Page 1712 

LEAD

Lead toxicosis is discussed in Chapter 35.

MERCURY

Mercury toxicosis has been associated with ingestion of seed treated with organic mercurial fungicides.505-507 Topical application or ingestion of inorganic mercurials, used as counterirritants (blistering agents), also can result in toxicosis.505,507,508 If the blistering agents are used concurrently with dimethyl sulfoxide (DMSO), the absorption of the mercury is enhanced and is more likely to cause toxicosis.508

Mercuric ions form covalent bonds with sulfhydryl groups and form mercaptides.507 The kidney is the primary target organ, but mercury also localizes in the GI mucosa. Mercury is excreted by the kidneys, partly by exfoliation of renal tubular epithelium.508 Metallothionein, a metal-binding protein, is synthesized within 48 hours of exposure to mercury. Metallothionein initially may protect the kidney by sequestering mercury; however, as the sequestered mercury is slowly released from the tubular epithelium, renal damage continues.505,508

Acute signs of mercury toxicosis are caused by the corrosive effect of mercury on mucous membranes. Ulceration of the mouth, esophagus, and rest of the GI tract may be followed by diarrhea and anorexia. If the animal survives, acute toxic nephrosis occurs. Anorexia, gastroenteritis, weight loss, nephritis, and alopecia have been reported in animals exposed to chronic, low doses of mercury.507

Clinical pathology testing may reveal elevated creatinine and blood urea nitrogen (BUN), as well as proteinuria, glucosuria, and isosthenuria, depending on the acuteness of the disease. Necropsy findings include an ulcerated and edematous GI tract, with possible intraluminal hemorrhage. The kidneys may be pale and swollen.505,508

Diagnosis of mercury toxicosis is made from history, clinical presentation, and mercury levels within the animal. Mercury concentrations can be measured in liver, kidney, brain, whole blood, and urine. Tissue samples and urine may be frozen after collection.

Chelation therapy can be done using BAL or sodium thiosulfate as for arsenic toxicosis. Remaining topical mercurials should be washed from the animal. Supportive care is necessary for the gastroenteritis and kidney failure. Oliguria indicates a poor prognosis.505,508

MOLYBDENUM

Excess dietary molybdenum (copper deficiency) is discussed in Chapter 32.

SELENIUM

See Toxic Plants.

SODIUM

Sodium (salt) toxicosis is discussed in Chapter 35.

SULFATE

Excess dietary sulfate is discussed with copper deficiency in Chapter 32 and with polioencephalomalacia in Chapter 35.

ZINC

Excess dietary zinc is discussed with osteochondrosis in Chapter 38.

TOXICOLOGY OF ORGANIC COMPOUNDS

KONNIE H. PLUMLEE

INSECTICIDES

Anticholinesterase Insecticides

The anticholinesterase insecticides include the organophosphates and the carbamates. Many organophosphate insecticides are used for insect control in crops. Several organophosphates also have been approved for use as insecticides or anthelmintics in domestic animals. Carbamate insecticides are used primarily for crops and other plants. Livestock are usually exposed to these insecticides as a result of drift, accidental ingestion, or improper treatment by owners.509

The mechanism of action of these insecticides is to bind with and inhibit acetylcholinesterase (AChE). As a result, acetylcholine (ACh) accumulates at nerve junctions, and repetitive firing of parasympathetic nerves occurs. Because carbamates bind with ACh on a reversible basis, inhibition of AChE is temporary.509,510 Therefore, carbamates have a shorter duration of action than organophosphate insecticides, which bind irreversibly with AChE.509

Clinical signs are predominantly described by the acronym SLUD: salivation, lacrimation, urination, and defecation. In addition, the animals may have miosis, diarrhea, muscle tremors, seizures, dyspnea, or bloating.509 Death can occur within minutes, depending on the toxicity of the specific compound and the amount of toxicant ingested.511 At postmortem examination, lesions may be minor or absent, depending on the acuteness of death. Pulmonary edema is a frequent finding, because death usually results from increased pulmonary secretions, bronchial constriction, and respiratory paralysis.510

Measuring the cholinesterase activity in brain, retina, or whole blood can be used to make a presumptive diagnosis of toxicosis.512-514 Some laboratories use serum rather than whole blood, depending on the testing method used; therefore it is best to check with the laboratory before submission. Whole blood can reliably be used for cholinesterase evaluation for up to 1 week after collection if the sample is refrigerated.515 Half the brain should be submitted for homogenization because cholinesterase activity varies among regions of the brain.512 Cholinesterase activity in whole blood or brain that is less than 50% of normal for the species being tested indicates excessive exposure to a cholinesterase inhibitor. Cholinesterase activities less than 25% of normal indicates toxicosis, as long as appropriate clinical signs are present.513

An insecticide screen should be performed on GI contents or the liver to identify the insecticide chemical that is involved. These samples should be stored in glass or metal, such as foil, and frozen as soon as possible after collection. If the sample is stored in plastic, the insecticide may leech from the sample into the plastic.

Because carbamates rapidly dissociate from AChE, a diagnosis is often difficult to make based on cholinesterase activity in brain or blood. The activities can be normal or near normal even though the animal has toxicosis.509,510 Because carbamates hydrolyze rapidly, parent compounds may be difficult to detect in ingesta and especially in tissue.509,510 Therefore, when carbamate toxicosis is suspected, multiple samples should be collected and tested as soon as possible.

  Page 1713 

Treatment for organophosphate toxicosis includes supportive care, activated charcoal, atropine, and oximes. Activated charcoal (1 to 2 lb [0.45 to 0.90 kg] PO for 500-kg animal) should be given even if the route of exposure is not oral, because these insecticides can act systemically even when given topically. If the route of exposure is dermal, the animal should be washed with soap and water to decrease absorption through the skin.513 Atropine competitively inhibits ACh at muscarinic nerve receptors. Therefore, muscarinic effects, but not nicotinic effects, are reduced. Atropine should be given at a dose of 0.10 to 0.25 mg/kg. One fourth of the initial dose is given by IV injection and the rest SC or IM. The dose can be repeated if the clinical signs reappear.513 GI motility should be monitored carefully because atropine can cause ileus, especially in horses. Oximes such as 2-pyridine aldoxime methiodide (2-PAM) can be used as a treatment to release the bond between AChE and an organophosphate. However, oximes are of little benefit after the AChE-organophosphate bond undergoes “aging,” meaning that the bond cannot be broken. The amount and time of “aging” that occurs vary among the different organophosphate compounds.513,516 Recommended doses for 2-PAM are 20 mg/kg IV twice daily517 or 10 to 15 mg/kg SC.518

Treatment of carbamate toxicosis is restricted to atropine, activated charcoal, and supportive care. Oximes such as 2-PAM are contraindicated with carbamate toxicosis because the carbamates bind reversibly, and thus oximes are not needed to release the carbamate from the AChE.509

Most anticholinesterase compounds are rapidly metabolized and excreted.519 Therefore these insecticides do not persist in tissues, and tissue levels are usually low. Animals that survive the toxicosis, however, should be analyzed for residues before consumption.520 Animals that have died from anticholinesterase insecticide toxicosis have been prohibited from being rendered.519

Chlorpyrifos is a chlorinated organophosphorus insecticide used for lice and fly control in cattle. It is applied as a pour-on caudal to the shoulders. This product causes a delayed toxicosis that occurs primarily in bulls and some exotic breeds of cattle. Toxicosis is associated with the testosterone levels in the animal.521 Larger and older bulls are affected more severely. The manufacturer recommends not treating bulls over 8 months old with chlorpyrifos.517 Clinical signs do not occur until at least 2 to 7 days after exposure and include depression, weakness, muscle fasciculations, anorexia, rumen stasis, and rumen distention.517,518,521 The clinical signs typically seen with acute organophosphate toxicosis are usually not present with chlorpyrifos toxicosis.

Animals with chlorpyrifos toxicosis will have depressed cholinesterase activities in brain and blood. After topical application, chlorpyrifos will undergo systemic distribution and may be detected in blood, rumen contents, fat, and hair for weeks. Animals with chlorpyrifos toxicosis should be washed with detergent and water as soon as possible.517,518,521 Oral treatment with activated charcoal reduces the signs of toxicosis by adsorption of the chlorpyrifos. The manufacturer recommends atropine as an antidote.517 Because most cases involve ruminal stasis and few muscarinic signs, however, atropine may be contraindicated. 517,518 Severity of the rumen stasis often requires removal of rumen contents by a large-bore stomach tube or rumenotomy.518 Pralidoxime (2-PAM) has decreased clinical signs in some animals when given within 4 days of insecticide exposure.517,518

Haloxon is an anthelmintic and toxicosis was reported in the 1970s in sheep that had a familial absence of a plasma A-esterase.522 This enzyme rapidly hydrolyzes haloxon.522 Clinical signs occur 5 to 90 days after exposure and predominantly include hindlimb ataxia or paresis.523 Gross lesions are not evident on necropsy. Microscopically, the white matter of the spinal cord was vacuolated, with swollen axons and increased glial cells.523 Cholinesterase activity can be decreased.522

Haloxon also has been associated with bilateral laryngeal paralysis in Arabian and part-Arabian foals.524 The foals were 23 to 35 days old and had been treated with haloxon every 2 weeks since 2 days of age. The clinical signs were noticeable only when the foals exercised or were stressed. Rhinolaryngoscopy revealed no abductor movement of the arytenoid cartilages.524 Active wallerian degeneration and loss of nerve fibers were seen in the recurrent laryngeal nerves.524 Tracheotomy relieved the dyspnea, and in foals that survived, function of the right arytenoid cartilage returned before the left.524

Organochlorine Insecticides

Chlorinated hydrocarbons are used to control insects and other pests. Examples include DDT, aldrin, heptachlor, and lindane. Although use of several of these products has been banned or restricted, toxicosis in domestic animals still occurs, either from improper use or from exposure to discarded products.525

The mechanism of actions of organochlorines is not completely understood and seems to vary among compounds. Interference with the kinetics of nerve sodium channels and inhibition of γ-aminobutyric acid (GABA) binding are two proposed mechanisms.

Initial clinical signs may include apprehension, hypersensitivity, and a belligerent attitude. Fasciculations of the face and cervical muscles are followed by spasms of the eyelids, forequarters, and finally hindquarters.525 Ataxia, hypersalivation, and diarrhea also have been reported.526 Intermittent convulsions are the major manifestation in most cases.520 Significant gross lesions usually are not found at necropsy.527

Organochlorines accumulate in fatty tissues. Therefore, diagnostic testing should be performed on samples such as brain, fat, milk, and liver.525 The compounds also may be found in whole blood and GI contents. Collected samples should be put in glass or metal containers525 and frozen, with the exception of blood, which should be refrigerated. As with other insecticides, plastic containers should be avoided if possible.

Treatment is mostly symptomatic because no antidote for organochlorine toxicosis exists.525 The animal should be washed with water and detergent if the exposure was dermal.525 Activated charcoal is beneficial if given immediately after oral exposure.525,526 Convulsing animals require treatment with sedatives and muscle relaxants.

Organochlorine residues persist in fat and are excreted in the milk of lactating animals.520 Some of these compounds cross the placental barrier and concentrate in fetal fat.528 Several strategies have been tried to reduce residues as quickly as possible to minimize economic loss. Treatment with compounds such as phenobarbital and butylated hydroxyanisole (BHA) to promote metabolism, cholestyramine to increase excretion, or thyroprotein to increase elimination has not been very successful in livestock.528

At this time, the only successful method of reducing organochlorine residues in livestock is by fat mobilization and removal. One method of accomplishing fat mobilization is by feeding a calorie-restricted diet.526 As the animal loses weight, the fat and organochlorine are mobilized and removed from the body. After removal of the fat, the animal should be fed a high-calorie diet that is free of organochlorine compounds to regain its body fat and decrease the whole-body concentration of the compound. As the body fat decreases, concentration of organochlorine may increase in other tissues as the chemical is mobilized.525 If the diet is too restrictive, the organochlorine mobilization may occur too rapidly, and the animal may develop acute toxicosis.526 The fastest method of reducing organochlorine residues is through milk secretion.528 Residue elimination is fastest during early lactation. Unfortunately, residue reduction may not be economically feasible because of extra costs of feed, labor, and loss of saleable milk.

  Page 1714 

HERBICIDES

Arsenics

See Metals and Other Inorganic Compounds.

Paraquat

Paraquat is in the class of bipyridyl herbicides, which act as dessicants by altering enzyme systems and reducing photosynthesis.529 Most concerns about this product occur when it is sprayed on foliage that will later be used for animal consumption or when it accidentally drifts from a sprayed field onto animals or their pasture. Paraquat quickly becomes irreversibly inactivated once it comes into contact with soil.529,530 It is a concern only when the animal ingests a concentrated form of the herbicide or grazes a field that is still wet from paraquat application.

Paraquat is concentrated against a gradient within type I alveolar pneumocytes. The herbicide accepts electrons to form free radicals.529,530 This reaction is catalyzed by nicotinamide adenine dinucleotide phosphate (NADP), reduced from cytochrome P450 reductase (NADPH).529 The result is destruction of cell membranes and subsequent cell death.

Acute signs generally occur 1 to 3 days after ingestion and involve anorexia, depression, and diarrhea. Several days later the animal develops respiratory distress, dyspnea, and pneumomediastinum. Death may be delayed until several weeks after ingestion. Gross and histopathologic lesions primarily consist of progressive lung fibrosis, but may include renal and liver damage.529,530

Treatment should include fuller’s earth or bentonite given orally as soon as possible after exposure and certainly within 24 hours of ingestion.529,530 These products inactivate the paraquat on contact and thus are more effective than activated charcoal. Oxygen therapy reportedly worsens the lung damage.531 Regardless of therapy, the prognosis is poor because of the progressive fibrosis of the lungs.

Chlorophenoxy Acids

Chlorophenoxy acid herbicides includes the compounds 2,4-D; 2,4,5-T; and silvex. These herbicides are plant growth regulators, so the mechanism of action is to alter the metabolism of the plant. Therefore they are relatively nontoxic to mammals unless ingested in a concentrated form.529,530 Indirect toxicosis may occur, however, because of the altered metabolism of the sprayed plants. Toxic plants that are normally untouched by grazing animals may become more palatable and result in plant toxicosis after application of some herbicides. In addition, the altered plant metabolism may cause some plants to accumulate higher levels of nitrate or cyanide or increase the level of their inherent toxins.530 Animals should be removed from treated pastures for at least 7 days after application to reduce the occurrence of plant toxicosis.530

Clinical signs of chlorophenoxy acid toxicosis include depression, anorexia, abdominal pain, and diarrhea.529 Weakness is especially profound in the hindlimbs. Gross findings include epicardial hemorrhage and hydropericardium. The liver is swollen and friable. The kidneys may be congested.532

Clorophenoxy acid herbicides are quickly absorbed from the GI tract, but dermal absorption is minimal.533 Treatment consists of activated charcoal and supportive care. The major route of elimination is the urine.533 Neither parent compounds nor metabolites have been found in the milk from orally dosed cattle.533

Triazines

Triazine herbicides inhibit photosynthesis by blocking electron transport.534 Products in this group of herbicides include atrazine, simazine, and propazine. Livestock should be held from pastures treated with simazine for 30 days and off hay for 60 days.530

Sheep have been reported to exhibit signs of generalized muscle tremors, which progressed to mild tetany and collapse of the hindlegs. Other sheep developed a short, prancing gait.535 Cattle have been reported to develop diarrhea after 12 hours, followed by salivation, ataxia, and stiffness.536 Gross lesions include myocardial degeneration, subcutaneous hemorrhages, and liver congestion. Histopathologic examination demonstrated focal degenerative myocardiopathy and mild nonsuppurative encephalitis.535,536 Rumen papillae are edematous and the reticulum, rumen, and omasum may contain black pigment.536

Triazine herbicides have no antidote, but treatment with activated charcoal once daily for 4 consecutive days after exposure has reportedly decreased lesions and death loss.536

RODENTICIDES AND OTHER PESTICIDES

Anticoagulant Rodenticides

Toxicosis can result from anticoagulant rodenticides when animals ingest baits made with anticoagulants. These products are often incorporated into grains or pellets that are palatable to livestock. Toxicosis also has been reported in horses that have been overdosed with warfarin, which has been used as a treatment for navicular disease and jugular phlebitis.537,538

First-generation compounds such as warfarin were developed originally, but rodents developed a resistance to these over time. The second-generation compounds, such as brodifacoum and bromadiolone, were created for rats that had developed a resistance to the first-generation compounds.539-541 In general, the second-generation compounds are more toxic and have longer half-lives.

All anticoagulant rodenticides function by interfering with vitamin K–epoxide reductase, the enzyme that converts inactive vitamin K to its active form. This interference results in a depletion of active vitamin K and subsequently the vitamin K–dependent clotting factors (II, VII, IX, and X). Factor IX is in the intrinsic coagulation pathway, factor VII is in the extrinsic pathway, and factors II and X are in the common pathway. Therefore, all three pathways are affected.539,540

Clinical signs may not be noticed until 3 to 5 days after ingestion. Affected animals may exhibit melena, epistaxis, hematuria, or excessive bleeding from a wound or injection site.540 Often the hemorrhaging occurs in body cavities, and the animal may show nondescript clinical signs such as depression, weakness, pallor, colic, dyspnea, or fever, depending on the location and extent of hemorrhage.539,540 Sometimes the onset is acute, and the animal dies suddenly from extensive hemorrhage and shock.539 Gross and histopathologic lesions primarily consist of unexplained hemorrhage, which may be generalized or localized.

