• Shuffle
    Toggle On
    Toggle Off
  • Alphabetize
    Toggle On
    Toggle Off
  • Front First
    Toggle On
    Toggle Off
  • Both Sides
    Toggle On
    Toggle Off
  • Read
    Toggle On
    Toggle Off
Reading...
Front

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/690

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

690 Cards in this Set

  • Front
  • Back
Opiate
-Drugs derived from opium
-Natural compounds
-Morphine
-Codeine
-Thebaine
Opioid
-All drugs with morphine-like properties
-Natural or synthetic compounds
-Produce analgesia without loss of touch, proprioception, or consciousness
Endogenous opioid peptides
-Naturally occurring ligands for opioid receptors
Narcotics
-Associated with opoioids
-Substances with abusive or addictive potential
Endogenous Opioid Peptides
-Act on all 3 opioid receptors
-Work all of the time, even during periods of stress
-Peptides synthesized from precursor molecules
-Each precursor has characteristic anatomic distribution
-Naturally produced in the CNS
--released from pituitary and adrenal glands
-ACTH and B-endorphin have common precursor
--allows for stress-induced analgesia
-Enkephalins, Dynorphins, Endorphins
-Nociceptin, Endomorphin 1 and 2 (recently discovered)
Endogenous Pain Suppressive System
-Each peptide group has variable affinities for opioid receptors
-No opioid peptide binds exclusively to one opioid receptor
-Endorphins inhibit release of NT from nerve terminals carrying nociceptive impulses
-Analgesia can be produced from electrical/mechanical stimulation of peripheral areas
--acupuncture
Phenanthrenes
-3 ring structure with amine group
--amine group allows metabolism in the liver
-Morphine
-Codeine
-Thebaine

Alkaloids of opium
Benzylisoquinolones
-Lack opioid activity
-Papaverine
-Noscapine
Semi-synthetic opioids
-Produced by making simple modifications to morphine molecule
Synthetic Opioids
-Morphinan series (levorphanol, butorphanol)
-Diphenylpropylamine series (methadone)
-Benzomorphan series (pentazocine)
-Phenylpiperidine series (meperidine, fentanyl)
-Contains phenanthrene nucleus of morphine
-Manufactured by synthesis, not chemical modification of morphine
Opioid Receptors
-Mu, Delta, Kappa
-Nociceptin/Orphanin FQ receptor
-All are G-protein coupled receptors
Mu opioid receptor
-Most important opioid receptor for analgesia
-Responsible for supraspinal and spinal analgesia
-Mu1: analgesia
-Mu2: bradycardia, hypoventilation, dependence
-Other sub-types have unclear mechanisms
-Endomorphins in brain have high affinity and selectivity for Mu receptors
Kappa Opioid receptor
-Provides mild analgesia
--not great for a painful surgery
-4 sub-types (K1a, K1b, K2, K3)
-Mediates analgesia in the CNS and PNS
-Opioid agonist-antagonists act on Kappa receptors
-Hard to distinguish Kappa analgesia from Mu analgesia
-High intensity pain may be resistant to analgesic effects of Kappa receptors
-Less respiratory depression vs. Mu receptors
--less chance of respiratory arrest
-Dynorphin acts mostly on Kappa receptors
-Agonists give inhibition of neurotransmitter release via Ca channels
--agonist binds to kappa receptor and decreases pain by inhibiting Ca channels that inhibits NT release
Delta Opioid Receptor
-Poor alangesic activity
-May modify Mu receptor-mediated anti-nociception
-Enkephalins act on delta receptors and modulate Mu receptor activity
-Involved in vomiting caused by analgesics?
N/OFq receptor
-Nociception receptor
-Inhibits pain facilitating and analgesia facilitating neurons in RVM
--Pain OR Analgesia
-Effects on pain response depend on pre-existing state of pain in the animal
-Can have pro-nociceptive effects
-Little to no affinity for conventional opioid ligands
-Precursor molecule is located in the dorsal and ventral horns of spinal cord, hippocampus, cortex, and numerous sensory sites
-Produces effects related to drug reward, reinforcement, stress responsiveness, feeding behavior, and memory process
-
Mu1 Receptor Stats
-Effects: Supraspinal and spinal analgesia, euphoria, miosis, bradycardia, hypothermia, urinary retention
-Low abuse potential
-Agonists: Endorphins, Morphine, Synthetic opioids
-Antagonists: Naloxone, Naltrexone, Nalmefene
Mu2 Receptor Stats
-Effects: Spinal Analgesia, Depression of ventilation, marked constipation
-Physical dependence
-Agonists: endorphins, morphine, synthetic opioids
-Antagonists: Naloxone, Naltrexone, Nalmefene
Kappa Receptor Stats
-Effect: Supraspinal and spinal analgesia, dysphoria, sedation, miosis, diuresis
-Low abuse potential
-Agonists: Dynorphins
-Antagonists: Naloxone, Naltrexone, Nalmefene
Delta Receptor Stats
-Effect: Supraspinal and spinal analgesia, depression of ventillation, minimal constipation, urinary retention
-Physical dependence
-Agonists: Enkephalins
-Antagonists: Naloxone, Naltrexone, Nalmefene
Mechanisms of Action for Opioid Receptors
-Opioid binds at stereospecific GPCRs
--can be Gi and/or Go
-Signal transduction mechanisms produce effect
--inhibits inward shift of K (hyperpolarizes cell)
--Inhibits adenylyl cyclase, decreases cAMP and inhibits Ca release
--Inhibits voltage-gated Ca channels, prevents Ca influx into cell
-Presynaptic and postsynaptic inhibition of NTs
--ACh, dopamine, NorEpi, Substance P, tachykinins all inhibited
-Main effect is to decrease neurotransmission via activation of opioid receptors
Pain pathways and Opioids
-Opioid receptors in periphery in supraspinal and spinal locations
-INhibit peripheral and central afferent nociceptive transmission
--Decrease pain transduction
--inhibits pathways up to brain AND pathways out of the brain
-Inhibits directly ascending transmission from spinal cord dorsal horn
-Activates pain control circuits that come down from brain via rostral ventral medulla to spinal cord dorsal horn
-Produce via direct action on spinal cord
-Produce neurally mediated action in region separate form site of administration
-Neuropathic pain responds poorly to opioids
--need higher doses
-Dosage, species, stimulus intensity, efficacy of opioid, character, and duration change analgesic effects
Opioid Agonist Receptor-Ligand Interaction
-Stimulates Mu, Kappa, and Delta receptors
-Full Mu agonists act on ALL receptors
--higher dose will increase effect
-Binds to both Mu and Kappa receptors
-Increased analgesic effect with increased dose
-Morphine, Oxymorphine, fentanyl
Partial Opioid Agonists Receptor-Ligand Interaction
-Binds to Mu receptors, not Kappa receptor
--only binds to Mu part of the receptor, not Kappa
-Produces only limited clinical effect
-Ex: buprenorphine at Mu receptor
-Prevents full agonist from fitting into the binding site, acts as partial agonist and partial antagonist
Opioid Agonist-Antagonists
-Competitive Mu receptor antagonists
-Analgesic effects from binding to Kappa receptor (agonist activity)
-Acts kind of like a partial agonist but still activates Kappa receptor
-Ex: butorphanol, buprenorphine, nalbuphine
Opioid Antagonists
-High affinity for opioid receptors
-Displace opioid agonists from Mu and Kappa receptors
-Just take up space and prevent agonist form binding
-Effect is based on concentration
-Ex: Naloxone
Neurophysiological Effects of Opioids
-Analgesic Action:
--analgesia, drowsiness, mental cloudiness, euphoria
--Pain-free individual may feel unpleasant
-Anesthetic action:
--Unconsciousness is unpredictable and inconsistent, may need propofol/fentanyl in combo

-Different species interact differently
--horses, cats, ruminants get excited by Mu full agonists
--dogs and primates get sedate with Mu full agonists
Specific Neurophysiological Effects of Opioids
1. Cerebral Metabolic Rate decreases
--neuroexcitation may cause regional increase
2. Cerebral Blood flow: depends on drugs being used, usually no influence or only small increase
3. Intracranial Pressure: generally minimal effect, depends on surgical procedure
4. Pupil Size: usually constriction by excitatory action (miosis)
--In cats cause mydriasis
5. Neuroexcitatory phenomena: can cause delirium or grand mal seizures

Safe for Neurologic Cases
Respiratory Effects of Opioids
1. Therapeutic Effects:
-Prevents hyperventilation from pain/anxiety
--decreases RR and turbulence
-Dyspnea
-Increases tolerance to endotracheal intubation
-Anti-tussive
2. Non-therapeutic Effects (side effects)
-Respiratory depression
--can be issue in brachycephalic dogs with stenotic ariways
-Decreases hypoxic ventilatory drive
-Decreases CO2 ventilatory drive, brain does not respond to increase [CO2]
-Anti-tussive
Respiratory Depression with Opioids
-Dose-dependent depression
--more drug has a bigger effect
-Mediated by Mu2 receptors
-Co-administration of other sedatives (benzodiazepines)
-Humans are more sensitive
-Decreased clearance
-reuptake from muscle, fat, lung, etc.
Cardiovascular Effects of Opioids
-Minimal cardiovascular effects, no major effect
-Can reduce sympathetic tone
-Can cause hypotension of patient is dependent on high sympathetic tone
--increases vagal tone
-Bradycardia due to medullary vagal stimulation
--Issue for neonates
-Histamine release leads to hypotension
--morphine and meperidine
Neonates and Opioids
-Cannot increase Stroke Volume
-Increasing Heart Rate is the only way for neonates to maintain cardiac output
-With opioids, neonates cannot compensate for decreased HR
Endocrine Effects of Opioids
-Inappropriate ADH response
-Inhibits pituitary-adrenal axis
-Endorphin and ACTH are co-secreted during stress
-Can give pre-emptively for decreasing stress response
Renal/Urologic Effects of Opioids
-Mu receptor activates anti-diuresis
-Kappa receptor activates diuresis
--inhibits ADH
-Urinary Retention
Opioids and GI motility
-Decrease GI motility, cause constipation and delayed gastric emptying
-Dogs will defecate initially
-Causes ileus and constipation
-Horses can colic
-Ruminants can end up with rumen tympani with full Mu agonist
-Mediated by Mu and Delta receptors
-Affects myenteric plexus of the GI tract
Opioids and Nausea and Vomiting
-Stimulate chemoreceptor trigger zone in area postrema of medulla
-Mediated by delta receptors
Pharmacokinetics of Opioids
-IV injection leads to plasma concentrations peaking
-Lung takes up a lot of opioids before it redistributes
--uptake by lung can be significant, leads to rapid release
-Rapid redistribution phase
-If administered to central compartment is eliminated or distributed to peripheral compartments
-Highly lipid soluble, high volume of distribution
--easily diffuses into membranes
--easily goes in and out of membranes, redistributed to fat and muscle
-Redistirbution has a high impact on plasma concentration
Methods of Administration for Opioids
-IV
-IM
-Subcutaneous
-Transdermal (fentanyl patch)
-Intra-articular
-Inhaled
-Oral
-Buccal mucosa in cats
-Rectal
-Neuroaxial
-Ocular
Neuroaxial Administration
-Into epidural or subarachnoid space
-Uptake into epidural fat
-Systemic distribution or diffusion across the dura into CSF
--lipophiic drug goes quickly into systemic vasculature
-Diffusion of drug across dura to CSF and opioid receptors
-Receptors in substantia gelatinosa of spinal cord
-Penetration of dura depends on lipid solubility and MW
-Poorly lipid soluble opioids (morphine) have longer duration than lipophilic opioids (Fentanyl)
--increased cephalid movement in CSF
Neuroaxial Side Effects
-
Neuroaxial Side Effects of Opioids
-Sedation
-CNS excitation due to cephalad migration of opioids to non-opioid receptors in brainstem or basal ganglia
--May block GABA mediated inhibition
-Viral reactivation: reactivation of herpes 2-5 days after epidural
-Neonatal Morbidity due to systemic absorption of opioids
--depression of ventilation in the newborn
-Vertigo, nystagmus, water retention, paresis, paralysis
Opioid Agonists
-Pure/Full agonists elicit maximal activation of opioid receptors
--Mu or Kappa agonists
-Full Mu agonists are superior analgesics
-Opioid agonists are drug of choice for moderate to intense pain
Morphine
-Full agonist at all opioid receptors (Mu, Kappa, and Delta)
-Sedation and analgesia
--IV or IM onset is 15-30 minutes
--Peak effect in 45-90 minutes
--Lasts 3-4 hours, epidural can last 12-24 hours
-Relatively hydrophilic
--delays onset of action
-Histamine release can occur after IV administration
--never give bolus of morphine IV! Will cause histamine release!
