine stomach is a large single compartment with two distinctly divided types of muco- sa. The dorsal third of the stomach is lined with non-glandular stratified squamous epithelium. The squamous epithelium is predisposed to ulceration due to acid injury because it lacks protec- tive mechanisms such as mucus and bicarbonate. The ventral two thirds of the stomach is lined with glandular mucosa. This portion can be further divided into the ventral glandular fundus and the distal pyloric antrum. The glandular mucosa secretes hydrochloric acid and pepsinogen. In contrast to the squamous mucosa, the glandular mucosa is highly vascularized and produces significant amounts of protective mucous and bicarbonate. The margo plicatus is the line of de- marcation between the squamous and glandular mucosa.

Clinical signs of EGUS are variable and frequently vague. Possible clinical signs include, but are not limited to, recurrent colic, poor appetite, poor performance, weight loss, behavioral changes and pain upon tightening of the girth. Horses with gastric ulcers may also seem to be asymptomatic, but show improvement in behavior, performance or appetite following treatment.

History and physical exam findings may raise suspicion for the presence of EGUS, but gas- troscopy is the only definitive method for diagnosis. Additionally, gastroscopy is necessary to characterize the location and severity of EGUS, as well as response to treatment on repeat exam- ination. A 3 meter gastroscope is used to visualize all portions of the stomach. Shorter 2 meter scopes may be used to visualize the squamous mucosa, but because the presence of ulceration in the squamous mucosa is not predictive of glandular or pyloric ulceration, failure to visualize the pylorus results in an incomplete exam. Patient preparation consists of withholding feed (hay and concentrate) for at least 16 hours prior to the exam. Water may be offered free choice or withheld for up to an hour prior to exam. An occasional horse may not completely empty the stomach for up to 24 hours, but the vast majority of horses will have a stomach empty enough to provide com- plete examination. Horses on lower roughage and higher concentrate diets will require less time for complete gastric emptying.

Gastroscopy is not a complicated procedure and becomes easier with practice. Attention should be paid as the scope is passed from the pharynx into the proximal esophagus to ensure proper advancement into the esophagus and prevent retroflexion in the pharynx or around the soft palate. Alternatively, a plastic tube with an inner diameter that will receive the gastroscope may

50 be pre-placed into the proximal esophagus to prevent damage to the scope. Once the scope is passed into the cardia the stomach should be distended with air using insufflation through the bi- opsy channel or through the air mechanism on the scope itself. After orienting the scope correctly with the fluid ventral on the image, the squamous mucosa should be observed to the left side and dorsally on the image. The scope should then be advanced until the lesser curvature becomes visible. The opening to the pyloric antrum is seen ventral to the lesser curvature and the scope should be slowly advanced while keeping the tip of the scope dorsal, pointing towards the pyloric antrum and above the level of the fluid for as long as possible. In many cases, the tip of the scope will fall into the fluid before rising again above the fluid once advanced into the pyloric antrum. While below the level of the fluid, the air function on the scope can be occasionally used to create bubbles in the fluid to help direct the operator as to which direction is dorsal, as the bubbles will float to the surface of the fluid. The pyloric antrum should be observed completely, followed by advancement of the scope into the proximal duodenum. Lesions of the proximal duodenum are uncommon so evaluation of this area is not considered critical. The proximal duodenum and bile duct papillae can, however, be easily observed by advancing the scope through the pylorus and then turning the scope to the left. Following examination, the scope can be withdrawn to the level of the cardia and air suctioned from the stomach if desired.

Findings should be graded, and more recently it has been recommended that separate grades be assigned to the squamous and glandular mucosa. Grade 0 is assigned for normal, intact squamous or glandular epithelium with no evidence of hyperkeratosis or hyperemia. Grade 1 is assigned for hyperkeratosis of the squamous mucosa or hyperemia of the glandular mucosa. Grade 2 is assigned for squamous or glandular mucosa with small, single or multifocal superficial lesions. Grade 3 is assigned for squamous or glandular mucosa with large single, deep or multi- focal superficial lesions. Grade 4 is assigned for squamous or glandular mucosa with extensive lesions with deep ulceration.

Therapeutic drug trials have been used as a method of diagnosis for EGUS. The therapy most commonly recommended is a 28 day course of Gastrogard at 4 mg/kg PO SID. Resolution of clinical signs is suggestive of gastric ulceration, however resolution of clinical signs does not necessarily indicate complete healing of ulcers and premature treatment withdrawal is possible. Thus, when a therapeutic trial is used and a response is seen, gastroscopy prior to withdrawing or decreasing treatment to ensure complete healing is recommended.

Diagnostic tests such as sucrose permeability testing and fecal occult blood have been used but remain unvalidated in clinical cases.

Squamous gastric ulceration occurs due to increased exposure of the relatively unprotected mucosa to a highly acidic environment. Disruption of the normal buffering capacity of the stom- ach, such as is provided by the presence of roughage, or increased acid production, such as occurs with high carbohydrate feeds or increased exercise, predispose the squamous mucosa to potentially ulcerogenic acidity. Squamous gastric ulceration most commonly occurs as a primary disease, and the prevalence of ulceration is directly proportional to exercise intensity. Although the prevalence has been shown to be very high in performance horses (>70% of racehorses and 93% of endurance horses), a proportion of these populations have mild (grade 1-2) ulceration which may not be clinically significant. Other predisposing factors for the development of squa-

51 mous ulceration include high concentrate/low roughage diets, stall confinement, restricted access to water, and transport. It should also be stated that ulceration of varying degrees has also been observed in horses on pasture without any of the listed predisposing management practices.

Removal of risk factors that predispose to the development of squamous ulceration will result in improvement of lesions but is frequently not sufficient for resolution. Constant availability of roughage is helpful to buffer the acidic environment of the stomach, and the minimization of car- bohydrate meals will reduce volatile acid production. The addition of corn oil to the diet is shown to decrease gastric acid production while providing calories so that the concentrate portion of the diet can be minimized. Feeding of a small amount of roughage prior to exercise has also been shown to help prevent squamous ulceration by providing buffering of acid and preventing the splashing of acidic fluid onto the squamous mucosa.

Medical treatment of squamous ulceration is achieved by decreasing acid production. Ome- prazole has been shown to be the most effective treatment to suppress acid production, which occurs through inhibition of the proton pump that secretes HCl. The formulation of omeprazole is important, as protection through a carrier or coating is necessary to prevent degradation in the acid environment of the stomach. Gastrogard is the most well-studied and contains a buffered paste for protection. Gastrogard at a dose of 4 mg/kg PO SID has been shown to result in healing of 70-77% of squamous ulcers after a treatment period of 28 days. Treatment failures occur and may be due to acid suppression that lasts for less than 24 hours or individual variation in absorp- tion. Alternative treatment options should be considered in cases of treatment failure or incom- plete resolution. The H2 blockers ranitidine and cimetidine have also been used for the treatment of EGUS by acid suppression. Ranitidine at a dosage of 6.6 mg/kg PO TID is effective in reducing acid suppression and is recommended in horses that are refractory to omeprazole treatment. Ant- acids are effective at reducing gastric acidity but are only effective for up to two hours making their use impractical for prolonged effects. A pectin-lecithin complex and salts of organic acids with B vitamins have been studied with variable results and remain of questionable clinical use.

Recommendations for prevention of squamous ulceration include reducing as many risk fac- tors as possible. When removal of risk factors is limited, treatment with mucosal protectants such as pectin-lecithin complexes may be useful when used in combination with antacids. Omeprazole at a reduced dosage of 1 mg/kg PO SID in a buffered preparation has been shown to be effective, though absorption and therefore effectiveness may be decreased in some horses.

In recent years as gastroscopy using 3 meter scopes has become routine, it has become clear that glandular mucosal ulceration, particularly in the region of the pyloric antrum, has different risk factors and a different response to treatment compared to ulcers in the squamous mucosa. The glandular mucosa is normally exposed to highly acidic conditions in the ventral portion of the stomach. The glandular mucosa normally contains extensive protective mucus and bicarbonate layers. It is likely that ulceration in the region is due to loss of these normal protective mecha- nisms. Although bacterial infection and NSAIDs remain risk factors, neither of these factors are present in cases of glandular mucosal ulceration in many horses. Thus, many cases of glandular ulceration in horses, as seen in humans, are considered idiopathic. The prevalence of glandular ulceration has not yet been extensively studied, but risk factors appear different from those of squamous ulceration. A high carbohydrate/low roughage diet is likely a risk factor, but other risk

52 factors remain unclear.

Well-defined treatment recommendations for glandular ulceration have not been established to date. It is clear that treatment recommendations for squamous ulceration are not appropriate for glandular ulceration, as resolution of glandular ulceration was only seen in a small proportion of cases after treatment with omeprazole at a dosage of 4 mg/kg PO SID for 28 days. Glandular ulcers demonstrate a slower response to treatment and a minimum of 6 weeks of treatment is recommended prior to re-evaluation. Sucralfate, a mucosal barrier protectant which adheres to affected mucosa and stimulates mucous and prostoglandin production, is a reasonable adjunct treatment to omeprazole. A low carbohydrate/high roughage diet is recommended, as is constant availability of hay and the addition of corn oil to the diet. As As the role of bacteria in the develop- ment of glandular ulceration remains controversial, antibiotic therapy is commonly recommended in addition to acid suppressing treatment, however recent studies have not demonstrated an im- provement in response to treatment with antibiotics and omeprazole when compared to treatment with omeprazole alone.

Recommendations for prevention of glandular ulceration are difficult to make due to the in- complete understanding of pathophysiology, risk factors and treatment strategies. Dietary chang- es to minimize soluble carbohydrates and maximize roughage in the diet, as well as the addition of corn oil, should be made. The development of glandular ulceration, and increase in severity of existing ulcers, have been demonstrated despite the use of prophylactic dosages of omeprazole. Despite this, however, treatment with omeprazole remains a recommendation for prevention until more definitive strategies are described. A pectin-lecithin complex in combination with an antacid and live yeast, as well as a formulation containing sea buckthorn berries have recently shown promise in the prevention of glandular ulceration.

Suggested reading:

Murray MJ, Nout YS, Ward DL. Endoscopic Findings of the Gastric Antrum and Pylorus in Horses: 162 cas- es (1996-2000). JVIM. 2001. 15: 401-406.

Sykes BW, Jokisalo JM. Rethinking equine gastric ulcer syndrome: Part 1- Terminology, clinical signs and diagnosis. EVE. 2014. 26(10): 543-547.

Sykes BW, Jokisalo JM. Rethinking equine gastric ulcer syndrome: Part 2- Equine squamous gastric ulcer syndrome (ESGUS). EVE. 2015. 27(5): 264-268.

Sykes BW, Jokisalo JM. Rethinking equine gastric ulcer syndrome: Part 3- Equine glandular gastric ulcer syndrome (EGGUS). EVE. 2015. 27(7): 372-375.

Videla R, Andrews FA. New Perspective in Equine Gastric Ulcer Syndrome. Vet Clin Equine. 25. 2009: 283- 301.

53 Evaluation and Management of the Recumbent Horse Rachel Gardner, DVM, DACVIM (LAIM) BW Furlong & Associates Oldwick, NJ

Evaluation and management of recumbent horses are challenging. Familiarity with diseases that can result in recumbency are important in reaching a diagnosis rapidly. Rapid and appro- priate diagnosis is necessary to formulate a reasonable prognosis and to develop an effective treatment and nursing plan.

Initial evaluation of the recumbent horse involves assessment of the entire situation, including the location of the horse and safety of the horse and personnel. The primary veterinarian must be observant, directive and methodical during the evaluation and initial treatment. A thorough history should be obtained, including information regarding vaccination status, travel history and activity prior to the onset of recumbency. All horses, regardless of vaccination status, should be treated as rabies suspects and appropriate biosecurity measures taken.

A physical exam, though limited by recumbency, should be as thorough as possible. The ini- tial goal of the physical exam is to determine if the patient is stable and determine a general cause for the recumbency. General causes of recumbency may be categorized as traumatic, neurologic, musculoskeletal, cardiopulmonary, abdominal discomfort or metabolic. Infectious, metabolic or toxic diseases should also be considered. A neurologic exam, thorough to the extent possible, should be performed following physical exam. The goal is to determine the presence or absence of neurologic signs, and if they are present, to determine a neuroanatomic diagnosis. Abnor- malities may initially be characterized as central nervous system (CNS) disorders involving the brain and/or spinal cord, peripheral nerve system, neuromuscular junction or multifocal disorders. When the physical and neurologic exams fail to elucidate a cause for the recumbency, the horse should be assisted to stand by tail support, sling or other means.

Diagnostic testing for a recumbent horse without a primary diagnosis should begin with a minimum database, including a complete blood count, serum biochemistry panel and urinalysis. Measurement of lactate and blood ammonia may also be helpful for diagnosis. Testing for spe- cific disease processes using titers, virus isolation and polymerase chain reaction (PCR) testing is commonly indicated. If neurologic signs are observed, a cerebrospinal fluid (CSF) tap may be performed. CSF is most easily obtained by atlanto-occipital (AO) puncture in the recumbent horse with the use of short-term general anesthesia. Myelography should be used to confirm cervical spinal cord compression if suspected. Radiography to locate the presence and extent of fractures should be used when trauma is suspected. Ultrasonography is useful for establishing soft tissue structures, bone integrity or body cavity effusion. Endoscopy may be used to evaluate the upper airway, guttural pouches and stylohyoid bones. An electrocardiogram should be performed in any recumbent patient with an arrhythmia. Transcranial magnetic stimulation is used to determine ab- normal nerve conduction along the descending motor tracts and determine the presence of spinal cord or peripheral nerve injury.

There are a myriad of disease or disorders that may result in recumbency, and reaching a diagnosis is important in forming a reasonable therapeutic plan and realistic prognosis. Disorders

54 can be broken down into several categories, including musculoskeletal, CNS, peripheral nervous or neuromuscular system, metabolic, respiratory or cardiovascular disorders.

Transport of the recumbent horse is challenging due to the size of the horse and because many recumbent horses become frightened and react violently to being moved. Thus, sedation and/or anesthesia may be needed. Recumbent horses may be moved on a flat surface with the coordinated effort of several people pulling on ropes tied to the down limbs. More effectively, a horse can be moved using a Large Animal Rescue Glide. The glide is a large sheet of conform- able plastic with handles and areas to hook ropes around the edges. The plastic material used for the glide slides easily over a variety of surfaces and the edges can be folded up to accommodate doorways. The UC Davis Large Animal Lift can be used to pull the horse onto the glide.

Management of the recumbent horse includes treatment of the primary disease and intensive supportive care. Many horses can bear some weight if assisted to stand and can spend consid- erable time upright with the assistance of a sling. By increasing time standing in a sling, adverse effects of recumbency can be minimized and the examiner is afforded an opportunity for more thorough exam. Nutritional support is an integral and challenging part of supportive care for the recumbent horse. Horses that are not dysphagic can be assisted into sternal recumbency and offered typical diets of long stem forage, concentrate feeds and water. Dysphagic or inappetant horses may be fed enteral diets or complete feed slurries through a nasogastric tube divided into multiple small feedings. In horses with gastrointestinal dysfunction in which enteral feeding is not possible, parenteral nutrition and intravenous fluid therapy should be considered.

Management of recumbent horses is challenging. In a recent evaluation of factors associ- ated with survival in recumbent horses, the presence of clinical signs for greater than 24 hours, the presence of a band neutrophilia, and the inability of horses to stand following treatment were associated with a higher risk of non-survival. Use of a sling was associated with lower risk of non-survival, and increasing cost of a case was associated with a lower risk of death.

Suggested reading:

Gardner RB. Evaluation and management of the recumbent adult horse. Vet Clin Equine. 2011. 27(3): 527-543.

Winfield LS, Kass PH, Magdesian KG, et al. Factors associated with survival in 148 recumbent horses. EVJ. 2014. 46: 575-578.

55 Nonexertional Rhabdomyolysis Stephanie Valberg, DVM, PhD, Diplomate ACVIM, ACVSMR

Immune Mediated Myopathies There are 3 newly recognized myopathies with an apparent immune-mediated origin that have recently been recognized in horses. The first myopathy manifests as acute, severe rhab- domyolysis1 the second presents as focal severe muscle swelling due to infarction;1,2 and the third myopathy is characterized by rapid muscle atrophy.1,3,4 Many, but not all, of the cases of immune-mediated myositis appear to be a sequella to infection with Streptococcus equi subspe- cies equi.

Acute rhabdomyolysis due to S. equi The small numbers of cases described in the literature are Quarter Horses less than 7 years of age. This may not reflect the prevalence of the disease, however, as many cases may not pres- ent to university hospitals or diagnostic laboratories.

Clinical signs: Affected horses usually have evidence of submandibular lymphadenopathy and/or guttural pouch empyema due to S. equi. Owners notice that horses develop a stiff gait which progresses rapidly to markedly firm, swollen, painful epaxial and gluteal muscles. Muscle pain becomes severe in spite of aggressive antimicrobial and anti-inflammatory treatment. The majority of reported cases became recumbent, was unable to rise, and developed unrelenting pain necessitating euthanasia within 24-48 hours of hospitalization.

Hematological abnormalities include mature neutrophilia, hyperfibrinogenemia, and marked elevations in creatine kinase (115,000 – 587,000 U/L), and aspartate aminotransferase activities (600- 14,500 U/L).Titers to the M protein of S. equi are low in affected horses, unless horses are recently vaccinated for strangles. Titers to another protein called myosin binding protein were high in a small number of horses that were tested.

At postmortem examination large, pale areas of necrotic muscle are evident in hindlimb and lumbar muscles. The histopathologic lesions are characterized by severe acute myonecrosis with a degreed of macrophage infiltration. Sublumbar muscles often show the most severe and chronic necrosis as indicated by greater macrophage infiltration of myofibers.

Pathogenesis: In human medicine, ß-hemolytic streptococci of Lancefield groups A, B, C, and G can cause severe myonecrosis manifested by severe myalgia, muscle swelling and sometimes toxic shock. Toxic shock arises as a result of profound non-specific T cell stimulation by strepto- coccal superantigens with the release of high levels of inflammatory cytokines. It is possible that horses with S. equi rhabdomyolysis also develop a toxic shock like syndrome as genes for four superantigens have recently been identified in S. equi. An alternative explanation for rhabdomy- olysis may be a bacteremia with local multiplication and production of exotoxins or proteases within skeletal muscle. S. equi virulence factors that may account for muscle necrosis include an unidentified cytotoxic protein, several proteases, streptokinase, and streptolysin S.Although, S.

56 equi has not been cultured in skeletal muscle from horses with rhabdomyolysis, S. equi bacteria have been identified in affected muscle using immunofluorescent stains for both Lancefield group C carbohydrate and S. equi M protein. There is currently no evidence that the S. equi involved is an atypical genetic strain of S. equi.

Treatment: A high mortality rate has been reported in horses receiving intravenous penicillin therapy once clinical signs of strangles and myopathy were well established. It is possible that early recognition of the signs of muscle stiffness in horses with S. equi infections and prompt aggressive treatment may be required for a successful outcome. Although streptococcal species are exquisitely susceptible to ß-lactam antibiotics, a mortality rate of 85% has been reported in human group A streptococcal myositis despite penicillin treatment. An antimicrobial that inhibits protein synthesis, such as rifampin, combined with intravenous penicillin, might enhance survival rates in horses with S. equi rhabdomyolysis. In addition flushing infected guttural pouches and draining abscessed lymph nodes will diminish the bacterial load. Nonsteroidal antiinflammatories and possibly high doses of short acting corticosteroids may assist in diminishing the inflammatory response. Control of unrelenting pain is a major challenge in horses with severe rhabdomyolysis. Constant rate infusion of lidocaine, detomidine or ketamine may provide better anxiety and pain relief than periodic injections of tranquilizers. Horses should be placed in a deeply bedded stall moved from side to side every 4 hours if they are unable to rise. Some horses may benefit from a sling if they will bear weight on their hind limbs when assisted to stand.

Infarctive Purpura Hemorrhagic

Prevalence: The prevalence of infarctive Purpura Hemorrhagica (PH) in one study was three out of a total of 53 PH cases reviewed.5 Five other cases of infarctive PH have been described in horses that were either exposed to S. equi within three weeks of presentation and or had marked- ly elevated serum ELISA M protein titers.2 Although horses with classic PH usually have a good prognosis, infarctive PH has a high fatality rate.

Clinical signs: The primary presenting complaint for horses with infarctive PH is often painful lameness, muscle stiffness and/or colic.2 Careful physical examination reveals classic signs of PH such as petechia and moderate well demarcated limb edema, however, in addition, horses with infarctive PH will have focal firm intramuscular swellings (figure 1). Horses with evidence of colic may have markedly decreased borborygmia and hemorrhagic gastric reflux.

57 Figure 1: infarctive hemorrhagic purpura. Arrow shows firm swelling in musculature.

Hematologic abnormalities usually include a leukocytosis characterized by a neutrophilia with a left shift and toxic change, hyperproteinemia, hypoalbuminemia and marked elevations in CK (47,000-280,000 U/L) and AST (960 -7,000 U/L) activities. Peritoneal fluid obtained by abdomi- nocentesis may be normal or may have an increased total protein, nucleated and red blood cell counts if gastrointestinal infarction is present.

Ultrasonographic examination of swollen muscle reveals focal hypoechoic lesions within mus- cle tissue. Biopsies of abnormal muscle show diffuse acute coagulative necrosis, whereas sam- ples from palpably normal muscle tissue show no pathological abnormalities.

Post-mortem findings of horses with infarctive PH show extensive infarction of the skeletal musculature, skin, gastrointestinal tract, pancreas, and lungs and S equi abscessation of a lymph node. Definitive histopathologic findings include leukocytoclastic vasculitis and acute coagulative necrosis resembling infarction in numerous tissues.

Pathogenesis: Infarctive PH resembles Henoch-Schönlein purpura in humans, which is char- acterized by infarctive vasculitis of the skin, kidneys and gastrointestinal tract due to IgA immune complex deposition. Immune complexes are present in the sera of horses with PH that appear to primarily be composed of IgM or IgA and streptococcal M protein. Deposition of complement near immune complexes in vessel walls may result in cell membrane destruction, cell death and vascular occlusion. The distinctive feature of infarctive PH in horses is the extensive infarction of skeletal muscle and consequently marked elevation in serum CK and AST activity.

Treatment: Early recognition of focal muscle swelling, abdominal discomfort, neutrophil- ia, hypoalbuminemia and marked elevations in CK activity combined with aggressive antibiotic and corticosteroid treatment may enhance the likelihood of a successful outcome. Treatment of Henoch-Schönlein purpura in humans, including cases with intestinal infarctions, involves high dose intravenous pulse therapy with methylprednisolone (1000mg/m2 every other day for three treatments) followed by oral corticosteroids plus immunosuppressive agents such as cyclophos-

58 phamide and azathioprine. One horse with infarctive PH was successfully treated with penicillin, nonsteroidal antiinflammatories and three weeks of dexamethasone (0.1- 0.07mg/kg) followed by a ten week tapering course of oral prednisolone (2 mg/kg initially).

Immune-mediated polymyositis Prevalence: Immune-mediated polymyositis (IMM) has recently been reported in horses.4,6 The affected horses are primarily of Quarter horse related bloodlines, although two ponies, one Icelan- dic horse and a Thoroughbred have been described with IMM. A bimodal age distribution seems to occur in affected horses with all horses identified to date either ≤ 8 yrs of age or ≥ 16 yrs of age. In approximately 1/3 of horses with IMM a triggering factor appears to have been exposure to S. equi or a respiratory disease.

Genetics: IMM in humans is believed to have a nonMendelian polygenic pattern of inheri- tance. The high prevalence of the disorder in Quarter horse suggests that there is the potential for a polygenic mode of inheritance in this breed.

Clinical Signs: The most prominent clinical sign of IMM in Quarter horses is rapid onset of muscle atrophy, particularly affecting the back and croup muscles (figure 2), accompanied by stiffness and malaise. Atrophy may progress to involve 50% of the horses’ muscle mass within a week and may lead to generalized weakness. Focal symmetrical atrophy of cervical muscles has been reported in a pony with IMM.

Figure 2. Marked atrophy of epaxial and gluteal muscles with immune-me- diated myositis.

Hematologic abnormalities are relatively minor in affected horses and are usually re- stricted to mild to moderate elevations in serum CK and AST activity.However, in some cases serum muscle enzyme activities are normal.

Diagnosis: Muscle tissue obtained from the epaxial and gluteal muscles contains many of the following abnormalities, lymphocytic vasculitis, anguloid atrophy, lymphocytic myofiber

59 infiltration, fiber necrosis with macrophage infiltration and regeneration. Biopsies of semitendino- sus or membranosus muscles may show some evidence of atrophy and vasculitis but significant inflammatory infiltrates may be absent in these tissues. The extent of the inflammatory infiltrates in epaxial muscles is such that a diagnosis can often be established from several formalin fixed Trucut samples.

Pathogenesis: The lymphocytic infiltrate seen in muscle samples from horses with IMM is distinct from that found in dogs and humans with immune-mediated polymyositis, in that the CD4:CD8 ratio in horses appears higher In contrast to immune-mediated masticatory muscle myositis which does have a higher CD4:Cd8 ratio, the specific binding of IgG to myofibers seen in canine masticatory muscle is not a feature of equine IMM. The reason why specific muscle groups are affected in horses with immune mediated polymyositis is unclear.