Warfarin is almost completely absorbed from the GI tract of horses and has a biologic half-life of 13.3 hours, similar to that in other species.538 Warfarin is mostly bound to plasma protein, and only the unbound drug is toxic. Therefore, concomitant use of other protein-bound drugs, such as phenylbutazone, can increase the risk of warfarin-induced hemorrhage.537,542

Diagnosis of anticoagulant toxicosis is based on history, clinical signs, clinical pathology, and response to treatment. The degree of anemia resulting from hemorrhage varies with the time since ingestion of the toxicant and severity of disease. A coagulation panel will reveal prolongation of prothrombin time (PT), partial thromboplastin time (PTT), and activated coagulation time (ACT).540,543 The fibrinogen, fibrinogen degradation products, and platelet count are not directly affected by the anticoagulants.

  Page 1715 

Determining which anticoagulant rodenticide was ingested is difficult in the live animal. Levels are often too low in blood to be detected. Postmortem detection is most promising in the liver. Although GI contents can be tested, the chemical is often completely absorbed before clinical signs appear or death occurs.

Vitamin K1 is the treatment for all the anticoagulant rodenticides. The duration of treatment, however, depends on the specific compound because the half-life varies considerably. Warfarin has the shortest half-life of the anticoagulants (13.3 hours in the horse).538 The half-lives of the other compounds have not been determined in the horse, but diphacinone has a half-life of 15 to 20 days in humans. Therefore, warfarin toxicosis may require treatment for only a few days, whereas toxicosis from the other compounds may have to be treated for several weeks. Vitamin K1 treatment for an adult horse has been recommended at a dose of 300 to 500 mg SC every 4 to 6 hours.537 The PT should return to normal within 24 hours. To monitor duration of treatment after the animal has stabilized, vitamin K should be discontinued for 48 hours and the PT retested.539 Intravenous injection of vitamin K has been associated with anaphylactoid reactions, and intramuscular injection can aggravate hemorrhage. Oral treatment is advocated for small animals but may be cost-prohibitive for large animals. Vitamin K3 is less expensive but not as effective and will cause renal disease in horses.

Clotting factor synthesis requires 6 to 12 hours; therefore animals may need to be transfused with whole blood if anemic, or with fresh plasma if the anemia is not severe enough to warrant blood transfusion.539 Once the packed cell volume (PCV) begins to decrease, it can continue to decrease at a rapid pace, so it should be monitored regularly. Activated charcoal can be given orally to decrease absorption of rodenticides.

Strychnine

Strychnine is used for mammals such as rats, gophers, moles, squirrels, and coyotes. Strychnine is sold as a powder, as pellets, or as treated seeds that are usually dyed bright green or red. Strychnine poisoning was reported in four horses that had eaten milo treated with it. Use of strychnine is restricted in some states to professionals or registered individuals; however, toxicosis in domestic animals still occurs.544

Strychnine competitively blocks glycine, the transmitter for inhibitory cells (Renshaw cells) of the spinal cord. This lack of inhibition results in rigidity and tetanic convulsions. Clinical signs include sweating, incoordination, recumbency, and tonic clonic seizures that are inducible by loud sounds, touch, or bright light. Signs appear 10 minutes to 2 hours after ingestion. Gross and histopathologic lesions are limited to those attributable to self-induced trauma.544

Diagnosis of strychnine toxicosis is based on clinical signs and detection of the toxicant in urine or serum. Urine is a good diagnostic sample because strychnine is absorbed from the GI tract and excreted in the urine. Stomach contents, liver, kidney, and urine should be collected postmortem and analyzed for strychnine.544

A lethal dose of strychnine can be eliminated in 24 to 48 hours. Because strychnine is an alkaloid, it becomes ionized in the acid of the stomach and is not absorbed until it reaches the intestine. Activated charcoal is a good treatment to prevent further absorption of the toxicant. Seizures should be treated with anticonvulsants and rigidity with muscle relaxants. Animals should be protected from excessive light and sound to reduce the incidence of convulsions.

Zinc Phosphide

Zinc phosphide is used to control mice, ground squirrels, rats, and moles. Zinc phosphide is a dark-gray powder that is mixed with grains such as bran, wheat, and oats. Therefore it is palatable to most livestock. The lethal dose for most animals is 20 to 40 mg/kg of body weight.545

Zinc phosphide produces phosphine gas under acidic conditions. Therefore, toxicosis occurs when the rodenticide comes into contact with an acidic stomach. Gastric acidity is an important factor in phosphine gas production. Ruminants should be more resistant because the rumen has a higher pH than a monogastric stomach. Studies show a higher survival rate in dogs that are dosed with zinc phosphide on an empty stomach rather than dosed after a meal when the stomach has a lower pH.545

Zinc phosphide affects the CNS, and clinical signs are similar to those caused by other toxicants, such as strychnine or the anticholinesterase insecticides.545 Gross and histopathologic lesions are not specific, although the GI and pulmonary systems may be red and irritated. Thus, diagnosis is difficult without chemical analysis.

Diagnosis of zinc phosphide toxicosis is based on detecting phosphine gas in the stomach contents. Because the gas dissipates rapidly in air, the collected sample of stomach contents should be placed in an airtight container and frozen immediately.545

Treatment is mostly supportive care following activated charcoal therapy. Food should be withheld until the zinc phosphide has been emptied from the stomach. It may be helpful to add an antacid to the activated charcoal therapy to reduce the amount of phosphine gas produced.

Metaldehyde

Metaldehyde is a tasteless ingredient used in slug and snail baits and as a solid fuel for some camp stoves.546 Baits may be liquid or dry, but most are dry pellets of metaldehyde mixed with soybeans, rice, oats, sorghum, or apples.546,547 Some baits also contain other toxicants, such as arsenate or insecticides, which can cause concurrent disease. Toxicosis occurs mostly in coastal and low-lying areas where there is a high incidence of snails or slugs.547

The mechanism of metaldehyde toxicosis is unknown.546 It may increase excitatory neurotransmitters or decrease inhibitory neurotransmitters.546,547

Clinical signs can occur immediately or can be delayed for up to 3 hours after ingestion. Affected animals will have convulsions, which may be continuous or intermittent. The animal may have muscle tremors and anxiety and may be hyperesthetic between convulsions. The seizures are not necessarily evoked by external stimuli. Elevated body temperature, up to 43° C (110° F), is a common finding and is probably caused by the excessive muscle activity. Other findings include tachycardia, defective vision, hyperpnea, hypersalivation, ataxia, cyanosis, acidosis, diarrhea, and dehydration. Death usually results from respiratory failure and occurs 4 to 24 hours after ingestion. Gross and histopathologic findings include petechiae and ecchymoses of various organs and subcutaneous edema.548-550

A diagnosis is based on testing for metaldehyde in GI contents or serum.551 Milk samples taken within 24 hours from two affected cows tested negative for metaldehyde.552 No antidote is available.552,553 Administration of oral activated charcoal may prevent further absorption from the GI tract. If the animal is convulsing, tranquilizers should be used to control seizures. Tranquilizers should be allowed to wear off periodically and the convulsive condition reevaluated. Horses have been treated successfully with tranquilizers and mineral oil.554

  Page 1716 

INDUSTRIAL TOXICANTS

Petroleum

Animals can become exposed to petroleum hydrocarbons during crude oil spills, pipeline breaks, or careless disposal of automobile or tractor oil. Most cases of petroleum toxicosis occur when the animal’s water supply is contaminated.555

Clinical signs vary but usually include anorexia, depression, GI stasis, bloat, and diarrhea or constipation. Oil may be seen in the feces within days after ingestion. The most common cause of death is aspiration pneumonia after regurgitation of the hydrocarbons. The petroleum product may be seen around the muzzle of the animal. Oil may be found in the GI tract or the lungs at postmortem examination.

Diagnosis of petroleum toxicosis can be made based on history, clinical findings, and detection of hydrocarbons in tissues. The product may be visible on the animal as well as in the GI contents and lungs. A quick method of checking for oil is to place the GI contents or lung in warm water; the oil should float to the top of the water. Samples also can be checked under a black light because many petroleum products will fluoresce yellow or yellow-green. Liver, kidney, lung, and GI contents can be analyzed for hydrocarbons and matched with available source material.555 Collecting suspect source material is especially important because these toxicoses may become legal cases.

The primary aim of treatment is to prevent aspiration pneumonia, which is best achieved by performing a rumenotomy and removing the contaminated ingesta. If surgery is not feasible, activated charcoal should be administered. The charcoal should be followed with a cathartic to enhance removal of the oil. Supportive care is needed for the GI stasis and diarrhea or constipation. Vegetable oil (500 to 1000 mL) may increase the viscosity of the ingested petroleum within the rumen and reduce the occurrence of aspiration pneumonia. The prognosis is generally good unless the animal aspirates or severely bloats.555 Some petroleum products may contain heavy metals such as lead, and a concurrent metal toxicosis can occur.

Ethylene Glycol

The most common cause of ethylene glycol toxicosis is ingestion of antifreeze. Although cats and dogs are the species most often affected because of their proximity to available sources, ethylene glycol toxicosis has occurred in cattle and goats.556 Some new brands of antifreeze contain propylene glycol rather than ethylene glycol. Although much less toxic than ethylene glycol, propylene glycol can still cause toxicosis.

Ethylene glycol is metabolized in the liver to several metabolic intermediates, especially glycolic acid. This reaction is catalyzed by alcohol dehydrogenase. Glycolic acid results in metabolic acidosis and hyperosmolality in the animal. This metabolite can be excreted or further metabolized into oxalic acid, which combines with ionic calcium and causes hypocalcemia and calcium oxalate formation, especially in kidneys. Ruminants may be more resistant to ethylene glycol toxicosis than monogastrics because the rumen microorganisms can metabolize some ethylene glycol before it is absorbed.556,557

Toxicosis results in an initial inebriation, followed by metabolic acidosis and renal damage.557 Clinical signs in ruminants include ataxia, depression, hypersalivation, and absence of a menace response. These signs progressed to recumbency and clonic-tonic seizures in a pygmy goat.556 Clinical pathology reveals azotemia, metabolic acidosis, and hyperosmolality. Postmortem findings include swollen kidneys and pulmonary edema. Dilated capsular spaces and birefringent crystals are found in renal tubules on histologic examination. These crystals may be arranged in sheaves or rosettes and are typical of oxalate crystals.556

Rumen or stomach contents can be analyzed for ethylene glycol. It is absorbed within 48 hours after ingestion by monogastrics; however, ethylene glycol has been detected in the rumen contents of a goat 4 days after clinical signs began.5 Urine, serum, and ocular fluid can also be analyzed for glycolic acid.

The classic treatment for ethylene glycol toxicosis is 20% ethanol given at 5 ml/kg at 4- to 8-hour intervals. The ethanol binds with the alcohol dehydrogenase so that it is not available to convert ethylene glycol to glycolic acid. This treatment is effective, however, only if initiated within a few hours of ingestion. The effectiveness of ethanol in treating livestock has not been addressed in the literature. Because the ethylene glycol appears to remain in the rumen for several days, treatment with activated charcoal may be beneficial even after the appearance of clinical signs.556

Chlorinated Naphthalene

Chlorinated naphthalene has been used in wood preservatives, asphalt roofing, insulating waxes, sealing compounds, and in condensers. Most toxicoses in cattle, however, have been from ingesting lubricating grease used on farm machinery or feed-pelleting machines. Chlorinated naphthalene is no longer used in lubricants, but toxicosis still occurs when animals have access to dumps and salvage yards. The toxic forms are tetra-, penta-, hexa-, hepta-, and octachloronaphthalenes, with the hexa and hepta forms being the most toxic.558

Chlorinated naphthalene interferes with the conversion of carotene to vitamin A. Serum vitamin A levels decrease significantly a few days after exposure and remain decreased for at least 4 weeks.558

Initial clinical signs include weight loss, anorexia, and depression. Excessive salivation and lacrimation occur because of the formation of papular stomatitis and keratinization of the meibomian glands. Several weeks later, nonpruritic thickening and fissuring of the skin occur. Hyperkeratosis involves the withers, neck, head, trunk, and medial thighs; but usually does not involve the lower legs. Diarrhea may occur late in the disease. Postmortem findings include epithelial hyperplasia or metaplasia of the gallbladder, bile ducts, salivary glands, pancreas, and genital tract.558

Diagnosis is based on clinical signs and a low vitamin A level. Suspected source material can be analyzed for chlorinated naphthalene. Treatment with vitamin A may minimize some clinical signs, but treatment of this toxicosis is usually unsuccessful, especially after the appearance of skin lesions.558

Pentachlorophenol

Pentachlorophenol (PCP) is used primarily as a wood preservative. Residues have been found in cattle exposed to wood troughs, silos, barns, and fences treated with PCP. Horses have developed toxicosis when bedded on wood shavings that contain PCP.559-561

The primary mechanism of action is uncoupling of oxidative phosphorylation.559,561 PCP is quickly absorbed from the GI tract and excreted in the urine. It is stored mainly in the liver and kidney and acts as a mild hepatotoxin.510 The half-life in cattle is 1.5 days.559

Acute clinical signs in cattle include weight loss, depression, anorexia, intense thirst, and decreased milk production.559,560 Chronic signs in cattle are dyspnea, hyperkeratosis, liver damage, and increased abortion rate.559 Horses have anorexia, dependent edema, weight loss, and alopecia. The skin has cracks and fissures that exude serum. Clinical pathology reveals hepatic changes, anemia, and thrombocytopenia. Horses also may develop colic or recurrent hoof problems.561

  Page 1717 

Liver, kidney, and serum can be analyzed for PCP. Because of the rapid rate of excretion, serum PCP concentrations may be useful only during acute toxicosis.561 No antidote exists, and treatment is usually unsuccessful. Residues may be a concern, especially from dioxin-related contaminants in the “penta”-treated wood.

Phosphatic Fertilizers

Phosphatic fertilizers selectively promote legume instead of grass growth. Therefore these are popular fertilizers for maintaining subterranean clover pastures. The major components of phosphatic or superphosphate fertilizers are calcium pyrophosphate, calcium orthophosphate, calcium sulfate, and sodium fluoride. Toxicosis is believed to be a result of the phosphate and fluoride. It usually occurs after a short pasture has been top-dressed recently.562 The fertilizer is not usually palatable, except to ravenous animals.

Sheep have developed ataxia, bruxism, depression, and diarrhea. Animals will be hypocalcemic, probably because of renal failure. Hyperphosphatemia does not occur until oliguria develops and the animal is near death. Gross lesions include hyperemic GI mucosa, pulmonary edema, and bloody intestinal contents. Histopathologic examination reveals acute proximal renal tubular necrosis.562

Diagnosis is based on history, clinical signs, and lesions. Treatment is limited to supportive care, which is generally successful if the disease is diagnosed early.562

Boron Fertilizer

Sodium borate is mildly toxic, and clinical signs may not be seen in ruminants until they consume a near-lethal dose. Toxicosis usually occurs only if animals eat concentrated fertilizer.563 The mechanism of action is unknown, but boron may have a stimulatory effect on serotonergic and dopaminergic neurons.564

Reported clinical signs in cattle include weakness, depression, muscle fasciculations, seizures, and a spastic gait. Most animals develop diarrhea and become dehydrated. Gross and microscopic lesions are not seen.563 Goats that were given a sublethal dose of boron developed anorexia and depression. Seizure-like activity consisted mostly of ear flicking and chomping motions; however, tremors, stargazing, head jerking, and extensor rigidity also were noted.564

Liver, kidneys, and rumen contents can be analyzed for boron content.563 Treatment is limited to supportive care.

THERAPEUTIC AGENTS

Vitamin K3

Vitamin K3 is the synthetic vitamin menadione sodium bisulfite. It has been used as a treatment for anticoagulant rodenticide toxicosis, sweet clover (dicumarol) toxicosis, and exercise-induced pulmonary hemorrhage. Its popularity stemmed from being much less expensive than vitamin K1. Studies demonstrate, however, that vitamin K3 is not an effective treatment for sweet clover disease in cattle.565 Furthermore, case reports indicate that vitamin K3 is toxic to horses, even when used at the manufacturer’s recommended dose.566-568

The mechanism for the toxicosis is unknown. Within 4 to 48 hours of administration of vitamin K3, horses become depressed, anorexic, and weak. They may develop muscle stiffness, laminitis, or colic.566 The horses develop renal failure as evidenced by increased BUN and creatinine concentrations. Proteinuria, hematuria, and low specific gravity are found on urinalysis. Serum electrolyte levels are consistent with renal tubular disease: hyponatremia, hypochloremia, and hyperkalemia. Some patients develop hypercalcemia.568 Grossly, the kidneys are enlarged.566 Microscopically, the kidneys have nephrosis with tubular dilation, epithelial degeneration, and necrosis.568 Diagnosis is based on history, clinical signs, and lesions. Treatment should include diuresis and maintenance of serum electrolyte concentrations.567

Propylene Glycol

Propylene glycol is used as a vehicle for drugs with poor water solubility, for treatment of bovine ketosis, and in some new antifreeze products.569 Toxicosis has been reported when cows are overdosed or when horses are accidentally dosed with propylene glycol.569,570

The median toxic dose of propylene glycol in cattle is 2.6 g/kg body weight.570 Ataxia develops in 2 to 4 hours and resolves by 24 hours after dosing. The cattle also become depressed and temporarily recumbent. Serum and cerebrospinal fluid osmolality increase.570

A horse mistakenly given propylene glycol rather than mineral oil developed ataxia and depression in 10 to 15 minutes. The horse also developed pain, excessive salivation, and sweating, but these signs disappeared within 5 minutes. The animal developed rapid, shallow breathing and cyanosis and died of respiratory distress the next day.569 Gross lesions were not seen. Histopathologic findings included myocardial perivascular edema, pulmonary edema, scattered hepatocyte necrosis, and peracute renal infarcts.569

Serum and urine can be analyzed to determine the presence of propylene glycol.569,570 Propylene glycol causes lactic acidosis in humans, which is treated with sodium bicarbonate. This treatment was unsuccessfully used in the horse described here569; if acidosis occurs, however, sodium bicarbonate may be beneficial if the disease is treated early.