-1st pass effect occurs after oral administration
--bioavailability is about 25%
Morphine Metabolism and Excretion
-Conjugates with glucuronic acid
--morphine 6-glucoronide is active metabolite
--Morphine 3-glucoronide
-Major metabolites are eliminated via glomerular filtration
-Renal failure may result in accumulation
-Liver failure has minimal impact on clearance
Oxymorphine
Hydromorphone
-Synthetic full opioid agonist
-Acts on Mu receptors, full Mu agonist
-Sedation and analgesia is comparable to morphine for onset of action and duration
-10x potency of morphine, takes smaller dose to get the same effect
-Relatively more lipid soluble than morphine
-Causes less vomiting than morphine
-Does not produce histamine response
--can use with mast cell tumors
-Mainly used in small animal species at the moment
Meperidine
-Full opioid agonist at Mu and Kappa receptors
--also has other binding at A2 receptors, blocks Na channels
-Sedation lasts 1-4 hours
--shorter acting than morphine
-Same dose as morphine causes similar analgesia and euphoria
-much less potent than morphine
-Absorbed better than morphine from GI tract
--50% bioavailability
-Epidural effects may be advantageous due to local anesthetic-like effects
Meperidine Metabolism and Excretion
-90% undergoes demethylation in liver to normeperidine
-Hydrolysis to meperidinic acid
-Excretion via renal and urinary system
--pH dependent, acidification speeds up elimination
--Decreased renal function can lead to accumulation
Meperidine Side effects
-Lots of side-effects
-Negative inotropic effects
-Atropine-like effects (tachycardia, mydriasis)
-Seizures and Delirium due to accumulation of normeperidine
-Histamine release occurs with IV administration
-Combination with MAO inhibitor leads to serotonin syndrome
Fentanyl
-Synthetic Full Mu receptor opioid agonist
-Highly lipid soluble, crosses BB faster than morphine
--gets into the brain quickly
-Redistributes quickly
--longer infusion results in longer lasting after 3 hours
--will have to re-bolus unless given as CRI
--Fentanyl is still in body, not metabolized
-75-125x more potent than morphine
Fentanyl Indications
-Most often given as CRI or transdermal patch
-For use in dogs and cats
-Also in horses, cows, sheep, goats, pigs
-Previously in combination with Droperidol
Fentanyl Metabolism and Elimination
-Highly lipid soluble
-Rapid redistribution to fat and skeletal muscle
-1st pass pulmonary uptake
--75% of initial drug
-Saturation of tissues leads to liver metabolism
--Substrate for CYP450 enzymes
-Elimination half-life of 2-3 hours after brief infusion or single bolus
-Context-sensitive half-life increases with duration
-Renal excretion
Remifentanil
-Opioid agonist
-Structurally similar to fentanyl
-More rapid onset of action and termination
-Metabolized rapidly by plasma esterases to inactive metabolites
-has ester bond instead of amide bond
-Need to give as CRI
-Hepatic or renal dysfunction has little impact on metabolism
-Expensive!!
Methadone
-Synthetic Mu agonist, full agonist
-Affinity for NMDA receptors
--NMDA agonist, helps with chronic pain
-Excellent oral absorption
-High potency
-No active metabolites
-Good for management of chronic pain or withdrawal
-Becoming more popular in vet med
Tramadol
-Synthetic analog of Codeine
-Centrally acting analgesic
-moderate activity at Mu receptor, weak affinity for Kappa and Delta receptors
--weak agonist
-5-10x less potent than morphine
-Metabolite IS active, cats make metabolite
--dogs do not make metabolite
-Spinal inhibition of pain by decreasing reuptake of NorEpi and Serotonin
-Naloxone only antagonizes 30% of effects
-Metabolized by hepatic CYP450 enzyme systems
-IV, IM, or PO administration
Opiate Agonist-Antagonist
-Varying opioid receptor profiles
-All occupy Mu opioid receptors
-Competitive Mu antagonists
-Analgesic actions at Kappa receptor agonists
-Less respiratory depression
-Lower addictive potential, less euphoria
-Ceiling analgesic effect
Butorphanol
-Synthetic agonist-antagonist Opioid
-Originally an anti-tussive agent, suppresses coughing for 8 hours
-Analgesic effects through Kappa receptors, provides mild analgesia
-Antagonist at Mu receptors
-Minimal sedation alone, is best in combination
-Analgesia for mild to moderate visceral pain
-Spares MAC in dogs and cats
-In horses is used in combination with A2 agonists
-Provides good analgesia for cattle, sheep, goats
-Lasts 1-3 hours
-Reversal of pure Mu agonist
-Metabolizes to inactive metabolite
-Excreted in bile and urine
Partial Agonist
-Binds to Mu receptors with high affinity
-Produces limited clinical effects
Buprenorphine
-Semi-synthetic Opioid Partial agonist
-highly lipophilic opioid
-Cannot elicit maximal response at Mu receptor
-Causes sedation and analgesia
--onset of action is delayed 30 min-1 hour after IM injection
-Lasts 6-12 hours
-Good for mild to moderate pain
-Difficult to antagonize the effects of the drug
-Antagonist action at Mu and Kappa receptors
-Excreted in bile and urine
-Oral transmucosal absorption in cats
-Transdermal patch is available
Opioid Antagonists
-high affinity for opioid receptors
-Displace opipid agonists from Mu and Kappa receptors
-Bind to receptor but do not activate
-Causes reversal of all effects, including analgesia
-Development of intense pain and activation of SNS
Naloxone
-Opioid Antagonist
-Reverses all opioid agonist effects at all receptors
-Can be used to treat:
--ventilatory depression post-operatively
--ventilatory depression in neonate
--overdose
--detect physical dependence
-Lasts 1 hour, then patient will fall asleep again
-Excitement or anxiety may develop
-Causes hypertension, tachycardia, pulmonary edema
Naltrexone
-Opioid antagonist
-Clinical effects last twice as long as Naloxone
-Advantageous in prevention of renarcotization
-Similar side effects as Naloxone
--hypertension, tachycardia, pulmonary edema
Cats and Opioids
-Opioids do not always cause Morphine Mania
-can cause opioid mediated hyperthermia
-Mydriasis due to overriding sympathetic nervous system response
--may make excitation worse
-Constipation of not adequately hydrated
Horses and Opioids
-Cause nervousness and excitement
-Locomotor stimulants
-Ileus and colic
-Used in combination with sedatives/tranquilizers
-Butorphanol is most common opioid used
-Morphine should not be given until fully sedate
-Epidural opioids provide long-lasting analgesia
--no CNS excitement
Pharmacological effects of Benzodiazepines
-Anxiolysis
-Sedation
-Anticonvulsive effects
-Spinal-cord mediated muscle relaxants
-Anterograde amnesia
Benzodiazepine Uses in Vet med
-Prevent muscle rigidity associated with ketamine
-Synergistc effect with other drugs
--induction of anesthesia with propofol or etomidate
--With opioids in neuro-leptic induction
--Has inhalant sparing properties
-First line of drug treatment in seizure cases
Ketamine
-Causes muscle ridigidy
Benzodiazepine Mechanism of Action
-Works on GABA-A receptors only
-Facilitates binding of GABA to GABAa receptor
--enhances affinity of receptor for GABA with a ceiling effect
--Makes Benzodiazepine safe, and barbituates not safe
-At molecular level increases Cl consuctance and hyperpolarizes cell
GABAa Recepto
-Large macromolecule
-Pentameric protein complex
--Different expression of subunits in different species in different levels of the body give different affinity and responses
-Separate binding sites for endogenous GABA, Benzodiazepines, barnituates, propofol, alcohol, inhaled anesthetics
--GABA binds to A and B subunits
--Benzodiazepines binds to A and Gamma units
-Binding changes conformation and gives GABA a different affinity
-Does not mimic GABA, just changes affinity
-Gives safety profile
GABAa Subunit
-Several subunits isolated
-A1: responsible for sedataive effects
--60% of receptors
--in Cerebral cortex, cerebellum, and thalamus
-A2/3: responsible for anxiolytic effects
--less abundant
--Hippocampus and Amygdala
-Only known in humans
-Accounts for different effects in different species
Pharmacodynamic Effects of Benzodiazepines
-Causes delirium at recovery
-Can have excitatory phase in dogs and cats if used alone
--excitatory phase followed by sedation phase when given IV
-Different receptor subunits and receptor distribution variation between species gives different effects
Benzodiazepines in Vet Med
-Diazepam: fast onset, long lasting
-Midazolam: fast onset, short duration
-Lorazepam: Immediate onset and duration
--mostly human drug
-Zolazepam: long-lasting
--used in conjunction with telazol
--ONLY used in vet med
Benzodiazepine Receptor Occupancy
-Binding to receptor is saturable
--has ceiling effect
-Lorazepam has greatest receptor affinity, then midazolam, least diazepam
-Chronic administration causes decreased receptor binding and function
--only important with chronic use or abuse
-20% occupancy gives anxiolytic effect
-30-50% occupancy gives sedation
-more than 60% occupancy gives unconsciousness
Benzodiazepine Pharmacokinetics
-Highly lipophilic
--allows for rapid onset of action and fast redistribution
--Also give shorter duration
-Midazolam is most lipophilic, then diazepam, then lorazepam
-Has large volume of distribution
--gives rapid CNS effects
-High protein binding to albumin
-Clearance rates differ between different benzodiazepines
Benzodiazepine Metabolism
-Hepatic microsomal oxidation
-Glucoronidation
--watch for cats and liver function
-Awakening comes from redistribution of drug from brain to other less perfused tissues