Treatment: Horses with concurrent evidence of streptococcal infection should be treated with antibiotics. It is likely prudent to avoid intramuscular injections. Administration of corticoste- roids appears to immediately improve signs of malaise and inappetence and prevented further progression of muscle atrophy. Recommended dosages are: dexamethasone (0.05 mg/kg) for three days, followed by prednisolone (1 mg/kg for 7 to 10 days) tapered by 100 mg/week over one month. Serum CK activity often normalizes after 7 – 10 days. Muscle mass will usually grad- ually recover over two-to-three months.

Horses that are not treated with corticosteroids may develop extensive muscle atrophy but in many cases muscle mass will gradually recover. Recurrence of atrophy in susceptible horses is common and may require reintroduction of corticosteroid therapy. Some horses devel- op focal residual muscle atrophy.

References (1) Sponseller BT, Valberg SJ, Tennent-Brown BS, Foreman JH, Kumar P, Timoney JF. Severe acute rhabdomyolysis associated with Streptococcus equi infection in four horses. J Am Vet Med Assoc. 2005;227:1800-1804. (2) Kaese HJ, Valberg SJ, Hayden DW et al. Infarctive purpura hemorrhagica in five horses. J Am Vet Med Assoc. 2005;226:1893-8, 1845. (3) Lewis S., Valberg S.J., and Nielsen I.L. Suspected Immune-mediated Myositis in Horses. J.Vet. Intern.Med. (in press) (4) Valberg SJ, Bullock P, Hogetvedt W, Ames TA, Hayden DW, and Ott K. Myopathies associated with Streptococcus equi infections in horses. Proc.Amer.Assoc.Equine Pract. 292-293. 1996. (5) Pusterla N, Watson JL, Affolter VK, Magdesian KG, Wilson WD, Carlson GP. Purpura haemor- rhagica in 53 horses. Vet Rec. 2003;153:118-121.

60 Seasonal Pasture Myopathy in the Midwestern USA

A retrospective case series (14 horses) was recently performed in Minnesota that described clin- ical signs, diagnostic findings, tissue tremetone concentrations, and clinical outcome or postmor- tem findings of horses evaluated for acute severe nonexertional rhabdomyolysis initially attributed to white snakeroot toxicosis.{Finno, 2006 438 /id}

Clinical signs: The most common clinical signs of this myopathy were myoglobinuria, gener- alized weakness, muscle fasiculations, lethargy, and prolonged recumbency. Elevated heart and respiratory rates were present and most horses were initially thought to have colic. Horses did not have firm painful muscles typical of exertional myopathies. Serum CK activity ranged from 46,487 to 959,499 U/L (reference range 82-449 U/L) and aspartate transaminase activity was > 1,500 U/L (reference range 162-316 U/L). While all horses suffered from skeletal muscle necrosis, 50% were also found to have myocardial degeneration. All horses were kept on pasture >12 h a day without snow cover when minimum daily temperatures were from 29oF to 56°F and weather was often inclement. Thirteen of 14 horses developed the disorder in the fall and many farms had a history of horses previously dying from similar clinical signs.

Post mortem findings: Only two horses survived with aggressive antoxidant and anti-inflam- matory treatment. Postmortem examination revealed acute severe myonecrosis primarily in neck, proximal fore and hind limb, intercostal, and diaphragm muscles. Frozen intercostal, diaphragm or muscles surrounding the hip joint stained darkly for the presence of lipid (oil red O stain), which was an unusual common finding suggesting a similar underlying mechanism for necrosis.

Pathophysiology: Vitamin E and selenium concentrations were within reference range in all horses evaluated suggesting that the myopathy was not related to nutitional myodegeneration. Clinical signs were similar to those reported for monensin toxicosis in horses, however, none of the horses received significant supplemental grain (the usual source of monensin). The seasonal occurrence of the myopathy as well as the acute clinical signs of muscle fasiculations, weakness, esophageal obstruction, colic, and pigmenturia were consistent with white snakeroot toxicosis. However, the toxic component of white snake root, tremetone, was not detected in liver or urine samples of any horses.Weather conditions, seasonality, clinical signs, clinicopathologic findings, mortality rate, and postmortem findings of minesota cases closely resembled those associated with atypical myopathy (AM) reported in European countries where white snakeroot does not grow. Atypical myopathy affects postural and respiratory muscles as well as the myocardium and produces lipid accumulation in these muscles, In Minnesota, the majority of affected horses are isolated cases, whereas as many as 115 horses in a pasture may be affected in an outbreak of AM. The cause of AM is suspected to be an ingested or enterically produced toxin (eg, bacterial toxin, mycotoxin, or phytotoxin). AM was reported to cause 51 deaths in horses in Europe this fall.

Treatment: Impaired antioxidant capacity may play a role in the pathogenesis of AM since the primary muscles affected by the disorder are highly oxidative and develop marked lipid accumu- lation. In our study, of the 2 horses that survived, both presented within 4 h of clinical signs de- veloping. One of these horses was treated with phenylbutazone (2.2 mg/kg [1 mg/lb], PO, q12h) and vitamin E (5,000 U, PO, q24h). Serum CK activity in that horse decreased during a period of 5 days (serum CK activity at admission, 46,487 U/L; 1 day after admission, 29,842 U/L; 5 days

61 after admission, 1,426 U/L). A follow-up examination performed in that horse 6 months after dis- charge revealed no residual clinical signs. The other horse responded to treatment with vitamin E and seleniumd (0.02 ml/kg [0.01 ml/lb], IM) once followed by administration of vitamin E (5,000 U, PO q24h), vitamin C (5,000 mg in 5 L of lactated Ringer’s solution given IV over 12 hours q24h), 2.5% solution of dimethyl sulfoxide in lactated Ringer’s solution given IV over 4 hours q24h for 3 days), flunixin meglumine (1.1 mg/kg [0.5 mg/lb], IV, q12h), and balanced polyionic fluids (2.2 mg/ kg/h [1.1 mg/lb/h], IV). Serum CK activity in this horse increased from 256,103 U/L at admission to 264,359 U/L one day after admission, then subsequently decreased to 31,524 U/L three days after admission.

Clinical Relevance: Cases of rhabdomyolysis have been attributed to the toxin trematone in white snakeroot; however, tremetone was not identified in our study of horses with a pasture myopathy. A seasonal myopathy, characterized by primary acute severe skeletal muscle necrosis and, in many cases, myocardial degeneration exists in Minnesota. numerous similarities exist among cases of seasonal pasture myopathy and AM in Europe.

References

1. Finno CM, Valberg SJ, Wunschmann A and Murphy M. Pasture Myopathy in 14 horses in the Midwest- ern USA. J Am Vet Med Assoc 2006;229 (7):1134-1141.

2. International Veterinary Information Service. Atypical myopathy (atypical myoglobinuria). Available at: http://www.ivis.org/reviews/rev/votion/chapter.asp. Accessed Jun 1, 2005.

3. Hosie BD, Gould PW, Hunter AR, et al. Acute myopathy in horses at grass in east and south east Scot- land. Vet Rec 1986;119:444-449.

4. Harris P. Differential diagnosis of an acute episode of primary myopathy out at pasture. Equine Vet Educ 1996;8:272-276.

5. Whitwell KE, Harris P, Farrington PG. Atypical myoglobinuria: an acute myopathy in grazing horses. Equine Vet J 1988;20:357-363.

6. Thompson LJ. Depression and choke in a horse: probable white snakeroot toxicosis. Vet Hum Toxicol 1989;31:321-322.

62 Glycogen Branching Enzyme Deficiency Stephanie J Valberg, DVM PhD, Diplomate ACVIM, and Jim Mickelson PhD

Glycogen branching enzyme deficiency (GBED) is a glycogen storage disorder that recently has been identified in neonatal Quarter Horse foals or aborted feti.1-3 GBED is an autosomal re- cessive trait in Quarter Horses and Paint Horses. Thoroughbreds have been screened for GBED without finding this genetic mutation. The disease is due to a nonsense mutation in exon 1 of the GBE1 gene which introduces a premature stop codon. In the homozygous state, this mutation markedly reduces the function of the glycogen branching enzyme.2, 4 Carriers of GBED trace back to the sire King P234 in most cases, however, King’s sire Zantanon may also have carried GBED. It is not possible to use pedigree analysis to diagnose GBED as the majority of Quarter Horses are descendants of these two stallions. GBED has likely been in the Quarter at least since its inception in 1940.

Prevalence: Approximately 8% of both Quarter and Paint Horses are carriers of GBED.5 GBED was detected in 2 - 4% of 2nd and 3rd trimester abortions submitted to two diagnostic labo- ratories.5 Breeding farms with stallions that are carriers of GBED could expect a higher incidence of abortion due to GBED. In addition, published reports indicate that at least 11 foals have been born with GBED.3, 6 The incidence of GBED in neonatal foals is likely much higher than this be- cause many foals have likely gone undiagnosed.

Diagnosis: Muscle biopsy specimens from foals with GBED often, but do not always, contain basophilic globules and eosinophilic crystalline material in hematoxylin and eosin stains.2, 5 Peri- od acid Schiff’s stains show decreased normal background staining for glycogen and PAS positive globular inclusions with, in some cases, additional smaller crystalline inclusions.2 At postmortem examination, globular inclusions are readily apparent in the Purkinje cells of the myocardium and inclusions may also be found in cardiac myocytes. Abnormal polysaccharide can be identified in neural tissue and is inconsistently found in the liver.3 The most accurate diagnosis of GBED can be obtained through genetic testing. The Veteri- nary Genetics Laboratory at the University of California, Davis (www.vgl.ucdavis.edu) is licensed by the University of Minnesota to test for GBED. Mane or tail hairs with roots intact or fetal liver tissue can be submitted to identify if horses are homozygous or heterozygous for GBED

Clinical Signs: Fetal abortion is a common presentation of GBED.2, 6, 7 Foals that survive to parturition are often hypothermic and weak but gain strength when given milk by bottle feeding or through assistance to stand and nurse. Correctable flexural deformities of all 4 limbs are common in GBED foals. Progression of signs can be highly variable. Some foals have early onset of venti- latory failure and die even with mechanical ventilation. Other foals show intermittent collapse due to hypoglycemia, particularly if access to suckling is restricted. Sudden death is reported in some foals whereas others are euthanized due to muscle weakness and inability to rise.1, 2, 6 Most GBED affected foals die or are euthanized by 8 weeks of age, however, one foal survived with nursing care to 18 weeks of age. Common hematological findings in foals with GBED include a low white blood cell count, of- ten about 4,000 cells/ul, as well as moderate elevations in serum creatine kinase (CK), aspartate transaminase (AST) and gama glutamyl transferase (GGT).2

63 Pathogenesis: Glycogen is an extremely important energy source for the rapidly growing and developing fetus and neonate. It is synthesized by two enzymes, glycogen synthase, which cre- ates straight chains of glucose with alpha 1,4-glycosidic linkages and GBE which creates branch- es of glucose through alpha 1,6-linkages. Glycogen thereby becomes a compact, highly-branched and energy-dense molecule. Tissues from GBED foals have no measurable GBE enzyme activity or immuno-detectable GBE and are therefore unable to form normally branched glycogen2. As a result, tissues such as cardiac and skeletal muscle, liver and the brain cannot store and mobilize glycogen to maintain normal glucose homeostasis. The absence of normally branched glycogen molecules in individuals that possess GBE1 mutations appears to cause a fatal flaw in glucose ho- meostasis and metabolism in the fetus as well as the newborn foal resulting in muscle weakness, hypoglycemia, seizures and death. GBED has a similar clinical, biochemical and molecular basis to Glycogen Storage Disease Type IV, which has been described in humans and Norwegian forest cats 8, 9

Treatment: There is no treatment for GBED. Early recognition and euthanasia can save con- siderable expense for owners of foals in neonatal intensive care units. It is important that veter- inarians and breeders recognize that GBED may present both as foals born alive which subse- quently succumb to GBED, as well abortion. Many stallion owners offer a free repeat breeding to owners that lose foals to GBED, and if a diagnosis is not established the owner will have a 25% chance of having another GBED affected offspring. Since histologic changes are not always present, PCR analysis for the GBE1 mutation would appear to be the most accurate addition to postmortem diagnostic tests currently used to evaluate aborted feti and neonatal foals of Quarter Horse-related breeds.5

References 1. Sponseller BT, Valberg SJ, Ward T, Williams AJ, Mickelson JR. Muscular weakness and recumbency in a quarter horse colt due to glycogen branching enzyme deficiency. Equine Vet Educ. 2003;14:182-188. 2. Valberg SJ, Ward TL, Rush B, et al. Glycogen branching enzyme deficiency in quarter horse foals. J Vet Intern Med. 2001;15:572-580. 3. Render JA, Common RS, Kennedy FA, Jones MZ, Fyfe JC. Amylopectinosis in fetal and neonatal quarter horses. Vet Pathol. 1999;36:157-160. 4. Ward TL, Valberg SJ, Lear TL, et al. Genetic mapping of GBE1 and its association with glycogen storage disease IV in american quarter horses. Cytogenet Genome Res. 2003;102:201-206. 5. Wagner ML, Valberg SJ, Ames EG, et al. Allele frequency and likely impact of the glycogen branch- ing enzyme deficiency gene in quarter horse and paint horse populations. J vet Int Med. 2006;Sep- Oct;20(5):1207-11 6. Ward TL, Valberg SJ, Adelson DL, Abbey CA, Binns MM, Mickelson JR. Glycogen branching enzyme (GBE1) mutation causing equine glycogen storage disease IV. Mamm Genome. 2004;15:570-577. 7. Render JA, Common RS, Kennedy FA, Jones MZ, Fyfe JC. Amylopectinosis in fetal and neonatal quarter horses. Vet Pathol. 1999;36:157-160. 8. Tsujino S. Glycogen branching enzyme deficiency (Andersen disease). Ryoikibetsu Shokogun Shirizu. 2001;(36):23-24. 9. Fyfe JC, Giger U, Van Winkle TJ, et al. Glycogen storage disease type IV: Inherited deficiency of branch- ing enzyme activity in cats. Pediatr Res. 1992;32:719-725.

64 Exertional Rhabdomyolysis - Causes of Poor Performance Stephanie J Valberg, DVM PhD, DACVIM, DACVSMR

Optimal function of skeletal muscle is essential for successful athletic performance. Even minor derangements in locomotor muscle function will impact power output, coordination, stamina and desire to work during exercise. While many myopathies are easy to recognize in the acute stages, low grade muscle strains or weakness and chronic myopathies may be difficult to diagnose. The challenges of identifying their contribution to poor performance in horses include; 1) differentiating between true pain with exercise and an uncooperative attitude 2) locating and assigning significance to focal muscle strain 3) determining the degree to which orthopedic pain contributes to muscle pain 4) determining the degree to which myopathies contribute to orthopedic pain 5) identifying muscle weakness as a component of gait alterations 6) differentiating myogenic from neurogenic weakness Classification of Exertional Myopathies Horses with exertional myopathies often fall into one of two main categories; 1) horses in which an intrinsic muscle defect does not appear to be present, but a temporary imbalance within the muscle cells causes a sporadic exertional myopathy and; 2) horses in which the primary underly- ing susceptibility appears to be the result of an intrinsic defect in the muscle resulting in a chronic exertional myopathy. Chronic Exertional Myopathies Known causes of chronic ER include recurrent exertional rhabdomyolysis, type 1 and type 1 PSSM and Malignant Hyperthermia. There may well be yet other unrecognized causes.

Recurrent Exertional Rhabdomyolysis (RER): RER refers to a subset of chronic exertional myopathies affecting approximately 5% of Thoroughbreds and Standardbreds.that is believed to be due to an abnormality in intracellular calcium regulation that is intermittently manifested during exercise. Although, several characteristics of RER muscle are very similar to those of humans and swine with malignant hyperthermia (MH), a defect in the ryandodine receptor associated with MH has not been identified in RER horses. At present the exact defect in intracellular calcium reg- ulation with RER is not known. Mares more commonly show signs of RER than males, however, no general correlation has been observed between episodes of rhabdomyolysis and stages of the estrus cycle. Nervous horses (usually young fillies) have a higher incidence of rhabdomyolysis than calm horses. Horses on a high grain diet are more likely to show signs of RER, and one study found a higher prevalence of rhabdomyolysis among horses with lameness.

Genetics: A genetic susceptibility to RER appears to exist in Thoroughbred horses where RER-afflicted horses may pass the trait along to 50% or more of their offspring. Studies of Stan- dardbred horses with RER suggest that there is potentially a heritable basis for this condition in this breed as well. There are anecdotal reports of higher prevalence of RER in certain families.

Diagnosis: A presumptive diagnosis of RER is based on clinical signs of muscle pain and

65 the presence of risk factors commonly associated with RER. Skeletal muscle biopsies from Thor- oughbred and Standardbred horses with active signs of RER often show an increased number of mature muscle fibers with centrally displaced nuclei, increased subsarcolemmal staining for gly- cogen, and a variable amount of muscle necrosis and regeneration.15 There is a notable absence of abnormal amylase-resistant polysaccharide in muscle biopsies from RER horses. Malignant Hyperthermia (MH): MH is due to an autosomal dominant mutation in the skel- etal muscle ryanodine receptor and is present in 1% or less of American Quarter Horses and Paints. Affected horses may intermittently show signs of tying up and high body temperatures. Some MH affected horses have died suddenly after an episode of tying up. The MH defect can co-exist with the GYS1 mutation for type 1 PSSM. The combination of these two mutations makes signs of tying-up more severe, increases recurrence of high serum CK and makes horses more resistant to improvement with changes in diet and exercise. In addition, horses with MH may de- velop classic signs under general anesthesia of excessive body temperature, rigor, metabolic aci- dosis and death. Genetic testing is recommended in Quarter Horse and Paint horses with difficult to manage forms of PSSM or a family history of post-anesthetic complications. Testing is available through the Veterinary Diagnostic Laboratory at the University of Minnesota http://www.vdl.umn. edu/vdl/ourservices/neuromuscular.html and University of California, Davis. Polysaccharide Storage Myopathy: Several acronyms have been used for polysaccha- ridestorage myopathy besides PSSM including EPSM and EPSSM and debate existed as to whether these acronyms encompassed one muscle condition. In 2008, the presence of amylase-resistant polysaccharide in skeletal muscle from Quarter Horses was found to be highly associated with a mutation in the glycogen synthase 1 gene (GYS1). Some cases previously diagnosed with PSSM by muscle biopsy, particularly those with amylase-sensitive glycogen, did not possess the genetic mutation suggesting that there are at least two forms of PSSM9. For clarity, the form of PSSM caused by a GYS1 mutation is now termed type 1 PSSM whereas the form of PSSM that is not caused by the GYS1 mutation and whose origin is yet unknown is now termed type 2 PSSM1.

Type 1 PSSM: The GYS1 mutation responsible for type 1 PSSM is present in over 20 different horse breeds. The highest prevalence of PSSM appears to occur in draft horses derived from Continental Europe- an breeds,in contrast, the prevalence of PSSM is very low prevalence in breeds such as Shires and Clydes- dales. Prevalence in Quarter Horses range from 6 to 10% and 6 to 8% for American Paint and Appaloosa horses. The highest frequency of Type 1 PSSM occurs in halter Quarter Horses (28% affected) and the lowest frequency in racing Quarter Horses. The GYS1 mutation has been identified in approximately 72% of Quarter Horses diagnosed with PSSM by muscle biopsy and in 18% of Warmbloods of a variety of types diagnosed with PSSM by muscle biopsy9. The prevalence of type 1 PSSM is very low in light horse breeds such as Arabians and Thoroughbreds.

Etiology: Glycogen is formed by the enzymes glycogen synthase and branching enzyme. The autosomal dominant GYS1 mutation produces a gene product, glycogen synthase, which has an arginine substitution for histidine at codon 309. The effect of this amino acid substitution is a higher than normal activity of glycogen synthase both at rest and when activated by glucose 6-phosphate. As a result, skeletal muscle of PSSM horses has 1.5 to 4 fold higher concentrations of glycogen than normal horse muscle. The accumulation of abnormal polysaccharide is in itself not the cause of muscle dysfunction in PSSM since foals as young as 1 month of age may show evidence of muscle damage prior to the formation of abnormal polysaccharide in skeletal muscle

66 fibers. Rather, the persistent glycogen synthase activity in type 1 PSSM horse muscle appears to disturb the normal flux of muscle energy metabolism during exercise. Acute clinical signs: Horses usually show signs of PSSM at on average 6 years of age, how- ever, this can range from 1 to 14 years of age and some horses are asymptomatic. In general, owners describe horses with type 1 PSSM as having a calm and sedate demeanor. Acute clinical signs include tucking up of the abdomen, fasciculations in the flank, muscle stiffness, sweating reluctance to move forward and overt muscle contractures. The hindquarters are frequently most affected, but back muscles, abdomen, and forelimb muscles may also be involved. Signs of pain can last for more than 2 hours and about 10% of cases becoming recumbent. Muscle pain often occurs with less than 20 minutes of exercise at a walk and trot especially when horses are unfit at the commencement of training or after horses have had a substantial period of rest. A diet high in NSC exacerbates theses signs. During an acute episode of ER, horses with type 1 PSSM often have markedly elevated serum CK activity of >35,000 U/L and myoglobinuria may be present. Severe colic-like pain post-exercise and myoglobinuric renal failure are less common presenting complaints. Some owners report a seasonal incidence to development of acute clinical signs which some have attributed to quality of grass available at the time. Chronic clinical signs: Light breeds: In riding horses a lack of energy when under , re- luctance to move forward, stopping and stretching out as if to urinate and a sour attitude towards exercise occur. Horses may have a combination of low grade reluctance to exercise, poor perfor- mance and repeated episodes of ER. The range of severity of clinical signs of PSSM can be wide with some horses being asymptomatic and others completely incapacitated. Serum CK activities are often elevated in untreated Quarter Horses, even when horses are rested. While horses are symptomatic, CK will usually increase by 1000 U/L or more 4 hours after 15 minutes of exercise at a trot. The median CK and AST activity for all PSSM Quarter Horses with muscle biopsies sub- mitted to the University of Minnesota was 2,809 and 1,792 U/L, respectively. Affected Quarter and Paint Horse foals and weanlings may develop rhabdomyolysis without exercise. and Draft Crosses: The average age of draft horses diagnosed with PSSM is about eight years of age18. Many draft horses with PSSM are asymptomatic. Signs of severe rhabdomyolysis and myoglobinuria may occur in horses fed high grain diets, exercised irregularly with little turn out or horses that undergo general anesthesia. Other signs of PSSM in draft horses include progressive weakness and muscle loss resulting in difficulty rising in horses with normal serum CK activity. Pronounced weakness is more prevalent in homozygotes for the GYS1 muta- tion. Gait abnormalities, such as excessive limb flexion, fasciculations, and trembling are also re- ported in draft horses. Although the condition Shivers was previously attributed to PSSM, a recent study found no causal association between these two conditions. The median serum CK and AST activities in draft horses from which biopsies were sent to the University of Minnesota was 459 and 537 U/L, respectively. Diagnosis: A genetic test for the GYS1 mutation can be performed on whole blood or hair root samples in North America at the University of Minnesota (http://www.vdl.umn.edu/vdl/ourservices/ neuromuscular.html) and in Europe by Laboklin (http://www.laboklin.co.uk/laboklin/GeneticDis- eases.jsp). MH testing is provided at the University of Minnesota and the University of California, Davis. Muscle biopsy can also provide a means to diagnose type 1 PSSM. The distinctive fea- tures of type 1 PSSM in muscle biopsy samples are numerous subsarcolemmal vacuoles and dense, crystalline periodic acid Schiff’s (PAS) positive, amylase resistant inclusions in fast twitch fibers. Genetic testing provides the gold standard for diagnosis because a false negative diagno-