Isopropyl Alcohol

Isopropyl alcohol is used as a topical antibacterial agent. When ingested, it is quickly absorbed from the GI tract and metabolized into acetone by the liver through alcohol dehydrogenase. The majority of the acetone is excreted by the kidney and to a lesser extent by the lungs.571

Toxicosis in a horse was reported when 2 L of alcohol was mistaken for mineral oil and administered by nasogastric tube. Initially the horse was depressed and reluctant to move before collapsing and becoming semicomatose. A menace response was not detected, and pupillary light reflexes were slow. Treatment consisted of repeated gastric lavage with 2-L aliquots of warm water, followed by activated charcoal and intravenous fluids. Activated charcoal was repeated the following day, and the horse recovered from the incident. However, the odor of acetone could be detected on the horse’s breath for about 4 days, and acetone was detectable in the horse’s serum during this time.571

Phenothiazine

This anthelmintic combined with piperazine and carbon disulfide has been used for horses in the past.572,573 Phenothiazine currently is being manufactured for horses in combination with piperazine and trichlorfon. Phenothiazine also has been used in mineral blocks and protein supplements for ruminants.574 Phenothiazine is toxic to both horses and ruminants but causes different diseases.

  Page 1718 

RUMINANTS

Phenothiazine causes primary photosensitization in ruminants. The rumen converts phenothiazine to a phototoxin, phenothiazine sulfoxide. This toxic metabolite can be converted by the liver to a nontoxic metabolite, leukophenothiazine; if the liver is overwhelmed, however, toxicosis can occur. Most cases occur in debilitated or young animals that do not have a fully functional liver.574

Clinical signs begin as erythema and edema combined with varying degrees of pruritus, photophobia, and pain. Vesicles and bullae form and progress to oozing, necrosis, and ulceration of the skin. These lesions are usually confined to areas that have white pigmentation or have little hair and are exposed to sunlight. The tail, ears, teats, feet, or ventral surface of the tongue may slough. The skin of black cattle does not usually slough, but these animals can develop epiphora, corneal edema, and blindness because the phototoxin is also secreted in tears and aqueous humor.574,575

Diagnosis is based on the history of phenothiazine consumption and the skin lesions. A skin biopsy may be helpful but usually is not necessary because the lesions are uniquely confined to lightly pigmented skin.

No antidote exists for phenothiazine toxicosis, and treatment is limited to supportive care. Antibiotics may be needed to treat secondary bacterial infections of the skin. Antiinflammatory drugs may be indicated for pain. When prescribing drugs for ruminants with photosensitization, those that can compromise the liver should be avoided. Affected animals should be housed and fed in areas out of direct sunlight to prevent further damage to the skin.

HORSES

Phenothiazine acts as an oxidant to produce hemolytic anemia in horses. Heinz bodies (precipitated denatured hemoglobin) damage the red blood cell membrane, which results primarily in intravascular hemolysis. Toxicosis has been noted primarily in horses that are in poor condition before exposure to the phenothiazine.576 Primary clinical signs include anorexia, depression, weakness, icterus, anemia, and hemoglobinuria.572,573 Colic, diarrhea, fever, and dependent edema are less frequently reported clinical signs.576 Clinical pathology reveals anemia and elevated indirect bilirubin levels.576

Diagnosis is based on a history of exposure to phenothiazine and ruling out other causes of hemolytic anemia in the horse. Treatment is basically supportive. A blood transfusion may be necessary if the anemia reaches a critical level.576

FEED ADDITIVES

Urea and Nonprotein Nitrogen

Nonprotein nitrogen (NPN) products are converted by ruminal microorganisms to ammonia, which is used to form amino acids. Therefore, ruminants can use the nitrogen from NPN for part of their diet rather than the more expensive natural proteins. Urea is the best known source of NPN, and it is also the most toxic.577

Toxicosis can result from overexposure or loss of acclimation to NPN. Too much NPN may be fed to animals because of a miscalculation or by contamination. Urea toxicosis has been reported in cattle that drank from a water source that was contaminated with urea fertilizer.578 Ruminants acclimate to NPN in their diets through the ruminal microorganisms. Animals that have been acclimated to a certain level of NPN in their diets and then go without NPN for more than 1 day can develop toxicosis. Ruminants quickly lose their adaptation to NPN, and toxicosis can occur when urea consumption resumes, even if NPN is fed at the same level as previously.

Ammonia that is not used by the ruminal microorganisms is absorbed from the rumen and detoxified by the liver back into urea for excretion. Toxicosis occurs when the microorganisms and the liver are overwhelmed by the level of ammonia.577-579 As the ammonia level increases in the rumen, the ruminal pH increases and creates a shift from charged ammonium ions to uncharged ammonia. The uncharged ammonia is absorbed readily across the rumen wall and increases the ammonia level in the blood.577

Clinical signs begin 30 minutes to 4 hours after ingestion of NPN and include weakness, dyspnea, salivation, bruxism, bloat, and convulsions. Because death can occur in a few hours, some animals may be found dead with no clinical signs observed. The rumen pH will be greater than 8.0 with NPN toxicosis. Rumen pH can decrease with time after death, so the pH should be determined on a recently dead animal.577,579

Diagnosis can be made by analyzing rumen contents, whole blood, or an eye for ammonia. Because ammonia is volatile, the samples should be frozen immediately after collection. Feed material and rumen contents can be analyzed for urea. Urea concentration in the rumen may not be indicative of the amount that was eaten because the microorganisms continue to convert urea to ammonia even after the animal has died. Therefore, a diagnosis should not be based solely on the urea level in the rumen, but in conjunction with the ammonia levels in the animal.

Treatment focuses on decreasing the amount of ammonia absorbed from the rumen. Adult cattle should be given 20 to 30 L of cold water orally to reduce the microorganisms’ ability to convert urea to ammonia. These animals also should receive 2 to 6 L of 5% acetic acid (vinegar) to decrease the pH in the rumen so that ammonia absorption is minimized.577 Rumenotomies can be used to remove the excess NPN source.

To prevent toxicosis, NPN in feed should not exceed 40% of the total nitrogen requirement, and the animals should be acclimated slowly to NPN. Urea should not be fed at a concentration higher than 3% of the grain ration or 1% of the total ration.577

Ammoniated Feed

Molasses, hay, and silage can be treated with aqueous or anhydrous ammonia to increase the dietary quality for ruminants. Under certain conditions, these ammoniated feeds can cause “bovine bonkers,” a disease characterized by hyperexcitability. Treatment conditions that predispose to the occurrence of the toxicosis are ammoniating feedstuffs with more than 20% moisture, treating feedstuffs that contain ample soluble sugars, overapplying the ammonia, or treating the forage during high ambient temperatures. High-quality hays, such as wheat, Sudan, alfalfa, orchardgrass, Bermuda grass, fescue, and sorghum, contain high levels of soluble sugars and are more often incriminated in toxicosis than are low-quality hays such as corn stalks, corn silage, and most small grain straws.577

The mechanism of bovine bonkers is not completely understood. Pyrazines and imidazoles are the primary byproducts formed during ammoniation. The two major imidazoles that are formed, 2-methylimidazole (2-me-I) and 4-methylimidazole (4-me-I), cause convulsions in mice, with 4-me-I being more potent. Administration of 4-me-I has experimentally reproduced the disease in a nursing cow without affecting the calf.577 Nursing calves in field cases have developed the disease,580,581 even though the dams did not. However, when orally dosed with 4-me-I, cows developed the disease but the calves did not, despite the imidazole being detected in the milk and colostrum.582 Therefore, more work is needed regarding the mechanism of this disease.

  Page 1719 

The most striking clinical sign is hyperexcitability, which can occur spontaneously or can be induced by excitement. Animals will suddenly stampede and run in circles or in a straight line while they collide with other animals or with buildings and fences. Other clinical signs include ear twitching, mydriasis, trembling, salivation, increased urination, increased defecation, and bellowing. Gross lesions are not significant other than bruising and broken bones that are self-inflicted.577,580,583

Diagnosis is based on clinical signs and a history of feeding ammoniated feed. Treatment is limited to sedation of animals.581 Thiamine also has been used as a treatment, with variable results.577 It is often not possible to approach an affected animal to treat it without endangering the handler. Most animals recover spontaneously once the ammoniated feed source is removed.

Prevention of ammoniated toxicosis focuses on properly treating the feed. Only poor-quality roughages with low levels of soluble sugars should be ammoniated. The amount of ammonia used should not exceed 3% of the dry weight of the forage. Ammoniate only during cool weather so that the processing temperature of the forage is less than 70° C (158° F).577,580

Ionophores

Ionophore antibiotics are used as coccidiostats and feed additives for poultry and cattle. Ionophores alter the rumen so that a higher level of propionic acid is produced for improved feed efficiency. They also are used to prevent rumen acidosis and emphysema in cattle.584-588 Toxicosis can result from calculation or mixing errors or from use in inappropriate species. Cattle and sheep have been poisoned by ingesting litter from poultry that had been treated with an ionophore.589

Ionophores form lipid-soluble complexes with cations to facilitate transport of the cations across lipid membranes. The monovalent ionophores are monensin, salinomycin, and narasin. Monensin preferentially complexes with sodium ions, whereas salinomycin and narasin preferentially bind with potassium ions. Lasalocid is a divalent ionophore that binds with divalent cations, such as calcium and magnesium ions.585,587

Horses are the most sensitive species to the ionophore antibiotics.586,590 The recommended dose of monensin for cattle can be lethal to a horse. Acute clinical signs in horses include colic, anorexia, weakness, and ataxia.586 Creatinine phosphokinase (CPK) significantly increases within 24 hours; AST and serum ALP activities are increased to a lesser degree.586 Unconjugated serum bilirubin will be slightly but consistently increased. Erythrocyte fragility increases.590 Serum enzymes can be used diagnostically but are poor prognostic indicators.591 If the animal dies peracutely, significant gross lesions may not be present. Acutely affected horses develop degeneration and necrosis of the cardiac muscle and to a lesser degree the skeletal muscles.587

If horses survive the acute episode of toxicosis, they often experience delayed toxicosis because of permanent damage to the heart. These animals have marked cardiac myopathy and fibrosis.588 The most noticeable clinical sign is exercise intolerance, and affected animals may collapse and die during exercise, making them hazardous to use as riding horses.

Whereas the heart is the most affected organ in horses, ionophores damage the cardiac and skeletal muscles more equally in ruminants.587 Clinical signs in feedlot cattle include anorexia, pica, diarrhea, hindlimb ataxia, and dyspnea. These animals have postmortem lesions of hydrothorax, ascites, and pulmonary edema besides hemorrhages and necrosis of the cardiac and skeletal muscles.592 Signs of weakness, increased respiratory rate, nasal discharge, and reddened noses have been reported in dairy calves. Postmortem examination revealed pulmonary edema accompanied by pleural and peritoneal effusions. The hearts had myocardial necrosis.585 Sheep reportedly develop depression, anorexia, diarrhea, and stiffness. CPK and AST activities are increased. Necrosis of the heart and diaphragm has been reported.584

Diagnosis relies on finding appropriate clinical signs and lesions. Currently, analysis for ionophores in tissue or serum is not reliable; however, the feed source can be analyzed for ionophores.587

Antidotes for ionophores are not available.586,587 Mineral oil or activated charcoal may decrease further absorption of the toxicant. Large volumes of intravenous fluids are needed to treat dehydration and shock.586 Serum electrolyte levels should be monitored frequently and fluid electrolytes adjusted accordingly.

References

1 Nielsen DB, James LF. The economic impacts of livestock poisonings by plants. In: James LF, et al, editors. Poisonous plants: Proceedings of the Third International Symposium. Ames, Iowa: Iowa State University Press; 1992:3.

2 Holchek JL. Do most livestock losses to poisonous plants result from “poor” range management?. J Range Mgmt. 2002;55:270.

3 Torell LA, et al. Perceptions and economic losses from locoweed in northeastern New Mexico. J Range Mgmt. 2000;53:376.

4 Osweiler GD, et al. Clinical and diagnostic veterinary toxicology. Dubuque, Iowa: Kendall/Hunt, 1985;460.

5 Galey FD. Poisonous plant diagnostics in California. In: Colegate SM, Dorling PR, editors. Plant-associated toxins: agricultural, phytochemical, and ecological aspects. Wallingford, UK: CAB International; 1994:101.

6 Ralphs MH. Ecological relationships between poisonous plants and rangeland condition: a review. J Range Mgmt. 2002;55:285.

7 Pfister JA, et al. Risk management to reduce livestock losses from toxic plants. J Range Mgmt. 2002;55:291.

8 Williams MC, James LF. Effects of herbicides on the concentration of poisonous compounds in plants: a review. Am J Vet Res. 1983;44:2420.

9 Osweiler GD, et al. Clinical and diagnostic veterinary toxicology. Dubuque, Iowa: Kendall/Hunt, 1985;455.

10 Galey FD, et al. Diagnosis of oleander poisoning in livestock. J Vet Diagn Invest. 1996;8:358.

11 Holstege DM, Seiber JN, Galey FD. A rapid mulitiresidue screen for alkaloids in plant material and biological samples. J Agric Food Chem. 1995;43:691.

12 Picchioni AL. Efficacy of activated charcoal. Vet Hum Toxicol. 1983;25:452.

13 Ralphs MH, et al. Plant-animal interactions in larkspur poisoning in cattle. J Anim Sci. 1988;66:2334.

14 Merrill LB, Schuster JL. Grazing management practices affect livestock losses from poisonous plants. J Range Mgmt. 1978;31:351.

15 Cordo HA, et al. Biology of Heilipodus ventralis (Coleoptera:Curculionidae), and Argentine weevil for biological control of snakeweeds (Gutierrezia spp.) in the United States. Biol Control. 1999;15:210.

16 Landau SY, et al. Conditioned aversion to minimize Ferula communis intake by orphaned lambs. J Range Mgmt. 1999;52:436.

17 Pfister JA, et al. Conditioning taste aversions to locoweed (Oxytropis sericea) in horses. J Anim Sci. 2002;80:79.

18 Ralphs MH. Lithium residue in milk from doses used to condition taste aversions and effects on nursing calves. Appl Anim Behavioral Sci. 1999;61:285.

19 Ralphs MH. Conditioning sheep to graze duncecap larkspur (Delphinium occidentale). Rangeland Ecol Mgmt. 2005;58:628.

20 Panter KE, et al. Reproductive losses to poisonous plants: influence of management strategies. J Range Mgmt. 2002;55:301.

21 Panter KE, James LF. Natural plant toxicants in milk: a review. J Anim Sci. 1990;68:892.

22 Cheeke PR. Natural toxicants in feeds, forages, and poisonous plants, ed 2, Danville, Ill: Interstate; 1998:76.

23 Kingsbury JM. Poisonous plants of the United States and Canada. Englewood Cliffs, NJ: Prentice-Hall, 1964.

24 Pfister JA, et al. Alkaloids as anti-quality factors in plants on western US rangelands. J Range Mgmt. 2001;54:447.

25 Cheeke PR, Shull LR. Natural toxicants in feeds and poisonous plants. Westport, Conn: AVI, 1985;5.

26 James LF, Hartley WJ, Van Kampen KR. Syndromes of Astragalus poisoning in livestock. J Am Vet Med Assoc. 1981;178:146.

27 Molyneux RJ, James LF. Loco intoxication: indolizidine alkaloids of spotted locoweed (Astragalus lentiginosus). Science. 1982;216:190.

28 Dorling PR, Huxtable CR, Colgate SM. Inhibition of lysosomal alpha mannosidase by swainsonine, and indolizidine alkaloid from Swainsona canescens. Biochem J. 1980;191:649.

29 Tulsiani DRP, Touster O. Swainsonine, a potent mannosidase inhibitor, elevates rat liver and brain lysosomal alpha-D-mannosidase, decreases Golgi alpha-D-mannosidase II, and increases the plasma levels of several acid hydrolases. Arch Biochem Biophys. 1983;224:594.

30 Tulsiani DRP, et al. The similar effects of swainsonine and locoweed on tissue glycosidase and oligosaccharides of the pig indicating that the alkaloid is the principal toxin responsible for the induction of locoism. Arch Biochem Biophys. 1984;232:76.

31 March CD. The locoweed disease. USDA Farmers Bulletin. 380, 1912.

32 Ralphs MH, Graham D, James LF. Social facilitation influences cattle to graze locoweed. J Range Mgmt. 1994;47:123.

33 James LF, et al. Locoweed disease. Clin Toxicol. 1969;2:13.

34 James LF, W, et al. Abortive and teratogenic effects of locoweed on sheep and cattle. Am J Vet Res. 1967;28:1379.

35 Panter KE, et al. The relationship of Oxytropis sericea (green and dry) and Astragalus lentiginosus with high mountain disease in cattle. Vet Hum Toxicol. 1988;30:318.

36 James LF, et al. Relationship between ingestion of the locoweed (Oxytropis sericea) and congestive right-sided heart failure in cattle. Am J Vet Res. 1983;44:254.

37 James LF, Van Kampen KR, Staker GR. Locoweed (Astragalus lentiginosus) poisoning in cattle and horses. J Am Vet Med Assoc. 1969;155:525.