--NOT metabolized, just left the brain
CNS effects of Benzo
-Decreases CMRO2 and Cerebral Blood Flow
--neuroprotective, protects against cerebral hypoxia
-Increases seizure threshold of local anesthetics
--can tolerate more of lidocaine before seizure
-Midazolam does not produce isoelectric EEG
--increases ICP when given rapidly IV
--give slowly and with other drugs
--DO not used Midazolam to treat seizures
Respiratory Depression with Benzodiazepines
-Minimal in animals
-Midazolam causes more respiratory effects than diazepam or lorazepam
-Respiratory depression with fast IV dose of midazolam
-Additive effects with opioids give respiratory effects
-Decreased tidal volume
-Flattened CO2 rose response curves
Cardiovascular effects Benzo
-Cardiovscularly stable
-Will have some reduction in BP
--Seen more with midazolam than diazepam
--Can have synergistic effects with other anesthetic drugs
Diazepam
-Insoluble in water, viscous solution
-pH 6.6-6.9
-5mg/ml
--contains 0.4ml propylene glycol (Toxic!), 0.1ml alcohol, and sodium benzoate
-Dilution with sterile water or saline
-Injection may be painful
-Not recommended to give IM
--absorption is not predictable and it stings
--have other drugs that work better
Diazepam Metabolism
-90% bound to protein
-Metabolized hepatic microsomal oxidation
-Has active metabolites! unpreditable
--Desmethyl-diazepam
--Oxazepam
-Decreases hepatic biotransformation
-Low extraction ratio drug, takes the liver a long time to get rid of
Diazepam in Propylene Glycol Formulation
-Has been reported to cause hypotension and ventricular arrhythmia
-40% propylene glycol is toxic
-Recommended to give slowly IV
Diazepam Clinical Pharmacology
-Less reliable sedation in veterinary medicine
-Can be used in ruminants and swine pretty reliably
-Causes arousal in dogs and excitement or aggression in cats
-Causes Ataxia and recumbency in horses in very small doses
--horse can fall down with a small dose
Midazolam
-More potent than diazepam (2-3x)
--Has 2-3 times affinity for GABA receptor
-Water-soluble
-Can be used IV or IM
Midazolam IM and Benzene Ring
-Changes to chemical structure at different pH
-IN bottle, pH 3.5
--Benzene ring is open, and very water-soluble
--great IM absorption without irritating
-At pH greater than 4, benzene ring closes
--more lipid soluble than diazepam
--gives fast onset and easily crosses BBB
Midazolam Metabolism
-Short duration of action
-Rapidly redistributed from the brain to inactive tissues
-Accumulates, but not as much as diazepam
-Hydroxylation by hepatic and small intestine
-Rapid renal clearance
-Metabolite 1-hydroxymidazolam has 20-30% potency
Midazolam in Exotic Species
-Very effective due to IM use and fast onset
-ferrets, rabbits, birds, swine
-Can give intranasally in swine
-Costs more than diazepam, but almost same price
Flumazenil
-1,4-imidazobenzodiazepine derivative
-Competitive antagonist against GABAa receptor
-Reverses benzodiazepines activity
-has high affinity for receptors
-Cannot overdose, just give to effect
-Minimal agonist activity at high doses
-Similar structure to midazolam
-Weak water solubility, moderate lipid solubility
-Rapidly cleared from plasma
--short half-life gives potential for re-sedation
-Lower protein binding, 36-46%
-Metabolized by the liver
--5 active metabolites
Alpha2 Agonists History
-Good reputation in large animals
-Have to be more careful in ruminants and cows
-Bad reputation in small animals, esp. cats
-Used a lot in wildlife capture
-Discovered during development of human anti-hypertensive agents
--used to lower blood pressure
-Xyalzine was first in 1962
-Detomidine in 1908s
-Romifidine and medetomidine in 1990s
-Dexmedetomidine in 2000s
A2 Agonist Imidazoline
-Clonidine
-Romifidine, Detomidine
-Medetomidine
-Dexmedetomidine
-Mivazerol
-Azepexole
A2 Agonist Thiazine
-Xylazine
Alpha2 Agonists
-Cause hypertension first, then hypotension due to central mediation
Alpha2 Agonist Reversal
-Not FDA approved for humans!
-A2 agonists are VERY potent, can cause massive hypotension
-A2 agonists in everyday practice are not super potent
--for wildlife are VERY potent, be careful!
A1/A2 ratio
-A1 and A2 receptors
-For every A1 that is activated, there are a given number for A2 that are also activated
-A2 agonists preferentially affect A2 receptors but also have A1 effects
-Reversibility of drug with higher A2 affinity is higher, easier to reverse
-Ratio is only an advantage as far as reversibility is concerned
A2 Receptor Subtypes
-3 subtypes
-Based on susceptibility to activation/inactivation by agonists and antagonists
-Confirmed by molecular Biology
A2a Receptor
-Sedation
-Hypnosis
-Analgesia
-Sympatholysis
A2b Receptor
-Vasoconstriction
-Anti-shivering action
-Analgesia
A2c receptor
-learning
-Startle Response
A2 receptor Structure
-Metabotropic receptor
-G-protein coupled receptors
-Different results in activation re due to coupling to different G-proteins
-Agonist binding site involves 3 contact points
--amino acid residues on at least 2 separate transmembrane domains
A2 receptor distribution
-ALL OVER the body
-Each subtype is distributed ubiquitously throughout the body
-Want analgesia, but have to deal with all other types of activation and body response
-Very important side effects due to distribution of receptors
Pre-synaptic A2 receptors
-In sympathetic nerve endings and noradrenergic neurons of CNS
-Also act in peripheral nerve endings
-Inhibit the release nor NorEpi and Epi
-Work on negative feedback loops
Post-synaptic A2 receptors
-Located in the CNS and many tissues
-Have been found in platelets, lung, adipose tissue, pancreas, salivary gland, kidney, spleen, vas deferens, ileum, and eye
Junctional vs. Extrajunctional A2 receptors
1. Extrajunctional: Vasculature
--controlled by A2 receptors
-Causes dramatic vasoconstriction
--heart slows down, is pumping against a larger pressure
--If bradycardia is concern, take patient off of A2 agonist
2. Junctional: in synapse
Signal Transduction of A2 receptors
-A2 agonist binds to receptor and causes activation of Ai subunit
--inhibits adenylyl cyclase and decreases accumulation of cAMP dependent protein kinase
--Results in decrease in phosphorylation of target regulatory proteins
-May not be responsible for clinical effect that is observed
-Results in alteration of function of ligand gated ion channels through direct coupling
CNS effects of A2 agonists
-A2a receptors located pre-synaptically on neurons in Locus Coeruleus
-G-protein mediated hyperpolarization
-Decreases sympathetic outflow from the brain
--decreases HR, decreases BP
--NOT the same as post-synaptic effect on blood vessels (will see increased BP and reflex decrease HR)
-Causes sedation/hypnosis, analgesia, decreased sympathetic activity
Xylazine Dosage
-Should not be used in dogs and cats anymore
--have better drugs like medetomidine and dexmedetomidine
-1.1 mg/kg
-Peak effect within 15 minutes
-Lasts 45-60 minutes
Medetomidine Dosage IM
-30 ug/kg
-Given IM
-Onset within 5 minutes
-Lasts 1-2 hours
Medetomidine Dosage IV
-20ug/kg IV
-Onset within 5 minutes
-Lasts 90 minutes
-Smaller dose has same onset and shorter duration
-After 40 ug/kg will not have a change in degree of sedation
--will just have longer effects
--may have to reverse, ideally do not overdose
Horses and Xylazine A2 agonists
-1.1mg/kg IV
-Peak within 5 minutes
-Heavy sedation for 20-30 minutes
-Some sedation for up to 60 minutes
-Good for short surgeries, field castrations
Horses and Romifidine A2 agonist
-80-120 ug/kg
-Peak within 10 minutes
-Heavy sedation for 1 hour
Horses and detomidine A2 agonist
-20ug/kg
-Peak within 15 minutes
-Lasts about 1 hour
Horses and Medetomidine A2 agonist
-Expensive for horses!
-10ug/kg
-Peak within 5 minutes
-Heavy sedation for 20-30 minutes
A2 agonist equipotent doses in Horses
1.0 mg/kg Xylazine
100 Ug/kg Romifidine
20 ug/kg Detomidine
10 ug/kg Medetomidine
5 ug/kg dexmetdetomidine

Duration of sedation for equipotent doses:
Xylazine is the shortest, Medetomidine and dexmedetomidine and equal, then detomidine, and romifidine lasts longest
A2 agonist Xylazine in Calves
-0.3 mg.kg IV
-Onset within 1 minute
-Lasts about 420 minutes (7 hours)
A2 agonists in Cows
-0.01-0.05 mg/kg
-Lower dose compared to dog or horse
-Onset within 1 minute
-lasts about 1 hour
Analgesic Effects of A2 agonists
-Supraspinal level: activation of descending antinociceptive pathways
--suppresses pain
-Spinal level:
--Presynaptic and postsynaptic inhibition of nociceptive transmission
--dorsal horn of spinal cord
--Good for tissues with chronic pain
A2 agonist cardiovascular effects
-Vasoconstriction
-Decrease in sympathetic tone
-As systemic vascular resistance increases, CO decreases
--SVR decreases, CO mirrors and increases
A2 agonist and Vasoconstriction
-A2 receptors in the vascular smooth muscle
-Hypertension and reflex bradycardia cause decrease in cardiac output
-Initial phase
-No Atropine
A2 agonist and Decreased Sympathetic Tone
-Causes hypotension, direct bradycardia, and decreased CO
-Secondary phase
A2 agonist and Increased Vagal Tone
-Causes direct bradycardia
-AV blocks
Medetomidine in Dogs Cardiovascular effects
-Increase in systemic and pulmonary artery pressure
-Decrease in HR by 20-63%
-Decrease in CO by 50%
--big deal
-Increase in systemic and pulmonary vascular resistance
-No direct myocardial depressant effects
-Decreases in coronary blood flow due to decrease in CO
-Can cause arrhythmia (maybe, evidence is shaky)
Anticholinergics in Hypertensive Patient
-Do not give!