67 sis of type 1 PSSM by muscle biopsy may occur if biopsy samples are small or if horses are less than 1 year of age. Type 2 PSSM: There is much less known about type 2 PSSM, because as it turns out, pre- vious research on PSSM has largely involved horses with type 1 PSSM. Current knowledge of type 2 PSSM is based on retrospective evaluation of cases diagnosed with PSSM by muscle biopsy that are now known be free of the GYS1 mutation and a few years of prospective clinical cases. Approximately 28% of cases of PSSM diagnosed by muscle biopsy in Quarter Horses do not have the GYS1 mutation. Type 2 PSSM seems to be more common in higher performance horses such as barrel racing, reining and cutting horses compared to the high prevalence of type 1 PSSM in halter horses. About 80% of cases of PSSM diagnosed by biopsy in Warmbloods have type 2 PSSM. Breeds affected include Dutch Warmbloods, Swedish Warmbloods, Hanoverians, Friesians, Selle Francais, Westfalian, Canadian Warmblood, Irish , Gerdlander, Hus- sien, and Icelandic horses. Many other light breeds have also been diagnosed with type 2 PSSM including Morgans, Standardbreds and Thoroughbreds. Type 2 PSSM also occurs in Arabians, however, in my experience this breed is distinct in that it often has amylase-resistant rather than amylase sensitive polysaccharide but is negative for the GYS1 mutation. Etiology: The cause of type 2 PSSM is currently unknown. It may well be that there are a group of conditions that have separate etiologies but share common findings of glycogen accu- mulation and poor performance. A heritable predisposition is suspected in Quarter Horses but yet to be proven. Acute Clinical signs: Horses with type 2 PSSM do not necessarily have the same calm tem- perament as horses with type 1 PSSM. In adults, acute clinical signs of rhabdomyolysis are similar between type 1 and type 2 PSSM. Muscle atrophy after rhabdomyolysis is a common complaint in Quarter Horses with type 2 PSSM. There are more Quarter Horses less than one year of age reported with type 2 PSSM than type 1 PSSM and these foals may present with an inability to rise or a stiff hind limb gait. Chronic clinical signs: Chronic signs of type 2 PSSM are often most closely related to poor performance rather than recurrent ER and elevations in serum CK activity. An undiagnosed gait abnormality, sore muscles and drop in energy level and willingness to perform after 5 -10 min of exercise are common complaints in Quarter Horses with type 2 PSSM. Warmbloods with type 2 PSSM have painful firm back and hindquarter muscles, reluctance to collect and engage the hind- quarters, poor rounding over fences, gait abnormalities, and slow onset of atrophy. The mean age of onset of clinical signs in Warmbloods is between 8 and 11 years of age with the median CK and AST activity being 323 and 331U/L, respectively. Diagnosis: Type 2 PSSM must be diagnosed by muscle biopsy where increased or abnormal PAS positive material that is usually amylase-sensitive is apparent particularly in subsarcolemmal locations. Determination of what constitutes an abnormal amount of amylase-sensitive glycogen can be subjective. False positive diagnosis is possible for type 2 PSSM in highly trained horses that normally have higher muscle glycogen concentrations or in formalin fixed sections which show a greater deposition of subsarcolemmal glycogen even in healthy horses. Other histopatho- logical features that may be present with both type 1 and type 2 PSSM include muscle necrosis, macrophage infiltration of myofibers, regenerative fibers, and fiber atrophy. Some laboratories grade polysaccharide accumulation as mild, moderate, and severe where mild accumulation rep- resents a category which has a higher chance of being a false positive diagnosis. Mild PSSM

68 cases in particular should receive a full physical examination to ensure that there are not other underlying causes for performance problems. Management of Exertional Myopathies Regular daily exercise and daily turn out for as long as possible are key to the health of horses with chronic forms of ER. Diet change alone is not effective in managing chronic ER and must be combined with regular exercise. The results of standardized nutrition trials for RER thoroughbreds and PSSM1 Quarter Hors- es show that provision of a diet with a reduced NSC content and higher fat content than conven- tional concentrates reduces serum CK activity. These dietary recommendations differ between RER and PSSM in that PSSM horses have lower caloric requirements and lower recommended starch and fat content as a % of daily DE. With RER this diet reduces indices of nervousness which appear to impact ER and with PSSM1 they enhance glycogen utilization and fat metabolism as an alternate energy source. Caloric balance: A nutritionally balanced diet with appropriate caloric intake and adequate vitamins and minerals are the core elements of treating chronic forms of ER. For RER Thorough- breds and Standardbreds in training, the challenge is usually supplying enough calories in a highly palatable form to meet their daily energy demands. This is in part because they often require >30 MCal of DE a day and because with their nervous temperament they may be more discriminating in their eating habits. Many horses with PSSM are easy keepers and may be overweight at the time of diagnosis. Adding excessive calories in the form of fat to an obese horse may produce metabolic syndrome and is contraindicated. If necessary, caloric intake should be reduced by us- ing a grazing muzzle during turn-out, feeding hay with a low nonstructural carbohydrate content (NSC) at 1 to 1.5% of body weight, providing a low calorie ration balancer and gradually introduc- ing daily exercise. Rather than provide dietary fat to an overweight horse, fasting for 6 h prior to exercise can be used to elevate plasma free fatty acids prior to exercise and alleviate any restric- tions in energy metabolism in muscle. Selection of forage: Thoroughbred horses do not appear to show the same significant in- crease in serum insulin concentrations in response to consuming hay with an NSC of 17% as seen in Quarter horses. This fact combined with the high caloric requirements of racehorses may mean that it is not as important to select hay with very low NSC content in RER thoroughbreds as it is in PSSM horses. The degree to which the NSC content of hay should be restricted below 12% NSC depends upon the caloric requirements of a PSSM horse. Feeding a low NSC hay of 4% provides room to add an adequate amount of fat to the diet of easy keepers without exceeding the daily caloric requirement and inducing excessive weight gain. Low starch high fat concentrates: A controlled trial using the first specialized feed developed for RER (Re-Leve 13% fat by weight and 9% NSC), determined that, NSC should be no greater than 20% of daily DE and 20-25% of daily DE should be provided by fat for optimal management of RER horses fed 30 MCal or more/day. The principle consideration for amount of fat fed to PSSM horses should be whether this provides excessive calories and additional weight gain. Fat can be added such that it constitutes 13% DE in the form of oil added to a ration balancer or hay cubes. Medication: Tranquilizers may be of value in treating excitable horses prone to RER. Dantro- lene when given to RER horses 60 to 90 minutes prior to exercise appears to attenuate muscle damage To date there are no tested management strategies for horses with MH, although dan-

69 trolene seems like a reasonable but expensive approach. Horses with both MH and PSSM have been shown to respond to high fat low starch diets although their response is not as favorable as horses with PSSM alone.

aRe-Leve Kentucky Equine Research Versailles KY 40383 Conflict of Interest statement: Drs. Valberg, Mickelson and McCue own the license for PSSM testing and receive sales income from its use. Their financial and business interests have been reviewed and managed by the University in accordance with its conflict of interest policies. A portion of the profits from the sale of Re-Leve go to Dr. Valberg and her research.

References at: http://www.cvm.umn.edu/umec/lab/home.html

70 Equine Genetic Diseases - Genetic Testing: What Is Available And When To Use It Stephanie J Valberg DVM Ph.D University of Minnesota

The selective breeding of animal populations may give rise to a common founder that can disseminate a genetic trait to many thousands of related offspring within a few years. The number of DNA mutation is gradually increasing and there will likely be many more identified in the near future with the rapid development of genetic tools specific for horses.

The pattern of inheritance of these traits is either autosomal dominant or autosomal reces- sive. Autosomal dominant traits: Require only one copy of the mutant gene to cause disease. Breeding of an affected heterozygous horse (one copy of the defective gene) to a normal horse results in a 50% chance of producing an affected horse. Breeding two heterozygous affected horses has a 50% chance of producing a heterozygous affected, 25% chance of a homozygous affected (2 copies of the defective gene) and a 25% chance of a homozygous normal being born. Autosomal recessive traits: Requires two copies of the mutant gene to cause disease. Breeding two affected horses results in a 100% chance of producing an affected horse. Breeding two car- riers results in a 25% chance of producing an affected horse, a 25% chance of a normal horse and a 50% chance of a carrier. Breeding and affected and a carrier results in a 50% chance of producing an affected horse and a 50% chance of producing a carrier. Equine Genetic Diseases that have commercially available tests

Defect Breed Mode of Inheritance Genetic Test Available

HYPP QH, Paint, APP A-Dominant yes

GBED QH, Paint A-Recessive yes

HERDA Quarter Horses A-Recessive yes

MH Quarter Horses A-Dominant yes

PSSM type 1 20 breeds: QH, Paints, Morgan, Belgian, Percheron, some Warmbloods… A-Dominant yes

OLWS Paint, Pinto A-Recessive yes

JEB Belgian A-Recessive yes

JEB Saddlebred A-Recessive yes

SCID Arabian A-Recessive yes

Lavender Arabian A-Recessive yes foal syndrome

Cerebellar Arabian A-Recessive yes abiotrophy

Fell Pony Fell Pony Syndrome A-Recessive yes Syndrome 71 Genetic Disorders of Quarter Horses

Five genetic diseases have been found in Quarter Horses that have genetic tests commer- cially available. The 5 known genetic mutations in Quarter Horses include Hyperkalemic Periodic Paralysis (HYPP), Glycogen Branching Enzyme Deficiency (GBED), Hereditary Equine Regional Dermal asthenia (HERDA), type 1 Polysaccharide Storage Myopathy (PSSM1) and Malignant Hyperthermia (MH). Information is available on our website --http://www.cvm.umn.edu/umec/lab/ home.html. In my opinion this breed distribution reflects the larger number of Quarter Horses in the USA relative to other breeds, a tendency to line breed, and the openness and dedication of the Quarter Horse Association to finance and support investigation into equine genetics. As of Feb 1 2012 the AQHA began offering a panel test that includes HYPP, HERDA, MH, GBED, and PSSM1. Hyperkalemic Periodic Paralysis (Hypp)

Breeds affected: Quarter horse-related bloodlines

Bloodlines: Horses descendant from Impressive.

Prevalence: 3% of the Quarter Horse breed is affected. 60% of halter horses

Age affected: Signs usually begin by 2 to 3 years of age.

Clinical signs: Range from asymptomatic to intermittent muscle tremors and weakness. Horses homozygous for HyPP may present with difficulty swallowing or respiratory distress.

Mode of inheritance: Autosomal dominant.

Mutation: A point mutation that results in a phenylalanine/leucine substitution in a key part of the voltage-dependent skeletal muscle sodium channel alpha subunit that controls channel activity ( SCN44).

Testing: Veterinary Genetics Laboratory at the University of California, Davis on mane or tail hair roots.

Glycogen Branching Enzyme Deficiency (Gbed)

Breeds affected: Quarter horse-related bloodlines

Bloodlines: Horses descendant from Zantanon and King

Prevalence: 8% of the Quarter Horse breed. And 28% of Western pleasure are carriers

Age affected: Signs usually present in utero or at birth

Clinical signs: Abortion or stillbirth, may be born alive and are weak at birth. With supportive care may live to up to 18 weeks of age. Death may be sudden when exercised on pasture, associated with weak respiratory

72 muscles or the result of euthanasia due to persistent recumbency. Treatable flexural deformities of all limbs and recurrent hypoglycemia (low blood sugar) and seizures occur in some affected foals.

Mode of inheritance: Autosomal recessive.

Mutation: A point mutation in exon 1 changes a tyrosine to a premature stop codon in the glycogen branching enzyme gene (GBE1) that is expressed in numerous tissues.

Testing: Histopathological tissue samples (muscle and heart) stained for Period ic acid Schiff’s (PAS) show a variable amount of abnormal PAS positive globular and crystalline intracellular inclusions. Genetic testing is done by Veterinary Genetics Laboratory at the University of California, Davis or Vetgen in Michigan on mane or tail hair roots or Animal Genetics, or Progressive Molecular Diagnostics Hereditary Equine Regional Dermal Asthenia (Herda) Breeds affected: Quarter horses

Bloodlines: Working cow and cutting horses

Prevalence: 3.5% of the Quarter Horse breed and 28% of cutting horses are carriers.

Age affected: Signs usually begin by 1.5 years of age

Clinical signs: Wounds or sloughing skin, loose easily tented skin that does not return to its original position, scars, and white hairs at areas of hair re-growth found along the back and saddle area or areas with trauma. Healing is slow.

Mode of inheritance: Autosomal recessive.

Mutation: Point mutation that results in a glycine to arginine substitution in the equine cyclophilin B gene (PPIB) that plays a role in the processing of collagen for the anchoring of the skin to underlying tissue.

Testing: University of California at Davis tests for this mutation.

Type 1 Polysaccharide Storage Myopathy

Two forms of PSSM appear to exist type 1 and type 2 PSSM. We have found the mutation for the type 1 in the GYS1 gene, the cause or causes of type 2 PSSM are under investigation but not yet known. Type 1 PSSM Breeds affected: Quarter horse-related bloodlines, Belgians, Percherons, Morgans, Mustangs and some Warmblood breeds.

Bloodlines: Present in founders of QHs and therefore widespread in all types of QHs with highest prevalence in halter and pleasure horses.

73 Prevalence: 36-50% of Belgians and Percherons, 8% of the Quarter Horse related breeds, 30% of halter horses

Age affected: Signs usually begin by 2 to 3 years of age but may occur in Weanlings. Some horses are subclinical.

Clinical signs: Firm painful muscles, stiffness, skin twitching, sweating, weakness and reluctance to move with light exercise. Sometimes gait abnormalities, mild colic and muscle wasting. Serum CK and AST activity elevated except in Drafts.

Mode of inheritance: Autosomal dominant.

Mutation: Point mutation that results in an arginine to histidine substitution in the GYS1 gene that codes for the skeletal muscle form of the glycogen synthase enzyme.

Testing: Muscle biopsy samples evaluated for presence of amylase-resistant crystalline polysaccharide.

Genetic testing on mane or tail hair roots, or unclotted blood samples at the Neuromuscular Diagnostic Laboratory at the University of Minnesota. http://www.vdl.umn.edu/vdl/ourservices/neuromuscular.html Malignant Hyperthermia

Breeds affected: Quarter horse-related bloodlines

Bloodlines: High frequency in one or two QH families often co-exists with PSSM

Prevalence: 0.1% of the Quarter Horse breed is affected.

Age affected: Adults

Clinical signs: High temperature, metabolic failure and death under anesthesia. Tying up and fever, Signs of PSSM are more severe when both mutations are present. Sudden death

Mode of inheritance: Autosomal dominant.

Mutation: Point mutation that results in an arginine to glycine substitution in the RYR1 gene

Testing: Genetic testing at Neuromuscular Diagnostic Laboratory at the University of Minnesota. http://www.vdl.umn.edu/vdl/ourservices/neuro muscular.html Or through Monica Aleman at UC Davis

74 Summary Table : Observed percentages of horses carrying a disease-causing allele for whole breeds (QH and APH paints) and for elite competitive subgroups.

Affected Carrier dominant (%) recessive (%)

Population HYPP PSSM GBED HERDA OLWS

QH 1.5 11.3 11.0 3.5 NO

APH 4.5 4.5 3.9 1.7 21.3

Halter 56.4 28.2 5.1 0.8 NO

Western pleasure 1.1 8.6 26.3 12.8 NO

Cutting NO 6.7 13.6 28.3 NO

Reining NO 4.3 3.1 9.3 NO

Working cow horse NO 5.7 9.5 11.5 NO

Barrel racing 1.2 1.4 1.2 1.2 NO

Racing NO 2.0 NO NO NO

NO = Not observed in the dataset.

from “Tryon RC, Penedo MCT, McCue, ME, Valberg SJ, Mickelson JR, Famula TR, Wagner M, Jackson M, Hamilton M, Noteboon S, Bannasch DL”. Allele frequencies of inherited disease genes in subpopulations of American Quarter Horses. JAVMA2009 Jan 1;234(1):120-5 Genetic Disorders of Arabians

Severe Combined Immunodeficiency (Scid)

Breeds affected: Arabian

Prevalence: 8% in USA CARRIERS

Age affected: < 6 months of age

Clinical signs: Recurrent infections, respiratory disease, eventual death.

Mode of inheritance: Autosomal recessive.

Mutation: 5 base pair deletion in the gene coding for DNA-dependent protein kinase (DNA-PK) catalytic subunit

Testing: Vetgen on mane or tail hair roots

Lavender Foal Syndrome (Lfs)

Breeds affected: Arabian, higher prevalence in Egyptian Arabians

75 Prevalence: 8 – 17% CARRIERS

Age affected: Signs present at birth

Clinical signs: A dilute coat color and a range of neurological signs, including recumbency, opisthotonous, paddling movements and extensor rigidity leading to euthanasia

Mode of inheritance: Autosomal recessive.

Mutation: A single base pair deletion in the MYO5A gene, which codes for the protein myosin-Va

Testing: Veterinary Genetics Laboratory, University of California, Davis

Cerebellar Abiotrophy

Breeds affected: Quarter horse-related bloodlines

Prevalence: 19% CARRIERS

Age affected: Signs usually between 1 to 6 months of age

Clinical signs: Clinical signs of CA usually develop between the ages of 6 weeks and 4 months and include ataxia, hypermetria and intention head tremors

Mode of inheritance: Autosomal recessive.

Mutation: A single nucleotidepolymorphism located adjacent to a potential binding site for GATA-2. GATA-2 is a transcription factor involved in expression of MUTYH, a post replication DNA glycosylase

Testing: Veterinary Genetics Laboratory, University of California, Davis, Animal Health Diagnostic Center at Cornell University.

Genetics Diseases in Other Breeds Junctional Epidermolysis Bullosa (Jeb)

Breeds affected: Belgian drafts, Saddlebreds

Prevalence: 36% CARRIERS for Belgians, 3% CARRIERS Saddlebreds

Age affected: Signs usually present within a few days after birth

Clinical signs: Ulceration of skin and oral cavity lesions develop which become more extensive with age. Extensive oral ulcerations

76 may also be present. Ulceration of the coronary band may proceed to sloughing of the hooves.

Mode of inheritance: Autosomal recessive.

Mutation: JEB1 (Belgians) stop codon terminating translation of LAMC2 gene. JEB2 (Saddlebreds) deletion in LAMA3 These genes encode subunits of Laminin-5 a structural protein that serves to anchor basal epithelial cells within the dermis.

Testing: Veterinary Genetics Laboratory, University of California, Davis, University of Kentucky

Conflict of interest statement: Drs. Valberg, Mickelson and McCue own the license for PSSM testing and receive sales income from its use. Their financial and business interests have been reviewed and managed by the University in accordance with its conflict of interest policies.

77 The Pleurals and the Peritoneals: Body Cavity Cytology Tracy Stokol DVM Cornell University

The objective of this session is to go over some tips and clues on preparing aspirates of body cavity fluids for cytologic analysis to optimize results and examining and interpreting the results of the prepared smears. Cytologic evaluation is a useful technique for the equine practitioner, because a potential defin- itive diagnosis or treatment plan can be rapidly established with minimal equipment and cost. However, obtaining an accurate diagnosis is entirely dependent on smear quality and cellularity and the proficiency of the cytologist examining and interpreting the smear. Becoming adept at cytologic diagnosis requires practice and comparison to a known “standard” (such as a clinical pathologist or comparing cytologic results to biopsy results). There are some inherent limitations to cytologic evaluations: • Smears of poor cellularity or quality (slowly dried, smudged cells) are rarely diagnostic. • Smears represent the aspirated site. Focal or multifocal lesions may be missed or a misdiagnosis may be obtained with a complex process (e.g. inflammation as part of a tumor).

• Aspirates do not evaluate tissue architecture, which can be crucial for diagnosing certain tumors and differentiating inflammation from neoplasia in some circumstances.

• Inflammation does strange things to tissues – back off calling cancer if there is inflammation. Give the horse the benefit of the doubt and biopsy if needed.

• Avoid collection of post-mortem samples – they usually are not diagnostic (bacteria from bowel leakage, cell deterioration and sloughed mesothelial cells).

• Cytologic (and histologic) diagnoses are opinions (however well-informed). They can (and should be) challenged if they do not fit the clinical picture. Always interpret provided results with what you know about your patient. A diagnosis may be changed entirely based on the clinical presentation.

Preparation and submission of body cavity fluids

Cells in fluids are not “happy”. They deteriorate rapidly with storage (particularly if not kept cool). Bacterial overgrowth can occur, lysing cells. Some cells, particularly macrophages, do stay alive to some extent and do what comes naturally, i.e. phagocytosis. Erythrophagocytosis (mimicking hemorrhage) and bacterial phagocytosis (mimicking bacterial infection) can both be an artifact of storage. In fact, these processes can occur with a few hours (even as quickly as 30 minutes) in the tube. So to ensure that erythrophagia and intracellular bacteria are really occurring in the animal and not just a consequence of sample storage, and to optimally preserve cells, always make smears of the fluid (ASAP after aspiration) and submit along with the fluid. Keep the fluid (but not the smears) cool and submit in break-proof containers overnight. Do not allow cells to freeze (no direct contact with that ice-pack). Avoid formalin at all costs. It messes

78 up staining making interpretation difficult.

• What type of tube for fluids? EDTA is preferable (preserves morphologic features, inhibits bacterial growth). If a need for culture is anticipated, provide an appropriate sample (microbio- logical transport media or sterile nonanticoagulant tube). Collection of fluid into a nonanticoagu- lant (red-top) tube is worthwhile, particularly if the fluid is bloody (clotting suggests blood con- tamination versus true in vivo hemorrhage) or if a need for biochemical tests is anticipated (e.g. glucose, creatinine).

• What type of smear?

o Can do direct (straight from the fluid) or concentrated smears (sediments using a urine centrifuge for instance). If making sediments, don’t use all the fluid – just a portion. eW need unconcentrated fluid to perform cell counts. To determine which of these smears will be best for a particular fluid, look at the clarity of the fluid.

• If clear (can read through it = most body cavity fluids): Cellularity is low. Make sediments.

• If hazy: Cellularity is likely high. Direct smears may be fine.

• If bloody or flocculent: Direct smears are fine.

o Do “contact” not “squash” smears. Gentle is best. Force squashes cells = unrecognizable = no diagnosis. Can do “wedge” smears (like blood smears).

• Stain or not to stain? Go ahead (one way to train yourself) but always leave one unstained for us (we really like our stainer and do not really like Diff-quik).

o Keep your stains clean: Scrub out with ethanol if stain precipitate builds up (mimics bacteria).

o Ensure staining is adequate: Under- or over-staining = potential erroneous diagnosis.

• Other tips?

o Use new, precleaned glass slides, preferably with frosted ends. Start the smears next to the frosted ends and they should not extend beyond ¾ of the length of the slide (if possible). A feathered edge (like a blood smear) is good.

o Rapidly dry using a hair dryer directed at the back of the slide (on high). Heat does not hurt cells. The purpose is to help cells spread so you can see what they are as well as inside them. Rounded up cells = unrecognizable (let alone look for bugs inside them) = erroneous diagnosis.

o Label slides (site, patient ID, type, i.e. sediment or direct) on frosted end in pencil. Ink from marker pens (e.g. permanent markers) dissolves in some stainers.

o Give us a good history (no history = limited diagnosis or misdiagnosis).

o Consider that we can do other tests (gram stain for bacteria, Prussian blue for hemosiderin = hemorrhage, Gomori-methamine silver for fungi, immunostaining for cancer).

79 Results to expect from a pleural or peritoneal fluid • Volume, color, turbidity: Lab-dependent. • Total protein (TP): Done by refractometry (preferred over specific gravity). • Total nucleated cell count (TNCC): Does not equal leukocytes as will include mesothelial and cancer cells, as well as non-cells such as food debris or bacteria/protozoa). • Red blood cell (RBC) count. • Smear evaluation: May include a gram or Prussian blue stain (if indicated) - laboratory dependent. • Others (e.g. biochemical tests): You have to request. Smear Examination

The most important thing is to be consistent. Develop a technique and style and avoid short- cuts. Don’t always expect a definitive diagnosis. Cytology can help define a disease process (in- flammation, sepsis), which can help with formulation of an immediate treatment plan. Due to the aforementioned caveats, there is always the potential to be misled, so always interpret in light of your knowledge of the patient. When in doubt as to the interpretation or diagnostic or pathologic relevance of any cytologic finding, always submit specimens to a clinical pathologist for evalua- tion.

A suggested technique:

• Scan the smear with a low objective (10x is ideal) to evaluate staining quality and cellularity, and identify the right area to look at (thin, adequately spread and intact cells).

o Start identifying processes (lots of blood? Inflammation? Lots of round cells?).

o Look for large objects (cell clusters, crystals, foreign material, parasites, fungi).

o Look for unique features, e.g. cells aggregating around something.

• Look in detail under higher power (40 to 100x) in the optimal area or large/unique feature (look at that something).

o Identify the cells: Are they normal for the tissue (e.g. mesothelial cells)? Is there inflammation? Is there cancer?

o Look for infectious agents.

• Tell fact from fiction – several things mimic organisms, e.g. ruptured nuclear material, stain precipitate, water artifact.

General disease processes

• Hemorrhage: Differentiate from blood contamination. If see platelets - could be either hemor-

80 rhage or contamination. If see erythrophages in a fresh sample - likely hemorrhage. If see hemo- siderin in macrophages - indicates hemorrhage. If see rhomboidal yellow crystals (hematoidin) - indicates hemorrhage in a low oxygen environment. If see erythrophages in a stored sample - cannot be certain (phagocytosis could have occurred in the animal or in the tube, hence make fresh smears). If aspirate a very bloody fluid and it clots in a red-top tube - likely reflects contam- ination (blood clots and defibrinates rapidly in a body cavity).

• Inflammation: Classify by type (e.g. neutrophilic) and look for cause (e.g. bacteria).

• Neoplasia: Uncommon in horses. Some tumors do exfoliate into fluids (e.g. gastric squamous cell carcinoma, lymphoma, melanoma, mesothelioma) so always worth an aspirate if concerned (cheap, quick test with good turnaround). Peritoneal fluid (PTF)

Fluid analysis can yield diagnostic information, such as identifying inflammation, sepsis, hem- orrhage, ischemia, gastrointestinal rupture. The cause of these changes is usually not evident. In some cases, fluid analysis may be diagnostic in horses, e.g. visible tumor cells, ruptured urinary tract (measure creatinine and compare to serum to confirm). Do not rely on cell counts or gross features of fluid to identify septic inflammation. I have seen a few cases of septic inflammation in PTF with normal TNCC and TP but have identified bacteria within degenerate or non-degenerate neutrophils in the smears.