38 McIlwraith CW, James LF. Limb deformities in foals associated with ingestion of locoweed by mares. J Am Vet Med Assoc. 1982;181:255.

39 James LF, Binns W. Blood changes associated with locoweed poisoning. Am J Vet Res. 1967;28:1107.

40 Panter KE, et al. Effects of Locoweed (Oxytropis sericea) on reproduction in cows with a history of locoweed consumption. Vet Hum Toxicol. 1999;41:282.

41 Panter KE, et al. Locoweeds: effects on reproduction in livestock. J Nat Toxins. 1998;8:53.

42 James LF, Van Kampen KR, Johnson AE. Physiopathologic changes in locoweed poisoning of livestock. Am J Vet Res. 1970;31:663.

43 Van Kampen KR, James LF. Pathology of locoweed poisoning in sheep. Vet Pathol. 1969;6:413.

44 Hartley WJ, James LF. Microscopic lesions in fetuses of ewes ingesting locoweed (Astragalus lentiginosus). Am J Vet Res. 1973;34:209.

45 James LF, Van Kampen KR. Acute and residual lesions of locoweed poisoning in cattle and horses. J Am Vet Med Assoc. 1971;158:614.

46 Alroy J, et al. Swainsonine toxicosis mimics lectin histochemistry of mannosidosis. Vet Pathol. 1985;22:311.

47 Stegelmeier BL, et al. Serum swainsonine concentration and alpha-mannosidase activity in cattle and sheep ingesting Oxytropis sericea and Astragalus lentiginosus (locoweeds). Am J Vet Res. 1995;56:149.

48 Ralphs MH, James LF. Locoweed grazing. J Nat Toxins. 1999;8:47.

49 James LF, Hartley WJ. Effects of milk from animals fed locoweed in kittens, calves, and lambs. Am J Vet Res. 1977;38:1263.

50 Stegelmeier BL, et al. The pathogenesis and toxicokinetics of locoweed (Astragalus lentiginosus) in livestock. J Nat Toxins. 1999;8:35.

51 Cheeke PR. Natural toxicants in feeds, forages, and poisonous plants, ed 2, Danville, Ill: Interstate; 1998:383.

52 Cheeke PR. Toxicity and metabolism of pyrrolizidine alkaloids. J Anim Sci. 1988;66:2343.

53 Segall HG, et al. Trans-4-hydroxy-2-hexenal: a reactive metabolite from the macro-cyclic pyrrolizidine alkaloid senecionine. Science. 1985;229:472.

54 Fu PP, et al. Pyrrolizidine alkaloids: genotoxicity, metabolism enzymes, metabolic activation, and mechanisms. Drug Metab Rev. 2004;36:1.

55 Nyska A, et al. The hepatic entothelial carcinogen riddelline induces endothelial apoptosis, mitosis, S phase, and p53 and hepatocytic vascular endothelial growth factor expression after short-term exposure. Tox Appl Pharmacol. 2002;184:153.

56 Huan JY, et al. Species differences in the hepatic microsomal enzyme metabolism of the pyrrolizidine alkaloids. Toxicol Lett. 1998;99:127.

57 Deol HS, Dorling PR, Thomas JB. Experimental copper and heliotrope intoxication in sheep: morphological changes. J Comp Pathol. 1991;105:49.

58 Mendel VE, et al. Pyrrolizidine alkaloid—induced liver disease in horses: an early diagnosis. Am J Vet Res. 1988;49:572.

59 Molyneux RJ, Johnson AE, Stuart CD. Delayed manifestation of Senecio-induced pyrrolizidine alkaloidosis in cattle: case reports. Vet Hum Toxicol. 1988;30:201.

60 Smith BP. Pyrrolizidine alkaloid-induced hepatic disease in a group of calves. Compend Cont Educ. 1982;4:S531.

61 Stegelmeier BL. The clinico-pathologic changes of Cynoglossum officinale, houndstongue intoxication in horses. Fourth International Symposium on Poisonous Plants. 1993. Freemantle, West Australia, 1993 (abstract)

62 Araya O, et al. Serum changes and histological liver lesions due to experimental ingestion of ragwort (Senecio erraticus) in sheep. Vet Hum Toxicol. 1983;25:4.

63 Seawright AA, et al. The identification of hepatotoxic pyrrolizidine alkaloid exposure in horses by the demonstration of sulphur-bound pyrrolic metabolites on their hemoglobin. Vet Hum Toxicol. 1991;33:286.

64 Schoch TK, Gardner DR, Stegelmeier BL. GC/MS/MS detection of pyrrolic metabolites in animals poisoned with the pyrrolizidine alkaloid riddelliine. J Nat Toxins. 2000;9:197.

65 Yan CC, Huxtable RJ. Relationship between glutathione concentrations and metabolism of the pyrrolizidine alkaloid monocrotaline in the isolated, perfused liver. Toxicol Appl Pharmacol. 1995;30:132.

66 Candrian U, et al. Transfer of orally administered [3H] seneciophylline into cow’s milk. J Agric Food Chem. 1991;39:930.

67 Kingsbury JM. Poisonous plants of the United States and Canada. Englewood Cliffs, NJ: Prentice-Hall, 1964;125.

68 Cronin EH, Nielsen DB. The ecology and control of rangeland larkspurs, Bulletin 499. Logan Utah: Utah Agricultural Experiment Station, 1979.

69 Olsen JD, Manners GD. Toxicology of diterpenoid alkaloids in rangeland larkspur (Delphinium spp). In: Cheeke PR, editor. Toxicants of plant origin, vol 1, Alkaloids. Boca Raton, Fla: CRC Press; 1989:291.

70 Majak W, McDonald RE, Binn MH. Isolation and HPLC determination of methyllycaconitine in a species of low larkspur (Delphinium nuttalianum). J Agric Food Chem. 1987;35:800.

71 Manners GD, et al. The toxic evaluation of norditerpenoid alkaloids in 3 tall larkspur (Delphinium) species. Fourth International Symposium on Poisonous Plants. 1993. Freemantle, West Australia, 1993 (abstract)

72 Pfister JA, et al. Larkspur (Delphinium spp.) poisoning in livestock. J Nat Toxins. 1999;8:81.

73 Cheeke PR, Shull LR. Natural toxicants in feeds and poisonous plants. Westport, Conn: AVI, 1985;140.

74 Ralphs MH, et al. Plant-animal interactions in larkspur poisoning in cattle. J Anim Sci. 1988;66:2334.

75 Manners GD, Pfister JA, et al. The phenological variation of diterpenoid alkaloids in tall larkspur (Delphinium barbeyi). In: James LF, et al, editors. Poisonous Plants: Proceedings of the Third International Symposium. Ames, Iowa: Iowa State University Press; 1992:309.

76 Olsen JD. Tall larkspur poisoning in cattle and sheep. J Am Vet Med Assoc. 1978;173:762.

77 Olsen JD, Sisson DV. Description of a scale for rating the clinical response of cattle poisoned by larkspur. Am J Vet Res. 1991;52:488.

78 Galey FD. Poisonous plant diagnostics in California. Fourth International Symposium on Poisonous Plants. 1993. Freemantle, West Australia

79 Gardner DR, Pfister JA. Late season toxic alkaloid concentrations in tall larkspur (Delphinium spp.). J Range Mgmt. 2000;53:329.

80 Ralphs MH, et al. Individual plant control of tall larkspur (Delphinium barbeyi) with tebuthiuron. Weed Technol. 2004;18:248.

81 Olsen JD, Ralphs MH. Feed aversion induced by intraruminal infusion with larkspur extract in cattle. Am J Vet Res. 1986;47:1829.

82 Ralphs MH, Jones WA. Population cycles of the larkspur mirid (Heteroptera: Miridae). J Entomol Sci. 2000;35:483.

83 Bush LP, Crowe MW. Nicotiana alkaloids. In: Cheeke PR, editor. Toxicants of plant origin, vol 1, Alkaloids. Boca Raton, Fla: CRC Press; 1989:87.

84 Panter KE, Keeler RF. Piperidine alkaloids of poison hemlock (Conium maculatum). In: Cheeke PR, editor. Toxicants of plant origin, vol 1, Alkaloids. Boca Raton, Fla: CRC Press; 1989:109.

85 Panter KE, et al. Radio ultrasound observations of the fetotoxic effects in sheep from ingestion of Conium maculatum (poison hemlock). Clin Toxicol. 1988;26:175.

86 Keeler RF. Teratogens in plants. J Anim Sci. 1984;58:1029.

87 Keeler RF, Crowe MW, Lambert EA. Teratogenicity in swine of the tobacco alkaloid anabasine isolated from Nicotiana glauca. Teratology. 1984;30:61.

88 Panter KE, Bunch TD, Keeler RF. Maternal and fetal toxicity of poison hemlock (Conium maculatum) in sheep. Am J Vet Res. 1988;489:281.

89 Keeler RF. Livestock models of human birth defects, reviewed in relation to poisonous plants. J Anim Sci. 1988;66:2414.

90 Baker DC, Keeler RF. Thermopsis montana–induced myopathy in calves. J Am Vet Med Assoc. 1989;194:1269.

91 Cheeke PR, Shull LR. Natural toxicants in feeds and poisonous plants. Westport, Conn: AVI, 1985;127.

92 Casper HH, et al. Lupin bean meal toxicosis in swine. J Vet Diagn Invest. 1991;3:172.

93 Galey FD, Holstege DM, Fisher EG. Toxicosis in dairy cattle exposed to poison hemlock (Conium maculatum) in hay: isolation of Conium alkaloids in plants, hay, and urine. J Vet Diagn Invest. 1992;4:60.

94 Cheeke PR, Shull LR. Natural toxicants in feeds and poisonous plants. Westport, Conn: AVI, 1985;115.

95 Keeler RF, et al. Teratogenicity and toxicity of coniine in cows, ewes, and mares. Cornell Vet. 1980;70:19.

96 Plumlee KH, et al. Nicotiana glauca toxicosis of cattle. J Vet Diagn Invest. 1993;5:498.

97 Panter KE, Keeler RF, Baker DC. Toxicosis in livestock from the hemlocks (Conium and Cicuta spp). J Anim Sci. 1988;66:2407.

98 Panter KE, Keeler RF, Buck WB. Congenital skeletal malformations induced by maternal ingestion of Conium maculatum (poison hemlock) in newborn pigs. Am J Vet Res. 1985;46:2064.

99 Gardner DR, Panter KE. Comparison of blood plasma alkaloid levels in cattle, sheep, and goats fed Lupinus caudatus. J Nat Toxins. 1993;2:1.

100 Lopez-Ortiz S. The effect of body condition on disposition of alkaloids from silvery lupine (Lupinus argenteus Pursh) in sheep. J Anim Sci. 2004;82:2798.

101 Panter KE, James LF. Natural toxicants in milk: a review. J Anim Sci. 1990;68:892.

102 Lopez TA, Cid MS, Bianchini ML. Biochemistry of hemlock (Conium maculatum L.) alkaloids and their acute and chronic toxicity in livestock: a review. Toxicon. 1999;37:841.

103 Cheeke PR. Natural toxicants in feeds, forages, and poisonous plants, ed 2, Danville, Ill: Interstate; 1998:393.

104 Knight AP. Veratrum californicum poisoning. Compend Cont Educ. 1989;11:528.

105 Kingsbury JM. Poisonous plants of the United States and Canada. Englewood Cliffs, NJ: Prentice-Hall, 1964;289.

106 Keeler RF, Binns W. Teratogenic compounds of Veratrum californicum. Teratology. 1968;1:5.

107 Sharma RP, Salunkhe DK. Solanum glycoalkaloids. In: Cheeke PR, editor. Toxicants of plant origin, vol 1, Alkaloids. Boca Raton, Fla: CRC Press; 1989:179.

108 Schlosberg A, et al. The effect of feeding dried tomato vines to beef cattle. Vet Hum Toxicol. 1996;38:135.

109 Binns W, Keeler RF, Balls LD. Congenital deformation in lambs, calves, and goats resulting from maternal ingestion of Veratrum californicum. Clin Toxicol. 1972;5:245.

110 Binns W, et al. Chronologic evaluation of teratogenicity in sheep fed Veratrum californicum. J Am Vet Med Assoc. 1965;247:839.

111 Keeler RF, Stuart LD. The nature of congenital limb defects induced in lambs by maternal ingestion of Veratrum californicum. Clin Toxicol. 1987;25:273.

112 Cheeke PR. Natural toxicants in feeds, forages, and poisonous plants, ed 2, Danville, Ill: Interstate; 1998:382.

113 Galey FD, et al. Residues of Datura species in horse. In: Auer D, Houghten E, editors. Proceedings of the Eleventh International Conference of Racing Analysts and Veterinarians. Queensland, Australia, Newmarket, England: R & W; 1996:333.

114 Schulman ML, Bolton LA. Datura seed intoxication in two horses. J S Afr Vet Assoc. 1998;69:27.

115 Piva G, Piva A. Anti-nutritional factors of Datura in feedstuffs. Nat Toxins. 1995;3:238.

116 Alden CL, et al. Japanese yew poisoning of large domestic animals in the Midwest. J Am Vet Med Assoc. 1977;170:314.

117 Smythies JR, et al. The action of the alkaloids from yew (Taxus baccata) on the action potential in the Xenopus medullated axon. Experientia. 1975;15:337.

118 Vidensek NA, et al. Taxol content in bark, wood, root, leaf, twig, and seedling from several Taxus species. J Nat Prod. 1990;53:1609.

119 Lowe JE, et al. Taxus cuspidata (Japanese yew) poisoning in horses. Cornell Vet. 1969;60:36.

120 Ogden L: Taxus(yews): a highly toxic plant. Vet Hum Toxicol. 1988;30:563.

121 Wilson CR, et al. Taxines: a review of the mechanism and toxicity of yew (Taxus spp.) alkaloids. Toxicon. 2001;39:175.

122 Kite GC, Lawrence TJ, Dauncey EA. Detecting Taxus poisoning in horses using liquid chromatography/mass spectrometry. Vet Hum Toxicol. 2000;42:151.

123 Culvenor CCJ, Dal Bon R, Smith LW. The occurrence of indolealkylamine alkaloid in Phalaris tuberosa L and P. arundinacea L. Aust J Chem. 1964;17:1301.

124 Cheeke PR. Natural toxicants in feeds, forages, and poisonous plants, ed 2, Danville, Ill: Interstate; 1998:275.

125 Gallagher CH, et al. The toxicity of Phalaris tuberosa for sheep. Nature. 1964;204:542.

126 Gallagher CH, Koch JH, Hoffman H. Deaths of ruminants grazing Phalaris tuberosa in Australia. Aust Vet J. 1967;43:495.

127 McDonald IW. A “staggers” syndrome in sheep and cattle associated with grazing Phalaris tuberosa. Aus Vet J. 1942;18:182.

128 East NE, Higgins RJ. Canarygrass (Phalaris spp) toxicosis in sheep in California. J Am Vet Med Assoc. 1988;192:667.

129 Colegate SM, et al. Suspected blue canary grass (Phalaris coerulescens) poisoning of horses. Aust Vet J. 1999;77:537.

130 Lean IJ, et al. Tryptamine alkaloid toxicosis in feedlot sheep. J Am Vet Med Assoc. 1989;195:768.

131 Cheeke PR, Shull LR. Natural toxicants in feeds and poisonous plants. Westport, Conn: AVI, 1985;148.

132 Lee HJ, Kuchel RD. The aetiology of Phalaris staggers in sheep. Aust J Agric Res. 1953;4:88.

133 Majak W. Review of toxic glycosides in rangeland and pasture forages. J Range Mgmt. 2001;54:494.

134 Cheeke PR, Shull LR. Natural toxicants in feeds and poisonous plants. Westport, Conn: AVI, 1985;9.

135 Kingsbury JM. Poisonous plants of the United States and Canada. Englewood Cliffs, NJ: Prentice-Hall, 1964;264.

136 Joubert JPJ. Cardiac glycosides. In: Cheeke PR, editor. Toxicants of plant origin, vol II, Glycosides. Boca Raton, Fla: CRC Press; 1989:61.

137 Adams HR. Digitalis and other inotropic agents and vasodilator drugs. In: Booth NH, McDonald LE, editors. Veterinary pharmacology and therapeutics. ed 6. Ames, Iowa: Iowa State University Press; 1988:495.

138 Szabuniewicz JD, McCrady JD, Camp BJ. Treatment of experimentally induced oleander poisoning. Arch Intern Pharmacodynam. 1971;189:12.

139 Maejima H, et al. Distinct sites regulating grayanotoxin binding and unbinding to D4S6 of Na(v)1.4 sodium channel as revealed by improved estimation of toxin sensitivity. J Biol Chem. 2003;278:9469.

140 Ansford A, et al. Oleander poisoning. Toxicon. 1983;3(suppl):15.

141 Knight AP. Oleander poisoning. Compend Cont Educ. 1988;10:262.

142 Knight AP. Rhododendron and laurel poisoning. Compend Cont Educ. 1987;9:F26.

143 Everist SL. Poisonous plants of Australia, ed 2, London: Angus and Robertson; 1981:77.

144 Mahin L, Marzou A, Huart A. A case report of Nerium oleander poisoning in cattle. Vet Hum Toxicol. 1984;26:303.

145 Galey FD, et al. Toxicity and diagnosis of oleander (Nerium oleander) poisoning in livestock. In: Garland T, Barr AC, editors. Toxic plants and other natural toxicants. Wallingford, UK: CAB International; 1998:215.