-Transient decrease in bradycardia
-Arrhythmia
-2nd degree AV block, tachycardia, pulsus alternans, premature ventricular contractions
-Decreased contractility and impairs myocardial oxygenation
-Patient actually does worse
A2 agonist Respiratory effects in Horses
1. Normal horses:
-A2 is muscle relaxant, horses have issues breathing
-Increased work of breathing
-Increased resistance to breathing
2. Horses with COPD:
--changes in respiratory function are not more severe
3. Ponies:
-Xylazine decreases pulmonary resistance and increases compliance
A2 agonist Respiratory effects in Cows
-Xylazine and Medetomidine in Calves
-Will decrease PaO2 from 90mmHg to 42mmHg
-After reversal will go back up, but not to normal levels
A2 agonist Respiratory effects in Sheep
-Damage capillary endothelium and pulmonary macrophages
-Degranulation of pulmonary alveolar macrophages
-Can result in sever hemorrhage, pulmonary edema, and hypoxemia
-Xylazine, medetomidine induced damage to capillary endothelium and PAM
-Even if drug is reversed, damage is still there
Xylazine
-A2 agonist
-IM injection
-Peak plasma concentration in 15 minutes
-LOW dose in bovines
--give 1/10 dose given to horses
Dexmedetomidine
-A2 agonist
-A1/A2 ratio of 1:1620
--MUCH more effect on A2 receptors
-Distribution half life of 6 minutes
-Terminal elimination half-life is 2 hours
-Metabolized by the liver
--methyl and glucuronide inactive conjugates
-Secreted by kidneys
-Large volume of distribtion
-Longer elimination half life than xylazine
-Sedation lasts 45 min-2 hours
A2 agonist reversal
-A2 antagonists do the job
-Yohimbine
-Tolazoline
-Atipamezole for dexmedetomidine due to high A1/A2 ratio
--if given IV on its own, can cause hypotension and excitement
Phenothiazine and Butyrophenone History
-Originally developed as schitzophrenia medication
--Did not really treat patient, just made them incapable
--Decreases arousal and motility
-Anxiolytic? Or just not showing anxiety?
Antipsychotic Agents
-Phenothiazines
-Butyrophenones
-Thioxanthenes
-Benzepines
-Dyphenylbutylpiperidines
-Indolones
Phenothiazines and Butyrophenones
-Phenothiazines: Acepromazine
-Butyrophenones: Azaperone

-Tranquilizers
Classification of Antipsychotics
-Neuroleptics or "Traditional Antipsychotics"
-Prominent D2 antagonism
--works on dopamine receptor
-Increased release of prolactin
-Increased risk of extrapyramidal side effects
-Older agents, 1st generation drugs
Neuroleptic Syndrome
-Suppression of spontaneous movements and complex behaviors
-Spinal reflexes and unconditioned nociceptive-avoidance behaviors remain intact
--can pull hand off of hot stove
-Used to describe the effects of chlorpromazine and reserpine in lab animals and psychiatric patients
Neuroleptic in Humans
-Reduced initiative and interest in the environment
-Reduced manifestation of emotions
--emotions still present, just less manifestation
-Possible drowsiness and diminished response to external stimuli
-Remain easily aroused
--can answer questions, retain intact cognition
-Also exert neurological effects
--bradykinesia, mild rigidity, tremor, subjective restlessness
Phenothiazine Structure and Activity
-Tricyclic Structure
-2 benzene rings linked by S and N atom
-Side chains at positions 2 and 10
-Substitutions at position 2 affect antipsychotic effects
-Substitutions at position 10 affect antidopaminergic effects, antimuscarinic effects, antihistaminergic effects, antiadrenergic effects, antiserotoninergic effects
Butyrophenone Structure
-Hetercyclic Structure
-Derivatives of phenylpiperidine opioid meperidine
Dopamine Receptors
-G-protein coupled receptors
-D1: excitatory
-D2: inhibitory
--inhibits adenylate cyclase intracellular formation
-5 types total
--D1-like: D1, D5
--increase cAMP
--Increase PIP2 hydrolysis, leading to Ca mobilization and PKC activation
--D2-like: D2, D3, D4
--Decrease cAMP
--Increase K currents
--Decrease voltage-gated Ca currents
Dopamine Receptor Function
-Agonist: promotes release of dopamine
--fosters symptoms of schizophrenia
--Amphetamine and Cocaine block reuptake of dopamine, keeping it in the synapse for longer, and fostering symptoms of schitzophrenia
-Antagonist: Blocks symptoms of schitzophrenia
--occupies dopamine site on D2 receptor, prevents dopamine from activating the receptor
Chlorpromazine
-Dopamine antagonist
Distribution and Characteristics of Dopamine Receptors in CNS
-D1: Striatum and Neocortex
-D5: Hippocampus and hypothalamus
-D2: Striatum, SNpc, Pituitary
-D3: Olfactory tubercle, nucleus accumbens, hypothalamus
-D4: Frontal Cortex, medulla, midbrain
Dopamine Pre-synaptic Receptors
-Autoreceptors
-D2 and D2-like receptors
-Coupled to Gi
-Decrease phosphorylation of Tyrosine Hydroxylase
--decreases dopamine synthesis
-Modulation of Ca and K currents leads to decreased dopamine synthesis
Dopamine Post-synaptic Receptors
-D1 and D1-like receptors
--coupled to Gs, activate adenylate cyclase and increase cAMP to activate PKA
-D2 and D2-like receptors
--coupled to Gi, inhibit adenylate cyclase and decrease cAMP
-D1/D2 heterooligomer
--coupled to Gq, activates Phospholipase C and causes PI hydrolysis
--Leads to intracellular Ca release and downstream signaling
--DAG activation allows for activation of protein kinases
Antidopaminergic effects of Phenothiazines and Butyrophenones
-What you rely on for sedation
-Works on Cerebral cortex, basal ganglia, and limbic system
-Cerebral cortex:
--prefrontal and deep temporal regions
--causes EEG abnormalities and seizures
-Basal ganglia:
--caudate nucleus, putamen, globus pallidus
--extrapyramidal effects
-Limbic System:
--amygdala, hippocampus, olfactory tubercle, septal nuclei
--schitzophrenia
Phenothiazine and Butyrophenone Mechanism of Action
-Hypothalamus and Pituitary:
--prolactin secretion
--impaired thermoregulation, can cause hypothermia in anesthetized patients
-Brainstem: impaired motor reflexes
-Medulla:
--CTZ, protection against nausea and emesis
Non-dopaminergic Effects of Phenothiazine and Butyrophenone
-ANS
--antimuscarinic M1 receptors
-Antiadrenergic A1 and A2 receptors
--Hypotension that is hard to treat
-Antihistaminergic H1 receptors
-Antiserotoninergic receptors
Absorption of Acepromazine
-Relatively slow absorption after IM administration
--15-30 minutes
-Poor bioavailability, 20%
--can be variable
Distribution of Acepromazine
-Mostly highly lipophilic, highly membrane or protein bound
--lends to wide distribution
-Volume of distribution is 4-6 L/kg in horses
-Accumulates in the brain, lungs, and other VRG tissues
-Can enter fetal circulation and breast milk
Metabolism of Acepromazine
-Very slow!
-Halflife in dogs is 6-8 hours, horses 2-3 hours
-Dose dependent, but overall long half-life
-Oxidative metabolism via CYP450 in liver
--Followed by glucuronidation, sulfation, and other conjugation
-Most metabolites are inactive
-Excreted by the kidneys
Acepromazine
-approved for use in many animals
--dogs, cats, cattle, horses, swine, sheep
-Has milk withdraw of 48 hours
-Meat withdraw of 7 days, long time!
Indications of Acepromazine in Dogs and Cats
-Used as preanesthetic agent
-In conjunction with opioid
--provides chemical restraint
-Use doses that are lower than label dose
--0.01 mg/kg
-Used extensively as preanesthetic agent
Indications of Acepromazine in Horses
-Supplement to preanesthetic medication
-Anti-anxiety effects?
-Antispasmodit effect to treat colic pain?
Indications of Acepromazine in Cows
-Anti-anxiety effects
-Not commonly used
-Can be alternative to A2 agonists
Indications fo Acepromazine in Small Ruminants
-Not used, need high doses
Acepromazine in CNS
-Sedation
-MAC reduction is needed (30-45%)
-Hypothermia
-Sedation onset is 15 minutes, peak effect in 30 min
-Lasts 3-4 hours
-Injectable induction agent is needed
-reduces MAC by 30-45%
-Can cause hypothermia
-No evidence to suggest that acepromazine lowers seizure threshold
-Prevents onset of halothane-induced Malignant hyperthermia in pigs
Acepromazine Complications
-Inconsistent calming
-Orthostatic Hypotension
-Extrapyramidal side effects
--anti-dopaminergic
Effects of Acepromaznie on Cardiovascular System
-Decreases Mean Aeterial Pressure by 20-30%
-decreased stroke volume and CO also reported in dogs and horses
-Hypotension is worse with isoflurane compared to halothane
-At clinical doses HR may not change significantly, ot may increase a little
-At very high doses may see bradycardia and SA block
-Prevents arrhythmogenic effects of Epi and halothane
Effects of Acepromazine on Respiratory System
-Minimal effects on pulmonary function
--oxygenation and ventilation stay the same
-Respiration rate may decrease in conscious animals
Effects of Acepromazine on Hematologic System
-Decreased hematocrit and PCV by 20-50% in dogs and horses
--RBCs go to the spleen, splenic sequestration
-Occurs within 30 min and last for at least 2 hours
-Magnitude and duration of effect is dose dependent
-Platelet aggregation is also inhibited
--minimal hemostatic significance
Effects of Acepromazine on GI system
-Antiemetic effect when administered 15 minutes before morphine, hydromorphone, or oxymorphone
-Decreases Lower esophageal sphincter tone and leads to increased risk of gastric reflux
-Decreased GI motility and delayed gastric emptying in horses
Effects of Acepromazine on Urinary system
-GFR and Renal blood flow are maintained in dogs on Acepromazine and isoflurane
-Decreased urethral pressure by 20% in cats under halothane anesthesia
-Penile protrusion/pariapism in stallions
--magnitude and duration of protrusion are dose dependent
--Mediated via A-adrenergic antagonism
Effects of Acepromazine on Integumentary System
-Antihistaminergic effects
-not suitable for use in allergic skin testing patients
Acepromazine in Boxers
-No scientific evidence of sensitivity
Azaperone
-Tranquilizer in swine
-Only used in pigs
-Used to reduce aggression when groups of animals are mixed
-not really a sedative
-May be used as a preanesthetic in pigs to produce sedation
--15 minute onset
--peak effect in less than 30 minutes
--lasts 2-3 hours
-Has similar cardiovascular effects as acepromazine
--Decreases BP and CO
--HR has been reported to decrease also
-Penile Prolapse reported
"Anesthesia"
-Greek for "Insensibility"
-Loss of sensation to entire body or any part of the body
-Regional anesthesia: insensibility of a large but limited body area
-General Anesthesia: drug-induced unconsciousness
--controlled but reversible depression of the CNS
--patient is not arousable by noxious stimulation