• Expected normal: Dialysate of plasma = low protein transudate, clear to slightly turbid (es- pecially if some blood), colorless to light yellow, <2.5 g/dL TP, low RBC count (<1000 cells/μL, un- less blood-contaminated) with no erythrophages (unless smears made from stored fluid), TNCC <5000 cells/μL in an adult horse and <1500 cells/μL in foals, mixture of non-degenerate neutro- phils (50% to 70%) and macrophages (30% to 50%, may see leukophages), with low numbers of lymphocytes, mast cells, and mesothelial cells (may be in clusters).

• Fiction/non-pathologic: Starch granules, skin squamous epithelial cells and flakes (keratin “bars”), post-surgical carboxymethylcellulose (“belly jelly”).

• Enterocentesis: Mimics acute gut rupture (look at patient to discriminate – if really sick, could be rupture). If obvious enterocentesis, the sample may be turbid, flocculent, and greenish brown. A partial enterocentesis may not be evident grossly (fluid is grossly normal). On smears, see a mixture of bacteria ± protozoa (if aspirated cecal or colonic contents) and plant debris. Cell counts may be inaccurate (counting non-cells). May see the above features with a mixture of nor- mal PTF cells if partial enterocentesis and abdominocentesis. If the latter has occurred, bacteria should not be phagocytized, neutrophils should not be degenerate and horse should not be that sick.

• Colic: Depends on stage and cause.

o In early stages, PTF may be normal.

o First changes with ischemia are increased RBC count and erythrophagia, often accom panied by increased TP. TNCC is usually normal. These findings can also be seen with serosal congestion with impaction colics (does not always equal ischemia).

81 o As ischemia worsens, see increased TNCC, TP and usually RBC count. Dark red-brown fluid has been associated with intestinal necrosis. Bacteria may not be seen untilthe intestinal wall is devitalized enough.

• Other inflammatory conditions: Primary bacterial peritonitis (not due to GI disorders, e.g., Actinobacillus infection), sterile peritonitis (uroperitoneum, seminoperitoneum, pancreatitis, post-abdominal surgery). Will see increased TNCC, variable RBC count, high TP and usually >85% neutrophils (depending on duration and antibiotic therapy, which makes it more mixed).

• Hemorrhagic effusions: These bloody fluids do not clot in a red-top tube and have a high PCV (>1%). Causes include trauma (e.g. splenic tears), ruptured abdominal vessels (e.g., uterine artery during foaling), neoplasia (hemangiosarcoma, ovarian tumors), coagulopathies (e.g., hemophilia A) and idiopathic.

• Chyloabdomen: Very uncommon in horses. Consists of lipid (does not sediment like cells if leave overnight in fridge). Can measure triglycerides in serum and PTF to confirm. Reported with abdominal adhesions, lymphatic abnormalities.

• Neoplasia: Can cause any type of effusion (transudative, i.e. too much of normal, sterile or septic inflammation, chylous, hemorrhagic). Can only make a diagnosis of neoplasia if you see cancer cells, with lymphoma being the most common tumor type seen in fluids. Do not mistake mesothelial cells for cancer (carcinomas are uncommon in horses), although mesotheliomas do occur (give the horse the benefit of the doubt or get another opinion or biopsy if concerned). Pleural fluid (PLF)

Like PTF, PLF can be aspirated in healthy or sick horses and is a dialysate of plasma (low protein transudate). Expected normal results are the same as PTF.

Some conditions causing fluid accumulation in the thorax:

• Pleuropneumonia: The fluid is cloudy to flocculent and yellow or red. TNCC is high and we usually see bacteria within degenerate neutrophils (depending on stage and if the horse has been treated). Other causes of exudative effusions in the pleural cavity are pericarditis and migrating parasites.

• Neoplasia: Lymphoma is the most common neoplasm diagnosed in PLF, but other tumors can be seen (e.g. mesothelioma, melanoma).

• Chylothorax: This has been reported in foals with presumed congenital lymphatic defects or diaphragmatic hernias and in adults with obstruction or destruction of pleural lymphatic vessels by a primary intrathoracic hemangiosarcoma.

Additional reading Cowell and Tyler: Diagnostic cytology of the horse www.eclinpath.com A Case-Based Approach to Equine Blood

82 A Case-Based Approach to Equine Blood Tracy Stokol DVM Cornell University

The objective of this session is to use two clinical cases to discuss how examination of hemogram results and particularly the blood smear can help facilitate a diagnosis in sick horses. The first case involves an anemic horse whereas the second case involves a horse with a marked leuko- cytosis.

The most common hematologic abnormality in horses is an inflammatory leukogram, secondary to sepsis (usually of gastrointestinal origin). We are also seeing more horses with tick-borne infectious diseases, particularly Anaplasma phagocytophilum, which most frequently causes a thrombocytopenia, followed by a mild-non-regenerative anemia. Some horses with this infectious agent can have normal hemograms.

The following changes in hemogram results are unusual in horses and trigger a “mental” flag when I am reviewing the results:

1) Moderate to marked leukocytosis: WBC > 15,000/uL 2) Thrombocytopenia: Due to increased incidence of Anaplasma infections (not just in fall or spring!) 3) Moderate to severe anemia (regenerative or non-regenerative) 4) Lymphocytosis in an adult horse 5) Bi- or pancytopenia (non-regenerative anemia, neutropenia, thrombocytopenia)

Note, the following artifacts are seen in mailed-in samples due to storage-associated changes: 1) High MCV and low MCHC (from RBC swelling): This could shift a low MCV into the normal range (i.e. results may not always be abnormal). 2) Low platelet count: From clumping. We verify all automated counts by examining a blood smear for clumps. 3) Low WBC and/or neutropenia: Neutrophils deteriorate with storage and are no longer counted by our analyzer.

To minimize the impact of these changes on hemogram results, make fresh blood smears and submit along with the EDTA blood when submitting blood to a referral laboratory for analysis. Keep EDTA blood cool at all times, but do not refrigerate the smears (cells lyse). Case 1 “Flash”

Flash is a 6 year old Arabian gelding. The horse presented with acute colic in November after being moved to new pasture 4 days earlier. On physical examination Flash was moderately de- hydrated, had brownish mucous membranes and was tachycardic (66 bpm). A rectal examination revealed no abnormalities and the horse had no evidence of gastric reflux. Diagnostic imaging revealed a fluid-filled hypermotile small intestine.

Provided below are hemogram results (large animal CBC plus fibrinogen) and represen- tative images of the blood smear from Flash.

83 HEMATOLOGY HCT HB HB (calc) RBC MCV % g/dL g/dL mill/μL fL (34-46) (11.8-15.9) (11.8-15.9) (6.6-9.7) (43-55) 17! 7.2! 6.4! 4.9! 46

CH CHCM RDW nRBC WBC pg g/dL % /100 WBC thou/μL (15-20) (34-37) (16.3-19.3) (0) (5.2.-10.1) 17 37 20.7! 1! 10.8

PMN BAND LYMPH MONO EOS thou/μL thou/μL thou/μL thou/μL thou/μL (2.7-6.6) (0) (1.2-4.9) (0-0.6) (0-1.2) 8.4! 1.4! 0.8! 0.2 0.0

BASO PLAT MPV PLAT SMEAR TP-REF thou/μL thou/μL fL g/dL (0) (94-232) (5.3-8.4) (5.2-7.8) 0.0 213 8.2 Normal CANC*

RBC MORPHOLOGY: Heinz bodies-moderate, Eccentrocytes-moderate, Pyknocytes-moderate, Macrocytes-few WBC MORPHOLOGY: Mild toxic change in neutrophils PARASITES: None seen PLASMA APPEARANCE: Moderate hemolysis Fibrinogen by heat precipitation: CANC* (100-200 mg/dL)

* Cancelled due to hemolysis. Note that the MCH and MCHC were inaccurate (and cancelled) and the CH and CHCM (directly measured from the intact RBCs) were provided, along with a HgB calculated from these values. Low power view of Flash’s blood smear (200x)

Arrows: Eccentrocyte, G: Ghost RBC, M: Macrocyte, P: Pyknocyte

84 Chemistry SPECIMEN SUMMARY: Serum

SODIUM POTASSIUM CHLORIDE BICARB ANION GAP mEq/L mEq/L mEq/L mEq/L mEq/L (134-141) (2.3-4.6) (95-106) (24-31) (8-19) 131! 4.7! 101 18! 17

UREA N CREAT CALCIUM PHOSPH MAGNES mg/dL mg/dL mg/dL mg/dL mg/dL (9-20) (0.9-1.8) (11.2-13.0) (2.1-4.7) (1.2-2.0) 58! 5.2! 12.9 1.8! 3.2!

TOT PROT ALBUMIN GLOBULIN A/G GLUCOSE g/dL g/dL g/dL mg/dL (5.7-7.8) (2.8-3.8) (2.4-4.4) (0.6-1.4) (75-117) 6.2 2.4! 3.8 0.6 152!

AST SDH GLDH GGT TOT BILI U/L U/L U/L U/L mg/dL (212-426) (2-11) (1-9) (8-33) (0.8-2.2) 507! <2 8 <3 3.6!

DIR BILI INDIR BILI CK IRON TIBC mg/dL mg/dL U/L μg/dL μg/dL (0.1-0.3) (0.5-2.1) (93-348) (105-277) (284-502) 0.3 3.3! 2753! 217 350

% SAT % (26-62) 62

Special Blood Chemistry

LIPEMIA HEMOLYSIS ICTERUS 71 1047 3 Lipemia: This is an approximate measure of the degree of turbidity in the sample as follows: <30 = no turbidity, 30-60 = mild turbidity, 61-120 = moderate turbidity, >120 = marked turbidity. This value weakly corresponds to the triglyceride (lipid) concentrations. Hemolysis: This is an approximate measure of the hemoglobin concentration in mg/dL. This value corresponds to visual estimates of hemolysis as follows: <20 = no hemolysis, 20-100 = mild hemolysis, 101-300 = moderate hemolysis, > 300 = marked hemolysis. This degree of he- molysis may affect the following results: Falsely decreased: Bicarbonate, direct bilirubin, GGT; Falsely increased: AST, CK, Mg, Ph, TP; Variable effect: TIBC.

85 Icterus: This is an approximate measure of the total bilirubin concentration in mg/dL. This value will be increased by non-bilirubin chromogens (e.g. carotenoids). 1) Does Flash have a regenerative anemia? 2) What is the likely mechanism of Flash’s anemia? 3) Are there clues in the blood smear as to the cause of Flash’s anemia? 4) What are the pertinent findings from Flash’s chemistry results? 5) What is the likely diagnosis and prognosis?

Anemia Anemia is defined as reduced red blood cell (RBC) mass and is recognized by a packed cell volume (PCV), hematocrit (HCT), hemoglobin concentration, or RBC count below established reference intervals. The tradition in veterinary medicine is to use PCV (a direct measured value) or HCT (a calculated value from hematologic analyzers and the default used in these notes) as the main indicator of RBC mass. Anemia is an uncommon hematologic abnormality in horses. There are three main pathophysio- logic mechanisms for anemia: Hemorrhage (loss), hemolysis (decreased lifespan) and decreased production. Distinguishing between these mechanisms is important for identifying the underlying disease or cause of anemia and is best accomplished by using a combination of signalment, historical infor- mation, clinical signs, imaging findings and clinical pathologic results. Anemia can be multifacto- rial in origin, which can complicate discovery of the underlying cause. There are always going to be those challenging and potentially frustrating cases, where the mechanism and cause of the anemia remain ambiguous.

Mechanisms ofAnemia If an anemia is regenerative, the underlying mechanism is hemorrhage or hemolysis. Distinguish- ing between these two mechanisms relies primarily upon clinical signs, physical examination, im- aging and other laboratory tests, i.e. specifically identifying a source of hemorrhage or a cause for hemolysis on a blood smear examination. It can be quite difficult in horses to detect hemorrhage, particularly when internal or occurring into the gastrointestinal system. Other testing, e.g. fecal occult blood, imaging studies, may need to be performed to document hemorrhage. 1) Hemorrhage: This can be internal (into a body cavity) or external (“lost’ from the body, e.g. from gastrointestinal hemorrhage). Low total protein (especially the combination of low albumin and globulins) would support an anemia due to hemorrhage, but is not specific for hemorrhage. A normal total protein also does not rule out hemorrhage, since peracute hemorrhage, internal bleeding or chronic intermittent external blood loss may not result in low protein concentra- tions. 2) Hemolysis: This is when RBCs have reduced lifespan and are prematurely removed from the circulation by macrophage phagocytosis. Some patients may have a component of intravas- cular hemolysis as well, i.e. when the RBCs rupture within vessels releasing free hemoglobin. When severe, intravascular hemolysis dominates the clinical picture. It is important to recog-

86 nize the presence of intravascular hemolysis, because this helps narrow the differential diag- nostic list for the cause of the anemia as only a few conditions result in intravascular hemolysis (see below). Intravascular hemolysis can be recognized by the presence of hemoglobinemia and hemoglobinuria. Examination of a blood smear for relevant (and diagnostic) RBC changes (e.g. oxidant injury) or pertinent infectious agents (e.g. Babesia equi or caballi) is essential. He- molytic anemia is usually associated with a normal or high total protein and bilirubinemia due to unconjugated (indirect) bilirubin. Note conjugated (direct) bilirubin may increase and even dominate in foals with neonatal isotherythrolysis. Liver injury enzymes (SDH, GLDH, AST) may be increased from hypoxic injury but can be normal. Iron and percentage saturation of trans- ferrin may be increased due to increased RBC turnover. None of these biochemical results are specific for hemolytic anemia and should be considered as supportive not confirmatory results. The lack of these findings does not rule out hemolysis. 3) Decreased production: If the anemia is non-regenerative, this indicates a problem with RBC production in the bone marrow, of which there are several causes (as long as the bone marrow has had time to respond). Clues as to the mechanism of a non-regenerative anemia can be gleaned from the severity of the anemia and other hematologic findings, particularly concurrent cytopenias (specifically, neutropenia and thrombocytopenia) and abnormal (neoplastic) cells. A mild normocytic normochromic anemia is the most common form of non-regenerative anemia in horses and is usually due to extramedullary disease, particularly anemia of inflammatory disease (aka anemia of chronic disease) or anemia of chronic renal disease. Other causes of non-regenerative anemia are quite rare in horses. A Mechanistic Approach To Anemia A stepwise approach can be used for distinguishing between mechanisms of anemia: 1) How acute is the anemia? This is important for assessing whether an anemia is regenerative (question 2 below). The bone marrow takes approximately 3-5 days to mount a regenerative response. An anemia of acute onset may appear “non-regenerative” if the bone marrow has had insufficient time to respond. Clinical signs and history can help with determining the dura- tion of anemia, but this can be difficult because anemia frequently manifests with vague clinical signs that may be missed by the owner, particularly in horses. Unless there has been a known traumatic event, a regenerative response should be manifest by the time of presentation in most affected patients. The expected marrow response will also depend on the degree of ane- mia. A robust regenerative response is expected in a moderate to severe anemia, however a weaker response would be expected in a mild anemia. 2) Is the anemia regenerative? Unfortunately, this is very difficult to judge in horses because they do not release many reticulocytes (immature anucleated RBC with RNA) in response to anemia. They do release macrocytes and we look for these larger RBCs (the same color as normal RBCs) in blood smears from anemic horses. However, low numbers of macrocytes can be missed (smear assessment is very subjective). The mean corpuscular volume (MCV) should also not be relied upon because the MCV will only increase in a horse with regenerative anemia if there are sufficient numbers of macrocytes to boost the mean above a reference interval. Also, macrocytes are not specific for regeneration. Macrocytes can also be produced if there is something wrong with bone marrow production of RBCs, particularly with nuclear de- fects. For example, we have seen macrocytosis in horses on sulphonamides because sulphur drugs can result in folate deficiency, which is needed for normal DNA metabolism. I have also

87 seen macrocytes in horses with acute leukemia causing abnormal RBC production. Several studies have shown that measurement of reticulocytes in bone marrow is helpful for identifying a regenerative anemia in horses. However, in reality, a bone marrow aspirate is not performed for the sole purpose of determining whether an anemia is regenerative or non-regenerative. Studies with newer more sensitive hematology analyzers have shown that reticulocytes are detectable and increased in equine blood after experimentally induced anemia, however the increase is very small (e.g. 2,000 to 4,000 reticulocytes/uL) and this test would have low sen- sitivity for detecting regeneration in naturally occurring anemia in horses (where we have no baseline or normal result for that horse to compare the anemic result to). Ultimately, we are faced with doing serial PCVs or HCTs (and blood smears to look for macrocytes) to assess for regeneration in a horse. 3) Is a cause for the anemia evident on a blood smear? Blood smear examination is an essen- tial part of assessment of an anemic patient and provides a wealth of information. Changes in morphologic features of RBC (and other cell lineages) and detection of infectious agents or neoplastic cells can provide clues to or specifically identify the cause of an anemia. Finding abnormal RBC morphologic features in a blood smear does not necessarily mean they are of pathologic relevance. Relevance is subjective and depends on both the type and number of abnormalities and should always be determined in context of the patient. 4) What additional tests are indicated? Results of complete clinical pathologic testing (hemo- gram, clinical chemistry) and imaging, combined with cytologic or histologic assessment of tissues, is essential in all anemic patients for identifying extramedullary disease that could be causing the anemia or suppressing the bone marrow’s response. Testing for infectious disease is worthwhile in horses with an unexplained regenerative or non-regenerative anemia, because blood smear examination can be insensitive for organism detection. Bone marrow aspiration is rarely need in horses to determine the cause of an anemia. Some flags in a blood smear that may reveal the need for bone marrow aspiration or evaluation of the patient for underlying dis- ease (especially hematopoietic neoplasia) is an unexplained moderate to severe non-regen- erative anemia, unexplained thrombocytopenia (i.e. infectious disease testing is negative) and bi- or pancytopenia (non-regenerative anemia, thrombocytopenia, neutropenia). Bone marrow aspiration could be considered if abnormal cells are detected in blood smears from anemic animals.

Causes of anemia 1) Hemorrhage: Causes of hemorrhagic anemia in horses are uncommon and include hemoabdomen and hemothorax from ruptured vessels (e.g. uterine artery secondary to foaling), hemostatic disorders (acquired factor VIII inhibitors, anticoagulant rodenticide toxicosis), splenic injury, drugs (phenylephrine) and cancer. In many cases of hemoperitoneum (up to 22% of cases in two studies), a specific cause was not identified. Causes of gastrointestinal hemorrhage include gastric ulcers and neoplasia. Chronic external blood loss (particularly if intermittent) can result in a secondary iron deficiency anemia, which is quite uncommon in horses. 2) Hemolysis: Intravascular hemolysis can be seen in causes that are bolded below. a) Immune-mediated: Primary (uncommon) or secondary to infectious agents (e.g. Streptococcus equi, equine infectious anemia), neonatal isoerythrolysis, cancer

88 (lymphoma) or drugs (penicillin). Helpful smear features: ± spherocytes (hard to conclusive- ly ID in horses), agglutination. b) Infectious agents: Babesia sp., Leptospirosis sp. Clostridia sp. Helpful smear features: ± organism (Babesia). c) Oxidant-induced: Red maple leaf, phenothiazine drenches, wild onions. Helpful smear features: Heinz bodies, eccentrocytes, pyknocytes. d) Inherited defects: Glucose-6-phosphate dehydrogenase deficiency, flavine adenine dinucleotide (FAD) deficiency (rare, rare, rare). Helpful smear features: Heinz bodies, eccentrocytes, pyknocytes, methemoglobinemia (FAD). e) Miscellaneous: DMSO, acute liver failure. 3) Decreased production: a) Bone marrow suppression from inflammatory disease (anemia of “chronic” disease): Liver disease, neoplasia, infectious agents (e.g. equine infectious anemia). b) Bone marrow suppression from chronic kidney disease. c) Bone marrow damage/injury/suppression from drugs: Recombinant human erythropoietin therapy can cause a pure red blood cell aplasia. d) Primary bone marrow disease: Leukemia, bone marrow aplasia (very uncommon).

Low power view of Xanadu’s blood smear (200x) =

89 Case 2 “Xanadu”

Xanadu is a 14 year old Clydesdale mare, who presented with weight loss, lethargy, inap- petance, fever and diarrhea. The horse had recently developed a bilateral serosanguineous nasal discharge, ventral edema and dyspnea.

On physical examination, the horse was depressed, lethargic and sweating profusely. Xanadu was markedly tachycardic (92 beats/minute) and febrile (103ºF). The horse had labored breath- ing, enlarged intermandibular and prescapular lymph nodes and pitting edema in the sternal area. Blood was taken for a hemogram and chemistry profile.

Provided below are hemogram results (large animal CBC plus fibrinogen) and represen- tative images of the blood smear from Xanadu.

Hematology HCT HB RBC MCV MCH % g/dL mill/μL fL pg (34-46) (11.8-15.9) (6.6-9.7) (43-55) (15-20) 24! 9.3! 4.9! 48 19

MCHC RDW NRBC/ WBC PMN g/dL % 100 WBC thou/μL thou/μL (34-37) (16.3-19.3) (0) (5.2.-10.1) (2.7-6.6) 39 17.4 0 110.4 ______

BAND LYMPH MONO EOS BASO thou/μL thou/μL thou/μL thou/μL thou/μL (0) (1.2-4.9) (0-0.6) (0-1.2) (0) ______

OTHER WBC PLAT MPV PLAT SMEAR TP-REF thou/μL thou/μL fL g/dL (0) (94-232) (5.3-8.4) (5.2-7.8) ______16! 11.0 Very low 10.0!

RBC MORPHOLOGY: ______WBC MORPHOLOGY: ______PARASITES: None seen PLASMA APPEARANCE: Normal

Fibrinogen by heat precipitation: 600 mg/dL (100-200 mg/dL)

90 High power view of Xanadu’s blood smear (500x)

Chemistry

Specimen Summary: Plasma SODIUM POTASSIUM CHLORIDE BICARB ANION GAP mEq/L mEq/L mEq/L mEq/L mEq/L (134-141) (2.3-4.6) (95-106) (24-31) (8-19) 126! 3.0 87! 19! 23!

UREA N CREAT CALCIUM PHOSPH MAGNES mg/dL mg/dL mg/dL mg/dL mg/dL (9-20) (0.9-1.8) (11.2-13.0) (2.1-4.7) (1.2-2.0) 20 1.2 10.4! 4.9! 1.1!

TOT PROT ALBUMIN GLOBULIN A/G GLUCOSE g/dL g/dL g/dL mg/dL (5.7-7.8) (2.8-3.8) (2.4-4.4) (0.6-1.4) (75-117) 9.0 2.4! 6.6! 0.4! 81

AST SDH GLDH GGT TOT BILI U/L U/L U/L U/L mg/dL (212-426) (2-11) (1-9) (8-33) (0.8-2.2) 534! 3 15! 16 3.3!

DIR BILI INDIR BILI CK IRON TIBC mg/dL mg/dL U/L μg/dL μg/dL (0.1-0.3) (0.5-2.1) (93-348) (105-277) (284-502) 0.3 3.0! 270 194 238

% SAT% (26-62) 91 2 LIPEMIA HEMOLYSIS ICTERUS 45 4 5

Lipemia: This is an approximate measure of the degree of turbidity in the sample as follows: <30 = no turbidity, 30-60 = mild turbidity, 61-120 = moderate turbidity, >120 = marked turbidity. This value weakly corresponds to the triglyceride (lipid) concentrations.

Hemolysis: This is an approximate measure of the hemoglobin concentration in mg/dL. This val- ue corresponds to visual estimates of hemolysis as follows: <20 = no hemolysis, 20-100 = mild hemolysis, 101-300 = moderate hemolysis, > 300 = marked hemolysis.

Icterus: This is an approximate measure of the total bilirubin concentration in mg/dL. This value will be increased by non-bilirubin chromogens (e.g. carotenoids).

6) Does Xanadu have a regenerative anemia? 7) What is the likely mechanism of Xanadu’s anemia? 8) Xanadu has a marked leukocytosis. What is the dominant cell type in the blood smear? 9) Are there relevant abnormalities in Xanadu’s RBCs? 10) Based on the hemogram results, what is your diagnosis? 11) Based on the hemogram and chemistry results, what other tests might you consider performing? 12) What type of prognosis would you give the owner?

Total protein electrophoresis was performed on Xanadu with the following results:

92 Leukemia Leukemia means that there are tumor cells of hematopoietic origin in the blood and/or the bone marrow. Most types of leukemia that arise in the horse have a poor prognosis. Horses usually present at an advanced stage of disease and treatment of these large animals is costly. Also, many of the drugs used to treat leukemia in small animals are of unknown efficacy or tox- icity in horses. Luckily for the horse, leukemias are quite uncommon and it is unlikely that most veterinarians will encounter a case of equine leukemia in their career. Classification of Leukemia Leukemias are classified on the basis of degree of differentiation and cell of origin: 1) Chronic leukemia consists of mature cells that have fully differentiated. Thus, they can be recognized by their morphologic appearance. They can be lymphoid (CLL) or myeloid (CML) in origin. Chronic leukemias are usually indolent and slowly progressive disorders. 2) Acute leukemia can be lymphoid (ALL) or myeloid (AML) and consists of undifferentiated cells that do not resemble normal counterparts (and are called “blasts”). They can, however, express markers that indicate which lineage they arose from (or were differentiating towards), e.g. a positive reaction for CD3 would support a T cell origin for a leukemia. These markers are used to help identify the cells and classify them as lymphoid or myeloid and their respec- tive subtypes. Acute leukemias are usually of acute onset and progression.