146 Tracqui A, et al. Confirmation of oleander poisoning by HPLC/MS. Int J Legal Med. 1998;111:32.

147 Puschner B, et al. Grayanotoxin poisoning in three goats. J Am Vet Med Assoc. 2001;218:573.

148 Osterloh J, Herold S, Pond S. Oleander interference in the digoxin RIA in a fatal ingestion. J Am Med Assoc. 1982;247:1596.

149 Haber E. Antibodies and digitalis: the modern revolution in the use of an ancient drug. J Am Coll Cardiol. 1985;5:111A.

150 Shumaik GM, Wu AW, Ping AC. Oleander poisoning: treatment with digoxin-specific Fab antibody fragments. Ann Emerg Med. 1988;17:732.

151 Camp BJ, et al. Isolation of a steroidal sapogenin from the bile of sheep fed Agave lechuguilla. Vet Hum Toxicol. 1988;30:533.

152 Kellerman TS, et al. Photosensitivity in South Africa. VI. The experimental induction of geeldikkop in sheep with crude steroidal saponins from Tribulus terrestris. Onderstepoort J Vet Res. 1991;58:47.

153 Miles CO, et al. Identification of a sapogenin glucuronide in the bile of sheep affected by Panicum dichotomiflorum toxicosis. NZ Vet J. 1991;39:150.

154 Miles CO, et al. Identification of insoluble salts of the β-D glucuronide of episarsapogenin and epismilagenin in the bile of lambs with aveld, and examination of Narthecium ossifragum, Tribulus terrestris, and Panicum miliaceum for sapogenins. J Agric Food Chem. 1993;41:914.

155 Miles CO. Role of saponins in hepatogenous photosensitization. NZ Vet J. 1993;41:221. (abstract)

156 Smith BL, Miles CO. A role for Brachiaria decumbens in hepatogenous photosensitization of ruminants. Vet Hum Toxicol. 1993;35:256.

157 Low SG, et al. Photosensitization of cattle grazing signal grass (Brachiaria decumbens) in Papua New Guinea. NZ Vet J. 1993;41:220.

158 Cornick JL, Carter GK, Bridges CH. Kleingrass-associated hepatotoxicosis in horses. J Am Vet Med Assoc. 1988;193:932.

159 Bridges CH, et al. Kleingrass (Panicum coloratum L) poisoning in sheep. Vet Pathol. 1987;24:525.

160 Mathews FP. Lechuguilla (Agave lechuguilla) poisoning in sheep and goats. J Am Vet Med Assoc. 1938;93:168.

161 Langseth W, et al. Isolation and characterization of 3 methoxy-2(5H) furanone as the principal nephrotoxin from Narthecium ossifragum (L) Huds. Nat Toxins. 1999;7:111.

162 Osweiler GD, et al. Clinical and diagnostic veterinary toxicology. Dubuque, Iowa: Kendall/Hunt, 1985;455.

163 Tewe OO, Iyayi EA. Cyanogenic glycosides. In: Cheeke PR, editor. Toxicants of plant origin, vol 2, Glycosides. Boca Raton, Fla: CRC Press; 1989:44.

164 Morgan SE, Johnson B, Brewer B. Sorghum cystitis ataxia syndrome in horses. Vet Hum Toxicol. 1990;32:582.

165 McKenzie RA, McMicking L. Ataxia and urinary incontinence in cattle grazing sorghum. Aust Vet J. 1977;53:496.

166 Bradley GA, et al. Neuroaxonal degeneration in sheep grazing sorghum pastures. J Vet Diagn Invest. 1995;7:229.

167 Burrows GE. Cyanide intoxication in sheep: therapeutics. Vet Hum Toxicol. 1981;23:22.

168 Stermitz FR, Norris FA, Williams MC. Miserotoxin, a new naturally occurring nitrocompound. J Am Chem Soc. 1969;91:4599.

169 Majak W, Benn MH. New glycosides of 3-nitropropanol from Astragalus miser var serotinus. In: James LF, et al, editors. Poisonous Plants: Proceedings of the Third International Symposium. Ames, Iowa: Iowa State University Press; 1992:523.

170 Majak W, et al. Analysis and metabolism of nitrotoxins in cattle and sheep. In: Seawright AA, et al, editors. Plant Toxicology: Proceedings of the Australia-USA Poisonous Plants Symposium. Brisbane, Australia, Yeerongpilly, Queensland: Queensland Department of Primary Industries; 1985:446.

171 Majak W, Pass MA. Aliphatic nitrocompounds. In: Cheeke PR, editor. Toxicants of plant origin, vol 2, Glycosides. Boca Raton, Fla: CRC Press; 1989:446.

172 James LF, et al. Field and experimental studies in cattle and sheep poisoned by nitro-bearing Astragalus or their toxins. Am J Vet Res. 1980;41:377.

173 Pass MA. Toxicity of aliphatic plant-derived nitrotoxins. In: Colegate SM, Dorling PR, editors. Plant-associated toxins: agricultural, phytochemical, and ecological aspects. Wallingford, UK: CAB International; 1994:541.

174 Majak W, et al. Toxicity and development of tolerance in cattle to timber milkvetch. J Range Mgmt. 2003;56:266.

175 Fenwick GR. Bracken (Pteridium aquilinum): toxic effects and toxic constituents. J Sci Food Agric. 1988;46:147.

176 Smith BL, et al. Study on DNA adduct formation by ptaquiloside: the carcinogen of bracken fern (Pteridium spp) and from a cultivated collection of bracken from worldwide sources. In: Colegate SM, Dorling PR, editors. Plant-associated toxins: agricultural, phytochemical, and ecological aspects. Wallingford, UK: CAB International; 1994:239.

177 Smith BL, et al. Ptaquiloside in bracken (Pteridium spp). In: Colegate SM, Dorling PR, editors. Plant-associated toxins: agricultural, phytochemical, and ecological aspects. Wallingford, UK: CAB International; 1994:45.

178 Hirono I. Carcinogenic bracken glycosides. In: Cheeke PR, editor. Toxicants of plant origin, vol 2, Glycosides. Boca Raton, Fla: CRC Press; 1989:239.

179 Schacham P, Philip RB, Gowdey CW. Antihematopoetic and carcinogenic effects of bracken fern (Pteridium aquilinum) in rats. Am J Vet Res. 1970;31:191.

180 Kingsbury JM. Poisonous plants of the United States and Canada. Englewood Cliffs, NJ: Prentice-Hall, 1964;105.

181 McKenzie RA. Bovine enzootic haematuria in Queensland. Aust Vet J. 1978;54:61.

182 Hirono I, et al. Induction of tumors in ACI rats given a diet containing ptaquiloside, a bracken carcinogen. J Natl Cancer Inst. 1987;79:1143.

183 Evans IA. Bracken carcinogenicity. Res Vet Sci. 1979;26:339.

184 Evans WC. Bracken poisoning of farm animals. Vet Rec. 1964;76:365.

185 Gerenutti M, et al. Effects of bracken fern (Pteridium aquilinum L Kuhn) feeding during the development of female rats and their offspring. Vet Hum Toxicol. 1992;34:307.

186 Singh RP, et al. Experimental bracken fern toxicity in calves: changes in blood and urine. Indian J Vet Med. 1987;7:96.

187 Alonso-Amelot ME. The link between bracken fern and stomach cancer: milk. Nutrition. 1997;13:694.

188 Alonso-Amelot ME, Avendano M. Human carcinogenesis and bracken fern: a review of the evidence. Curr Med Chem. 2002;9:675.

189 Adams NR. Phytoestrogens. In: Cheeke PR, editor. Toxicants of plant origin,vol 4, Phenolics. Boca Raton, Fla: CRC Press; 1989:23.

190 Livingston AL. Forage plant estrogens. J Toxicol Environ Health. 1978;4:301.

191 Bickoff EM, et al. Coumestrol, a new estrogen isolated from forage crops. Science. 1957;126:969.

192 Bickoff EM, et al. Relative potencies of several estrogen-like compounds in forages. J Agric Food Chem. 1962;10:410.

193 Lundh TJO, Pettersson H, Kiessling KH. Liquid chromatographic determination of the estrogens daidzein, formononetin, coumestrol, and equol in bovine blood plasma and urine. J Assoc Off Anal Chem. 1988;71:938.

194 Welshons WV, et al. A sensitive bioassay for detection of dietary estrogens in animal feeds. J Vet Diagn Invest. 1990;2:268.

195 Galey FD, et al. Estrogenic activity in forages: diagnostic use of the classical mouse uterine bioassay. J Vet Diagn Invest. 1993;5:603.

196 Cheeke PR. Natural toxicants in feeds, forages, and poisonous plants, ed 2, Danville, Ill: Interstate; 1998:294.

197 Benson ME, Casper HH, Johnson LJ. Occurrence and range of dicoumarol concentration in sweet clover. Am J Vet Res. 1981;42:2014.

198 Cole RJ, Cutler HG, Stuart BP. Carboxyatractyloside. In: Cheeke PR, editor. Toxicants of plant origin, vol 2, Glycosides. Boca Raton, Fla: CRC Press; 1989:253.

199 Martin T, Stair EL, Dawson L. Cocklebur poisoning in cattle. J Am Vet Med Assoc. 1986;189:562.

200 Witte ST, et al. Cocklebur toxicosis in cattle associated with the consumption of mature Xanthium strumarium. J Vet Diagn Invest. 1990;2:263.

201 Cheeke PR. Natural toxicants in feeds, forages, and poisonous plants, ed 2, Danville, Ill: Interstate; 1998:436.

202 Rowe LD, et al. Photosensitization of cattle in Southwest Texas: identification of phototoxic activity associated with Cooperia pedunculata. Am J Vet Res. 1987;48:1658.

203 Campero CM, Odriozola E. A case of Solanum malacoxylon toxicity in pigs. Vet Hum Toxicol. 1990;32:238.

204 Krook L, et al. Hypercalcemia and cacinosis in Florida horses: implication of the shrub Cestrum diurnum as the causative agent. Cornell Vet. 1975;65:26.

205 Krook L, et al. Cestrum diurnum poisoning in Florida cattle. Cornell Vet. 1975;65:557.

206 Gorniak SL, et al. Evaluation in rabbits of fetal effects of maternal ingestion of Solanum malacoxylon. Vet Res Commun. 1999;23:307.

207 Cheeke PR. Natural toxicants in feeds, forages, and poisonous plants, ed 2, Danville, Ill: Interstate; 1998:281.

208 Nachman RJ, Olson JD. Ranunculin: a toxic constituent of the poisonous range plant bur buttercup (Ceratocephalus testiculatus). J Agric Food Chem. 1983;31:1358.

209 Olson JD, et al. Bur buttercup poisoning of sheep. J Am Vet Med Assoc. 1983;183:538.

210 Cheeke PR, Shull LR. Natural toxicants in feeds and poisonous plants. Westport, Conn: AVI, 1985;224.

211 Hooper PT, Best SM, Campbell A: Axonal dystrophy in the spinal cords of cattle consuming the cycad plant Cycas media. Aust Vet J. 1974;50:146.

212 Hooper PT. Cycad poisoning in Australia: etiology and pathology. In: Keeler RF, et al, editors. Effect of poisonous plants in livestock. New York: Academic; 1978:337.

213 Reams RY, et al. Cycad (Zamia puertoriquensis) toxicosis in a group of dairy heifers in Puerto Rico. J Vet Diagn Invest. 1993;5:488.

214 Eizirik DL, Kisby GE. Cycad toxin-induced damage of rodent and human pancreatic beta-cells. Biochem Pharmacol. 1995;50:355.

215 Morgan SE. Gossypol as a toxicant in livestock. Vet Clin North Am Food Anim Pract. 1989;5:251.

216 Haschek WM, et al. Cottonseed meal (gossypol) toxicosis in a swine herd. J Am Vet Med Assoc. 1989;195:613.

217 Reyes J, Borriero L, Benos DJ. A bioenergetic model of gossypol action: effects of gossypol on adult rat spermatogenic cells. Am J Physiol. 1988;254:C564.

218 Taitzoglou IA, et al. Gossypol-induced inhibition of plasminogen activator activity in human and ovine acrosomal extract. Andrologia. 1999;31:355.

219 Smalley SA, Bicknell EJ. Gossypol toxicity in dairy cattle. Compend Cont Educ. 1982;4:S378.

220 Hudson LM, Kerr LA, Maslin WR. Gossypol toxicosis in a herd of beef calves. J Am Vet Med Assoc. 1988;192:1303.

221 Morgan S, et al. Clinical, clinicopathologic, pathologic, and toxicologic alterations associated with gossypol toxicosis in feeder lambs. Am J Vet Res. 1988;49:493.

222 Patton CS, et al. Heart failure caused by gossypol poisoning in two dogs. J Am Vet Med Assoc. 1985;187:625.

223 Holmberg CA, et al. Pathological and toxicological studies of calves fed a high concentration cottonseed meal diet. Vet Pathol. 1988;25:147.

224 Randel RD, Chase CC, Wyse SJ. Effects of gossypol and cottonseed products on reproduction of mammals. J Anim Sci. 1992;70:1628.

225 Pattanaik AK, et al. Effect of gossypol from cottonseed meal diets on some clinico-biochemical parameters and humoral immune response of crossbred calves fed barley or sorghum. Asian-Australasian J Anim Sci. 2003;16:1291.

226 Vranova J, et al. Inhibitory effect of gossypol on basal cell luteinization factor-stimulated progesterone synthesis in porcine granulosa cells. Physiol Res. 1999;48:119.

227 Hassan ME, et al. Reversibility of the reproductive toxicity of gossypol in peripuberal bulls. Theriogenology. 2004;61:1171.

228 Velasquez-Pereira J, et al. Long-term effects of feeding gossypol and vitamin E to dairy calves. J Dairy Sci. 1999;82:1240.

229 Eisele GR. A perspective on gossypol ingestion in swine. Vet Hum Toxicol. 1986;28:118.

230 Osweiler GD, et al. Clinical and diagnostic veterinary toxicology. Dubuque, Iowa: Kendall/Hunt, 1985;468.

231 Pigeon RF, Camp BJ, Dollahite JW. Oral toxicity and polyhydroxyphenol moiety of tannin isolated from Quercus havardi (shin oak). Am J Vet Res. 1962;23:1268.

232 Dollahite JW, Pigeon RF, Camp BJ. The toxicity of gallic acid, pyrogallol, tannic acid, and Quercus havardi in the rabbit. Am J Vet Res. 1962;23:1264.

233 Oelrichs PB, et al. The isolation, structure elucidation and toxicity of hepatotoxic and nephrotoxic principles in Terminalia oblongota. In: Colegate SM, Dorling PR, editors. Plant associated toxins. Wallingford, UK: CAB International; 1994:245.

234 Spier SJ, et al. Oak toxicosis in cattle in Northern California: clinical and pathologic findings. J Am Vet Med Assoc. 1987;191:958.

235 Hart GH, et al. Acorn calves, Bulletin 699, California Agricultural Experiment Station. 1947.

236 Basden KW, Dalvi RR. Determination of total phenolics in acorns from different species of oak trees in conjunction with acorn poisoning in cattle. Vet Hum Toxicol. 1987;29:305.

237 Householder GD, Dollahite JW. Some clinical biochemical changes in the blood serum of calves fed Quercus havardi. Southwest Vet. 1963;16:107.

238 Kingsbury JM. Poisonous plants of the United States and Canada. Englewood Cliffs, NJ: Prentice Hall, 1964;235.

239 Cheeke PR, Shull LR. Natural toxicants in feeds and poisonous plants. Westport, Conn: AVI, 1985;314.

240 Van Kampen KR, James LF. Acute halogeton poisoning of sheep: pathogenesis of lesions. Am J Vet Res. 1969;30:1779.

241 Allison MJ, Cook HM, Dawson KA. Selection of oxalate-degrading rumen bacteria in continuous cultures. J Anim Sci. 1981;53:810.

242 Lincoln SD, Black B. Halogeton poisoning in range cattle. J Am Vet Med Assoc. 1980;176:717.

243 Beier RC, Normal JO. The toxic factor in white snakeroot: identity, analysis, and prevention. Vet Hum Toxicol. 1990;32(suppl):81.

244 Beier RC, et al. Microsomal activation of constituents of white snakeroot (Eupatorium rugosum) to form toxic products. Am J Vet Res. 1987;48:583.

245 White JL, et al. White snakeroot (Eupatorium rugosum) poisoning: clinical effects associated with cardiac and skeletal muscle lesions in experimental equine toxicosis. In: Seawright AA, et al, editors. Plant Toxicology: Proceedings of the Australia-USA Poisonous Plants Symposium. Brisbane, Australia, Yeerongpilly, Queensland: Queensland Department of Primary Industries; 1985:411.

246 Olson CT, et al. Suspected tremetol poisoning in horses. J Am Vet Med Assoc. 1984;185:1001.

247 Kingsbury JM. Poisonous plants of the United States and Canada. Englewood Cliffs, NJ: Prentice-Hall, 1964;397.

248 Stotts R. White snakeroot toxicity in dairy cattle. Vet Med. 1984:p, 18.

249 Smetzer DL, et al. Cardiac effects of white snakeroot intoxication in horses. Equine Pract. 1983;5:26.

250 Cheeke PR. Natural toxicants in feeds, forages, and poisonous plants, ed 2, Danville, Ill: Interstate; 1998:233.

251 James LF, et al. Pine needle abortion in cattle: a review and report of 1973-1984 research. Cornell Vet. 1989;79:39.

252 Stuart LD, et al. Pine needle abortion in cattle: pathological observations. Cornell Vet. 1989;79:61.

253 Call JW, James LF. Effect of feeding pine needles on ovine reproduction. J Am Vet Med Assoc. 1976;169:1301.

254 James LF, Call JW, Stevenson AH. Experimentally induced pine needle abortion in range cattle. Cornell Vet. 1977;67:294.