Elements of the Anesthetic State
-Unconsciousness
-Amnesia
-Analgesia
-Immobility (skeletal muscle relaxation)
Stages of Anesthesia
1. Analgesia
2. Excitement
--want to pass through rapidly
3. Surgical Anesthesia
--desired stage, where you want to be
4. Medullary paralysis
--too deep
--animal cannot regulate BP or RR
--want to get out of this stage as soon as possible
Theories of Anesthesia
-CNS site of action
-Cellular site of action
-Molecular site of action
CNS sites of action in Anesthesia
-Many sites involved
-Cerebral cortex
-Reticular Activating System
-Thalamus, amygdala, vestibular system
-Areas are inhibited, depressed
--do not send out impulses
Cellular Sites of Action
-Interference with synaptic mechanisms
-Pre-synaptic release by inhibiting synthesis or release of NT
-In synapse by altering NT clearance
-On post-synaptic membrane by changing membrane potential or NT receptor binding
-Inhibit axonal nerve impulse conduction
Molecular Sites of Action
-Ion channels and neuronal cell membrane proteins
-Can activate lipid bilayer as a while
-Lipids at protein/lipid interface
-Protein site bounded by lipid
-Protein site exposed to aqueous environment
Lipid Theory of Anesthesia
-Hydrophobic Site
-Can cause membrane volume expansion
-Membrane fluidisation
Protein Theory of Anesthesia
-Interaction of anesthetics with specific NT receptor proteins
-GABA receptor plays a role
--Has sites on receptor for almost every inhibitory drug
--Barbiturate site
--benzodiazepine site
--General anesthetics site
How GABA receptor works
-Spans membrane with 5 subunits
--subunits form pore
-GABA binds to site, opens channel, chloride molecules go through
-Hyperpolarizes the cell membrane
Properties of the Ideal Injectable Anesthetic
-Minimal cardio-respiratory effects
-Rapid onset
-Short duration of action
-Smooth emergence afterwards
-Water soluble
-Long shelf-life
-Small volume required for induction of anesthesia
-Wide safety margin, hard to mess up
-No effect on vital organ functions except CNS
-No histamine release elicited
-No anaphylactic reactions
-No local toxicity
Benefits of Injectable Agents
-Good for induction, start of anesthesia
-Rapid loss of consciousness
--inhalants take a long time for induction
-Quick control of airway
--important for hypoxic or regurgitating animals
-No environmental pollution from waste gasses
-Some may be administered intramuscularly
--easy to give to fractious or wild animals
Negatives of Injectable Agents
-Some have low margin of safety
--have moved away from most really bad ones
-Induce some degree of hypotension, respiratory, cardiovascular depression
-Some can have long-lasting effects compared to inhalant
--have to be metabolized to leave the body
-Patient physiology will influence pharmacokinetics and pharmacodynamics of agent
-Once it’s in, can’t get it out
--have to wait for effects to wear off
Rapidly Acting Injectable Anesthetics
-Barbituates (Thiopental
-Phenols (Propofol)
-Imidazoles (Etomidate)
-Cyclohexanones (Ketamine, Tiletamine)
-Steroids (Alfaxalone)
Slower Acting Injectable Anesthetics
-Not used often anymore
-Chloral hydrolase
-Chloralose
-Urethan
-Potent Opioids
-Neuroleptanalgesics
Barbituates as Injectable drugs
-Formed from Urea and Malonic acid
-No longer used here in US
Oxybarbituates
-Oxygen at C2
-Delayed onset of action
-Long effect
-Hypnotic Action
-Barbital, pentobarbital, phenobarnital
Methylated oxybarbituates
-Oxygen at C2
-Rapid onset of action
-rapid recovery
-Some excitatory effects
-Methohexital
Thiobarbituates
-Sulfur at C2
-Rapid acting
-Smooth fast recovery
-Thiopental
-Thiamylal
Methylated thiobarbituates
-Sulfur at C2, Methyl at C1
-Rapid onset of action
-Very rapid recovery
-High incidence of excitatory effects
Structure Activity Relationship of Barbituates
-Lipid solubility increases action
-2-5 carbon side-chains increase lipid solubility
-Sulfur on 2 carbon increases lipid solubility, faster action
-Changes that increase lipid solubility decreases duration of action
--also decrease latency of onset of action
-Accelerate degradation
-Increases albumin binding
-Increases hypnotic potency
Mechanism of Action of Barbituates
1. Depress transmission in CNS at synapse level
-Presynaptic inhibition of NT release
--ACh, glutamate
-Inhibits post-synaptic impulse transmission
-Targets thalamus and ascending Reticular Activating System
Main Mechanism of Action of Barbituates
-Alloesteric modulation of GABAa receptor
--increase binding of benzodiazepines and GABA receptors
-Enhance action of GABAa receptor
--inhibtory on CNS
-Allow longer duration of Cl to enter cell
-Increase average duration of GABA activated Cl-ion channel opening
CNS actions of Barbituates
-Direct activation of GABAa Cl channel
-Interaction with central Ca and Na channels and glutamate and nicotinic ACh receptors
-Depresses autonomic ganglia
PNS actions of barbituates
-Blocks ACh effects at nicotinic synapses
-Inhibits transmembrane Ca fluxes
Pharmacological effects of Barbituates
-Depresses activity in CNS
-Gradual depression of CNS activity
-Sedation leads to hypnosis leads to anesthesia
-Decrease in EEG activity at anesthetic doses leads to global CNS depression
Analgesic effects of Barbituates
-No effects
-No Anti-nociceptive effect
-Need to give something else to give analgesia
Non-hypnotic effects of Barbituates on CNS
-Decreases Cerebral Blood Flow, ICP, and CMRO2
-Decreases convulsive activity
Respiratory effects of Barbituates
-Respiratory depression
-Central and peripheral chemoreceptor sensitivity to CO2 is decreased
-Plasma concentration needed for respiratory arrest is less than what is needed for cardiac arrest
-Treatment for barbituate overdose is ventilatory support
Cardiovascular effects of Barbituates
-Decreases BP
-Decreases CO
-Decreases contractility by decreasing intracellular Ca release
-May cause ventricular bigeminy
--every other beat is a premature ventricular complex
-Decreases occur in a dose-dependent manner
Adverse effects of barbituates
-Affects other organs
-Decrease in renal blood flow
-Decreases hematocrit due to splenic sequestration of RBCs
--normal finding
-Decreases WBC
-CYP450 enzymes are induced
--speeds up clearance of other drugs used
-Cannot go out of the vein! Causes severe phlebitis with perivascular injection
--make sure catheter is well-placed
--due to basic pH
Correlation between lipid solubiity and onset of anesthesia
-more lipid soluble gives faster acting anesthetic
-Barbital is least lipid soluble, loss of consciousness occurs in 22 minutes
-Thiopental is most lipid soluble, gives immediate loss of consciousness after injection
Pharmacokinetics of Barbituates
-As concentration in blood decreases, concentration in other tissues increases
-Brain and viscera is first in distribution
-Lean tissues are moderate
-Fat has long increase in concentration
--poorly perfused
Contact sensitive half-life of thiopental
-As duration of use increases, time it takes for drug levels in the body increase
-Need to pay attention to how much dose over how much time
-All about how drug redistributes in the body
Pharmacokinetics of Barbituates
-Hepatic Metabolism
-Oxidated and converted in glucaronic acid
--Conjugated with glucaronic acid
-Excreted in bile or in urine
--less than 1% of intact drug is excreted in urine
-Changed in tissues and excreted
Greyhounds and Barbituate Pharmacokinetics
-May have different CYP450 metabolism
-Have very little fat and extensive muscle
--different volume of distirbutiuon
-Takes much longer for animal to recover
--especially with Thiopental
Thiopental
-Most commonly used barbiturate
-Used for induction and short procedures
-Can get tissue necrosis if it goes out of the vein (perivascular injection)
-Biotransformaiton in liver, slow metabolism
-Waking up from anesthesia is NOT slow
--recovery is due to redistribution, not metabolism
--context sensitive half-life
When to NOT use Thiopental
-Patients with cardiovascular dysfunction
--Cardiac failure and low contractility
--Hypovolemia
--Tachyarrhythmias
-Patients with metabolic acidosis
-Greyhounds
-Cachectic patients
-Total Invravenous Anesthesia
--drug can build up over time with continued use
Phenols
-Propofol
-Sedative-hypnotic drug
-Unrelated to barbituates
-Alkylated phenol, 1% in oil-in-water emulsion
-Has short half-life, discard within 6 hours
--due to soybean, glycerol, and egg phosphatide solution
--No preservatives, lots of possibility for microbial growth in medium
Propophol Mechanism of Action
-Allosteric binding to GABAa receptor
-Synergism with benzodiazepines
Propophol Pharmacologic Effects
-Gradual depression of CNS activity
--sedation leads to hypnosis leads to anesthesia
-General anesthesia within 1 minute
--rapid!
-Rapid recovery of action without residual CNS depression
--Ideal situation!
-Can maintain with repeated IV boluses or CRI, do not need inhalant
-Can use IV for long-term sedation, low risk of drug accumulation
--rapidly metabolized
-Non-hypnotic CNS effects
--decrease CBF, ICP, and Oxidation
-Antiemetic effects, can be very helpful
Principle Pharmacologic Effects of Propophol
-Intravenous long-term sedation
-Low risk for drug accumulation
-Non-hypnotic CNS effects
-Decreases cerebral blood flow, ICP, and CMRO2
-Antiemetic effects
Respiratory Effects of Propophol
-Central Respiratory Depression
-Bronchodilation (usually helpful)
-Inhibition of laryngeal reflexes
--good for intubation
Cardiovascular Effects of Propophol
-Causes hypotension
-Vasodilation
--decreases sympathetic vasomotor tone centrally
--Peripherally has negative inotropic effect and direct vasodilation
-Can have effect on contractility, negative inotropy
-Usually no change in HR
Adverse effects of Propophol
-Excitatory effects sometimes (transient)
--10% of dogs and cats
-Limb paddling, nystagmus, muscle twitching
-not that common and not that big of a deal
Pharmacokinetics of Propophol
-2-3 compartment open model
-Starts in blood, goes to brain, then muscle and other tissues
-Rapidly reaches peak CNS concentrations
-Rapidly redistributed, halflife is 7 minutes
-Elimination is slower, takes a while to metabolize
-Highly protein-bound (97-99% bound to albumin and Hb)
-Can cross placenta in pregnant animals but will be removed from fetal circulation very quickly
-Significant 1st pass uptake into lungs (28-61%)
-Extrahepatic metabolism in kidneys and lungs
Extrahepatic metabolism of Propophol
-Extrahepatic metabolism in kidneys also
-Significant 1st pass into the lungs
--28-61%
--big dose goes into lungs!