Specialized diagnostic techniques for leukemia Two techniques are used to determine the type of leukemia, immunophenotyping and cy- tochemistry. These techniques are used primarily for acute leukemia, where neoplastic cells cannot be distinguished based on morphologic features, but are also used CLL to determine the phenotype of the neoplastic lymphocytes (they are not used for CML, because the involved cell lineage can be identified based on morphologic features alone). The major limitations with these techniques are their relative unavailability and the paucity of equine-specific reagents. 1) Immunophenotyping: This involves the application of antibodies raised against markers expressed on cells. In some cases, these markers are lineage-specific (e.g. CD3 is aT cell marker), whereas in others they are expressed on more than one cell type (e.g. CD172a is a neutrophil and monocyte marker). The antibodies can be applied to cells in suspension or cells on slides. With suspended cells (e.g. venous blood or bone marrow aspirates), positive reactions (antibody binding) are detected with fluorescence–based techniques using flow cytometry. When antibodies are applied to cells on cytology or histology slides (called im- munocyto- and immunohistochemistry, respectively), they are usually detected by colorimet- ric-based methods. More antibodies can be used with flow cytometry, because it is not limited by slide numbers or formalin fixation (destroys antigens). More antibodies are available for lymphoid versus myeloid antigens.

2) Cytochemistry: Here, substrates are applied to cells to detect enzyme activity in the cell, e.g. myeloperoxidase, or a particular protein (e.g. Sudan B Black). Enzymes are mostly expressed in myeloid cells and this technique is most useful to diagnose acute myeloid leu- kemias (particularly granulocytic and monocytic). They are less useful for lymphoid leukemia because lymphoid cells usually lack these enzymes. Cytochemistry can only be done on cyto- logic samples or blood.

93 Types of leukemia in horses Leukemia in general is uncommon in horses. 1) Chronic leukemia: All types of chronic leukemia are quite rare in the horse, however there have been more reports of CLL than CML. The main differential diagnosis for a CML is a cytokine-driven response due to neoplasia (not leukemia) or inflammation. For example, a high HCT has been reported with lymphoma, hepatocellular tumors, carcinomas and phaeo- chromocytomas in horses. In the few reports of CLL, affected horses were generally older and presenting signs were non-specific (weight loss, inappetance, fever, edema, lymphadenopa- thy). The diagnosis of CLL was based on markedly increased numbers of small to intermedi- ate lymphocytes in blood, ranging from 26,800 to 208,300/uL. Other cytopenias were absent or mild, except in advanced cases. The main differential diagnosis for CLL is a leukemic phase of a small cell lymphoma. These two disorders are difficult to distinguish (based on main site of involvement, i.e. blood or/or marrow versus extramedullary tissue) and there is substantial overlap between them in clinical signs, hemogram results and postmortem findings. Indeed, distinguishing between these two neoplasms may be an academic exercise, because they may represent different clinical manifestations of the same neoplasm. Both B- and T-cell CLLs have been identified in horses. Several horses with B-CLL have a concurrent hyperglobulin- emia that is often due to a monoclonal gammopathy (usually IgG), so serum electrophoresis can be helpful in these horses. Bone marrow may not be infiltrated by the tumor, hence it is not always needed for a diagnosis. 2) Acute leukemia: There have been several individual reports of acute leukemia in horses, with both ALL and AML being identified (using the specialized techniques described above). Affected horses are usually younger than those with CLL, but clinical signs are still non-spe- cific (weight loss, inappetance, fever, edema, lymphadenopathy). Affected horses usually have moderate to severe bi- or pancytopenia with rare or no circulating tumor cells, which are usually “blasts” (big blue cells). Specialized testing is required to differentiate between AML and ALL. As for CLL, the main differential diagnosis for an ALL is a large cell lymphoma with a leukemic phase. Bone marrow aspiration is required for this distinction, with >25% “blasts” being compatible with an ALL versus lymphoma.

A step-wise approach to leukemia diagnosis This approach is mostly used for cells that are mononuclear, since mature differentiated myeloid cells (platelets, RBCs, neutrophils etc) can be recognized readily by their appearance. Further- more, CMLs are exceedingly rare in horses. 1) Are there large mononuclear cells in blood (aka “big blue cells”)? If yes, decide if they are monocytes, reactive lymphocytes or “other WBC” (“blasts”). This is best done by a clinical pathologist. 2) If the cells are blasts, are there other cytopenias (neutropenia, non-regenerative anemia, thrombocytopenia)? a) If yes, a diagnosis of acute leukemia is favored, particularly with moderate to se- vere bi- or pancytopenia. Bone marrow aspiration and additional diagnostic testing is indicated.

94 b) If no cytopenias are present or the cytopenias are mild, the horse could have lym- phoma with neoplastic cells in blood. A search for lymphoma (lymph node enlargement, organomegaly, peritoneal or pleural fluid analysis, skin lesions, gastrointestinal dis- ease) is prudent. If no lymphoma is evident, re-evaluate assessment of the large cells in blood or consider bone marrow aspirate (if still consider neoplasia based on clinical signs and other results). 3) Is there a leukocytosis of mononuclear cells in blood? If yes, decide on how big the cells are (usually compare their nuclei to a neutrophil). a) If the cells are larger than a neutrophil – likely indicates an acute leukemia or large cell lymphoma (see #1 above) b) If the cells are the same size (intermediate lymphocyte) or smaller than a neutrophil (small lymphocyte) – these are likely lymphoid in origin and could indicate a CLL or leukemic phase of small cell lymphoma. Immunophenotyping of the cells and a hunt for extramedullary tumors is warranted.

95 The ABCs of Acid-Base Analysis or the View from the Eye of a Clinical Pathologist Tracy Stokol DVM Cornell University

The objective of this session is to use a clinical case to work through how a clinical pathologist uses laboratory data from blood gas analysis and a chemistry panel to identify acid-base disturbances.

Some general principles of interpretation of results: 1) The body reacts to a acid-base abnormality through buffering the response with endogenous body buffers and compensatory responses. a) Compensation opposes the primary disorder, i.e. compensation for a metabolic acidosis is a respiratory alkalosis. b) Respiratory compensation for a primary metabolic disorder occurs within minutes. Metabolic compensation for a primary respiratory disorder takes several days and requires a normal functioning kidney. c) Compensation does not usually return the blood pH to normal. d) Over-compensation does not occur. 2) The body attempts to maintain electroneutrality at all times! a) Sum of positive charges = sum of negative charges (see gamblegram to the right) b) Positive charges: sodium (Na+), potassium (K+), unmeasured cations (UC+) c) Negative charges: chloride (Cl-), bicarbonate (HCO3-), unmeasured anions (UA-). Note that unmeasured anions include albumin (however this contributes a small amount to unmeasured anions compared to other anions, such as phosphate). 3) Always interpret blood gas with chemistry results. 4) Do not over-interpret mild changes. No analyzer or test is 100% accurate! 5) Strong ion principles: Essentially means to look at electrolytes (strong ions), minerals (strong ions) and proteins (weak ions) when evaluating for acid-base disturbances.

Some definitions Acidemia: A decrease in pH (increased H+) below the reference interval. Alkalemia: An increase in pH (decreased H+) above the reference interval. Acidosis: A pathophysiologic process causing accumulation of an acid or loss of a base, which lowers the pH. This may or may not result in acidemia. Alkalosis: A pathophysiologic process causing accumulation of a base or loss of an acid, which increases the pH. This may or may not result in alkalemia.

96 Blood gas analysis Ideally this should be done on arterial samples. This provides the following: 1) pH: a) Low = academia. b) High = alkalemia. c) Normal: Does not rule out an acid-base disturbance. 2) Partial pressure of carbon dioxide (mmHg): Only indicator of the respiratory component of acid-base status. a) Low (hypocapnea) = respiratory alkalosis. b) High (hypercapnea) = respiratory acidosis. 3) Partial pressure of oxygen (mmHg): Only reliable in arterial blood samples. a) Low = hypoxemia. b) High = hyperoxemia. Can be seen in animals given supplemental oxygen. 4) Bicarbonate (HCO3-) and total carbon dioxide (mEq/L): With base excess, these indicate the metabolic component of acid-base status. Total carbon dioxide = HCO3- + dissolved CO2 (usually 1-2 mEq/L higher than HCO3-). a) Low = metabolic acidosis. b) High = metabolic alkalosis. 5) Base excess (BE): Indication of the metabolic component of acid-base status (reflects non-carbonic acids). a) Low = insufficient base or metabolic acidosis. b) High = excess base or metabolic alkalosis.

What a blood gas analysis does not tell you: • The type of metabolic disturbance (mixed disorders may be missed) • The type of acidosis (titration by acid or bicarbonate loss) • Strong ions and anion gap: Helps identify alkalosis and acidosis (including type).

Chemistry panel This provides information on metabolic disturbances only and the type of metabolic disturbance. 1) Sodium: Na+ is a strong ion and behaves like a base. Changes in Na+ generally reflect changes in water status. a) Low = Metabolic acidosis (dilutional). b) High = Metabolic alkalosis (contraction). 2) Chloride: Chloride is also a strong ion and behaves like an acid. The Cl- concentration should always be assessed after taking into account changes in Na+. Na+ and Cl- will change proportionally when there is a gain or loss of water or concomitant losses of both electrolytes. Disproportionate changes in Cl- with respect to Na+ indicate an acid-base abnormality. Thus, Cl- concentrations should always be interpreted with Na+ by either eyeballing the data or “correcting” the Cl- (for changes in water or Na+). a) Eyeballing the data: Compare the direction and degree of change in Na+ and Cl-. b) Correct the chloride = (Na+(average)/ Na+(measured)) x Cl-(measured), where average = midpoint of reference interval; measured = actual value obtained. i) Low Cl-corr = metabolic alkalosis (usually see with high bicarbonate). ii) High Cl-corr = metabolic acidosis (usually see with low bicarbonate).

97 3) Bicarbonate: Indicates the metabolic component of acid-base status (but is a dependent variable unlike base excess). a) Low = metabolic acidosis. b) High = metabolic alkalosis. c) Normal: Does not rule out an acid-base disturbance. In mixed disturbances, due to opposing effects (alkalosis increases and acidosis decreases HCO3-), the HCO3- may be normal. Evaluate Cl-corr and AG results or will miss a mixed disturbance. 4) Anion gap (AG): The anion gap helps distinguish the type of metabolic acidosis. AG = (Na+ + K+) – (Cl- + HCO3-) = UA – UC Since unmeasured anions are more abundant than cations, the AG is usually positive and can be assumed to be an indicator of unmeasured anions. In the horse, these are usually renal acids (phosphate, sulfates etc), L-lactate or proteins (primarily albumin). Unmeasured cations include the ionized versions of calcium and magnesium (hence pretty darn low). a) Low = decreased UA or increased UC (rare). Often indicates a measurement error, but low albumin may decrease the AG. b) High AG + low HCO3- = increase in UA or a titration metabolic acidosis. c) Normal anion gap + low HCO3- = no change in UA or UC. This usually indicates a bicarbonate loss metabolic acidosis, which is accompanied by high Cl-corr. 5) Proteins: Applied in Stewart’s principles. Albumin is an unmeasured anion (think of it as a weak acid) and most abundant in blood. a) Low albumin = hypoproteinemic alkalosis. b) High albumin = hyperproteinemic acidosis. 6) Minerals: Phosphate in particular is a strong unmeasured ion (aka acid because it binds H+). a) Low phosphate = hypophosphatemic alkalosis. b) High phosphate= hyperphosphatemic acidosis. 7) Renal tests: Helps identify the cause of an acid-base disturbance. A normally functioning kidney is required to compensate for a primary respiratory disturbance or to correct a primary metabolic disturbance. Types of acid-base disturbances 1) Metabolic acidosis: This is the most common acid-base disturbance encountered in sick horses and is usually a primary disturbance (not in com- pensation for a respiratory disturbance). a) Blood gas: Low HCO3- and BE. pH may be low if primary. b) Chemistry: See gamblegrams to the right. i) Titration acidosis: Low HCO3- and high AG. Accumulation of an acid, e.g. lactic and uremic acidosis. Always primary.

98 ii) Bicarbonate loss: Low HCO3-, normal AG and high Cl-corr. Causes include secretory diarrhea, proximal renal tubular disease. Can be primary or secondary in compensation for a primary respiratory alkalosis. iii) A normal HCO3- does not rule out an acidosis or alkalosis and suggests a mixed disturbance if AG and Cl-corr are abnormal. 2) Metabolic alkalosis: This is usually caused by loss of HCl or loss of chloride (a strong anion). Can be primary (e.g. gastric reflux, excess sweating) or in compensation for a primary respiratory acidosis. a) Blood gas: High HCO3- and BE. pH may be high if primary. b) Chemistry: See gamblegrams to the right. i) High HCO3-, usually normal AG, low Cl-corr. ii) Remember, a normal HCO3- does not rule out an acidosis or alkalosis and suggests a mixed disturbance if AG and Cl-corr are abnormal. 3) Respiratory acidosis: Can be primary (e.g. severe pneumonia, tracheal obstruction) or secondary in compensation for a primary metabolic alkalosis (uncommon). a) Blood gas: High pCO2. b) Chemistry: Will only see compensatory responses if primary and sufficient time. 4) Respiratory alkalosis: Can be primary (e.g. hyperventilation from pain) or secondary in compensation for a primary metabolic acidosis (common). a) Blood gas: Low pCO2. b) Chemistry: Will only see compensatory responses if primary and sufficient time. Summary of Biochemical Panel Changes in Primary Metabolic Acid-Base Disturbances

Primary disturbance HCO3- AG Cl-corr Titration metabolic acidosis ↓ ↑ Normal Bicarbonate loss metabolic acidosis ↓ Normal ↑ Metabolic alkalosis ↑ Normal ↓

Compensation This opposes the primary disturbance. Below is a table listing the primary acid-base distur- bances with expected compensatory responses. As can be seen, the change in the prima- ry result (HCO3- for metabolic and pCO2 for respiratory) is paralleled by the compensatory response.

99 Primary disorder pH [H+] Primary change Compensation

Metabolic acidosis β β β HCO3- ↓ pCO2

Metabolic alkalosis β β ↑ HCO3- ↑ pCO2

Respiratory acidosis β β β pCO2 ↑ HCO3-

Respiratory alkalosis β β β pCO2 ↓ HCO3-

Mixed Acid-Base Disorders

A mixed acid-base disturbance is defined as the presence of more than 1 primary disturbance. There could be 2 or even 3 primary acid-base disturbances (but not 4 as you cannot have more than one primary respiratory acid-base abnormality). Note that it is incorrect to use this term for a single primary disturbance with the appropriate compensatory response.

A mixed acid-base disturbance, although not that common in horses, should be suspected in these situations:

1) The change in pH is greater than can be attributed to one disorder alone. Remember, com- pensation will usually not normalize the pH.

2) The pCO2 and HCO3- change in opposite directions (see table above).

3) The expected compensatory response is: a) Not present, b) Opposite to that which is expected, or c) Exceeds that which is expected.

For example, in a primary metabolic acidosis, the expected response is a respiratory alkalosis (low pCO2), which should occur within minutes. If the pCO2 is high normal or increased, there is a concurrent respiratory acidosis. The pH would be lower than expected for a metabolic acido- sis alone, because the combined respiratory and metabolic acidosis has an additive effect on lowering the pH.

4) The degree of change in acid-base biochemical test results is not proportional. a) In an uncomplicated high anion gap (titration) metabolic acidosis, the increase in AG is roughly proportional to the decrease in HCO3- and Cl-corr is normal. b) In an uncomplicated hyperchloremic metabolic acidosis (bicarbonate loss), the decrease in HCO3- is roughly proportional to the increase in Cl-corr and the AG is normal. c) In an uncomplicated metabolic alkalosis, the increase in HCO3- is roughly proportional to the decrease in Cl-corr and the AG is usually normal.

100 Any deviations from that listed above suggest the likelihood of a mixed-acid disturbance. Re- member that changes in serum proteins (mostly albumin) may impact the AG (and should be considered when using these guidelines).

Remember, the body tries to correct or compensate for changes in pH (H+ rules and is BAD when high or low). Also remember that the kidney is the main organ responsible for compen- sating for a primary respiratory disturbance and correcting a primary metabolic disturbance. So compensation (primary respiratory) or correction (primary metabolic) requires a normal function- ing kidney (in terms of acid-base handling, not necessarily GFR).

101 Non-traditional therapies for endometritis: science or voodoo? Dr. Charles F. Scoggin, DVM, MS, DACT Rood and Riddle Equine Hospital, 2150 Georgetown Road, Lexington, KY 40511, [email protected].

Academic Involvement: Affiliate faculty member, Department of Clinical Sciences, Colorado State University; current member, Grayson Jockey Club Research Advisory Committee.

Conflicts of Interest: None

Take Home Message: I consider the uterus a tomb where, aside from semen and a devel- oping conceptus, nothing else should inhabit it. In problem or susceptible mares, this tomb can be raided by pathogens, such as bacteria and/or various inflammatory mediators—all of which can adversely affect pregnancy and live-foal rates. While conventional treatments (e.g., uterine irrigation and infusion with antimicrobial agents) are often successful, there are some mares in which these approaches are not enough. This talk will provide a review of endometritis, as well as discuss both routine and non-conventional therapies for treating this condition.

Introduction: Inflammation of the uterus following breeding is considered a normal yet transient physiologic response in mares (Troedsson, 2006). Most mares (~90%) are capable of managing this inflammation with little to no intervention; these individuals are considered repro- ductively normal or resistant mares. The other 10% either have or are prone to endometritis and are considered susceptible mares (Hughes and Loy, 1969). Causes include: infiltration of pathogenic organisms, anatomic and functional defects, aberrant local immune response, and method of breeding. These are not mutually exclusive and can work in tandem to adversely affect a mare’s reproductive efficiency.

Common Causes of Endometritis: In the author’s practice, the two most common causes of endometritis are infectious endometritis (IE) and post-mating-induced endometritis (PMIE). With respect to IE, the most frequently encountered pathogens are Streptococcus equi ssp. zooepidemicus (ß-Strep) and Escherichia coli (Nielsen, 2005). Less common but certainly no less important agents include the bacteria Pseudomonas aeruginosa, Klebsiella spp. and the yeast Candida spp. These agents are identified by microbiologic techniques, cytologic evalu- tions, and histologic studies of the endomentrium.

The primary clinical attribute of PMIE is the presence of an abnormal amount of fluid within the uterus 24-36 hours after breeding (Troedsson, 1999). Normal mares should have only a trace amount of anechoic fluid on transrectal ultrasound following mating or insemination. In contrast, susceptible mares will have a significant amount of echogenic fluid several centimeters tall by several centimeters wide. Endometrial edema will often surround the fluid, indicating an exaggerated local inflammatory response.

Other characteristics of IE and PMIE include inflammation (hyperemia) of the cervix and cranial vaginal vault as seen with a vaginal speculum exam. Severe cases may have vulvar discharge. Uterine irrigation will often yield a cloudy efflux, comprised of inflammatory cells and proteinaceous material. Risk factors for IE and PMIE include advancing age, multi-parity, poor

102 perineal conformation, urine pooling, cervical dysfunction (e.g., fibrosis, incompetence, or failure to dilate), and a low-slung or pendulous uterus, all of which will be discussed below.

Other Causes of Endometritis: In 1937, Dr. E.A. Caslick wrote the seminal paper regard- ing the importance of perineal conformation on reproductive efficiency in broodmares (Caslick, 1937). He found that air aspiration or “wind-sucking” through the vulva can lead to pneumova- gina and potential contamination of the uterus. Dr. Caslick’s findings have been verified through the years, and the commonly performed vulvoplasty bears his name in honor of the technique. This procedure is often performed in performance fillies and usually maintained through their broodmare career. We typically replace Caslick’s sutures 4-7 days after foaling, which has been shown to reduce the incidence of PMIE in foaling mares and improve first-cycle conception rates (Hemberg et al., 2005).

Endometrosis or chronic degenerative endometritis (CDE) is a consequence of advanc- ing age and parity (Allen, 1993). A definitive diagnosis is made by uterine biopsy, which com- monly shows periglandular fibrosis, glandular nesting, lymphangectasia, and inflammatory cell infiltrates within the stratum compactum. Cytologies are often inflammatory and may contain copious amounts of mucus or proteinaceous debris in the background. Bacteria may or may not be evident. Clinical findings in these mares are similar to those described for IE and PMIE. Additionally, these mares often have a pronounced “sag” to their uterus in that it will lay cranial and ventral to the pelvic brim creating dependent fluid pooling in the uterine horns.

Biofilm: Biofilm is a hot-topic in equine reproduction and deserves its own section for discussion. As a review, bacteria can exist in one of two states. The first is the free-floating or planktonic state, while the second is the biofilm state produced by a community of bacteria. Bio- film is thought to provide an added barrier of protection against the host’s immune system and antibiotic agents. The presence of biofilm has been found in various areas, including the blad- der and skin wounds, and can lead to persistent infections, making treatment and resolution dif- ficult. The actual presence of biofilm within the mare’s uterus has not been confirmed as of this writing (Ferris 2015, personal communication), but we appear to be on the precipice of answer- ing that question. Pathogenic bacteria implicated in biofilm formation are: E. coli, K. pneumonia, P. aeruginosa, and ß-Strep. As will be discussed in the next section, various agents have been studied and can be used in cases of suspect biofilm formation. In my opinion, a presumptive diagnosis of uterine biofilm infection can be made in mares that fail to respond to conventional treatments as determined by repeated “dirty” cultures and inflammatory cytologies.

Therapeutics for Endometritis: In mares that must have a functional uterus either to yield an embryo or a live foal, my overarching goal is to make the uterus as hospitable as possible for a developing conceptus. The first step is to identify and address potential sources of contami- nation and inflammation. The clinician should also evaluate the mare frequently, monitoring for proper follicular development, endometrial edema, and cervical relaxation.

In cases of IE, treatment should ideally be based on culture and sensitivity testing. For a first time offender, my preference is to irrigate the uterus with 1-4 L of fluids and then infuse with an appropriate antibiotic. Future flushes will be dependent on character of uterine effluent and subsequent exams. The majority of ß-Strep and E. coli isolates I encounter are sensitive to ticarcillin and clavulonic acid (Timentin®)a or ceftiofur (Naxcel)b, both of which are often suitable uterine infusates. This protocol also works well as an empirical treatment when culture results

103 are pending or not available. Intrauterine therapy will be continued for another 2-4 days or until the mare’s cervix has closed (usually no later than 2 days post-ovulation). In most cases, we will forgo mating the mare on this cycle and instead administer a luteolytic dose of prostaglandin (Estrumate®)c five days after ovulation and repeat diagnostics during the ensuing estrus. In chronically infected mares, giving them a period of sexual rest (e.g., 2-3 cycles) may also be of benefit to quiet down inflammation.

In mares with CDE, my primary goal is assisting the mare with clearance of intraluminal uterine fluid. Judicious use of ecbolic agents is at the front-line of therapy. Oxytocin (10-20 IU, IM, q 6 h) or cloprostenol (Estrumate®;c 250 ug, IM, q 24 h) through 2 days post-ovulation have been shown to be effective treatments in normal mares (Nie et al., 2002). In mares in which the cervix fails to dilate sufficiently (usually secondary to fibrosis or scarring), manual dilation can be performed prior to insemination or during subsequent vaginal exams. Some colleagues report reasonable success with topical application of compounded misoprostol or butylscopolamine, but I have no first hand experience with these agents for cervical dilation.

The use of corticosteroids in the management of susceptible mares has become increas- ingly popular over the past several years. The clinical benefit of corticosteroids was described by Bucca et al. (2008) in which they reported improved pregnancy rates in susceptible mares treated with 50 mg (IM) of dexamethasone prior to breeding. Prednisolone (0.1 mg/kg, PO, BID) beginning two days prior to breeding has also been used to manage acute inflammation when breeding with frozen semen (Dell Aqua et al., 2006).

Shown in Table I are various therapeutics and their indications in the management of mares with endometritis. Some are relatively new treatments (and some are not) with various degrees of efficacy and evidenced-based research supporting their use. By no means do I con- sider this a comprehensive list, and the objective of this table and ensuing discussion is not to discredit the use of traditional antibiotics or other published treatments; rather, it is to offer some novel options that I have found either useful or intriguing in the management of problem mares. Lactated Ringers Solution (LRS)

Obviously far from a novel treatment, LRS is nonetheless a great choice for uterine irrigation. No adverse affects on semen viability have been reported, and irrigation with LRS immediately prior to insemination (Livini et al., 2013) or 4-6 hours after breeding (Brinsko et al., 1991) has been shown not to have detrimental effects on early pregnancy rates. Physiologic saline (0.9%) can also be used and is what we relied on this past season (2015) due to a shortage of LRS and other polyionic solutions in the USA. The goal of uterine irrigation is to assist with clearance of or—at the very least—dilute out inflammatory debris within the uterus. Irrigation is usually performed 1-2 times daily and concluded once the mare has ovulated and/or the cervix is clos- ing down. Total volume used is case-dependent but averages 2-4 L per treatment.