255 Gardner DR, et al. Ponderosa pine needle induced abortion in beef cattle: identification of isocupressic acid as the principal active compound. J Agric Food Chem. 1994;42:756.

256 Gardner DR, et al. Abortifacient effects of lodgepole pine (Pinus contorta) and common juniper (Juniperus communis) in cattle. Vet Hum Toxicol. 1998;40:260.

257 Ford SP, et al. Abortifacient effects of a unique class of vasoactive lipids from Pinus ponderosa needles. J Anim Sci. 1999;77:2187.

258 Ford SP, et al. Effects of ponderosa pine needle ingestion on uterine vascular function in late-gestation beef cows. J Anim Sci. 1992;70:1609.

259 Panter KE, Keeler RF, Baker DC. Toxicosis in livestock from the hemlocks (Conium and Cicuta spp). J Anim Sci. 1988;66:2407.

260 Smith RA, Lewis D. Cicuta toxicosis in cattle: case history and simplified analytical method. Vet Hum Toxicol. 1987;29:240.

261 Knight AP. Milkweed poisoning. Compend Cont Educ. F348, 1987.

262 Panter KE, Baker DC, Kechele PO. Water hemlock (Cicuta douglasii) toxicosis in sheep: pathologic description and prevention of lesions and death. J Vet Diagn Invest. 1996;8:474.

263 Cheeke PR, Shull LR. Natural toxicants in feeds and poisonous plants. Westport, Conn: AVI, 1985;348.

264 Johnson AE. Predisposing influence of range plants on Tetradymia-related photosensitization in sheep: work of Drs AD Clawson and WT Huffman. Am J Vet Res. 1976;35:1583.

265 Johnson AE. Experimental photosensitization and toxicity in sheep produced by Tetradymia glabrata. Can J Comp Med. 1974;38:406.

266 Osweiler GD, et al. Clinical and diagnostic veterinary toxicology. Dubuque, Iowa: Kendall/Hunt, 1985;460.

267 Crawford RF, Kennedy WK, Davison KL. Factors influencing the toxicity of forages that contain nitrate when fed to cattle. Cornell Vet. 1966;56:3.

268 Schneider NR, Carlson MP. Implications and significance of excessive nitrate/nitrite exposure in animal health. Proceedings of the California Animal Nutrition Conference, Fresno, Calif. 1991:121.

269 Johnson JL, et al. Post-harvest change in cornstalk nitrate and its relationship to bovine fetal nitrite/nitrate exposure. In: James LF, et al, editors. Poisonous Plants: Proceedings of the Third International Symposium. Ames, Iowa: Iowa State University Press; 1992:423.

270 T-Dargatz JL, Hamar DW. Selenium toxicity in horses. Compend Cont Educ. 1986;8:771.

271 Harrison LH, et al. Paralysis in swine due to focal symmetrical poliomalacia: possible selenium toxicosis. Vet Pathol. 1983;20:265.

272 Blodgett DJ, Bevill RF. Acute selenium toxicosis in sheep. Vet Hum Toxicol. 1987;29:233.

273 Janke BH. Acute selenium toxicosis in a dog. J Am Vet Med Assoc. 1989;195:1114.

274 Osweiler GD, et al. Clinical and diagnostic veterinary toxicology. Dubuque, Iowa: Kendall/Hunt, 1985;132.

275 Stowe HD, et al. Selenium toxicosis in feeder pigs. J Am Vet Med Assoc. 1992;201:292.

276 Casteel SW, et al. Selenium toxicosis in swine. J Am Vet Med Assoc. 1985;186:1084.

277 O’Toole D, Raisbeck MF. Pathology of experimentally induced chronic selenosis (alkali disease) in yearling cattle. J Vet Diagn Invest. 1995;7:364.

278 Yaeger MJ, et al. The effect of subclinical selenium toxicosis on pregnant beef cattle. J Vet Diagn Invest. 1998;10:268.

279 Davidson-York D, et al. Selenium elimination in pigs after an outbreak of selenium toxicosis. J Vet Diagn Invest. 1999;11:352.

280 Raisbeck MF, McAllister MM, Gould DH. Toxic syndrome associated with sulfur. In: Howard JL, Smith RA, editors. Current veterinary therapy 4: food animal practitioner. Philadelphia: Saunders; 1999:280.

281 Peckham JC, et al. Atypical interstitial pneumonia in cattle fed moldy sweet potatoes. J Am Vet Med Assoc. 1972;160:169.

282 Kerr LA, Linnabary RD. Atypical bovine pulmonary emphysema: another cause (abstract). Proceedings of the Thirty-first Annual Meeting of the American Association of Veterinary Laboratory Diagnosticians, Little Rock, Ark. 1988:74.

283 Low SG, et al. Sweet potato (Ipomoea batatas) poisoning of pigs in Papua New Guinea. NZ Vet J. 1993;41:218.

284 Cheeke PR, Shull LR. Natural toxicants in feeds and poisonous plants. Westport, Conn: AVI, 1985;261.

285 Kerr LA, Johnson BJ, Burrows GE. Intoxication of cattle by Perilla frutescens (purple-mint). Vet Hum Toxicol. 1986;28:412.

286 Carlson JR, et al. Pulmonary edema and emphysema in cattle after intraruminal and intravascular administration of 3-methyl indole. Am J Vet Res. 1975;36:1341.

287 Boyd MR, et al. Lung toxic furanoterpenoids produced by sweet potatoes (Ipomoea batatas) following microbial infection. Biochem Biophys Acta. 1974;337:184.

288 Atkinson G, et al. Effects of 3 methyl indole in cattle. Br J Pharmacol. 1977;61:285.

289 Rajendran S, et al. Oxidative stress in rat liver and lung induced by furanoterpenoids isolated from Fusarium solani infected sweet potatoes. Indian J Exp Biol. 1996;34:57.

290 Garst JE, et al. Species susceptibility to pulmonary toxicity of 3 furyl isoamyl ketone (perilla ketone): in vivo support for involvement of the lung monooxygenase system. J Anim Sci. 1985;60:248.

291 Kubow S, Bray TM. The effect of lung concentrations of glutathione and vitamin E on the pulmonary toxicity of 3 methyl indole. Can J Physiol Pharmacol. 1988;66:863.

292 Li X, Castleman WL. Ultrastructural morphogenesis of 4-ipomeanol-induced bronchiolitis and interstitial pneumonia in calves. Vet Pathol. 1990;27:141.

293 Durham SR, Boyd MR, Castleman WL. Pulmonary endothelial and bronchiolar epithelial lesions induced by 4-ipomeanol in mice. Am J Pathol. 1985;118:61.

294 Dorster AR, et al. Effects of 4-ipomeanol, a product from mold-damaged sweet potatoes, on the bovine lung. Vet Pathol. 1978;15:367.

295 Bradley BJ, Carlson JR. Ultrastructural pulmonary changes induced by intravenously administered 3-methylindole in goats. Am J Pathol. 1980;99:551.

296 Turk MAM, Thomas DE. Effects of phenobarbital on 3 methyl indole toxicosis in ponies. Am J Vet Res. 1986;47:901.

297 Cheeke PR, Shull LR. Natural toxicants in feeds and poisonous plants. Westport, Conn: AVI, 1985;276.

298 Pierce KR, et al. Acute hemolytic anemia caused by wild onion poisoning in horses. J Am Vet Med Assoc. 1972;160:323.

299 Van Kampen KR, James LF, Johnson AE. Hemolytic anemia in sheep fed wild onions. J Am Vet Med Assoc. 1970;156:328.

300 Hutchison TWS. Onions as a cause of Heinz body anemia and deaths in cattle. Can Vet J. 1977;18:358.

301 Stair EL, et al. Suspected red maple (Acer rubrum) toxicosis with abortion in two Percheron mares. Vet Hum Toxicol. 1993;35:229.

302 George LW, et al. Heinz body anemia and methemoglobinemia in ponies given red maple (Acer rubrum) leaves. Vet Pathol. 1982;19:521.

303 Plumlee KH. Red maple toxicity in a horse. Vet Hum Toxicol. 1991;33:66.

304 Knight AP, et al. Adaptation of pregnant ewes to an exclusive onion diet. Vet Hum Toxicol. 2000;42:1.

305 Osweiler GD, et al. Clinical and diagnostic veterinary toxicology. Dubuque, Iowa: Kendall/Hunt, 1985;476.

306 Sizelone W, et al. Perirenal edema in a calf. Vet Hum Toxicol. 1988;30:265.

307 Kerr LA, Kelch WJ. Pigweed (Amaranthus retroflexus) toxicosis in cattle. Vet Hum Toxicol. 1998;40:216.

308 Colvin BM, et al. Cassia occidentalis toxicosis in growing pigs. J Am Vet Med Assoc. 1986;189:423.

309 Henson JB, et al. Myodegeneration in cattle grazing Cassia species. J Am Vet Med Assoc. 1965;147:142.

310 Simpson CF, Damron BL, Harms RH. Toxic myopathy of chicks fed Cassia occidentalis seeds. Avian Dis. 1971;15:284.

311 Martin BM, et al. Toxicity of Cassia occidentalis in the horse. Vet Hum Toxicol. 1981;23:416.

312 Ohara PJ, Pierce KR, Read WK. Degenerative myopathy associated with ingestion of Cassia occidentalis L: clinical and pathologic features of the experimentally induced disease. Am J Vet Res. 1969;30:2173.

313 Rowe LD, et al. Experimentally induced Cassia roemeriana poisoning in cattle and goats. Am J Vet Res. 1987;48:992.

314 Robles MR, et al. Cytotoxic effects of repen, a principal sesquiterpene lactone of Russian knapweed. J Neurosci Res. 1997;47:90.

315 Roy DN, Peyton DH, Spencer PS. Isolation and identification of two potent neurotoxins, aspartic acid and glutamic acid, from yellow star thistle (Centaurea solstitialis). Nat Toxins. 1995;3:174.

316 Cheeke PR. Natural toxicants in feeds, forages, and poisonous plants, ed 2, Danville, Ill: Interstate; 1998:385.

317 Thomsen C, Williams WA. Yellow starthistle control. Components. 1992;3:2.

318 Calhoun MC, et al. Experimental prevention of bitterweed (Hymenoxys odorata) poisoning of sheep. Am J Vet Res. 1989;50:1642.

319 Cheeke PR, Shull LR. Natural toxicants in feeds and poisonous plants. Westport, Conn: AVI, 1985;359.

320 Rowe LD, et al. Hymenoxys odorata (bitterweed) poisoning in sheep. Southwest Vet. 1973;26:287.

321 Kim HL, Rowe LD, Camp BJ. Hymenoxin, a poisonous sesquiterpene lactone from Hymenoxys odorata (bitterweed). Res Commun Chem Pathol Pharmacol. 1975;11:647.

322 Hill DW, Camp BJ. Reactions of hymenoxin: base conversion to psilotropin and greenein and formation of a “Michael adduct” with cysteine. J Agric Food Chem. 1979;27:882.

323 Galey FD, et al. Black walnut (Juglans nigra) toxicosis: a model for equine laminitis. J Comp Pathol. 1991;104:313.

324 Minnick PD, et al. The induction of equine laminitis with an aqueous extract of the heartwood of black walnut (Juglans nigra). Vet Hum Toxicol. 1987;29:230.

325 True RG, et al. Black walnut shavings as a cause of acute laminitis. Proc Am Assoc Equine Pract. 1978;24:511.

326 Geor RJ, et al. Toxicosis in horses after ingestion of hoary alyssum. J Am Vet Med Assoc. 1992;210:63.

327 Kingsbury JM. Poisonous plants of the United States and Canada. Englewood Cliffs, NJ: Prentice-Hall, 1964;220.

328 Bermudez MW, et al. Experimental intoxication with fruit and purified toxins of buckthorn (Karwinskia humboldtiana). Toxicon. 1986;24:1091.

329 Jaramillo-Juarez F, et al. Renal failure during acute toxicity produced by tullidoro ingestion (Karwinskia humboldtiana). Gen Pharmacol. 1995;26:649.

330 Galey FD, Hullinger PJ, McCaskill J. Outbreaks of stringhalt in Northern California. Vet Hum Toxicol. 1991;33:176.

331 Huntington PJ, et al. Australian stringhalt: epidemiological, clinical, and neurological investigations. Equine Vet J. 1989;21:266.

332 Cahill JI, Goulden BE, Jolly RD. Stringhalt in horses: a distal axonopathy. Neuropathol Appl Neurobiol. 1986;12:459.

333 Cheeke PR. Natural toxicants in feeds, forages, and poisonous plants, ed 2, Danville, Ill: Interstate; 1998:193.

334 McKenzie RA, Brown OP. Avocado (Persea americana) poisoning of horses. Aust Vet J. 1991;68:77.

335 Craigmill AL, et al. Toxicity of avocado (Persea americana; Guatemalan) leaves: review and preliminary report. Vet Hum Toxicol. 1984;26:381.

336 Craigmill AL, et al. Pathological changes in the mammary gland and biochemical changes in the milk of the goat following oral dosing with leaf of the avocado (Persea americana). Aust Vet J. 1989;46:206.

337 Sani Y, Atwell RB, Seawright AA. The cardiotoxicity of avocado leaves. Aust Vet J. 1991;68:150.

338 Hargis AM, et al. Avocado (Persea americana) intoxication in caged birds. J Am Vet Med Assoc. 1989;194:64.

339 Galey FD, et al. Facial edema and myopathy in horses exposed to avocado (Persea americana) plants. Am Assoc Vet Lab Diagn. 1993;36:33. (abstract)

340 Burger WP, et al. Avocado (Persea americana) poisoning in ostriches. In: Colgate SM, Dorling PR, editors. Plant-associated toxins: agricultural, phytochemical, and ecological aspects. Wallingford, UK: CAB International; 1994:546.

341 Oelrichs PB, et al. Isolation and identification of a compound from avocado (Persea americana) leaves which causes necrosis of the acinar epithelium of the lactating mammary gland and the myocardium. Nat Toxins. 1995;3:344.

342 Cheeke PR, Shull LR. Natural toxicants in feeds and poisonous plants. Westport, Conn: AVI, 1985;240.

343 Ma L, Hsu CH, Patterson E. Ricin depresses cardiac function in the rabbit heart. Toxicol Appl Pharmacol. 1996;138:72.

344 Panciera RJ, Mosier DA, Ritchey JW. Hairy vetch (Vicia villosa Roth) poisoning in cattle: update and experimental induction of the disease. J Vet Diagn Invest. 1992;4:318.

345 Johnson B, et al. Systemic granulomatous disease in cattle in California associated with grazing hairy vetch (Vicia villosa). J Vet Diagn Invest. 1992;4:360.

346 Woods LW, et al. Systemic granulomatous disease in a horse grazing pasture containing vetch (Vicia spp). J Vet Diagn Invest. 1992;4:356.

347 Anderson CA, Divers TJ. Systemic granulomatous inflammation in a horse grazing hairy vetch. J Am Vet Med Assoc. 1983;183:569.

348 Claughton WP, Claughton HD. Vetch seed poisoning. Auburn Vet. 1954:125.

349 Cook WE, et al. Xanthoparmelia chlorochroa intoxication in wapiti (Cervus canadensis). Logan, Utah: International Symposium on Poisonous Plants, 2005. (abstract)

350 Galey FD, et al. Blue-green algae (Microcystis aeruginosa) hepatotoxicosis in dairy cows. Am J Vet Res. 1986;9:1415.

351 Beasley VR, et al. Diagnostic and clinically important aspects of cyanobacterial (blue-green algae) toxicosis. J Vet Diagn Invest. 1989;1:359.

352 Ross C, et al. Toxin release in response to oxidative stress and programmed cell death in the cyanobacterium Microcystis aeruginosa. Aquat Toxicol. 2006;78:66.

353 Hooser SB, et al. Actin filament alterations in rat hepatocytes induced in vivo and in vitro by microcystin LR, a hepatotoxin from the blue-green algae Microcystis aeruginosa. Vet Pathol. 1991;28:259.

354 Hooser SB, et al. Toxicity of microcystin LR, a cyclic heptopeptide hepatotoxin from Microcystis aeruginosa to rats and mice. Vet Pathol. 1989;26:246.

355 Hooser SB. Fulminant hepatocyte apoptosis in vivo following microcystin-LR administration to rats. Toxicol Pathol. 2000;28:726.

356 Carmichael WW, Biggs DF, Gortiam DR. Toxicology and pharmacological action of Anabaena flos-aquae toxin. Science. 1975;187:542.

357 Cook WO, et al. Comparison of effects of anatoxin a(s) and paraoxon, physostigmine, and pyridostigmine on mouse brain cholinesterase activity. Toxicon. 1988;26:750.

358 Puschner B, et al. Blue-green algae toxicosis in cattle. J Am Vet Med Assoc. 1998;213:1605.

359 Orr PT, et al. Exposure of beef cattle to sub-clinical doses of Microcystis aeruginosa: toxin bioaccumulation, physiological effects and human health risk assessment. Toxicon. 2003;41:613.

360 Cheeke PR, Shull LR. Natural toxicants in feeds and poisonous plants. Westport, Conn: AVI, 1985;393.

361 Osweiler GD, et al. Clinical and diagnostic veterinary toxicology. Dubuque, Iowa: Kendall/Hunt, 1985;409.

362 Casteel SW, et al. Liver disease in cattle induced by consumption of moldy hay. Vet Hum Toxicol. 1995;37:248.

363 Cheeke PR, Shull LR. Natural toxicants in feeds and poisonous plants. Westport, Conn: AVI, 1985;402.

364 Hsieh DPH, Atkinson DN. Bisfuranoid mycotoxins: their genotoxicity and carcinogenicity. In: Witmer CM, editor. Biological reactive intermediates IV. New York: Plenum; 1990:525.