-Also metabolized in the liver via CYP450 and glucuronide conjugation
Propophol concentration
-Does not really concentrate at all
-Metabolism is fast
Cats and Propophol Metabolism
-No glucuronide system
-Slower elimination
-Repeated dosing can cause prolonged recovery
-Reduced conjugation capacity can lead to oxidative injury in RBCs
--Heinz body formation
--mostly occurs with repeated uses
When to NOT use Propophol
(or use with caution)
-Cardiovascular dysfunction
--cardiac failure
--decreases contractility and vasodilation
--Hypovolemia
--Hypotension
-Patients with Sepsis or systemic infections
--great medium for bacteria due to suspension
-Cats with Anemia
--heinz body formation
Etomidate
-Imidazole
-Class of sedative-hypnotic drugs chemically unrelated to other anesthetics
-VERY Short-acting
-very safe, high therapeutic window
-Great in high-risk patients
--cardiovascular or respiratory unstable patients
-Mostly used off-label in small animal anesthesia
Chemistry of Etomidate
-Carboxylated imidazole derivative
-Racemic mixture, R and S isoforms
--R is active isoform
-Dissolved in 35% propylene glycol
--can cause irritation and kidney issues
Etomidate Mechanism of Action
-Acts through GABAa receptor, allosteric modulation
--has own binding site
-Depression of ascending Reticular Activating System
-Synergism with Benzodiazepine and GABA binding to GABA receptor
Etomidate Phamacologic effects
-Ultra-rapid onset of CNS depression
--less than 20 seconds
-No analgesic effects
-Non hypnotic CNS effects:
--decreases EEG activity
--Vasoconstricts cerebral vasculature and decreases CBF, direct cerebral vasoconstriction
--Decreases ICP and CMRO2
Adverse effects of Etomidate
-Minimal respiratory and cardiovascular side effects
-Excitation on induction
--vomiting
--Need to have a very sedate patient before giving
-Inhibits adrenal steroid output, suppresses steroid formation
-Hemolysis due to high osmolality of solution
--pulls blood into RBCs and bursts cells
--Can dilute to reduce effects
-Can be painful with IV injection if it goes outside of the vein
Etomidine Pharmacokinetics
-2-3 compartment open model
-Rapidly reaches peak CNS concentrations
-Immediate and extensive redistirbtion
-Lots of uptake into peripheral tissues
-Moderately protein-bound (76%)
-Will go into placenta but incomplete transfer
--safe for pregnant animals
--fast decline once in fetal plasma
Etomidine Metabolism
-Extensive heptic metabolism via CYP450 system
-Extra-hepatic metabolism in plasma esterases
-Renal excretion of metabolites
Uses for Etomidine in Clinical Practice
-Great for patients with hemodynamic issues
--cardiac failure
--hypovolemic shock
--sepsis/endotoxemia
-Patients with cerebral disease and increased ICP
--seizures
Cautions for Etomidine Use
-Adrenal insufficiency, Addison’s disease
-Patients with Sepsis
Cyclohexanones
-Dissociative anesthetics
-Ketamine, Tiletamine
-Produces anesthesia-like state
--Dissociative Anesthesia
-Intense analgesia, light sleep, amnesia, catalepsy (rigid muscles)
--do not want to use by itself
-Provides analgesia
Chemistry of Ketamine
-Cyclohezylamine
-Highly lipid soluble, more than thiopental
-Racemic Mixture
--S isoform is 3-4x more potent than R isoform
--S isoform is 2x as long active
-Dysphoria is major issue, more with R isoform
Molecular Mechanism of Action of Ketamine
-Blocks NMDA receptors an sub-anesthetic doses
--NMDA receptor antagonist
--"Use dependent block"
--"Closed" channel block
-Decrease in central glutamatergic activity leads to analgesia and anesthesia
NMDA receptor Mg block
-Mg blocks ion channel
-Activation removes Mg and allows ions to move through channel
-Use-dependent block, has to be activated first
-Ketamine blocks channel
Ketamine CNS action
-Inhibits nicotinic ACh receptors
-Interacts with central opioid receptors
--Mu, delta, and Kappa opioid receptors
--Monoaminergic receptors
--Muscarinic ACh receptors
Ketamine Other Mechanisms of Action
-Blocks NorEpi reuptake
-Blocks voltage-sensitive Ca channels
-Blocks peripheral nerve conduction
Ketamine Pharmacologic effects
-Works on thalamic/neocortical area
-Dissociative because it dissociates area of brain from the spinal cord
--not depressing
--makes impulses disconnected, more “unaware”
-Selective suppression of thalamo-neocortical projection system
-Provides analgesia at sub-anesthetic doses
Dissociative Anesthesia Features
-Loss of consciousness despite neuronal activity outside neo-cortex
-Catalepsy, catatonia
-Maintains protective reflexes
--airway reflexes maintained
-Skeletal Muscle Movements still possible
--need to use with muscle relaxant
CNS effects of Ketamine
-increases EEG activity
--Common in dogs and non-domestic cats
-Increases cerebral blood flow, cerebral vasodilation
--increases ICP and CMRO2
-May see emergence delirium or dysphoria with recovery
--violent recovery
Respiratory Effects of Ketamine
-Mild, transient decrease in RR and Tidal volume
-Apneustic, shallow, irregular breathing pattern
--holds breath at peak of inspiration
-Bronchodilation, useful in asthmatics
Cardiovascular effects of Ketamine
-Ketamine Stimulates sympathetic nervous system and causes effects on heart
-indirect stimulation
-increased HR, CO, and BP
--Good for vasodilatory patients
-Inhibits neuronal NorEpi reuptake
-Pro-arrhythmogenic effects
-Direct negative inotropic and vasodilatory action
-Do not want to give to an animal in heart failure! Will get negative inotropic effects
Ketamine Adverse Effects
-increases salivation
-Increases muscular rigidity
-Decreases nerve conduction to muscles
Pharmacokinetics of Ketamine
-2-compartment open model
-Rapidly reaches peak CNS concentrations
-Rapidly redistributed due to lipid solubility
-Slower elimination phase, longer metabolism
--effects last longer than other drugs
Ketamine Metabolism
-very dependent on hepatic metabolism
-In cats is metabolized by the kidney also
--not in dogs
--cats do not metabolize to inactive compounds
-Metabolized to norketamine
--20-30% residual activity
-Hydroxylated to hydroxynorketamine (inactive metabolite)
-Glucuronide conjugation
-Renal elimination of metabolites
Norketamine
-Active metabolite of Ketamine
-Ketamine is transformed into Norketamine
-Norketamine levels increase continually as time goes on
--get a big build-up
Ketamine Administration
-Not used as a CRI at anesthetic levels, use as CRI in balanced anesthesia
-Can give IM or IV
-Common to give to difficult animals
-Dogs, pigs, horses are more susceptible to excitatory effects
-Co-administed with tranquilizers and/or central muscle relaxants
Ketamine and Muscle relaxant
-Give together
-Co-administed Ketamine with tranquilizers or central muscle relaxants
--guaifensesin, benzodiazepines
-Use as CRI
When to use Ketamine with Caution
-Animal is in cardiac failure and does not have sympathetic reserve
-Decompensated shock, causes hypotension
-Animals with tachyarrhythmias
-Pheochromocytoma (adrenal medullary neoplasia)
-Hyperthyroidism
-CNS excitatory signs
Tiletamine
- Telazol
-Analog of ketamine (similar chemistry and pharmacology)
-Dissociative anesthetic
-Longer lasting than ketamine in cats and horses
-May cause severe catalepsy and convulsions
-Violent recoveries in many species if not combined with tranquilizers
-Commercially available ONLY in combination with zolazepam (Telazol)
Potent Opioids
-Fentanyl, sufentanil, alfentanil, remifentanil
-Can be used in high dose in really debilitated animals to get a procedure done
-Will not give all anesthesia stages
--will give very sedate animal
-No adverse cardiovascular effects
-Short plasma half-life, fast redistribution and metabolism
-Slow onset, not an anesthetic
-Combine with muscle relaxant, otherwise will get CNS excitement
--will be reactive to light and noise still
-Usually combined with benzodiazepines
Alfaxalone
-Neurosteroid, progesterone-related steroid
-CNS depressant
-Hypnotic effects
-Modulates GABAa receptor
-Minimal cardiovascular effects
-Provides muscle relaxation
-Can give IM (water soluble)
-Smooth recovery
-Recently approved by FDA in US
Serotonin
-5-hydrocytryptamine (5-HT)
-Causes vasoconstrictor activity in serum after clotting
-Gut stimulating factor
--present in invertebrates and invertebrates
--Indole alkylamine
-Widespread distribution with multiple functions
-Hard to generalize, does so many different things
-Role as NT and role in psychopharmacology
Indole Alkylamine
-Serotonin Gut Stimulating Factor
-Enteramine
Serotonin Distribution
-Widely distributed
-In plants, animals, tissues,
-Present in venoms, stings
-90% is present in enterochromaffin cells in GI tract
--Kulchitsky cells
-Concentrated in platelets in blood
--platelets are active carrier, not synthesized in platelets
-Acts as CNS NT
-In mast cells of rodents and cattle, not humans
-Present in diet but rapidly metabolized
--active serotonin is synthesized in the body
Kulchitsky Cells
-IN gut
-Where most serotonin in the body hangs out
-Enterochromaffin cells of GI tract
Serotonin in Diet
-Present but rapidly broken down
-Active serotonin is synthesized in the body NOT taken from diet
Serotonin synthesis and Inactivation
-Tryptophan precursor
--tryptophan-5-hydroxylase forms 5-hydroxytryptophan
-Broken down by MAO-a and excreted in urine
-Most common breakdown product is 5-HIAA
-Serotonin is also precursor of the pathway to make melatonin in pineal body
--via separate pathway
Tryptophan-5- hydroxylase
-Forms 5-hydroxytryptophan from tryptophan
-Rate-limiting step in Serotonin synthesis
Melatonin
-Hormone
-Serotonin is precursor for melatonin
-Important for circadian rhythms, biological clock, sleep regulation
-Also acts as antioxidant
Storage and release of Serotonin
-Serotonin is Stored in secretory vesicles
--complex with ATP allows vesicles to manage acid/base control
-Exocytosis results in release
--"release reaction" of platelets
Serotonin Metabolism
-Metabolized by MAO (deamination)
--then oxidized or reduced and excreted in urine
-High affinity uptake in nerve terminals and platelets
Serotonin Receptor Diversity
-Many subtypes
--25 receptor subtypes in 7 major classes
-Mostly GPCR
-5-HT3 receptor is an ion channel (only ion gated channel)
-Identified based on affinities for 5-HT, Selective angatonists, distribution, and species-specificity
-Most receptors have been cloned
5-HT3 Receptor
-Ion-gated channel for serotonin
-Only ion gated serotonin receptor
-Monovalent cation channel
-Mediates fast depolarization responses
CNS functions of Serotonin
-Involved in making melatonin in pineal body, percursor of melatonin
-Important NT in raphe nuclei of brain
--Pain perception, Sleep, Autonomic functions (temperature, BP), Mood, sexuality, food intake, emesis
--low levels are associated with mood disorders
-Action is somewhat species-dependent
Serotonin Actions in CNS
-Acts as NT
-Perception of Pain
-Sleep
-Autonomic function, regulates temp and BP
-Mood
-Sexuality
-Food intake