Antimicrobial peptides (AMPs)

Natural occurring AMPs are produced and used by neutrophils to degrade bacteria. Cer- agynTM is a commercially available wound-healing medical deviced that is marketed as an AMP mimic. According to CeragynTM’s website, AMPs need only come in contact with the cell membrane to exert a bactericidal effect. Other attributes of AMPs include: a broad-spectrum of

104 activity; less risk of developing resistance to microbial defense mechanisms; and direct activity against biofilm. CeragynTM is also being distributed as a uterine lavage device labeled for use either 4 hours prior to or 6-48 hours after breeding. Preliminary studies evaluating its activity against certain biofilm-producing bacteria have been promising. Ferris (2014) showed that Cer- agynTM was effective in degrading biofilm biomass produced in vitro by E. coli, K. pneumonia, P. aeruginosa, and beta-Strep, and it was also capable of killing beta-Strep with a biofilm. More research is necessary, but CeragynTM thus far appears to be a promising treatment option in susceptible mares, especially those with suspect biofilm infections.

Hydrogen peroxide (H2O2)

Long-used as an anti-septic, H2O2 has shown a broad-spectrum of activity against various microbes, including bacteria, virus, fungi, yeast, and spores. In mares, the recommended dose is 20 mL of a 3% solution diluted to 60 mL in LRS (LeBlanc and McKinnon, 2011). An in vitro study demonstrated that 1% H2O2 is very useful against most pathogens in both their plankton- ic and biofilm state, except for P. aeruginosa, which was capable of inactivating H2O2 (Ferris, 2014). In cases of known ß-Strep infections, I will add 100 mL of 3% H2O2 to 1 L of fluids and lavage it in and out, usually followed by 1 L of plain LRS. This obviously represents a significant dilution to the reported dosage but has led to resolution of numerous clinical cases of ß-Strep in- fections when used in conjunction with other treatments, such as ecbolics and systemic antimi- crobials. There is minimal inflammatory response following either infusion or lavage with H2O2. Bacterial resistance to H2O2 has been documented in human cases but research is lacking in horses.

N-acetylcysteine

N-acetylcysteine (NAC) is a mucolytic agent and useful adjunct when treating cases of en- dometritis. Treatment involves diluting a 20% (200 mg/mL) solution in 150-250 mL of saline and instilling it into the uterus (LeBlanc and McKinnon, 2011). This can be done 24-48 prior to or 6-24 hours after breeding. Serial uterine lavages are often performed 12-24 hours after infusion and repeated until the efflux is clear. In a clinical study, mares treated with NAC prior to breed- ing had higher fertility rates than those that were not, and treatment had no adverse effects on the endometrium as judged by uterine biopsy (Gores-Lindholm et al., 2013). My personal expe- rience with NAC has been primarily as a pre-breeding lavage and it is not uncommon to obtain a cloudy and even chunky effluent. The exact source is unknown but could be inspissated material within the glands and lymphatics, which is a common histologic finding in uterine biopsy specimens from susceptible mares. Another source could be inflammatory cells and biofilm, but these observations are purely speculative.

Tricide®

Buffered chelator solutions, such as Tricide, have proven useful in cases of refractory bacterial and fungal infections. Tricide has been shown to potentiate the effects of certain antimicrobial agents, presumably by altering cell-wall permeability (LeBlanc, 2010). Amino- glycosides are one such example, and the two can be mixed together. Assuming appropriate sensitivity results, my preference is to add two (2) grams of gentamicin sulfate to Tricide® and instill it into the uterus. I will follow with daily lavages 12-24 hours later and repeat as necessary for 3-5 days. My clinical experience with Tricide is highly favorable. It is often my treatment of

105 choice in E. coli and P. aeruginosa infections. In vitro studies evaluating the use of a similar buffered chelator solution, tris-EDTA, showed it to be highly effective on free-floating bacteria (Ferris, 2014). However, tris-EDTA was inconsistent in disrupting biofilm production from clinical isolates of P. aeruginosa (Ferris, 2014), so it is by no means a “silver bullet” for suspect biofilm infections.

Kerosene

Kerosene is a clear liquid formed by fractional distillation of petroleum. Primarily used a fuel, it is also a useful solvent for removing grease and tenacious mucilage, as well as a pes- ticide for killing lice and bed bugs. Intrauterine infusion of kerosene in subfertile mares was first reported by Bracher et al. (1991). They reported relatively high early pregnancy (87.5%) and live-foal (62.5%) rates in mares treated with kerosene on the cycle prior to mating. Despite inducing an intense and, in some cases, diphtheroid inflammation, no long term side effects— such as cervical or uterine adhesions—were observed in mares treated with kerosene. My personal experience suggests that kerosene can be used at any stage of the estrous cycle, in- cluding diestrus and up to 8 hours prior to breeding. Mares who have gone more than one year barren, have not responded favorably during multiple cycles of other IU therapies, or have se- vere intrauterine fluid accumulation are candidates for uterine infusion of kerosene. Treatment is thought to perform a chemical curettage of the endometrial epithelium, effectively stripping the epithelium of debris, inspissated material, and microorganisms (Bradecamp et al., 2014).

My treatment protocol involves instillation of a fairly large volume of kerosene (500 mL in a typical 500 kg Thoroughbred), followed by uterine lavage the next day (and successive days if indicated). Effluent character ranges from mostly clear (yet strongly odorous of kerosene) to hemorrhagic with the consistency of curdled milk. Caution should be taken when handling and using kerosene due to it being flammable, and I recommend turning mares out in a paddock overnight. Of all the uterine therapies I have used for CDE, kerosene is the most reliable and successful treatment option I’ve used in the past three years. In collaboration with two other cli- nicians, we are in the midst of preparing a study involving ~35 clinical cases in which kerosene treatment was associated with increased pregnancy and live-foal rates in mares barren for ≥ 2 years (Cook et al., unpublished).

Regenerative Therapies

Relatively popular for musculoskeletal injuries and loosely falling under the umbrella of “regenerative therapies”, autologous and allogenic tissues are being explored as adjunct thera- pies for PMIE. Two decades ago, autologous plasma in conjunction with antibiotics was shown to increase both pregnancy and live-foal rates in lactating mares (Pascoe, 1995). More recently, the use of platelet-rich plasma (PRP) was explored. Preliminary studies have shown that IU infusion of PRP caused down-regulation of inflammatory cytokines and improved pregnancy rates in susceptible mares (Metcalf et al., 2012; Metcalf, 2014). Researchers at Colorado State University evaluated the use of autologous conditioned serum (ACS) and mesenchymal allogen- ic stem cells (MSCs) in reproductively normal mares (Ferris et al., 2014). Mares treated with ACS 24 hours prior to a dead-sperm challenge had significantly lower neutrophil counts 6 and 24 hours after this challenge compared to control mares. Those treated with MSCs had sig- nificantly lower neutrophil counts 6 h after sperm challenge relative to controls. More research and clinical studies are necessary to explore further the efficacy and indications for these treat-

106 ments. My personal experience is very limited with all of these therapies, although their use appears intriguing and potentially beneficial in the treatment of PMIE

Oxytocin

Intrauterine infusion of oxytocin appears to be a viable means for inducing uterine con- tractions. A study by Campbell and England (2002) demonstrated the efficacy of an intrauterine infusion of 30 IU oxytocin for inducing uterine contractions: specifically, in the horns. To my knowledge, the exact mode of action is unknown. Oxytocin as a relatively quick onset of action regardless of site of administration, so perhaps it rapidly absorbs or diffuses to the myometrium. I have been incorporating it into my antibiotic infusions or into my uterine irrigation fluids for the past 5 years with good clinical results.

Conclusion: The above discussion revisits some tried-and-true treatments and offers other relatively new options when dealing with problem mares. It is important to emphasize that no matter how innovative or novel a treatment is, it can all be for not without first performing a thor- ough physical and reproductive exam. Careful evaluation of past history, stallion fertility, perine- al conformation, and reproductive organs will help identify problem areas. This information can then be used to formulate a treatment plan to optimize fertility and produce a live foal.

107 The impact of aging on fertility, pregnancy, and offspring vigor in broodmares Dr. Charles F. Scoggin, DVM, MS, DACT Rood and Riddle Equine Hospital, 2150 Georgetown Road, Lexington, KY 40511, [email protected].

Take Home Message: There is a significant association between increasing mare age and decreasing reproductive efficiency. Age-related changes can occur to the external and inter- nal reproductive tract that can affect pregnancy and live-foal rates. Evidence also exists that increasing mare age can adversely influence offspring vigor and performance. Recognizing risk factors affecting reproductive efficiency can be useful during pre-purchase examinations and in providing proper care and management of older mares. Not only will it prolong their reproduc- tive careers, but it may also improve the chances of their offspring reaching their athletic poten- tial and/or market value.

Introduction: Many factors are associated with the likelihood of producing quality equine athletes. Genetics obviously casts a long shadow, and genome mapping has allowed research- ers to identify certain genes associated with speed in Thoroughbred racehorses (Bower et al., 2012). Epigenetic events can also influence the way an individual develops and expresses certain phenotypes. The age of the dam is one such epigenetic effect and is the current topic of interest. Age Is Not Just a Number

When is it Consensual?

While exceptions do occur, most horses reach sexual maturity between 12-18 months of age. In some breeds, such as Thoroughbreds, fillies start their breeding careers at relatively young ages (e.g., 3-6 years). In other disciplines, mares do not begin their breeding careers until much later (> 12 years) owing to lengthy training and performance schedules. The age when a mare starts her broodmare career is a useful piece of history to obtain when performing a reproductive exam. As will be reviewed below, age-related changes can occur regardless of parity, which can adversely affect fecundity.

How Old is Too Old?

There is no hard-and-fast rule for the age at which a mare will experience a decline in fertil- ity. Individual and environmental factors can also contribute to a mare’s reproductive efficiency, and these can be difficult either to account for or control. Nevertheless, a review of the litera- ture clearly demonstrates that age is one of the most consistent factors dictating a mare’s repro- ductive capacity. Various publications indicate that fertility begins to decline around 14 years of age (Allen et al., 2007; Bosh et al., 2009; Nath et al., 2010). The effect of age becomes even more extreme around 18 years when the incidence of early embryonic and fetal loss signifi- cantly increases (Allen et al., 2007). Nevertheless, not all mares are washed up by the time they reach 18 years of age. Somethingroyal—the dam of Secretariat—was that age when she foaled “Big Red” and had two more stakes horses at 19 and 20 years of age.

108 The Aging Process and Effects on Reproductive Efficiency

Anatomic Changes Related to Age

Age-related changes to the reproductive tract include endometrial degeneration (en- dometrosis), pendulous uterus, dysfunctional cervix, and poor perineal conformation. All of these—alone or in combination—can adversely affect reproductive performance in broodmares (LeBlanc and Causey, 2009). One of the most well documented changes in broodmares is worsening of perineal conformation with age. First described by Caslick (1937), poor vulvar conformation has been shown to adversely affect pregnancy and live-foal rates. The effect on fertility is related to aspiration of air into and contamination of the vagina, which causes inflam- mation and/or colonization of bacteria in the reproductive tract. The most common clinical pre- sentation is a decreased angle of declination of the vulva with respect to the horizontal plane, a shortened vulva, and a sunken anus. Treatment entails performing a Caslick vulvoplasty. The need for this surgery can be determined by calculating the Caslick index (Pascoe 1979) and can be used in mares with a history of subfertility. As a general rule, once a mare has a vulvo- plasty, she will need one during the remainder of her reproductive career (Bradecamp, 2011).

Regardless of how well managed a mare is or how many (or few) foals she has had in the past, she will develop progressive changes to her endometrium with advancing age. These changes can adversely affect pregnancy and live-foal rates (Kenney, 1978). Inflammatory cell infiltrates, glandular fibrosis and nesting, and lymphangiectasia are common histopathologic findings that, when combined with a history of subfertility, comprise the disease complex known as chronic degenerative endometritis (CDE; Allen, 1993). During gestation, the relative intimacy of the fetal-maternal interface is instrumental for nutrient exchange and promoting proper devel- opment of the fetus. A possible consequence of CDE is reduced development and efficiency of the fetal membranes (Bracher et al., 1996; Wilsher and Allen, 2003), which could increase the risk of either fetal demise or growth restriction and birth of a weak foal. The former is a problem due to lost revenue from stallion seasons and expenses incurred from maintaining a barren mare for another year. The latter is equally problematic due to added veterinary fees, increased risk of mortality, and reduced offspring vigor and performance.

Certain equestrian disciplines demand a lengthy and intense period of training and perfor- mance for the athletes to achieve success and prove their worth as a breeding animal. Conse- quently, some mares may not assume their broodmare careers until their mid- to late- teenage years. There is a subpopulation of these mares that prove to be difficult breeders due to cervi- cal dysfunction. Often referred to as the “Older Maiden Mare Syndrome,” the most consistent finding is a cervix that fails to dilate appropriately when the mare is in estrus (Pycock, 2006). These mares will often have a dominant follicle and adequate uterine edema, but, due to a closed cervix, retain a significant amount of intrauterine fluid following breeding.This can lead to persistent inflammation, which creates a poor environment for a developing conceptus. Ad - ditionally, and despite never carrying a pregnancy, endometrial biopsies from these mares can reveal glandular degeneration and fibrosis (Rickets and Alonso, 1991). These are also common findings in older multiparous mares, thus suggesting that the aging process itself can be just as detrimental to the uterus as the relative parity of the mare. Treatment usually involves judicious use of ecbolics and uterine irrigation to assist with clearance of fluid. Other treatments that can be of benefit include systemic corticosteroids (Dell Aqua et al., 2006; Bucca et al., 2008) and

109 prostaglandin-F2alpha (PGF). Direct application of either compounded misoprostol or butylsco- polamine can also be used to assist with cervical dilation. Finally, I am a firm believer in exer- cise. Keeping these mares active, whether through turnout or riding, can be of great benefit in keeping the mare and her uterus happy.

Physiologic Changes Related to Age

Reduction in gamete and zygote quality is another possible sequela of increasing mare age. Despite having similar fertilization rates in vivo, embryos collected from older mares demonstrated delayed development and altered morphology compared to young mares (Carne- vale et al., 1993a). Furthermore, when oocytes were collected from both young and old mares and subsequently transferred into young, inseminated recipients, embryo development rates were significantly lower in those recipients that received oocytes from old compared to young donors (Carnevale and Ginther, 1995). Poor oocyte quality is likely to blame (Carnevale et al., 1999; Carnevale et al., 2005; Rambags et al., 2006; Rambags et al., 2014), and it may height- en the risk of aneuploidy and early embryonic loss (Nagaoka et al., 2012). These findings are important considerations from a clinical standpoint because they allow the veterinarian to deter- mine a mare’s ability to carry a foal to term or her suitability as an embryo or oocyte donor. This information also helps in managing owners’ expectations regarding the relative fertility of their mare.

In addition to reduced quality, older mares may also have a lower quantity of viable oo- cytes. Female mammals are born with a fixed number of follicles and oocytes. As mares reach their geriatric (18-20) years, the number of follicles recruited per follicular wave can decrease, thereby leading to a relative depletion of oocytes available for fertilization. In time, a mare may effectively use up all of her follicles, leading to ovarian senescence. This phenonomen likely occurs in all mares, provided they live long enough.

The ability to determine how long a mare can remain productive would be a useful tool to have when performing breeding soundness evaluations. One potential means of rubbing the pro- verbial crystal ball is via measuring anti-müllerian hormone (AMH). In females, this protein is produced exclusively by granulosa cells in antral follicles. Measuring serum concentrations of AMH has been used in other species to predict: (a) the response to and relative success of in vitro fertilization techniques; and (b) the timing of menopause in women. As concerning equids, a recent abstract from researchers at the University of Kentucky reported lower antral follicle counts (AFCs) in mares > 18 years of age relative to younger mares (Claes et al., 2014). They also found a positive correlation between AFCs and AMH concentrations in the older group of mares. Circulating AMH concentrations were thus predictive of AFCs, suggesting that AMH may be useful for determining follicular reserve and reproductive efficiency in broodmares. More research is necessary to fully elucidate the benefit of measuringAMH concentrations in mares for the specific purpose of suitability for breeding.

Aging can also alter the length of the estrous cycle in mares. When pony mares ap- proached their mid-teens, researchers noted a reduction in cycle length, leading to ovulation of smaller follicles (Carnevale et al., 1993b). As the age of these mares increased to late-teens, cycle length increased to where some mares had only sporadic ovulations and, eventually, ovarian senescence (Carenevale et al., 1994). Irregular cycle length has important clinical considerations with respect to induction of ovulation and timing of insemination in older mares.

110 These mares often require more frequent monitoring, and, if an ovulatory agent is given, the clinician should pay careful attention to multiple ovulations to avoid twinning. Dam Age and the Association with Offspring Vigor

Advancing Mare Age Adversely Affects Offspring Athletic Potential and Performance Regardless of the discipline, one of the main premises on which breeding programs are based is to raise horses in a manner that allows them to realize their full athletic potential. Breeding is also big business, and the money involved at public auctions generates added pressure on breeders and consignors to present a commercially appealing product. Producing a quality athlete and/or marketable prospect involves a confluence of factors. Advancing mare age serves as one of the main influences affecting offspring vigor and value and will be dis- cussed below. The first year of life is one of the more formative times of a horse’s life. Prevention and management of disease is crucial during this phase. Illness can have lasting and even disas- trous consequences on athletic performance. Bacteremic Thoroughbred neonatal foals have been shown to have fewer wins and total earnings than healthy maternal siblings (Sanchez et al., 2008). When evaluating risk factors for foal disease, researchers identified increasing mare age as a major factor for morbidity and mortality in both neonates (≤ 14 days of age) and foals between the ages of 15 days and one year (Morley and Townsend, 1997). Mares ≥ 12 years of age were at a greater risk of producing sick foals compared to younger mares, and consequences of juvenile illness were pronounced: sick neonates were five times more likely to be unsuitable racing prospects assuming they reached one year of age, while older foals that experienced health issues were seven times more likely to be deemed unacceptable for athletic performance by one year of age. Based upon these findings, preventative measures should be taken to reduce the risk of illness in neonates and foals out of the older mares. These include routine monitoring of the mare during gestation, ensuring adequate passive transfer of the neo- nate, and serial evaluations of the foal for systemic health and distal limb conformation.

Various measures exist for evaluating the quality of equine athletes. Speed ratings are commonly used in racehorses and utilize mathematical equations to express the racing merit of an individual horse. With respect to age of the dam, ratings peaked for offspring out of mares that were 9 years of age and declined significantly thereafter (Barron, 1995). Another measure of athletic potential and performance is class. Class describes both tangible and intangible virtues of a racehorse that, collectively, serves as an indicator for that athlete’s relative skill level. In racehorses, quantitative measures of class are calculated similar to speed ratings and are based primarily on past performances, such as timed workouts and races. More esoteric indicators of class are pedigree, physical stature, tactical speed, grit, and heart. Class is thus a fluid and somewhat amorphous concept that can change throughout an individual racehorse’s career. One of the capstones of class is winning a stakes race. In a large study involving 1,641 broodmares who produced 18,931 foals—an astounding 11.5 foals per mare—Finocchio (1985) reported that a mare’s stakes-producing ability declined in a nearly linear fashion after seven years of age. Moreover, the likelihood of producing a stakes winner progressively waned beginning with the sixth parity. Statistical analyses of this data revealed that mares ≤ 10 years produced a greater proportion of stakes winners than those > 10 years, while a larger propor- tion of foals with birth-ranks < 6 were stakes winners compared to foals with higher birth-ranks

111 (Lema and Finocchio, 1985). When taken as a whole, these results provide testimony to the perception within the Thoroughbred breeding business that class begets class.

Unfortunately—and as most breeders and owners would attest to—there is no “sure thing” when it comes to breeding for performance and profit. The element of uncertainty looms large when breeding horses, thereby making it an unpredictable process. The inexact nature of equine reproduction has been shown to have direct consequences on economic returns and profitability. As demonstrated by Bosh et al. (2009b), Thoroughbred broodmares are long-term investments with positive gains are dependent on their reproductive efficiency over the course of several years. Profits and rates of return were greatest for maiden mares that produced a registered foal for seven consecutive years prior to being sold or for those that were bred for nine years and only barren one time four years into their production history. Not surprisingly, these researchers identified increasing mare age as a significant factor that limited the odds of producing a registered foal and, summarily, a positive financial yield. This data would indi- cate that—in Thoroughbreds—mares should start their reproductive careers early and remain consistently productive. Doing so would optimize the likelihood of producing a viable foal and reaping a positive investment.

Conclusion As discussed above, causes for the drop in mares’ fertility with advancing age are numerous and multifactorial. Changes occur to the mare’s reproductive anatomy and physiology that can adversely affect reproductive efficiency. Knowledge of these alterations can be of great benefit to the clinician to maximize a mare’s reproductive efficiency and managing clients’ expectations. Also, preemptive strategies can be employed to foals out of older mares owing to them being more prone to morbidity and mortality during the first year of life.

References 1. Allen, W.R. (1993). Proceedings of the John P. Hughes International Workshop on Equine Endome- tritis. Equine Vet. J. 25, 184-193. 2. Barron, J.K. (1995). The effect of maternal age and parity on the racing performance of Thorough- bred horses. Equine Vet. J. 27, 73-75. 3. Bower, M.A., McGivney, B.A., Campana, M.G., Gu, J., Andersson, L.S., Barrett, E., Davis, C.R., Mikko, S., Stock, F., Voronkova, Bradley, D.G., Fayhey, A.G., Lindgren, G., MacHugh, D.E., Sulimova, G., and Hill. E.W. (2012). The genetic origin and history of speed in the Thoroughbred racehorse. Nature communications 3, 643. 4. Bracher, V., Mathias, S., and Allen, W.R. (1996). Influence of chronic degenerative endometritis (endometrosis) on placental development in the mare. Equine Vet. J. 28, 180-188. 5. Carnevale, E.M, Griffin, P.G., and Ginther, O.J. (1993a). Age-associated subfertility before entry of the embryo into the uterus in mares. Equine Vet. J., Suppl. 15, 31-35. 6. Carnevale, E.M., Bergfelt, D.R., and Ginther, O.J. (1993b). Aging effects on follicular activity and concentrations of FSH, LH, and progesterone in mares. Anim. Reprod. Sci. 31, 287-299. 7. Carnevale, E.M, Bergfelt, D.R., and Ginther, O.J. (1994). Follicular activity and concentrations of FSH and LH associated with senescence in mares. Anim. Reprod. Sci. 35, 231-246. 8. Carnevale, E.M., Uson, M., Bozzola, J.J., King, S.S., Schmitt, S.J., and Gates, H.D. (1999). Com- parison of oocytes from young and old mares with light and electron microscopy. Theriogenology, 51, 299. 9. Carnevale, E.M., Coutinho da Silva, M.A., Panzani, D., Stokes, J.E., and Squires, E.L. (2005). Fac- tors affecting the success of oocyte transfer in a clinical program for subfertile mares. Theriogenology 64,

112 519-527. 10. Caslick, E.A. (1937). The vulva and the vulvo-vaginal orifice and its relation to genital health of the Thoroughbred mare. Cornell Vet. 27, 178-187. 11. Claes, A., Ball, B.A., Scoggin, K.E., Esteller-Vico, A., Kalmar, J.J., Conley, A.J., Squires, and E.L., Troedsson, M.H. (2014). Anti-müllerian hormone predicts follicular reserve in aged mares. In ‘Proceed- ings from the 31st annual convention of the American Association of Equine Practitioners,’ pp. 48-49. 12. Dell Aqua, J.A., Papa, F.O., Araujo, J.P., Jr., Alvarenga, M.A., Zahn, F.S., and Lopes, M.D. (2006). Modulation of acute uterine inflammatory response with prednisolone in mares bred with frozen semen. Anim Repro Sci, 94, 270-273. 13. Finocchio, E.J. (1985). Race performance and its relationship to birthrank and maternal age – I. In ‘Proceedings from the 31st annual convention of the American Association of Equine Practitioners.’ pp. 571-577. 14. Hemberg, E., Lundeheim, N., and Einarsson, S. (2005). Retrospective study on vulvar conforma- tion in relation to endometrial cytology and fertility in Thoroughbred mares. J. Vet. Med. 52, 474-477. 15. Kenney, R.M. (1978). Cyclic and pathologic changes of the mare endometrium as detected by biop- sy, with a note on early embryonic death. J. Am. Vet. Med. Assoc. 172, 241-262. 16. LeBlanc, M.M., and Causey, R.C. (2009). Clinical and subclinical endometritis: both threats to fertili- ty. Reprod. Dom. Anim. 44, 10-12. 17. Lema, J.-A.S, and Finocchio, E.J. (1985). Race performance and its relationship to birthrank and maternal age – II. In ‘Proceedings from the 31st annual convention of the American Association of Equine Practitioners.’ pp. 577-578. 18. Morley, P.S., and Townsend, G.G. (1997). A survey of reproductive performance in Thoroughbred mares and morbidity, mortality and athletic potential of their foals. Equine Vet. J. 29, 290-297. 19. Nie G.J., Johnson, K.E., Wenzel, J.G.W., and Braden, T.D. (2002). Effect of periovulatory ecbolics on luteal function and fertility. Theriogenology 58, 461-463. 20. Pascoe, R.R. (1979). Observations on the length and the angle of declination of the vulva and its relation to fertility in the mare. J. Reprod. Fertil. Suppl. 27, 299. 21. Pycock, J.F. (2006). How to maximize the chances of breeding successfully from the older maiden mare. In ‘Proceedings from the 52nd annual convention of the American Association of Equine Practi- tioners.’ pp. 577-578. 22. Rambags, B.P.B. van Boxtel, D.C.J., Tharasanit, T., Lenstra, J.A., Colenbrander, B., and Stout, T.A.E. (2006). Oocyte mitochondrial degeneration during reproductive ageing in the mare. Havemeyer Foundation Monograph Series 18, 25-27. 23. Rambags, B.P., van Boxtel, D.C.J., Tharasanit, T., Lenstra, J.A., Colendbrander, B., and Stout T.A.E. (2014). Advancing maternal age predisposes to mitochondrial damage and loss during maturation of equine oocytes in vitro. Theriogenology 81, 959-965. 24. Rickets, S.W., and Alonso, S. (1991). The effect of age and parity on the development of chronic endometrial disease. Equine Vet. J. 23, 189-192. 25. Sanchez, L.C., Giguere, S., and Lester, G.D. (2008). Factors associated with survival of neonatal foals with bacteremia and racing performance of surviving Thoroughbreds: 423 cases (1982- 2007). J. Am. Vet. Med. Assoc. 233, 1446-1452. 26. Wilsher, S., and Allen, W.R., (2003). The effects of maternal age and parity on pla- cental and fetal development in the mare. Equine Vet. J. 35, 476-483.