365 Klein PJ, et al. Biochemical basis for the extreme sensitivity of turkeys to aflatoxin B-1. Toxicol Appl Pharmacol. 2000;165:45.

366 Angsubhakorn S, et al. Aflatoxicosis in horses. J Am Vet Med Assoc. 1981;178:274.

367 Connaughton D. The threat of aflatoxicosis. J Am Vet Med Assoc. 1989;194:743.

368 Coppock RW, et al. Acute aflatoxicosis in feeder pigs, resulting from improper storage of corn. J Am Vet Med Assoc. 1989;195:1380.

369 Osweiler GD, Trample DW. Aflatoxicosis in feedlot cattle. J Am Vet Med Assoc. 1985;187:636.

370 Ray AC, et al. Bovine abortion and death associated with consumption of aflatoxin-contaminated peanuts. J Am Vet Med Assoc. 1986;188:1187.

371 Colvin BM, et al. Aflatoxicosis in feeder cattle. J Am Vet Med Assoc. 1984;184:956.

372 Panangala VS, et al. Effects of aflatoxin on the growth performance and immune responses of weanling swine. Am J Vet Res. 1986;47:2062.

373 Cook WO, et al. Clinical and pathologic changes in acute bovine aflatoxicosis: rumen motility and tissue and fluid concentrations of aflatoxins B1 and M1. Am J Vet Res. 1986;47:1817.

374 Agnes VF, Akbarsha MA. Spermatotoxic effect of aflatoxin B-1 in the albino mouse. Food Chem Toxicol. 2003;41:119.

375 Van Vleet TR, et al. Metabolism and cytotoxicity of aflatoxin B-1 in cytochrome P-450-expressing human lung cells. J Toxicol Environ Health A. 2002;65:853.

376 Helferich WG, et al. Feedlot performances and tissue residues of cattle consuming diets containing aflatoxins. J Anim Sci. 1986;62:691.

377 Black RW, et al. Distribution of aflatoxicosis in tissues of growing pigs fed an aflatoxin-contaminated diet amended with a high-affinity aluminosilicate sorbent. Vet Hum Toxicol. 1990;32:16.

378 Harvey RB, et al. Prevention of aflatoxicosis by addition of hydrated sodium aluminosilicates to the diets of growing barrows. Am J Vet Res. 1989;50:416.

379 Harvey RB, et al. Effects on aflatoxin M1 residues in milk by addition of hydrated sodium calcium aluminosilicate to aflatoxin-contaminated diets of dairy cows. Am J Vet Res. 1991;52:1556.

380 Phillips TD. Dietary clay in the chemoprevention of aflatoxin-induced disease. Toxicol Sci. 1999;52(2 suppl):118.

381 Afriyie-Gyawu E, et al. Chronic toxicological evaluation of dietary NovaSil clay in Sprague-Dawley rats. Food Add Contam. 2005;22:259.

382 Cheeke PR. Natural toxicants in feeds, forages, and poisonous plants, ed 2, Danville, Ill: Interstate; 1998:116.

383 Cote LM, et al. Survey of vomitoxin-contaminated feed grains in midwestern United States, and associated health problems in swine. J Am Vet Med Assoc. 1984;184:189.

384 Pang VF, et al. Myocardial and pancreatic lesions induced by T-2 toxin, a trichothecene mycotoxin in swine. Vet Pathol. 1986;23:310.

385 Trenholm HL, et al. Feeding trials with vomitoxin (deoxynivalenol)–contaminated wheat: effects on swine, poultry, and dairy cattle. J Am Vet Med Assoc. 1984;185:527.

386 Pollmann DS, et al. Deoxynivalenol-contaminated wheat in swine diet. J Anim Sci. 1985;60:239.

387 Lun AK, Young LG, Lumsden JH. The effects of vomitoxin and feed intake on the performance and blood characteristics of young pigs. J Anim Sci. 1985;61:1178.

388 Gentry PA, Ross ML. Effect of T-2 toxin on bovine hematological and serum enzyme parameters. Vet Hum Toxicol. 1984;26:24.

389 Poppenga RH, et al. Assessment of a general therapeutic protocol for the treatment of acute T-2 toxicosis in swine. Vet Hum Toxicol. 1987;29:237.

390 Coppock RW, et al. Tissue residues of diacetoxy scirpenol in pigs and calves after intravenous dosing. Am J Vet Res. 1988;49:1997.

391 Beasley VR, et al. Pharmacokinetics of the trichothecene mycotoxin, T-2 toxin, in swine and cattle. Toxicon. 1986;24:13.

392 Marasas WFO, et al. Leukoencephalomalacia in a horse induced by fumonisin B1 isolated from Fusarium moniliforme. Onderstepoort J Vet Res. 1988;55:197.

393 Thiel PG, et al. Levels of fumonisins B1 and B2 in feeds associated with confirmed cases of equine leukoencephalomalacia. J Agric Food Chem. 1991;39:109.

394 Wang E, et al. Inhibition of sphingolipid biosynthesis by fumonisins. J Biol Chem. 1991;266:14486.

395 Sharma N, et al. Tumor necrosis factor alpha—mediated activation of c-Jun HN2-terminal kinase as a mechanism for fumonisin B-1–induced apoptosis in murine primary hepatocytes. J Biochem Mol Toxicol. 2005;19:359.

396 Stockmann-Juvala H, et al. Fumonisin B-1–induced toxicity and oxidative damage in U-118MG glioblastoma cells. Toxicology. 2004;202:173.

397 Sadler TW, et al. Prevention of fumonisin B1–induced neural tube defects by folic acid. Teratology. 2002;66:169.

398 Oswald IP, et al. Mycotoxin fumonisin B-1increases intestinal colonization by pathogenic Escherichia coli in pigs. Appl Environ Microbiol. 2003;69:5870.

399 Bermudez AJ, Ledoux DR, Rottinghaus GE. Effects of Fusarium moniliforme culture material containing known levels of fumonisin B1 in ducklings. Avian Dis. 1995;39:879.

400 Osweiler GD, et al. Effects of fumonisin-contaminated corn screenings on growth and health of feeder calves. J Anim Sci. 1993;71:459.

401 Ross PF, et al. Experimental equine leukoencephalomalacia, toxic hepatosis, and encephalopathy caused by corn naturally contaminated with fumonisins. J Vet Diagn Invest. 1993;5:69.

402 Wilson TM, et al. Fumonisin B1 levels associated with an epizootic of equine leukoencephalomalacia. J Vet Diagn Invest. 1990;2:213.

403 Colvin BM, Cooley AJ, Beaver RW: Fumonisin toxicosis in swine: clinical and pathologic findings. J Vet Diagn Invest. 1993;5:232.

404 Casteel SW, et al. Chronic toxicity of fumonisin in weanling pigs. J Vet Diagn Invest. 1993;5:413.

405 Turner PC, et al. Fumonisin contamination of food: progress in development of biomarkers to better assess human health risks. Mutat Res Genet Toxicol Environ Mutagen. 1999;443:81.

406 Marasas WFO, et al. Primary liver cancer and oesophageal basal cell hyperplasia in rats caused by Fusarium moniliforme. Int J Cancer. 1989;34:383.

407 Prelusky DB, et al. Biological fate of fumonisin B1 in food-producing animals. In: Jackson, et al. Fumonisins in food. New York: Plenum; 1996:265.

408 Meyer K, et al. Residue formation of fumonisin B1 in porcine tissues. Food Add Contam. 2003;20:639.

409 Galey FD, et al. Staggers induced by consumption of perennial ryegrass in cattle and sheep from Northern California. J Am Vet Med Assoc. 1991;199:466.

410 Valdes JJ, Cameron JE, Cole RJ. Aflatrem: a tremorgenic mycotoxin with acute neurotoxic effects. Environ Health Perspect. 1985;62:459.

411 Porter JK, Bacon CW, Robbins JD. Major alkaloids of a Claviceps isolated from toxic Bermuda grass. J Agric Food Chem. 1974;22:838.

412 Cole RJ, et al. Paspalum staggers: isolation and identification of tremorgenic metabolites from sclerotia of Claviceps paspali. J Agric Food Chem. 1977;25:1197.

413 Gallagher RT, White EP, Mortimer PH. Ryegrass staggers: isolation of potent neurotoxins lolitrem A and lolitrem B for staggers-producing pastures. NZ Vet J. 1981;29:189.

414 Mantle PG. Amino acid neurotransmitter release from cerebrocortical synaptosomes of sheep with severe ryegrass staggers in New Zealand. Res Vet Sci. 1983;34:373.

415 Cheeke PR. Natural toxicants in feeds, forages, and poisonous plants, ed 2, Danville, Ill: Interstate; 1998:261.

416 Mortimer PH. Perennial ryegrass staggers in New Zealand. In: Keeler RF, et al, editors. Effects of poisonous plants in livestock. New York: Academic; 1978:353.

417 Hunt LD, Blythe L, Holtan DW. Ryegrass staggers in ponies fed processed ryegrass straw. J Am Vet Med Assoc. 1983;182:285-286.

418 Gallagher RT, et al. Ryegrass staggers: the presence of lolitrem neurotoxins in perennial ryegrass seed. NZ Vet J. 1982;30:183.

419 Smith BL. Mycotoxicoses of sheep in Australia and New Zealand. Proceedings of University of Sydney Post-Graduate Committee in Veterinary Series 103, Veterinary Annual Toxicology, Sydney. 1987:279.

420 Munday BL, Mason RW. Lesions in ryegrass staggers in sheep. Aust Vet J. 1967;43:598.

421 Mason RW. Axis cylinder degeneration associated with ryegrass staggers in sheep and cattle. Aust Vet J. 1968;44:428.

422 Gallagher RT, Hawkes AD. Estimation of neurotoxin levels in perennial ryegrass by mouse bioassay. NZ J Agric Res. 1985;28:427.

423 Jackson JAJr, et al. Lolium alkaloids in tall fescue hay and seed and their relationship to summer fescue toxicosis in cattle. J Dairy Sci. 1984;67:104.

424 Martin T, Edwards WC. Protecting grazing livestock from tall fescue toxicity. Vet Med. 1986;81:1162.

425 Easton HS, Lane GA, Tapper BA. Ergovaline in endophyte-infected ryegrass pastures. NZ Vet J. 1993;41:214.

426 Schilto KK JA, et al. Effects of endophyte-infected fescue on concentrations of prolactin in blood sera and the anterior pituitary and concentration of dopamine metabolism in brains of steers. J Anim Sci. 1988;66:713.

427 McCann JS, et al. Influence of endophyte-infected tall fescue on serum prolactin and progesterone in gravid mares. J Anim Sci. 1992;70:217.

428 Dennis SB, et al. Influence of Neotyphodium coenophialum on copper accumulation in tall fescue. J Anim Sci. 1998;76:2687.

429 Garner GB, Cornell CB. Cattle response to the endophyte of tall fescue. Am Assoc Vet Lab Diagn. 1985;26:145.

430 Carr SB, Jacobson DR. Bovine physiological responses to toxic fescue and related conditions for application in a bioassay. J Dairy Sci. 1969;52:1792.

431 Jacobson DR, et al. Nature of fescue toxicity and progress toward identification of the toxic entity. J Dairy Sci. 1963;46:416.

432 Browning RJr. Physiological responses of Brahman and Hereford steers to an acute ergotamine challenge. J Anim Sci. 2000;78:124.

433 Browning RJr, et al. Effect of ergotamine and ergonovine on plasma concentrations of thyroid hormones and cortisol in cattle. J Anim Sci. 1998;76:1644.

434 Oliver JW, et al. Endophytic fungal toxin effect on adrenergic receptors in lateral saphenous veins (cranial branch) of cattle grazing tall fescue. J Anim Sci. 1998;76:2853.

435 Cunningham IJ. A note on the cause of tall fescue lameness in cattle. Aust Vet J. 1949;25:25.

436 Garner GB, et al. “Fescue foot” induction from experimental pastures. J Anim Sci. 1972;35:228.

437 Williams M, et al. Induction of fescue foot syndrome in cattle by fractionated extracts of toxic fescue hay. Am J Vet Res. 1975;36:1353.

438 Poppenga RH, et al. Mare agalactia, placental thickening, and high foal mortality associated with the grazing of tall fescue: a case report. Assoc Vet Lab Diagn. 1984;25:325.

439 Putnam MR, et al. Effects of the fungal endophyte Acremonium coenophialum in fescue on pregnant mares and foal viability. Am J Vet Res. 1991;52:2071.

440 Burke JM, et al. Interaction of endophyte-infected fescue and heat stress on ovarian function in the beef heifer. Biol Reprod. 2001;65:260.

441 Schuenemann GM, et al. Effects of administration of ergotamine tartrate on fertility of yearling beef bulls. Theriogenology. 2005;63:1407.

442 Burke JM, et al. Reproductive responses of ram lambs under short-term exposure to endophyte-infected tall fescue seed. Small Ruminant Res. 2006;66:121.

443 Nihsen ME, et al. Growth rate and physiology of steers grazing tall fescue inoculated with novel endophytes. J Anim Sci. 2004;82:878.

444 Watson RH, et al. Productivity of cow-calf pairs grazing tall fescue pastures infected with either the wild-type endophyte or a nonergot alkaloid-producing endophyte strain AR542. J Anim Sci. 2004;82:3388.

445 Rice RL, et al. Oral and parenteral vaccination of mice with protein ergotamine conjugates and evaluation of protection against fescue toxicosis. Vet Immunol Immunopathol. 1998;61:305.

446 Cross DL, Redmond LM, Strickland JR. Equine fescue toxicosis: signs and solutions. J Anim Sci. 1995;73:899.

447 Cheeke PR. Natural toxicants in feeds, forages, and poisonous plants, ed 2, Danville, Ill: Interstate; 1998:88.

448 Blaney BJ, et al. Sorghum ergot (Claviceps africana) associated with agalactia and feed refusal in pigs and dairy cattle. Aust Vet J. 2000;78:102.

449 Cheeke PR, Shull LR. Natural toxicants in feeds and poisonous plants. Westport, Conn: AVI, 1985;422.

450 Sundlof SF, Strickland C. Zearalenone and zeranol: potential residue problems in livestock. Vet Hum Toxicol. 1986;28:242.

451 Malekinejad H, et al. Species differences in the hepatic biotransformation of zearalenone. Vet J. 2006;172:96.

452 Crump MH. Slaframine (slobber factor) toxicosis. J Am Vet Med Assoc. 1973;163:1300.

453 Hagler WM, Benlow RF. Salivary syndrome in horses: identification of slaframine in red clover hay. Appl Environ Microbiol. 1981;42:1067.

454 Fowler ME. Veterinary zootoxicology. Boca Raton, Fla: CRC Press, 1993.

455 Iyaniwura TT. Snake venom constituents: biochemistry and toxicology. I. Vet Hum Toxicol. 1991;33:468.

456 Iyaniwura TT. Snake venom constituents: biochemistry and toxicology. II. Vet Hum Toxicol. 1991;33:475.

457 Russell FE. Snake venom poisoning. Vet Hum Toxicol. 1991;33:584.

458 Moss ST, et al. Association of rattlesnake bite location with severity of clinical manifestations. Ann Emerg Med. 1997;30:58.

459 Walton RM, et al. Mechanisms of echinocytosis induced by Crotalus atrox venom. Vet Pathol. 1997;34:442.

460 Kerrigan KR, et al. Antibiotic prophylaxis for pit viper envenomation: prospective, controlled trial. World J Surg. 1997;21:369.

461 Clark RF, et al. Successful treatment of crotalid-induced neurotoxicity with a new polyspecific crotalid Fab antivenom. Ann Emerg Med. 1997;30:54.

462 Ray AC, et al. Etiologic agents, incidence, and improved diagnostic methods of cantharidin toxicosis in horses. Am J Vet Res. 1989;50:187.

463 Hutchison WD, Murphy MJ, Tufte GN. Blister beetles in alfalfa: management options to minimize poisoning in horses. Minn Ext Serv. AGF05510-D, 1990.

464 Schmitz DG. Cantharidin toxicosis update. Clabough D, Marold A, Lyons R, editors. Equine Vet Data. 1988;9:87.

465 Zhu YC, et al. Factors influencing quantity of cantharidin transferred to alfalfa from Epicauta occidentalis Werner (Coleoptera: Meloidae). J Econ Entomol. 1997;90:1665.

466 Helman RG, Edwards WC. Clinical features of blister beetle poisoning in equids: 70 cases (1983-1996). J Am Vet Med Assoc. 1997;211:1018.

467 Morgan SE, Morgan GL, Edwards WC. Pinpointing the source of arsenic poisoning in a herd of cattle. Vet Med. 1984;79:1525.

468 Thatcher CD, et al. Arsenic toxicosis and suspected chromium toxicosis in a herd of cattle. J Am Vet Med Assoc. 1985;187:179.

469 Bahri LE, Romdane SB. Arsenic poisoning in livestock. Vet Hum Toxicol. 1991;33:259.

470 Boosinger TR, Riviere JE, Everson RJ. Arsenic-induced hemorrhagic enterocolitis in cattle. Proc Am Assoc Vet Lab Diagn. 1980:397.

471 Kahrs RF, et al. Fatal lead arsenate toxicosis resembling acute bovine viral diarrhea-mucosal disease. Proc Am Assoc Vet Lab Diagn. 1979:321.