-Emesis
Emesis and Serotonin
-Bad food ingested, enterochromaffin cells release Serotonin
-Platelets take up and release more serotonin
-high serotonin levels act on 5-HT3 receptors and cause vomiting
Neuroendocrine Effects of Serotonin
-Precursor for melatonin produced in pineal body
-Hypothalamic-hypophyseal secretion control
-Endocrine gland co-storage and release with polypeptide hormones
--thyroid
--pancreas
--Serotonin is released along with other hormones
Serotonin in GI system
-Peripheral nervous system, enteric nervous system
-Stored and released from Kulchitsky cells
-Contributes to enteric nevous system to control gastric secretion and motility
-Stimulates GI smooth muscle contraction
--facilitates tone, peristalsis, motility
--motility enhancing effect on 5-HT2 receptors, 5-HT2 agonists
-Basal release is augmented by mechanical stimulation, hypertonicity, acidity, NorEpi, and vagal tone
-Overproduction of serotonin in carcinoid tumor can lead to severe diarrhea
Serotonin in Cardiovascular System
-Promotes vascular smooth muscle contraction
-Acts on 5-HT2 receptor
--causes vasoconstriction of vascular smooth muscle
-No contraction in skeletal or heart muscle
--causes vasodilation of skeletal and heart muscle
Serotonin in Platelets
-Regulation of vascular tone
--Serotonin antagonists cause Anti-hypertensive action
-Bleeding time, clotting time, capillary resistance are not by serotonin depletion
How to alter Serotonin Levels
1. Target synthesis
2. Prolong activity by preventing breakdown
3. Change receptor activity
Behavior-modifying Activity of serotonin
-MAO inhibitors within presynaptic terminal prevent serotonin degradation
--prolong serotonin
-Tricyclic antidepressants act in synaptic cleft, reduce re-uptake
-Selective Serotonin Reuptake Inhibitors in synaptic cleft
Tryptophan and Serotonin
-Precursor to serotonin
-Tryptophan can cross BBB and get into brain, Serotonin cannot
-Serotonin is made in the brain by conversion of tryptophan
-Need enough tryptophan to get into brain to make serotonin
-Useful with PKU because tryptophan amine concentration can outbalance phenylalanine concentration
Chloramphetamine
-Blocks serotonin synthesis
-Will decrease serotonin levels in the brain
Serotonin re-uptake inhibitors
-Increase serotonin activity by prolonging the time serotonin is in the synaptic cleft
--leads to higher concentration of serotonin in synaptic cleft
-Tricyclic antidepressants
-SSRIs
-Ecstasy prevents uptake and induces high serotonin secretion
--post-synaptic overload/flooding effect
MAO inhibitors
-Prevent monoamine NT breakdown in synaptic terminal
-Increase serotonin availability
Tricyclic Antidepressants
-Block uptake of serotonin at pre-synaptic terminal
-Inhibit reuptake by transporter protein on presynaptic cell
-Also partially inhibits NorEpi reuptake by blocking amine pump
-Amitryptiline HCl
--treats behavioral conditions, separation anxiety, spraying, self-mutilation
--Tx for pruritus
Amitryptiline HCl
-Tri-cyclic anti-depressant, serotonin uptake inhibitor
-Treatment for behavioral conditions
--separation anxiety, spraying, self-mutilation, excessive grooming
-Adjunctive treatment for pruritus
-Treatment for neuropathic pain
Clomipramine HCl
-Tri-cyclic serotonin uptake inhibitor
-Anti-depressant
-Used for treatment of obsessive-compulsive disorders
--urine marking, mounting, inter-male aggression
Imipramine HCl
-Tricyclic Serotonin uptake inhibitor
-Treats anxiety disorders in animals
-Separation anxiety
-Urinary incontinence
-Ejaculatory dysfunction
-Cataplexy (narcolepsy attacks)
Specific Serotonin Reuptake Inhibitors
-more selective for serotonin
-Little to no effect on other NT (Dopamine, NorEpi)
-Good response for reversing depression
-Fluoxetine HCl
-Paroxetine HCl
-Sertraline
Fluoxetine HCl
-SSRI
-Treatment for anxiety disorders in dogs
-Can change blood levels of other medications due to CYP450 inhibition
--esp. diphenhydramine and propanolol
-Treats aggression, Obsessive compulsive disorders, destruction, excessive barking
-treatment for inappropriate elimination and pruritus
MAO inhibitors
-Prevent monoamine NT breakdown within the synaptic terminal
-Not very specific to serotonin, will break down NorEpi also
-MAO-a
-MAO-b
MAO-a
-Mostly serotonin and NorEpi degradation
-Selectively inhibited by Clorgyline
--irreversible inhibition
-Moclobemide is similar to SSRI
--reversible inhibition
MAO-b
-Mostly dopamine degradation
-Selective inhibition of dopamine
-Inhibitors can slow down degradation of neurons with age
Serotonin Receptor Agonists
-Cisapride: Stimulates 5-HT4 receptors, GI disorders
-Sumatriptan
-Buspirone: 5-HT1a, alleviates anxiety and behavior disorders
-Ergot Alkaloid Derivatives: helpful for migraine headaches
Cisapride
-5-HT4 antagonist
-Was used in treatment for GI reflux and GI stasis
-Since found to be toxic
"Triptans"
-Sumatriptan
-5-HT1D and 5-HT1B agonists
-Expensive
Buspirone
-5-HT1A agonist
-Anxiolytic
-Treatment for behavior disorders related to fear or phobias
Ergot Alkaloid Derivatives
-Serotonin receptor agonists
-Partial agonists and antagonists at numberous 5-HT receptor sub-types
-Effective treatment for acute migraines
Phenoxybenzamine
-A-adrenergic blocker and 5-HT2 antagonist
-Treats functional urethral obstructions
--Decreases sympathetic-mediated urethral tone
-Short-term treatment for hypertension
-Can treat laminitis in the developmental phase
Cyproheptadine
-H1 agonist
-5-HT2 antagonist
-Anti-histamine
-Treats skin allergies and pruritus
-Acts as appetite stimulant
-Reduces cortisol overproduction in equine cushing's disease
--overproduction of ACTH release due to pituitary tumor
Ketanserin
-Serotonin Receptor blocker 5-HT1 and 5-HT2
-Blocks platelet aggregation promoted by 5-HT
-Anti-hypertensive (experimental)
-Unclear mechanism of action
Dolasetron Mesylate
-Serotonin receptor antagonist, 5-HT3 antagonist
-Anti-emetic drug
-Blocks 5-HT3 receptors
Ondansetron
-Blocks 5-HT3 receptors, serotonin receptor antagonist
-Anti-emetic,
Psilocybin
-5-HT1a and 5-HT2 agonist
-Compound stimulates receptors
-Alters consciousness
Serotonin Syndrome
-Serotonin poisoning caused by combination of drugs that have additive or synergistic effects on serotonin levels
-Contraindicated Combination of antidepressants and opiates
-CNS stimulants in combination with something else
-Treatment: Benxodiazepines
--stimulate GABA activity in brain, counteract hyperstimulation of serotonin system
Autacoids
-“Local Hormones”
-Biologically active amines
-Present in many tissues throughout the body
-Histamine, Serotonin, endogenous peptides, prostaglandins, leukotrienes, cytokines
-Complex physiologic and pathologic effects through multiple receptor types
-Can be released together locally
Biosynthesis of Histamine
-Histidine is converted into Histamine via histamine decarboxylase
-2 breakdown pathways
--histamine to N-methylhistamine via N-methyltransferase
--histamine to Imidazoleacetic acid via Diamine oxiase/histaminase
-Broken down pretty quickly
-Has robust biological actions, do not want it to linger
-Orally ingested histamine is quickly metabolized and eliminated
Clinical Importance of Autacoids
-Broad and undesirable effects
-No clinical application for histamine or serotonin
-Imbalances in synthesis or release cause pathological conditions
--inflammation, allergies, hypersensitivity, ischemia-reperfusion injury, cardiovascular hypertension
-Compounds selectively activate/atagonize receptor sub-types that are clinically useful
Histamine
-2-(4-imidazolyl)ethylamine
-Hydrophilic vasoactive amine
-Naturally present in plants and animals
-In mast cells and stomach mucosa
--concentrations vary
--species and tissue specific
-4 different GPCR receptors
Histamine Major Physiological roles
1. Immune response and inflammation
2. Regulates gastric acid formation
3. NT or neuromodulator
Histamine Metabolism and Elimination
-2 breakdown pathways
-All excretion is renal
Histamine Receptor Subtypes
-H1, H2, H3, H4
H1 Histamine Receptors
-G-protein coupling to Gq
-Increases IP3, DAG
--increases Ca and cAMP in the cell
-On smooth muscle cells and endothelial cells
-in CNS
-2-methyl-histamine is agonist
-Pyrilamine is antagonist
H2 Histamine Receptor
-G-protein coupled receptor via Gs
--increases cAMP in the cell
-On gastric parietal cells, cardiac muscle, mast cells, and in CNS
-Agonist is Dimaprit, impromidine, and anthamine
-Antagonist is Ranitidine, cimetidine
H3 Histamine Receptor
-GPCR, activates Gi
-Decreases cAMP levels in cell
-In CNS, presynaptic terminals, in myenteric plexus of enteric NS
-important for arousal in the brain
-Agonist is R-a-methyl-histamine, imetite, and immepip
-Antagonist is Thioperamine, clobenpropit, iodophenpropit
H4 Histamine Receptor
-GPCR, acts via Gi
--increases Ca and decreases cAMP in cells
-On Eosinophils, neutrophils, and CD4-T cells
-Agonist is Clobenproprit, imetit, clozapine
-Antagonist is Thioperamide
Histamine Release
-Synthesized in cells and packaged into granules
-Stored in specialized vesicles/granules
--mast cells
--basophil granulocytes
--enterochromaffin-like cells in stomach muscosa
-Rapid release of granules into the interstitium, stimulated by tissue injury or trauma, physical agents, chemical compounds, or immunological factors
Mechanisms of Histamine Release
-Tissue injury or trauma
-Physical agents: heat, cold, x-rays
-Chemical Compounds: morphine, tubocurarine
--displace histamine in granules causing release
-Immunological response: Fc epsilon RI high-affinity receptor for IgE
--respont to tissue damage or immune actions
Histamine Triple response
-Released from mast cells
-Causes vasodilation, allows increased blood flow and increased infiltration
1. Small red dot: local reaction, local dilation of blood vessels
--immediate site of injection
2. Large red flare surrounding original red spot
3. Wheal around red spot
--Edema due to increase of vascular permeability
--separation of endothelial cells

Response can be inhibited by prior administration fo H1 antagonists (anti-histamines)
Local effects of Histamine Release
-Dilation of capillary venules (H1 receptor)
-Activates endothelium, increases blood vessel permeability
--also increases permeability for WBCs
-Chemotactic attraction of inflammatory cells
--neutrophils, basophils, eosinophils, monocytes, lymphocytes
-Irritates nerve endings
Roles of mast Cells in Disease
1. Allergic diseases: asthma, eczema, allergic rhinitis or conjunctivitis
--Histamine causes more problems than initial insult
2. Anaphylaxis: Strong systemic activation as reaction to allergens
--body-wide degranulation of mast cells
--causes mass vasodilation and shock
--Other factors also involved
3. Immunity: Recruitment of inflammatory cells to skin or joints
--integral part of innate immunity
Disorders of Mast Cells
-Mastocytosis: rare over-proliferation of mast cells
--can be cutaneous or systemic