113 The benefit of routine monitoring and supplementation of proges- terone in early pregnancy Dr. Charles F. Scoggin, DVM, MS, DACT Rood and Riddle Equine Hospital, 2150 Georgetown Road, Lexington, KY 40511, [email protected].

Academic Involvement: Affiliate faculty member, Department of Clinical Sciences, Colorado State University; current member, Grayson Jockey Club Research Advisory Committee.

Take Home Message: Early and routine evaluation of serum progesterone concentrations may reduce the likelihood of embryonic and fetal loss by early intervention with supplemental proges- tin therapy. Primary indications for supplementation are: (1) serum progesterone concentrations ≤ 4 ng/ml; (2) declining circulating concentrations of progesterone on sequential evaluations; (3) history of poor reproductive performance; (4) mare illness; and (5) fetal stress. This paper will discuss certain methods of progesterone monitoring and indications for progestin supplementa- tion.

.Introduction: As the name implies, progesterone (P4) is the primary hormone responsible for maintenance of pregnancy in female mammals. Major target tissues include the tubular genitalia (to regulate tone and secretory activity) and the anterior pituitary (to regulate gonadotropin secre- tion). In horses, the source of P4 and other progestins for maintenance of pregnancy varies with the stage of gestation. During the embryonic period (Days 1 through 40 of gestation), the corpus luteum (CL) of pregnancy serves as the primary source of P4. The early portion of this period be- gins with ovulation (designated as Day 0) and is marked by increasing serum P4 concentrations as the site of the ovulation luteinizes and forms the CL. Circulating P4 concentrations typically peak 5-8 days post-ovulation (Allen, 2000). Maternal recognition of pregnancy (MRP) in horses occurs Days 14-15. The exact mechanism by which MRP occurs in horses is unknown; however, in normal pregnancies, this time period is associated with serum progesterone concentrations > 4 ng/ml (Allen, 2000). During the transition to the fetal stage (Day 41), secretion of equine chorionic gonadotropin (eCG) from endometrial cups induces formation of secondary CLs on the ovaries to “boost” endogenous P4 production. By Day 70, the fetal-placental unit (FPU; i.e., placenta or fetal membranes) begins to produce P4 and other progestogens. Upon reaching Days 100-120 of gestation, the FPU is able to manufacture enough progestogens to maintain the pregnancy without any further luteal support (Holtan et al., 1979). The CLs begin to regress at this time, and, by 180 days of gestation, the FPU serves as the sole source of endogenous progestogens. Circulating concentrations of P4 can vary in the second and third trimesters, but will drop precip- itously before foaling. Interestingly, most mares experience a small spike in progestogens just prior to foaling. This may or not always be significant, but, as will be discussed in another section, may be the cause for prolonged Stage II of labor seen in mares treated with altrenogest during the peripartum period. Biologically Active Progestins in Mares

Currently, there are only two proven progestins that are effective in maintaining pregnancies in broodmares. The first is native P4 that can be either mixed in oil and given at a dose 0.2-0.3 mg/kg, IM, q 24 hours or a long-acting formulation containing 1.5 g of P4 administered intramus-

114 cularly once every 7-10 days (Vanderwall, 2011). Both of these preparations require compound- ing, so the reader is cautioned to abide by state and federal regulations governing the use of these products in horses.

The other progestin with known activity in mares is altrenogest, which are commercially available in the USA as either ReguMate®a or Altresyn®b. This synthetic progestin is typically administered at 0.044 mg/kg, PO, q 24 h (~10 ml for an 1,100 lb mare). In cases where stress on the pregnancy is of concern (e.g., chronic orthopedic conditions, colic surgery, or placentitis), the dose of altrenogest can be doubled to 0.088 mg/kg, PO, q 24.

While there are drawbacks to both of these progestins—such as injection site reactions, frequency of administration, and human health hazards—precautions can be taken to avoid un- toward side effects. These include switching to a new needle prior to injections and wearing non-permeable gloves when handling these substances. For a review of safety in handling re- productive hormones, the clinician is directed to the paper prepared by Dr. Dirk Vanderwall in the Proceedings from the Annual Conference for the Society for Theriogenoloy (2012). Also of note is that both of these progestins are administered on an “extra-label” basis since there are no commercially available formulations labeled for use in pregnant mares. Other progestogens (e.g., Synovex® and Depo-Provera®) have been anecdotally reported to be of use for estrus suppression, but research has shown them to be either ineffective for this purpose (McKinnon et al., 2000) or biologically inactive (Gee et al., 2009; for a review, see McCue, 2003) in horses. Finally, it should be noted that altrenogest administration does not interfere with endogenous P4 testing, but P4 in oil or in the bio-release formulation will falsely elevate serum values. These are important considerations when monitoring P4 concentrations in mares.

Monitoring Endogenous P4

Evaluation of endogenous P4 is most commonly performed by obtaining a serum sample from the mare and submitting it to a veterinary laboratory for automated analysis via enzyme-linked immunabsorbent assay (ELISA). Turnaround time is rapid with results usually available within a few hours after submission. Values < 1 ng/ml indicate lack of a functional CL or other endog- enous P4 source. Other means of measuring P4 concentrations are radioimmunoassay (RIA) and competitive protein-binding assay (CPBA). The RIA is highly specific for P4 but has largely been replaced by the ELISA because of convenience. The CPBA lacks specificity and will detect numerous progestins, including P4 metabolites, and is used infrequently (McKinnon and Pycock, 2011).

The main benefit of evaluating endogenous P4 during early pregnancy is determining if the mare is producing adequate quantities of P4 to maintain her pregnancy. A serum concentration of > 4 ng/ml has long been the watermark for determining adequacy for pregnancy maintenance (McCue and McKinnon, 2011). Anything lower than this value is considered insufficient for main- taining a pregnancy. The term “luteal insufficiency” was originally used to describe this phe- nomenon, but the more contemporary phrase is “uteropathic abbreviation of the corpus luteum” (UACL). Case reports and anecdotal evidence suggests UACL is a real phenomenon (Canisso et al., 2013; Scoggin, unpublished data), but most theriogenologists are of the opinion that it is likely secondary to a primary issue, such as failure of MRP, low-grade endometritis, or embryonic defects. As will be discussed below, exogenous progestin supplementation can help abate UACL and, in some cases, “rescue” pregnancies despite low circulating concentrations of P4. It should

115 also be noted that endogenous P4 can also be used as a secondary sign of pregnancy, but el- evated circulating P4 concentrations are not very specific [i.e., prone to false positives] for the presence of a pregnancy and thus cannot be used as a stand-alone test to confirm pregnancy; other diagnostics, such as transrectal palpation and ultrasonography, are necessary to confirm pregnancy (and thus considered primary signs of pregnancy).

In terms of active monitoring, various protocols have been described demonstrating the use- fulness of evaluating endogenous P4 during early pregnancy. In embryo transfer (ET) recipients, blood was collected at the time of transfer and, if found pregnant, collected again seven days post-transfer (Foss and Crane, 2004). This method was useful for identifying recipient mares with low initial P4 serum concentrations and showed a strong correlation between elevated pro- gesterone concentrations and subsequent embryo survival. This study also demonstrated that P4-sampling at the time of transfer had low predictive value for identifying recipients that would eventually become pregnant, which was likely due to the expected rise in P4 at the time of trans- fer due to an active CL.

In the author’s practice, serum P4 concentrations are measured at least twice in all brood- mares. The first time is 5-6 days post-ovulation and the second occurs on the 45-day pregnancy check. The reasons for utilizing this testing schedule are as follows: P4 concentrations peak around the time of the initial check (Days 5-6 post-ovulation), thereby providing an accurate re- flection of CL activity and whether or not progestin supplementation is necessary. Sampling on the 45-day check coincides with formation of accessory CLs, which should keep circulating P4 concentrations above 4 ng/ml. Over the past five (5) years, we have submitted a total of 2,163 samples for P4 testing. Of these samples, 1,322 (61.12%) were collected from mares at Day 5 or 6. In this subset of samples, 26 of them were found to be ≤ 4.0 ng/ml, thereby providing a UACL rate of 1.97%. Progestin supplementation was initiated in all of these mares, and 14/26 (53.8%) pregnancies were effectively rescued to develop to 45-day pregnancies and beyond. While the incidence of UACL is relatively low, and success of rescuing pregnancies from these cases is slightly better than a coin-flip, the subsequent value of the offspring makes routine monitoring and supplemental progestin treatment a likely worthwhile endeavor. More information will be forthcoming as this data is further evaluated to include live-foaling rates.

Serum concentrations of P4 can also be evaluated at the time of the first pregnancy check. This is often done between Days 12-15 following ovulation. The primary benefit is providing owner or client reassurance with respect to the current status of the pregnancy. The main disad- vantage for waiting until this time period is the delayed response in initiating therapy should the mare be experiencing UACL. It is for this reason why I prefer to check P4 concentrations earlier (e.g., Days 5-6) after ovulation as described previously.

As pertaining to Day 45 P4 samples, it has been my observation values twice that of Day 5-6 values are a positive indication of a healthy pregnancy. Furthermore, preliminary data suggest that Day 45 P4 values > 14 ng/ml were associated with a higher likelihood of producing a live foal (Scoggin, unpublished data). More evaluation of the data is necessary to determine the strength of this association and whether it could be a useful predictive measure for determining the likeli- hood of the mare producing a live foal.

Finally, it is important to remember that progestins should be used judiciously. Mares main- tained on altrenogest that experience fetal loss may retain the pregnancy, leading to mummifica-

116 tion or maceration. Also, if supplementation is started too soon (prior to or just after ovulation), mares should be monitored closely for endometritis since the supplemental progesterone can inhibit uterine drainage. Fortunately, altrenogest does not appear to adversely affect endogenous P4 production in mares carrying their own pregnancy (Jackson, et al., 1986), and it may even be indicated in older mares experience delayed early embryonic development (Wilmann, et al., 2011).

Case selection for P4 Supplementation

The value of determining endogenous P4 concentrations during early pregnancy is extreme- ly useful for deciding if an when to supplement. Previous work in ovariectomized mares showed the benefits of exogenous progestin supplementation in maintaining pregnancies despite a lack of an endogenous source (Shideler et al., 1982; Ball et al., 1992; Knowles et al., 1993; Vanderwall et al., 2007). Other indications for progestin supplementation include a history of reproductive loss, advancing age, chronic orthopedic conditions, and cases of placentitis. Addressing the un- derlying disease process is also necessary for either resolution or production of a healthy foal.

Unknowns of supplemental P4 administration include: how long to give and how to withdraw it. Even though the FPU takes over the majority of progestin production by 120 days of gestation, it is not uncommon to continue supplemental P4 well past this time, especially when a mare is carrying a high-dollar pregnancy. In my practice, we will discontinue treatment at Day 45 if serum concentrations are double the value of the Day 5 concentration or if ≥ 14 ng/ml. However, in mares with a history of reproductive loss, it is not uncommon for us to continue altrenogest up to 300 days of gestation. Progestin concentrations drop precipitously before foaling, but there is a spike just prior to parturition. Hypothetically, exogenous progesterone could inhibit this spike. In addition, I have always stopped treatment “cold-turkey,” but other schedules have been described that use a tapering-dose. There is likely no harm in continuing therapy up until foaling, and I have seen several mares foal through altrenogest therapy. Side effects are minimal but may include increased duration of Stage II of labor and mild clitoral hypertrophy in neonatal fillies.

Conclusion

In summary, monitoring of endogenous P4 concentrations during early pregnancy can be use- ful for identifying mares that may require supplemental progestin during gestation. While the incidence of UACL is generally considered low, early identification may effectively rescue some pregnancies. Future studies will be aimed at determining the benefit of progestin monitoring and supplementation in terms of live-foal rates and efficacy in combating mid- and late-gestation re- productive loss.

117 Management of mares for breeding with frozen semen: pre- and post-insemination considerations for maximum fertility Kristina Lu VMD, DACT Hagyard Equine Medical Institute Lexington, KY

An ever increasing number of clients are interested in breeding with frozen semen. Obvious advantages include access to a global market, access to deceased stallions or the flexibility to breed to stallions that are otherwise unavailable due to show schedule or injury. Offering frozen semen breeding or participating in readying a mare for successful breeding can be a practice builder. The following is a review of a few factors that may impact success with frozen semen.

Client communication with regards to mare selection for frozen semen breeding is critical for success and/or meeting the client’s expectations. Mare factors include age, fertility, diet, and exer- cise. Evaluation of fertility should include at least a physical examination, perineal exam, palpation and ultrasonography of the reproductive tract per rectum, endometrial culture, and endometrial cytology. Further discussion regarding diagnostics is included in another lecture.

Mare age at the time of breeding is an important factor no matter what the breeding modality, however it is a factor the veterinarian may have little control over other than managing the clients’ expectations. Decreased per cycle and seasonal pregnancy rates may result from degenerative changes of the oocytes (Rambags 2014) and reproductive tract (Kilgenstein 2015), wear and tear of reproductive manipulations, and foaling. Below is a table adapted from Bosh et al. (2009) illus- trating the impact of age on pregnancy rates. This data was collected from Thoroughbred mares. An important note is that many mares, often warmbloods, present for frozen semen breeding after establishing their show careers and thus often present in the 9 to 18 year range at the time of first breeding.

Mare age group 2-8 years 9-13 years 14-18 years >18 years

Day 15 pregnancy rate per cycle 66.3% 65.6% 61.0% 48.1%

Day 15 pregnancy rate per season 94.5% 93.1% 88.2% 75.9%

% pregnancies lost days 15-40 4.6% 10.1% 16.7% 23.1%

% pregnancies lost day 40-foaling 12.1% 11.2% 16.8% 20.0%

Live foal rate per cycle 55.6% 52.4% 42.3% 29.6%

Live foal rate per season 82.9% 80.1% 68.4% 55.2%

Mare diet, feeding patterns and supplements are frequently a focus of conversation with many mare owners. Ultimately, the diet needs to meet the needs of both horse and owner. A couple of recent papers suggest worthy dietary topics.

Supplementation with docosahexaenoic acid, an omega-3 fatty acid, has received atten-

118 tion for its health benefits in several species. One study evaluated the efficacy of an omega-3 fatty acid product in modulating the uterine inflammatory response in response to frozen semen breeding in mares resistant to post breeding endometritis (n=10, mean age=5 years) and mares susceptible to post breeding endometritis (n=5, mean age 16 years). Mares were in inseminat- ed with a single dose of 1 billion killed sperm close to the time of ovulation. In this study, sperm were intentionally killed to exacerbate the inflammatory response to breeding. All mares were inseminated prior to supplementation and inseminated again after 60 days of supplementation with 14,400 mg of omega-3 fatty acids. This small study reported decreased intrauterine fluid in all mares after supplementation and breeding with a more notable decrease in intrauterine fluid and neutrophils in mares susceptible to post breeding endometritis. Increased pro-inflammatory cytokine mRNA expression was noted after dietary supplementation with no difference between mare groups (Brendemuehl 2014).

Time restricted feeding patterns versus continuous feeding may also impact reproductive success. Benhajali et al. (2013) evaluated this in 100 Arab mares divided into two groups, both groups receiving equivalent roughage, with one group only receiving hay overnight while the other group received hay overnight as well as during the day. Free access to water was provided at all times. This study found a significant difference between groups with mares fed continuously experiencing few estrous abnormalities, increased fertility (Benhajali 2013).

Owners breeding their competition horses often ask about the impact of exercise. Mortensen et al. (2009) evaluated this by comparing mares (n=16) that were exercised or not exercised over 2 estrous cycles for embryo flushing prior to crossing over the other group for another 2 estrous cycles for embryo flushing with a rest estrous cycle between study groups. Exercise consisted of 30 minutes of long trot or canter under ambient conditions of >30 °C and >50% humidity. Follicle diameter the day before ovulation was significantly greater in non-exercised mares compared to exercised mares. The rate of embryo recovery was greater in non-exercised mares with a higher proportion of the embryos being grade 1 (excellent). No significant residual effects of exercise were observed when mares transitioned from the exercised group to the non-exercised group (Mortensen 2009).

The World Breeding Federation for Sport Horses recommendation for the number of sperm in a frozen semen dose is a minimum of 250 million progressively motile sperm post-thawing with a minimum of 35% progressively motile sperm post-thaw (www.wbfsh.org). Doses lower than that described in recommendations are available for purchase and are often presented to veterinarians to use for breeding. Additionally, it is quite common to have only one dose available per cycle. As such, providing guidance during the purchase period may improve chances for pregnancy.

Inducing ovulation to time frozen semen breeding is commonly practiced. Advantages in- clude increasing the accuracy of breeding near ovulation and decreasing the number of doses required. Commonly used ovulation induction agents include deslorelin acetate and human cho- rionic gonadotrophin. Further discussion regarding induction of ovulation and fixed time frozen semen insemination is included in another lecture.

Deep horn insemination methods may be valuable for increasing pregnancy rates in the face of low insemination doses. Hysteroscopic insemination can be advantageous with very low

119 sperm numbers (3 x 106), however this technique requires more equipment and people than may be readily available. Variability in fertility of frozen thawed semen was still evident among com- mercially available stallions with much higher doses (50 x 106 to 100 x 106) despite use of hys- teroscopic insemination (Morris 2002). Transrectally guided deep horn insemination was found to yield similar pregnancy rates compared to hysteroscopic insemination and breeding doses of 0.5 x 106 and 1 x 106 sperm (Hayden 2012).

Post breeding examination is just as critical in frozen semen breeding as monitoring for ovu- lation up to breeding. Few controlled studies exist evaluating the inflammatory response to frozen semen. In one study evaluating various methods of insemination found the highest post breed- ing neutrophil count to be associated with frozen semen or concentrated fresh semen. Bacterial studies performed in parallel led the authors to conclude that the neutrophilia was likely induced by sperm rather than by bacteria (Kotilainen 1994). A more recent, retrospective study evaluating post insemination uterine fluid in 283 warmblood mares over 496 cycles, 25% of cycles were as- sociated with >2 cm of fluid with the highest incidence in barren mares and mares >10 years old. Per cycle pregnancy rates were lower in mares with uterine fluid (45%) compared to those without (51%), though this may also be due to mare age. Fluid management comprised primarily of oxy- tocin administration and uterine lavage. The author points out that the incidence of post-breeding fluid in this study was lower than they reported previously, likely due to stricter mare selection (Barbacini 2003).

Sieme et al. (2004) evaluated different insemination techniques and sperm doses in Hanoveri- an mares with (n=85) and without (n=187) abnormal reproductive histories (barren after multiple breeding attempts the previous season, embryonic loss, abortion, stillbirth). In problem mares, pregnancy rates were lower with frozen semen (16/43, 37%) compared to fresh semen (25/42, 60%). Frozen semen pregnancy rates of both mare groups were not influenced by technique, site of insemination, sperm dose or volume. Thus, with regards to frozen semen, fertility of the mare appeared to be the most important factor.

Pycock (2006) provided a review regarding breeding the older maiden mare, which is a com- mon frozen semen breeding patient. The mare’s reproductive health should be carefully evaluat- ed prior to breeding with correction of anatomical or physiological defects. Degenerative endome- trial changes and potential for poor cervical dilation are two key considerations and often manifest themselves as intrauterine fluid before and/or after breeding as well as post breeding induced endometritis. Ideally, the older maiden mare is bred only once in a cycle to minimize the potential inflammatory response and intrauterine fluid. Breeding once with a single dose of frozen semen just after ovulation poses the added risk that an already poorly dilated cervix may be closing when (post-breeding) fluid clearance is most important.

Another session in this meeting will address endometritis treatment. With regards to frozen semen, lavage with isotonic solutions 4 to 6 hours after breeding can aid in removal of excess sperm and debris, since sperm used for fertilization are in the oviduct at this time. This is often used in conjunction with ecbolics such as oxytocin. Cloprostenol (250 μg IM, Estrumate, Merck Animal Health, Summit, NJ) is sometimes recommended for prolonged uterine contractility though should be used with caution after ovulation to spare the CL. Acupuncture may also be useful in older mares. Intrauterine infusions of antimicrobials are often performed though the risk of intro-

120 ducing additional fluid to be cleared may outweigh the benefit if an infectious cause of post-breed- ing fluid is not identified.

Modulation of the immune response to breeding can be beneficial and several treatments have been evaluated in the context of improving pregnancy rates. Glucocorticoid use has been re- ported as a successful method of controlling post breeding endometritis. Compounds investigated include prednisolone acetate (0.1 mg/kg) (Papa 2008) and dexamethasone (50 mg IV per horse) (Bucca 2008). In a fertility trial evaluating the use of prednisolone, thirty cycles of fifteen mares were evaluated comparing frozen semen insemination without immunomodulatory treatment fol- lowed by insemination with corticosteroid therapy. Corticosteroid therapy consisted of 0.1 mg/kg prednisolone acetate administered when a 35 mm follicle and edema were present, followed by repeat administration every 12 hours until ovulation was detection. In this trial, none of the mares achieved pregnancy without prednisolone treatment and 64.5% achieved pregnancy with prednis- olone treatment (Papa 2008).

Dexamethasone (0.1 mg/kg IV) administered once at the time of breeding was evaluated for efficacy in reducing breeding induced inflammation and increasing pregnancy rates in susceptible mares. Dexamethasone decreased post-breeding small volume lavage efflux turbidity and endo- metrial edema. Dexamethasone did not alter the pregnancy per cycle rate, however in mares that were determined to be at high risk of developing post-breeding endometritis with at least three of the study’s risk factors, pregnancy rates were significantly improved. Risk factors included: ab- normal reproductive history, positive endometrial culture, at least 2 cm endometrial fluid prior to breeding, abnormal perineal conformation or unrepaired Caslicks after foaling, abnormal cervix, greater than 1.5 cm post-breeding fluid, post-breeding fluid persisting beyond 36h, and abnormal- ities of the reproductive tract (Bucca 2008).

NSAIDs are commonly used in clinical practice to manage inflammation associated with breeding, embryo transfer, and placentitis. In a study evaluating the COX-2 inhibitor vedaprofen, barren mares (n=8) with a history of post-breeding endometritis were treated (2 mg/kg PO initial dose, 1 mg/kg PO BID) beginning the day before artificial insemination and continuing until 1 day after ovulation. These mares were compared to an untreated group though all mares received oxytocin. This study found a significantly improved pregnancy rate among mares treated with the COX-2 inhibitor around the time of breeding (Roger 2010).

Immunostimulants containing Propionibacterium acnes (EqStim, Neogen Corp, Lexington, KY) has been shown to induce a non-specific cell-mediated response predominantly by macro- phage activation and cytokine release. In a study evaluating its efficacy in a clinical setting, 95 mares with cytological evidence of endometritis were randomly administered P. acnes or placebo additional to other veterinary treatments. This study found improved pregnancy and live foal rates (Rohrbach 2007).

The use of platelet rich plasma is widely practiced in orthopedic cases in attempt to expedite healing with growth factors and inflammatory modulators. Recently, practitioners have described its use in the uterus for treatment of post-breeding endometritis. One study evaluated the efficacy of infusion of 20 mL autologous PRP (containing on average 257,000 platelets per microliter) 4 hours after insemination in 15 mares resistant to post-breeding endometritis and 8 mares sus-

121 ceptible to post-breeding endometritis. Parameters evaluated included nitric oxide concentration in uterine fluid, percent neutrophils in uterine cytology, and uterine fluid quantity 24 hours after insemination. In resistant mares, the only significant difference was in percent neutrophils in cytology. In susceptible mares, a significant difference was seen in percent neutrophils, nitric oxide quantity and uterine fluid leading the group to conclude that PRP reduced the inflamma- tory response after breeding (Reghini 2014). Another study described improved pregnancy and decreased post-breeding fluid in mares susceptible to post-breeding endometritis following PRP infusion (Metcalf 2014).