472 Riviere JE, et al. Environmental arsenic toxicosis in cattle. Proc Am Assoc Vet Lab Diagn. 1984:355.

473 Galey FD. Arsenic toxicosis. In: Howard JL, editor. Current veterinary therapy 3 (food animal practice). Philadelphia: Saunders; 1993:394.

474 Stair EL, Kirkpatrick JG, Whitenack DL. Lead arsenate poisoning in a herd of beef cattle. J Am Vet Med Assoc. 1995;207:341.

475 Galey FD, et al. Copper toxicosis in two herds of beef calves following injection with copper disodium edetate. J Vet Diagn Invest. 1991;3:260.

476 Bohman VR, et al. The toxicology and composition of bovine tissues after parenteral administration of high levels of copper salts. Vet Hum Toxicol. 1987;29:307.

477 Bulgin MS, et al. Death associated with parenteral administration of copper disodium edetate in calves. J Am Vet Med Assoc. 1986;188:406.

478 Vaala WE. Copper toxicity in sheep. Proc Am Coll Vet Intern Med. 1991:559.

479 Gough J. Copper poisoning in lambs fed horse and cattle feed. Can Vet J. 1991;32:750.

480 Junge RE, Thornburg L. Copper poisoning in four llamas. J Am Vet Med Assoc. 1989;195:987.

481 Perrin DJ, Schiefer HB, Blakley BR. Chronic copper toxicity in a dairy herd. Can Vet J. 1990;31:629.

482 Parada R, Gonzalez S, Bergqvist E. Industrial pollution with copper and other heavy metals in a beef cattle ranch. Vet Hum Toxicol. 1987;29:122.

483 Gummow, et al. Copper toxicity in ruminants: air pollution as a possible cause. Onderstepoort J Vet Res. 1991;58:33.

484 Banton MI, et al. Copper toxicosis in cattle fed chicken litter. J Am Vet Med Assoc. 1987;191:827.

485 Kerr LA, McGavin HD. Chronic copper poisoning in sheep grazing pastures fertilized with swine manure. J Am Vet Med Assoc. 1991;198:99.

486 Sullivan JM, Janovitz EB, Robinson FR. Copper toxicosis in veal calves. J Vet Diagn Invest. 1991;3:161.

487 Jenkins KJ, Hidiroglou M. Tolerance of the calf for excess copper in milk replacer. J Dairy Sci. 1989;72:150.

488 Humphries WR, Morrice PC, Mitchell AN. Copper poisoning in Angora goats. Vet Rec. 1987;121:231.

489 Tremblay RRM, Baird JD. Chronic copper poisoning in two Holstein cows. Cornell Vet. 1991;81:205.

490 Humphries WR, Morrice PC, Bremner I. A convenient method for the treatment of chronic copper poisoning in sheep using subcutaneous ammonium tetrathiomolybdate. Vet Rec. 1988;123:51.

491 Symonds HW, Ke Y. Enhancement of tetrathiomolybdate-induced copper excretion in bile of sheep by the α2-agonistic action of xylazine. Res Vet Sci. 1989;46:349.

492 Botha CJ, et al. The cupruretic effect of two chelators following copper loading in sheep. Vet Hum Toxicol. 1993;35:409.

493 Araya O, et al. Bovine fluorosis following volcanic activity in the southern Andes. Vet Rec. 1990;126:641.

494 Araya O, Wittwer F, Villa A. Evolution of fluoride concentrations in cattle and grass following a volcanic eruption. Vet Hum Toxicol. 1993;35:437.

495 Shupe JL, et al. Relationship of cheek tooth abrasion to fluoride-induced permanent incisor lesions in livestock. Am J Vet Res. 1987;48:1498.

496 Shupe JL, Olsen AE. Clinicopathologic aspects of chronic fluoride toxicosis in cattle. Bovine Pract. 1987;22:184.

497 Olson WG, et al. Iodine toxicosis in six herds of dairy cattle. J Am Vet Med Assoc. 1984;184:179.

498 House JK, et al. Hemochromatosis in Salers cattle. J Vet Intern Med. 1994;8:105.

499 Norrdin RW, Hoopes KJ, O’Toole D. Skeletal changes in hemochromatosis of Salers cattle. Vet Pathol. 2004;41:612.

500 Edens LM, Robertson JL, Feldman BF. Cholestatic hepatopathy, thrombocytopenia and lymphopenia associated with iron toxicity in a thoroughbred gelding. Equine Vet J. 1993;25:81.

501 Divers TJ, et al. Toxic hepatic failure in newborn foals. J Am Vet Med Assoc. 1983;183:1407.

502 Ruhr LP, et al. Acute intoxication from a hematinic in calves. J Am Vet Med Assoc. 1983;182:616.

503 Holter JA, Carson TL, Witte ST. Acute iron intoxication in a herd of young bulls. J Vet Diagn Invest. 1990;2:229.

504 Mullaney TP, Brown CM. Iron toxicity in neonatal foals. Equine Vet J. 1988;20:119.

505 Markel MD, Dyer RM, Hattel AL. Acute renal failure associated with application of a mercuric blister. J Am Vet Med Assoc. 1984;185:92.

506 Smith R, Morden BB, Ellis LB. Diphenylmercury poisoning in cattle. Can Vet J. 1992;33:135.

507 Short SB, Edwards WC. Are your patients safe from unnecessary mercury poisoning. Vet Med. 1988;83:287.

508 Schuh JCL, Ross C, Meschter C. Concurrent mercuric blister and dimethyl sulphoxide (DMSO) application as a cause of mercury toxicity in two horses. Equine Vet J. 1988;20:68.

509 Kerr LA, et al. Aldicarb toxicosis in a dairy herd. J Am Vet Med Assoc. 1991;198:1636.

510 Dorman DC, et al. Aldicarb toxicosis in a flock of sheep. J Vet Diagn Invest. 1992;4:45.

511 Osheim DL, et al. Carbofuran toxicosis in cattle: case history and analytical method. Vet Hum Toxicol. 1985;27:306.

512 Harlin KS, Hamdy S, Beasley VR. Preliminary studies with bovine retina cholinesterase determinations in organophosphorus insecticide poisoning. J Vet Diagn Invest. 1989;1:356.

513 Meerdink GL. Organophosphorus and carbamate insecticide poisoning in large animals. Vet Clin North Am Food Anim Pract. 1989;5:375.

514 Boermans HJ, et al. Terbufos poisoning in a dairy herd. Can Vet J. 1984;25:335.

515 Plumlee KH, et al. Effect of time and storage temperature on cholinesterase activity in blood from normal and organophosphorus insecticide-treated horses. J Vet Diagn Invest. 1994;6:247.

516 Mosha RD, Gyrd-Hansen N. Toxicity of ethion in goats. Vet Hum Toxicol. 1990;32:6.

517 Scarratt WK. Chlorpyrifos intoxication in a beef bull. Compend Cont Educ (Pract Vet). 1985;7:S719.

518 Scarratt WK, Blodgett DJ. Chlorpyrifos intoxication in a bull. J Am Vet Med Assoc. 1986;188:1444.

519 Cook WO, Carson TL. Fonofos toxicosis and milk residues in dairy cattle. Vet Hum Toxicol. 1985;28:281.

520 Frank R, et al. A review of insecticide poisonings among domestic livestock in southern Ontario, Canada, 1982-1989. Can Vet J. 1991;32:219.

521 Haas PJ, et al. Effect of chlorpyrifos on Holstein steers and testosterone-treated Holstein bulls. Am J Vet Res. 1983;44:879.

522 Baker NF, et al. Neurotoxicity of haloxon and its relationship to blood esterases of sheep. Am J Vet Res. 1970;31:865.

523 Williams JF, Dade AW. Posterior paralysis associated with anthelmintic treatment of sheep. J Am Vet Med Assoc. 1976;169:1307.

524 Rose RJ, Hartley WJ. Laryngeal paralysis in Arabian foals associated with oral haloxon administration. Equine Vet J. 1981;13:171.

525 Raisbeck MF, Kendall JD, Rottinghaus GE. Organochlorine insecticide problems in livestock. Vet Clin North Am Food Anim Pract. 1989;5:391.

526 Casteel SW, et al. Aldrin intoxication and clearance of associated dieldrin residues in a group of feedlot cattle. J Am Vet Med Assoc. 1993;202:83.

527 Beck B, Groom S. Lindane toxicity in beef cattle. Can Vet J. 1989;30:833.

528 Raisbeck MF, et al. Heptachlor contamination of dairy, beef, and swine: the experience in Missouri. Am Assoc Vet Lab Diagn. 1986;29:161.

529 Smith EA, Oehme FW. A review of selected herbicides and their toxicities. Vet Hum Toxicol. 1991;33:596.

530 Hovda LR. Herbicide toxicity in large animals. Proceedings of the Twelfth ACVIM Forum, San Francisco. 1994.

531 Onyeama HP, Oehme FW. A literature review of paraquat toxicity. Vet Hum Toxicol. 1984;26:494.

532 Osweiler GD, et al. Clinical and diagnostic veterinary toxicology. Dubuque, Iowa: Kendall/Hunt, 1985;253.

533 Arnold EK, Beasley VR. The pharmacokinetics of chlorinated phenoxy acid herbicides: a literature review. Vet Hum Toxicol. 1989;31:121.

534 Duke SO. Overview of herbicide mechanisms of action. Environ Health Perspect. 1990;87:263.

535 Allender WJ, Glastonbury JW. Simazine toxicosis in sheep. Vet Hum Toxicol. 1992;34:422.

536 Kobel W, et al. Protective effect of activated charcoal in cattle poisoned with atrazine. Vet Hum Toxicol. 1985;27:185.

537 Byars TD, Greene CE, Kemp DT. Antidotal effect of vitamin K1 against warfarin-induced anticoagulation in horses. Am J Vet Res. 1986;47:2309.

538 Thijssen HHW, et al. Warfarin pharmacokinetics in the horse. Am J Vet Res. 1983;44:1192.

539 Dorman DC. Anticoagulant, cholecalciferol, and bromethalin-based rodenticides. Vet Clin North Am Small Anim Pract. 1990;20:339.

540 Mount ME. Diagnosis and therapy of anticoagulant rodenticide intoxications. Vet Clin North Am Small Anim Pract. 1988;18:115.

541 Mount ME, Feldman BF, Buffington T. Vitamin K and its therapeutic importance. J Am Vet Med Assoc. 1982;180:1354.

542 Vrins A, Carlson G, Feldman B. Warfarin: a review with emphasis on its use in the horse. Can Vet J. 1983;24:211.

543 Scott EA, Sandler GA, Byars TD. Warfarin: effects on anticoagulant, hematologic, and blood enzyme values in normal ponies. Am J Vet Res. 1979;40:142.

544 Morgan S, et al. Investigating a case of strychnine poisoning. Vet Med. 1987;82:1044.

545 Guale FG, et al. Laboratory diagnosis of zinc phosphide poisoning. Vet Hum Toxicol. 1994;36:517.

546 Von Burg R, Stout T. Metaldehyde. J Appl Toxicol. 1991;11:377.

547 Booze TF, Oehme FW. Metaldehyde toxicity: a review. Vet Hum Toxicol. 1985;27:11.

548 Harris WF. Metaldehyde poisoning in three horses. Mod Vet Pract. 1975;56:336.

549 Sutherland C. Metaldehyde poisoning in horses. Vet Rec. 1983;112:64.

550 Simmons JR, Scott WA. An outbreak of metaldehyde poisoning in sheep. Vet Rec. 1974;95:211.

551 Williams BM, Thomas AI. Metaldehyde poisoning in cattle. Vet Rec. 1976;98:358.

552 Longbottom GM, Gordon ASM. Metaldehyde poisoning in a dairy herd. Vet Rec. 1979;104:454.

553 Stubbings DP, et al. Three cases of metaldehyde poisoning in cattle. Vet Rec. 1976;98:356.

554 Miller RM. Metaldehyde poisoning in horses. Vet Med. 1972;67:1141.

555 Edwards WC. Toxicology of oil field wastes. Vet Clin North Am Food Anim Pract. 1989;5:363.

556 Boermans HJ, Ruegg PL, Leach M. Ethylene glycol toxicosis in a pygmy goat. J Am Vet Med Assoc. 1988;193:694.

557 Hewlett TP, et al. Ethylene glycol and glycolate kinetics in rats and dogs. Vet Hum Toxicol. 1989;31:116.

558 Panciera RJ, et al. Bovine hyperkeratosis: historical review and report of an outbreak. Compend Cont Educ (Pract Vet). 1993;15:1287.

559 Exon JH. A review of chlorinated phenols. Vet Hum Toxicol. 1984;26:508.

560 Osweiler GD, Van Gelder GA, Zumwalt RW. Toxicologic and residue aspects of pentachlorophenol (PCP). Proc Am Assoc Vet Lab Diagn. 1977;20:159.

561 Kerkvliet NI, et al. Dioxin intoxication from chronic exposure of horses to pentachlorophenol-contaminated wood shavings. J Am Vet Med Assoc. 1992;201:296.

562 East NE. Accidental superphosphate fertilizer poisoning in pregnant ewes. J Am Vet Med Assoc. 1993;203:1176.

563 Sisk DB, Colvin BM, Bridges CR. Acute, fatal illness in cattle exposed to boron fertilizer. J Am Vet Med Assoc. 1988;193:943.

564 Sisk DB, et al. Experimental acute inorganic boron toxicosis in the goat: effects on serum chemistry and CSF biogenic amines. Vet Hum Toxicol. 1990;32:205.

565 Casper HH, et al. Evaluation of vitamin K3 feed additive for prevention of sweet clover disease. J Vet Diagn Invest. 1989;1:116.

566 Maxie G, et al. Menadione toxicity in six horses. Can Vet J. 1992;33:756.

567 Schmitz DG. Toxic nephropathy in horses. Compend Cont Educ (Pract Vet). 1988;10:104.

568 Rebhun WC, et al. Vitamin K3–induced renal toxicosis in the horse. J Am Vet Med Assoc. 1984;184:1237.

569 Dorman DC, Haschek WM. Fatal propylene glycol toxicosis in a horse. J Am Vet Med Assoc. 1991;198:1643.

570 Pintchuk PA, Galey FD, George LW. Propylene glycol toxicity in adult dairy cows. J Vet Intern Med. 1993;7:150.

571 Somerville BA, Plumlee KH. Acute isopropyl alcohol intoxication in a horse. Can Vet J. 1996;37:359.

572 Glenn MW, Burr WM. Toxicity of a piperazine—carbon disulfide—phenothiazine preparation in the horse. J Am Vet Med Assoc. 1972;160:988.

573 Blevins DI, Miller CC, Kleckner MD. Effects of a piperazine—carbon disulfide—phenothiazine preparation on hemoglobin and packed cell values of horses. J Am Vet Med Assoc. 1971;159:1260.

574 Rowe LD. Photosensitization problems in livestock. Vet Clin North Am Food Anim Pract. 1989;5:301.

575 Casteel SW, et al. Photosensitization: an investigation and review of the problem in cattle of south Texas. Vet Hum Toxicol. 1986;28:251.

576 McSherry BJ, Roe CK, Milne FJ. The hematology of phenothiazine poisoning in horses. Can Vet J. 1966;7:3.

577 Haliburton JC, Morgan SE. Nonprotein nitrogen—induced ammonia toxicosis and ammoniated feed toxicity syndrome. Vet Clin North Am Food Anim Pract. 1989;5:237.

578 Caldow GL, Wain EB. Urea poisoning in suckler cows. Vet Rec. 1991;128:489.

579 Casteel SW, Cook WO. Urea toxicosis in cattle: a dangerous and avoidable dietary problem. Vet Med. 1984;79:1523.

580 Henderson J, Dempsey DD. Toxicosis caused by ammoniated hay in calves. Can Vet J. 1991;32:180.

581 Kerr LA, Groce AW, Kersting KW. Ammoniated forage toxicosis in calves. J Am Vet Med Assoc. 1987;191:551.

582 Morgan SE, Edwards WC. Pilot studies in cattle and mice to determine the presence of 4-methylimidazole in milk after oral ingestion. Vet Hum Toxicol. 1986;28:240.

583 Morgan SE, Edwards WC. Bovine bonkers: new terminology for an old problem. Vet Hum Toxicol. 1986;28:16.

584 Anderson TD, et al. Acute monensin toxicosis in sheep: light and electron microscopic changes. Am J Vet Res. 1984;45:1142.

585 Blanchard PC, et al. Lasalocid toxicosis in dairy calves. J Vet Diagn Invest. 1993;5:300.

586 Rollinson J, Taylor FGR, Chesney J. Salinomycin poisoning in horses. Vet Rec. 1987;121:126.

587 Novilla MN. The veterinary importance of the toxic syndrome induced by ionophores. Vet Hum Toxicol. 1992;34:66.

588 Muylle E, et al. Delayed monensin sodium toxicity in horses. Equine Vet J. 1981;13:107.

589 Fourie N, et al. Cardiomyopathy of ruminants induced by the litter of poultry fed on rations containing the ionophore antibiotic, maduramicin. Onderstepoort J Vet Res. 1991;58:291.

590 Amend JF, et al. Equine monensin toxicosis: useful ante-mortem and post-mortem clinicopathologic tests. Proc Am Assoc Equine Pract. 1986;32:361.

591 Doonan GR, et al. Monensin poisoning in horses: an international incident. Can Vet J. 1989;30:165.

592 Schweitzer D, et al. Accidental monensin sodium intoxication of feedlot cattle. J Am Vet Med Assoc. 1984;184:1273.