Barbacini S, Necchi D, Zavagia G, Squires EL. Retrospective study on the incidence of postinsemination uterine fluid in mares inseminated with frozen/thawed semen. J Equine etV Sci 2003;23:493-496.

Benhajali H, Ezzaouia M, Lunel C, Charfi F, Hausberger M. Temporal feeding patter may influence repro- duction efficiency, the example of breeding mares. PLOS One 2013;8:1-5.

Bosh KA, Powell D, Neibergs JS, Shelton B, Zent W. Impact of reproductive efficiency over time and mare financial value on economic returns among Thoroughbred mares in central Kentucky. Equine Vet J 2009;41:889-894.

Brendemuehl JP, Kopp K, Altman J. Influence of dietary algal N-3 fatty acids on breeding induced inflamma- tion and endometrial cytokine expression in mares bred with frozen semen. J Equine Vet Sci. 2014;34:123- 4.

Bucca S, Carli A, Buckley T, et al. The use of dexamethasone administered to mares at breeding time in the modulation of persistent mating induced endometritis. Theriogenology 2008;70:1093-100.

Hayden SS, Blanchard TL, Brinsko SP, Varner DD, Hinrichs K, Love CC. Pregnancy rates in mares in- seminated with 0.5 or 1 million sperm using hysteroscopic or transrectally guided deep-horn insemination techniques. Theriogenology 2012;78:914-920.

Kelley DE, Gibbons, JR, Smith R, Vernon KL, Pratt-Phillip SE, Mortensen CJ. Exercise affects both ovarian follicular dynamics and hormone concentrations in mares. Therio 2011;76:615-622.

Kilgenstein, HJ, Schöniger S, Schoon D, Schoon HA. Microscopic examination of endometrial biopsies of retired sports mares: an explanation for the clinically observed subfertility? Res Vet Sci 2015;99:171-179

Kotilainen T, Huhtinen M, Katila T. Sperm-induced leukocytosis in the equine uterus. Theriogenology 1994;41:629-636.

Metcalf ES. The effect of Platelet-Rich Plasma (PRP) on intraluminal fluid and pregnancy rates in mares susceptible to Persistent Mating-Induced Endometritis (PMIE). Journal of Equine Veterinary Science 2014;34:128.

Morris LHA, Allen WR. An overview of low dose insemination in the mare. Reprod Dom Anim 2002;37:206- 210.

Mortensen CJ, Choi YH, Ing NH, Kraemer DC, Vogelsang SG, Vogelsang MM. Embryo recovery from ex- ercised mares. Anim Reprod Sci 2009;110:237-244

122 Papa FO, Dell'aqua JAJ, Alvarenga MA, et al. Use of corticosteroid therapy on the modulation of uterine in- flammatory response in mares after artificial insemination with frozen semen. Pferdeheilkunde 2008;24:79- 82.

Pycock JF. How to maximize the changes of breeding successfully from the older maiden mare. AAEP Proceedings San Antonio, TX 2006;52:245-249.

Rambags BPB, van Boxtel DCJ, Tharasanit T, Lenstra JA, Colenbrander B, Stout TAE. Advancing maternal age predisposes to mitochondrial damage and loss during maturation of equine oocytes in vitro. Therio 2014;81:959-965.

Reghini MFS, Bussiere MCC, Neto CR, et al. Effect of use of platelet rich plasma on post-breeding uterine inflammatory response of mares. Journal of Equine Veterinary Science 2014; 34:127.

Rohrbach BW, Sheerin PC, Cantrell CK, et al. Effect of adjunctive treatment with intravenously administered Propionibacterium acnes on reproductive performance in mares with persistent endometritis. Journal of the American Veterinary Medical Association 2007;231:107-13.

Rojer H and Aurich C. Treatment of Persistent Mating-Induced Endometritis in Mares with the Non-Steroid Anti-Inflammatory Drug Vedaprofen. Reproduction in Domestic Animals 2010;45:e458-e460.

Sieme H, Bonk A, Hamann H, Klug E, Katila T. Effects of different artificial insemination techniques and sperm doses on fertility of normal mares and mares with abnormal reproductive history. Theriogenology 2004;62:915-928.

123 How to successfully integrate assisted reproductive techniques in your busy practice Kristina Lu VMD, DACT Hagyard Equine Medical Institute Lexington, KY

Incorporating assisted reproductive techniques in a busy practice is time and timing manage- ment. The goal is to minimize the numbers of intensive breedings per pregnancy, not just for the sake of the client, but also for the purposes of managing time. This includes initial groundwork such as assessing the reproductive health of the mare, identifying the fertility of the stallion in the modality being used, and constructing a plan to optimize potential success in conjunction with preparation for failure.

Advancing the onset of the breeding season is less common in sport horse mares taking advantage of assisted reproductive techniques as compared to breeds such as Thoroughbreds. However, there are times that a client is quite keen to breed on the first observed cycle (first ovu- lation) of the season, commonly leading to questions regarding this cycle’s fertility. Is it worth the time and effort versus “programming” the mare or short-cycling after ovulation is detected? In attempt to evaluate this, Cuervo-Arango et al. compared the pregnancy rates of Thoroughbred mares being bred on the first ovulation of the year (n=46) with mares being bred on subsequent ovulations (n=50) and mares that had received progesterone prior to being bred on the first ovu- lation of the year (n=29). In this study, the first service pregnancy rates were not significantly different between the groups. Notably, the percent of mares bred more than once in the same cycle was significantly higher (21.7%) in the mares bred on the first ovulation of the year without progesterone priming. In conjunction with this, 58.7% of mares bred on the first ovulation with- out progesterone priming ovulated within 48 hours of hCG administration compared to 93.1% of mares that received progesterone priming (Cuervo-Arango 2010).

Administration of prostaglandin F2a to “short cycle” mares can be a time saver. In a compar- ison of pregnancy rates among mares that were administered 125 to 250 μg cloprostenol during behavioral diestrus (n=231) with mares that received no luteolytic treatment (n=230), no difference in pregnancy rate was observed (Metcalf 2010). A case report exists regarding the increased incidence of hemorrhagic anovulatory follicles following cloprostenol sodium (Estrumate, Merck Animal Health, Summit, NJ) administration to induce estrus in two mares (Cuervo-Arango 2009).

Follicle size at the time of prostaglandin F2a administration is significant for estimating the number days until breeding. A retrospective study of 275 mares and 520 estrous cycles sought to evaluate this. Mares received 250 μg cloprostenol IM 5 to 12 days after ovulation. The average interval to ovulation was 8.4 ± 2.5 days with the days to ovulation inversely proportional to the diameter of the largest follicle. If a follicle ≥35 mm was present at the time of cloprostenol administration, 73.1% of mares ovulated after 48 hours usually demonstrating endometrial edema, 13.4% of mares ovu- lated within 48 hours demonstrating variable endometrial edema, and 13.4% of mares regressed the follicle. In this study, incidence of hemorrhagic anovulatory follicles was 2.5% (Burden 2015).

124 Does the interval from prostaglandin F2a administration to ovulation effect pregnancy rate? A recent publication describes this in two scenarios, one in which mares (n=215 mares, 513 estrous cycles) were bred with frozen thawed semen with pregnancy detection at 14 days, and second in which mares (n=65 mares, 375 embryo flushes) were bred with fresh semen and embryo flushed 8 or 9 days after ovulation. The interval from prostaglandin F2a administration to ovulation was divided into three groups and compared to mares that were not treated with prostaglandin F2a (spontaneous luteolysis). Pregnancy rates are listed below. Of the factors evaluated in this study, interval from luteolysis to ovulation was the only factor that had significant impact on pregnancy rate (Cuervo-Arango 2015).

<6 days 6-8 days >8 days Natural

Frozen, carry 26.6% 39.4% 55.9% 42.5%

Fresh, embryo 55.0% 62.6% 73.7% 75.0%

Another potential application of prostaglandin F2a is antiluteogenesis. Antiluteogenesis was achieved with administration of 10 mg of dinoprost (Lutalyse, Pfizer, New York, NY) IM twice daily on days 0, 1, and 2 days after ovulation and once daily on days 3 and 4. Pregnancy rates were similar when compared to mares receiving 10 mg dinoprost at 10 days post-ovulation. The mean interovulatory interval was 13.1 ± 3.7 days with antiluteogenesis treatment compared to 18.5 ± 2.0 days with day-10 luteolytic treatment (Coffman 2014).

Reliable induction of ovulation is a mainstay in breeding time management to optimize sched- uling of semen ordering, breeding, and/or the veterinarian. Two of the more commonly used prod- ucts include human chorionic gonadotrophin (hCG, Chorulon, Merck Animal Health, Summit, NJ) and deslorelin acetate (SucroMate, Thorn-Biosciences, Louisville KY). The efficacy of SucroMate (1 mL, 1.8 mg deslorelin acetate, IM) has been compared to hCG (2.5 mL, 2,500 IU IV) in 256 Quarter Horse mares over 302 estrus cycles. There was no difference in the percentage of mares that ovulated within 48 hours after deslorelin compared to hCG. There was, however, a significant increase in the percentage of mares that ovulated within 24 hours after administration of hCG compared to deslorelin. In this study, the average intervals to ovulation were 41.4 ± 9.4 hours for deslorelin and 44.4 ± 16.5 hours for hCG (Ferris 2012).

Relatively recent publications suggest other potential benefits from hCG and deslorelin that may improve reproductive efficiency. Köhne et al reported that progestin concentration 5 to 15 days after ovulation was higher in mares whose ovulations were induced with hCG (n=14) com- pared to mares whose ovulations were not induced (n=28). Mares that double ovulated or mares that did not ovulate within 48 hours of hCG administration were excluded from the study (Köhne 2014). Nagao et al. evaluated the use of deslorelin to reliably induce double ovulation in order to optimize embryo production. After luteolysis 8 days after ovulation, mares were monitored until two follicles were detected, one no larger than 25 mm in diameter and one no smaller than 20 mm in diameter. At such time, 100 μg deslorelin acetate was administered q 12 hours. Ovulation was induced with 2,500 IU hCG. The number of days until ovulation was 3.5 days in the deslorelin treated group and 6.7 days in the control (saline treated) group. The interval from hCG to ovula-

125 tion was shorter in deslorelin treated mares (35 ± 6.1 hours) compared to saline treated mares (42 ±5.1 hours). The average number of ovulations per estrous cycle and the number of recovered embryos per estrous cycle was greater in the deslorelin treated group. There was no difference in the number of embryos recovered per ovulation (Nagao 2012).

Breeding with frozen semen is often perceived as a cumbersome breeding modality that fits poorly in a busy veterinarian’s schedule. In actuality, breeding management and timed insemina- tion can make frozen semen breeding quite feasible. Stallion fertility is a variable that is often out of the veterinarian’s control. Avanzi et al (2014) evaluated post-ovulation insemination and fixed- time insemination with semen from a fertile and a subfertile stallion. Twenty-nine mares were bred twice, once with a fixed time protocol and once with a post-ovulation protocol. Mares were bred with semen from either a fertile stallion or a subfertile stallion with both breedings occurring with semen from the same stallion. For post-ovulation insemination, mares were inseminated once with 800x106 total sperm no more than 6 hours after ovulation. For fixed-time insemination, mares were inseminated twice with 400x106 total sperm 24 and 40 hours after receiving deslorelin ac- etate. Pregnancy rates were similar for both protocols regardless of stallion, though a significant effect of the stallion was observed. Intrauterine fluid accumulation 24 hours after insemination was more frequently observed in mares bred once post-ovulation (Avanzi 2014).

Newcombe et al (2011) evaluated the effect of real life veterinary practice on frozen semen breeding. Though the intent was to evaluate and breed all mares within 6 hours of ovulation, this was not always possible and thus they evaluated the effect of extending the ovulation detection window on pregnancy rate. Data was evaluated from 14 breeding seasons, 867 cycles and over 700 mares of varying ages, breeds. Breeding was performed once per estrous cycle with a sin- gle dose of frozen semen of variable qualities. The data was evaluated by grouping ovulation detection to insemination time into 3 hour intervals ranging from 0 hours (breeding as ovulation was occurring) to 15 hours. Pregnancy rates were not statistically different between groups. The overall pregnancy rate of the study was 47.9% (Newcombe 2011).

Embryo transfer is another technique that can be integrated into a busy veterinary practice, though potentially more variables require attention compared to breeding for the mare to carry to term. Variables in addition to donor mare management and reproductive health include recipient management, ovulation synchronization, operator skill, embryo transfer method, embryo quality and recipient management after transfer. Camargo et al (2013) evaluated the effects of embryo size, embryo developmental stage, embryo quality, and donor and recipient synchrony on preg- nancy rates with 396 embryos from 120 Brazilian Jumper Horse mares. Factors that significantly improved pregnancy rates included embryos of a diameter ranging from 400-1199 μm compared to smaller embryos, embryos that had developed into blastocysts or expanded blastocysts, embry- os that were of the best (grade 1) quality, and transfer into recipients that ovulated the same day to 4 days after the donor (Camargo 2013).

Oocyte aspiration and intracytoplasmic sperm injection present a larger hurdle to integrate into an otherwise busy veterinary practice without significant practice on non-client horses and some investment in equipment and supplies. A recent presentation provided an overview of the procedure (Ortis 2013). Mature oocytes can be aspirated from dominant gonadotrophin-stimu- lated follicles or immature oocytes can be aspirated from follicles that are 10 to 20 mm in diameter.

126 Intracytoplasmic sperm injection (ICSI) in the horse in only available in a few locations. Thus, shipment of oocytes would increase the availability of this technique. A recent short report de- scribes the effect of potential oocyte transport protocols on blastocyst rates after ICSI. Immature oocytes can be shipped in a commercially available embryo holding solution at room temperature yielding a 35 to 37% blastocyst production. A higher blastocyst production rate (70%) can be achieved with mature oocytes from dominant gonadotrophin-stimulated follicles, however these oocytes are more time and temperature sensitive (Foss 2013).

Avanzi BR, Ramos RS, Araujo GHM, Fioratti EG, Trinca LA, Dell’Aqua Jr JA, Melo-Oña CM, Zahn FS, Mar- tin I, Alvarenga MA, Papa FO. Fixed-time insemination with frozen semen in mares: is it suitable for poorly fertile stallions? 2014, doi: 10.1016/j.theriogenology.2014.07.007.

Burden CA, McCue PM, Ferris RA. Effect of cloprostenol administration on interval to subsequent ovulation and anovulatory follicle formation in Quarter Horse mares. J Equine Vet Sci 2015;35:531-535.

Camargo CE, Weiss RR, Kozicki LE, Duarte MP, Duarte MCG, Lunelli D, Weber S, de Abreu RA. Some factors affecting the rate of pregnancy after embryo transfer derived from the Brazilian Jumper Horse breed. J Equine Vet Sci 2013;33:924-929.

Coffman EA, Pinto CRF, Snyder HK, Leisinger CA, Cole K, Whisnant CS. Antiluteogenic effects of serial prostaglandin F2α administration in cycling mares. Therio 2014;82:1241-1245.

Cuervo-Arango J, Newcombe JR. The effect of cloprostenol on the incidence of multiple ovulation and anovulatory hemorrhagic follicles in two mares: a case report. J Equine Vet Sci 2009;29:533-539.

Cuervo-Arango J, Clark A. The first ovulation of the breeding season in the mare: The effect of progesterone priming on pregnancy rate and breeding management (hCG response rate and number of services per cycle and mare). Anim Reprod Sci 2010;118:265-269.

Cuervo-Arango J, Mateu-Sánchez S, Aguilar JJ, Nielsen JM, Etcharren V, Vettorazzi ML, Newcombe JR. The effect of the interval from PGF treatment to ovulation on embryo recovery and pregnancy rate in the mare. Therio 2015;83:1272-1278.

Ferris RA, Hatzel JN, Lindholm ARG, Scofield DB, McCue PM. Efficacy of deslorelin acetate (Sucromate) on induction of ovulation in American Quarter Horse mares. J Equine Vet Sci 2012;32:285-288.

Foss R, Ortis H, Hinrichs K. Effect of potential oocyte transport protocols on blastocyst rates after intracy- toplasmic sperm injection in the horse. AAEP Proceedings Nashville TN 2013;59:525

Köhne M, Kuhl J, Ille N, Erber R, Aurich C. Treatment with human chorionic gonadotrophin before ovula- tion increases progestin concentration in early equine pregnancies. Anim Reprod Sci 2014;149:187-193.

Metcalf ES, Thompson MM. The effect of PGF2a-induction of estrus on pregnancy rates in mares. J Equine Vet Sci 2010;30:196-199.

Newcombe JR, Paccamonti D, Cuervo-Arango J. Reducing the examination interval to detect ovulation

127 below 12 h does not improve pregnancy rates after postovulatory insemination with frozen/thawed semen in mares. Anim Reprod Sci 2011;123:60-63.

Ortis H, Foss R. How to collect equine oocytes by transvaginal ultrasound-guided follicular aspiration. AAEP Proceedings Nashville TN 2013;59:519-524.

128 Diagnostic methods and interpretation for endometritis Kristina Lu VMD, DACT Hagyard Equine Medical Institute Lexington, KY

The standard methods for diagnosing endometritis are a uterine swab for microbial culture, endometrial cytology to evaluate for the presence of inflammation, and an endometrial biopsy for histopathology and/or microbial culture. Bacterial uterine infections are associated with signifi- cant time and monetary loss in the equine breeding industry. The incidence of identified bacte- rial uterine infection is estimated at 25 to 60% of barren mares (Causey 2006). Beta-hemolytic Streptococcus equi subspecies zooepidemicus, remains the most common bacterial reproductive pathogen in horses. One study identified beta-hemolytic Streptococcus in 3.8% (78/2044) of all uterine swabs and in 34% (78/234) of uterine swabs from which a microorganism was isolated, with the second most common isolate being E. coli (17%) (Riddle 2007). Another study found the incidence of positive bacterial cultures among a population of mares presented for fertility assess- ment to be 39% (84/212), of which 77% (or 29% of all mares) grew beta-hemolytic Streptococcus (Nielsen 2005). In 2014, the two most commonly isolated organisms from endometrial swabs at the Hagyard Diagnostic Laboratory were beta-hemolytic Streptococcus followed E. coli. A more detailed list of isolated organisms and their frequency of sensitivity to various antimicrobials are enclosed (Elam, 2014)

In a comparison of the uterine swab with endometrial biopsy for assessment of any bacterial growth, one study found that in 2% of all positive cultures, bacterial growth was observed from the swab and not from the biopsy, while in 55% of positive cultures the swab was negative for bacterial growth and the biopsy positive. This study went on to calculate the sensitivity (ability of the test to correctly identify patients with the disease) and specificity (ability of the test to correctly identify patients without the disease) in two additional scenarios: culture using a swab compared to cul- ture from an endometrial biopsy as the gold standard, and culture from either a swab or a biopsy compared to inflammatory cells seen in the biopsy as the gold standard (Nielsen 2005).

Swab with culture from biopsy as gold standard:

Sensitivity Specificity Positive Predictive Value Negative Predictive Value Swab culture 0.44 0.98 0.95 0.74

Tests with inflammatory cells in the biopsy as the gold standard3:

Sensitivity Specificity Positive Predictive Value Negative Predictive Value

Swab culture 0.34 1.00 1.00 0.44

Cytology 0.77 1.00 1.00 0.62

Biopsy culture 0.82 0.92 0.97 0.67

In an assessment of subclinical endometritis as defined by polymorphonuclear cells (PMNs) present during diestrus in the stratum compactum of the endometrial biopsy in the absence of

129 intrauterine fluid, another study compared the value of a uterine swab, a cytology brush, and an endometrial biopsy for cytological and bacteriological diagnosis of endometritis, with the following results (Overbeck 2011):

Sensitivity Specificity

Cytology Bacteriology Cytology Bacteriology

Swab 0.00 0.33 0.93 0.83

Brush 0.17 0.25 0.83 0.80

Biopsy 0.25 0.25 0.85 0.95

Brush: cytology+bacteriology 0.42 0.70

These studies are suggestive that uterine swab for microbial culture used by itself is insuffi- cient for diagnosis of endometritis. Diagnostic ability is improved with addition of at least endome- trial cytology though other studies suggest the brush technique may be superior to a swab.

Another valuable diagnostic technique is low volume lavage for culture and cytology. The advantage of this method is the ability to sample a larger surface area compared to swab and brush techniques. A recent study evaluated cytology samples obtained via double guarded swab, double guarded brush and low volume lavage with either 0.9% saline or lactated Ringer’s solution in reproductively normal mares. The swab method was the only method that occasionally yielded non-diagnostic slides and most frequently yielded slides of inferior quality compared to the other methods. Brush sampling yielded the best quality samples. Regardless of collection technique, samples contained <1 neutrophil/400x field though low volume lavage samples were more likely to contain neutrophils. Background mucus was most consistent with brush samples and minimal in low volume lavage samples (Bohn 2014).

Cocchia et al. compared the utility of the cytobrush, cotton swab and low volume lavage for endometrial cytology from 20 mares with a history of subfertility. Both cytobrush and low volume lavage were deemed superior than cotton swab for diagnosing PMNs with poor agreement be- tween the methods for diagnosing acute endometritis in this study (Cocchia 2012). The stage in the estrous cycle that the sample is obtained is an important component of cytological interpre- tation. Kozdrowski et al. evaluated this in 127 barren mares in estrus (n=46), diestrus (n=48) or anestrus (n=33) comparing endometrial biopsy to cytology obtained with a brush. Additionally they evaluated 2 methods of cytology interpretation: 1) percentage of PMNs out of 300 cells with >2% considered to be positive for endometritis, or 2) 0-2 PMNs per high power field indicating no inflammation, 2-5 PMNs/HPF indicating moderate inflammation and >5 PMNs/HPF indicating severe inflammation, averaging from 10 randomly chosen HPFs. When compared to endometrial biopsy, sensitivity values were highest with method 1 with samples collected during estrus using a brush (Kozdrowski 2015).

A polymerase chain reaction (PCR) assay is another method for diagnosing endometritis. Ferris et al. (2014) described a semi quantitative PCR that was capable of detecting 33 bacteria considered to be potential equine uterine pathogens. Currently the primary application of this as-

130 say is identification of pathogenic bacteria in the uterus when the mare is suspected of having an infectious endometritis despite a negative result from traditional microbial culture (Ferris 2014).

Endometrial biopsy for histopathological evaluation can provide significant information re- garding endometrial health and potential future management. This technique aids in detection of clinical and subclinical inflammation, degeneration, and glandular differentiation disorders. Age, more than parity, plays a prominent role in chronic degenerative endometrial disease. Suggested guidelines are that mares up to nine years of age should have no signs of degenerative endome- trial disease, mares 9 to 13 years old should only have mild changes, mares 13 to 15 years old should have less than moderate changes, and mares 17 years and older may have severe degen- erative changes (Ricketts 1991).

A retrospective study was performed on 189 retired sport mares in attempt to assess reasons for lower than expected initial pregnancy rates often seen among older maiden mares recently retired from competition. This study included maiden mares (n=68), barren mares (n=84), and mares that had foaled (n=37). Not surprisingly, the incidence and severity of endometrosis (chron- ic degenerative endometrial disease) was associated with higher mare age. Endometritis was least commonly diagnosed among unbred mares. Glandular differentiation disorders including en- dometrial inactivity and irregular glandular differentiation were observed in 50% of mares biopsied during the breeding season, with the most frequent occurrence in maiden mares that were most recently retired from sports (Kilgenstein 2015). Bohn AA, Ferris RA, McCue PM. Comparison of equine endometrial cytology samples collected with uterine swab, uterine brush, and low-volume lavage from healthy mares. Vet Clin Path 2014;43:594-600.

Causey, RC. Making sense of equine uterine infections. The Veterinary Journal 2006; 172:405-421.

Cocchia N, Paciello O, Auletta L, Uccello V, Silvestro L, Mallardo K, Paraggio G, Pasolini MP. Comparison of the cytobrush, cottonswab, and low-volume uterine flush techniques to evaluate endometrial cytology for diagnosing endometritis in chronically infertile mares. Theriogenology 2012; 77:89-98.

Elam, T. Hagyard laboratory 2014 uterine sensitivity report. Hagyard Equine Medical Institute. www.hag- yard.com, 859-259-3685. Kilgenstein, HJ, Schöniger S, Schoon D, Schoon HA. Microscopic examination of endometrial biopsies of retired sports mares: an explanation for the clinically observed subfertility? Res Vet Sci 2015;99:171-179

Kozdrowski R, Sikora M, Buczkowska J, Nowak M, Ras A, Dziecio) M. Effects of cycle stage and sampling procedure on interpretation of endometrial cytology in mares. Anim Repro Sci 2015;154:56-62.

Nielsen, JM. Endometritis in the mare. Theriogenology 2005; 64:510-518.

Overbeck W, Witte TS, Heuwieser W. Comparison of three diagnostic methods to identify subclinical endo- metritis in mares. Theriogenology 2011; 75:1311-1318.

Ricketts SW, Alonso S. The effect of age and parity on the development of equine chronic endometrial dis- ease. Equine Vet J 1991;23:189-192.

Riddle, WT, LeBlanc MM, Stromberg AJ. Relationships between uterine culture, cytology and pregnancy rates in a Thoroughbred practice. Theriogenology 2007; 68:395-402

131 132