-Mastocytoma: Mast cell tumor, often seen in dogs and cats
General Histamine Effects
-Powerful effects on smooth muscle and cardiac muscle
-Effect on nerve cells
-Endothelial cells
-Stomach secretory cells, promotes acid secretion
-Sensitivity to histamine varies species to species
-Guinea pigs, dogs, cats, humans have high response to histamine
-Mouse and rats have low response to histamine
Histamine Effects in Nervous System
-Stimulation of sensory nerve endings to cause pain and itching
--nettle and insect stings
--H1 mediated response
-Modulation of respiratory neurons
--breathing, sleep, wakefulness
--H1 and H3 mediated response
-Presynaptic H3 receptors modulate transmitter release
--Agonists reduce release of ACh, amines, peptides in brain and peripheral nerves
Histamine Effects on Cardiovascular System
-Decreases systolic and diastolic blood pressure
--due to dilation of arterioles and NO release
-Increased HR, tachycardia
--stimulates heart and reflex tachycardia
-Increased contractility in myocardium escept for atrial muscle
-Flushing, sense of warmpth in peripheral tissues due to extra blood flow
-Can cause headaches due to vasodilation
-Local sites of increased blood flow, Urticaria (Hives)
-Some cardiovascular effects are involved in anaphylaxis, other effects are more important
Time response of histamine on Cardiovascular System
1. Drop in cardiac output and blood pressure
2. Breif compensatory tachycardia, then consistently increased HR
3. Increase in peripheral resistance
4. Can cause death within hours
--50% lethality after 4-6 hours
Effect of histamine on the bronchiolar Smooth Muscle
-Causes bronchoconstriction via H1 receptors
--can cause death in guinea pigs
-Asthma pateints are much more sensitive to histamine, 100-1000x more sensitive
-Causes bronchodilation in rabbits and some other species
--H2 receptor effects predominate over H1 receptors
Histamine effects on GI tract smooth muscle
-Contraction of smooth intestinal muscle
-Mediated by H1 receptors
Histamine effects on Uterine Smooth Muscle
-Some species are sensitive
-Can have histamine-induced contractions
--may lead to abortion
Secretory effects of Histamine
-Powerful stimulant of gastric acid secretion
-increases production of pepsin and intrinsic factor
-H2 receptor activated action on gastric parietal cells
--increases cAMP, Ca
-Stimulates intestinal secretion
-H3 agonists can inhibit gastric acid secretion
--stimulated by food or pentagastrin
Role of Histamine in Gastric Acid Secretion
-Parietal cells secrete HCl into lumen
-Histamine acts of H2 receptors on parietal cells
--increases in cAMP and Ca are blocked by H2 antihistamine drugs, NOT blocked by H1 antihistamine drugs
-Parietal cells also respond to Ach, Gastrin, and SSK
Histamine Toxicity
-Bacteria can over-produce histamine
--spoiled fish or meat can have increased bacteria
-Symptoms:
--flushing, hypotension, Tachycardia, headache, hives, wheals, urticaria, bronchoconstriction, GI upset
-Animals with asthma are hyper-sensitive to histamine, should not be given histamine ever
-Patients susceptible to GI bleeding are also susceptible
Histamine Antagonists
1. Physiological Antagonists
2. Release Inhibitors
3. Receptor Antagonists (antihistamines)
4. Histidine decarboxylase inhibitors (in the works)
Physical antagonists of Histamine
-Ideally works through a different mechanism to have opposing effect
-Ex: Epi in anaphylaxis
--Causes smooth muscle action opposite of histamine
-Different receptor
-Clinically has life-saving action in acute analhylacis and rapid bronchodilation
-H1 receptor antagonists are only suitable prophylactically, need to use epi for acute cases
Release Inhibitors of Histamine
-Reduce immunologically triggered mast cell degranulation
-Reduce histamine release
-Cromoglicate, nedocromil for asthma treatment
-Beta-2 adrenergic receptor agonists also reduce histamine release
Histamine Receptor Antagonists
Anti-histamines
-Directly block histamine receptors
-Mostly H1 receptor antagonists
--little to no effects on H2 and H3 receptors
-Side effects intensity varies with compound
Common side effects of Antihistamines
-Sedation, drowsiness
-Dryness of mucous membranes
--mouth, nose, throat
-Skin allergies with topical treatment
-GI motility is affected
-Effects on muscarinic, cholinoreceptors, serotonin, or local anaesthesia receptors
-Intensity varies with compound
Categories of H1 receptor Antagonists
1st generation
-Ethanolamines: Diphenhydramine (Benadryl)
-Ethylaminediamines: Pyrilamine (Histall, H)
-Piperazine derivatives: Hydroxyzine (Ifosfamide, Ifex)
-Alkylamines: Chlorpheniramine Maleate
--alone or in combination with analgesics or antibiotics
--less sedating than other 1st generation agents

Strong sedative side-effects, anticholinergic
Categories of H1 receptor Antagonsts
2nd Generation
-Less lipid-soluble, less entry into CNS (or no entry)
-Less sensitive action
-Ethylenediamine: Tripelennamine (TBZ, Fenistil)
-Chlorpheniramine, Serotonin/Epi re-uptake inhibitors
--mild anti-depressants
-Loratadine (Claritin)
-Cetirizine (Zyrtec)
-Fexofenadine (Allegra), Terfenadine (Seldane)
Uses for H1 receptor antagonists
-Prevents bronchoconstriction
--NOT a bronchodilator! Use epi as acute bronchodilator!
-H1 receptor antagonists do NOT block gastric secretion
--only H2 receptor antagonists
-Treatment of allergic reactions
-Motion sickness and vestibular disturbances
-Do not block gastric secretion
--H2 receptors, need H2 antagonists
H1 receptor Antagonist Side effects
-Anticholinergic side effects
-Hyperthyroidism
-Cardiovascular Disease
-Use with caution in patients with angle closure glaucoma, prostatic hypertrophy, pyloroduodenal or bladder neck obstructions, hyperthyroidism, cardiovascular disease, or hypertension
Diphenhydramine Uses
-H1 receptor antagonist, Benadryl
-Bug bite allergies
-Bee stings
-Dermatitis in dogs
-Reverse sneeze syndrome
-Agitation associated with pruritus allergies
-Transient behavior management (car travel)
-Prevention of motion sickness, antiemetic
-Aseptic laminitis
-Adjunctive therapy of anaphylaxis in cattle and horses
H2 receptor Antagonist Uses
-Cimetidine, nizatidine, ranitidine
-Treatment for duodenal ulcers or stress-induced gastric ulcers
-Upremic gastritis, stress-related erosive gastritis and esophagitis
-Duodenal gastric reflux
--promotes gastric emptying and GI motility
-Reduces gastric acid secretion and helps with gastric acid reflux
-Prevents secretion from endocrine adenomas
H2 receptor Antagonist Side effects
-Toxicity is rare
-Can cause issues with older patients or patients with hepatic or renal insufficiency
Ranitidine
-Reduces gastric acid production
-Can be used to treat ulcers, esophagitis, chronic gastritis, gastrinoma, hyperhistaminemia due to mast cell tumors, hypergastrinemia due to chronic renal failure
-Pathologic hypersecretory conditions can be reduced
-Ranitidine is more effective than cimetidine
Local Anesthetic Molecule
-Weakly basic in nature
-Water-soluble salts
-low pH, 4.4-6.4
-Usually already diluted
-Lipophilic benzene ring and hydrophilic quaternary amine
-Can have an ester or amide linkage
--Esters have been moved away from, amides are mainly used
--longer chain leads to increased potency and toxicity
Local Anesthetics Tertiary Form
-Uncharged, free base
-Poorly soluble in water
-Crosses lipid barriers
Important details of Local Anesthetic Molecules
-Potency, Onset, and Duration are most important
-Potentcy is related to lipid solubility
-Onset time is related to degree of ionization
--closer the pKa of the anesthetic is to the tissue pH, more rapid onset
--important clinical application
-increased protein binding prolongs duration of anesthetic in body
Local Anesthetic Site of Action
-Work by blocking Inotropic Na channels
--Na channels are all over the body, how body propagates AP in nerves
-Overdose of local anesthetic will block how nervous system functions
--can cause major issues!
Local Anesthetic Site of Action Modern Theory
-traverse membrane, get protonated, protonated form blocks Na channel from the inside
-Local anesthetic acts like a cork, does not let Na through the channel
-Binding impedes propagation of action potentials
-Cause conduction blockade
Na Channel Mechanism of Action
Channel States
-Na channel is either "Open" or "Closed"
-If closed, can be resting or inactivated
-States depend on membrane potential
-Ion conducting state is "open"
--open for less than 1 millisecond
-Non-conducting/resting/inactivated exists for several milliseconds
Na Channel "Open" State
-Channel is open, ions can flow through
Na Channel "Inactivated/Resting/Non-conducting" state
-Cannot readily re-open form inactivated states
-Have to recover, or re-prime after hyperpolarization membrane potentials to become available
-Channels bind drug when they are depolarized and inactivated
-Channels release drug when they regain hyperpolarization and reach rested state
Vasomoter tone
-Keeps body functioning
-Baseline sympathetic tone on organs in the body
-Tone always changes but is always present
-Maintains pressure
-Fibers are always transmitting
--easy to catch in a state where drugs will have an effect
--always active, always have the chance to affect them
Repetitive depolarization of Na channels
-High affinity with repetitive depolarization
-"Use-dependent block" or "phasic block"
-More intense block
-Affects fibers that are most active
--stronger block in areas that are painful
-Excitable tissues show aberrant late Na currents responsible for neuropathic pain, dysrhythmias, epilepsy
Lidocaine as anti-arrhythmic drug
-Blocks foci in heart that are repetitively firing
Vasomotor tone and phasic block
-Reason why current local anesthetics have cardiovascular toxicity
-Lidocaine has vascular
-Bupivacaine is more cardiovascular
-Affect vasomotor fibers and change impulse transmission through sympathetic trunk
-Blocks a sector of spinal cord that controls transmission of impulses to a certain splanchnic area
--area will collect more venous blood
--removes blood from circulation
-Impedes venous return to the heart
--decreases CO, hypotension, cardiovascular instability
-Young animals can tolerate, older/sick animals cannot
Lidocaine vs Bupivacane with Cardiac Dysrhythmias
-Cardiac coltage gated channels are 100-1000x more sensitive to lidocaine than neural channels
-No systemic neuronal interruption with antiarrhythmic effects
-High activity of cardiac channels promotes high-affinity slow-inactivated sate and actions of local anesthetics on them
Local Anesthetics Effects besides Na Channels
-Serotonergic and cholinergic receptors also contribute to epidural and spinal anesthesia and behavioral changes
-Anesthesia/sedation
-Drowsiness and euphoria
--inhibitory actions on excitatory synapses
--inhibitory actions on inhibitory neurons
-Cocaine and procaine can antagonize behavioral actions of nicotine
-Cocaine and other local anesthetics can induce seizures
--inhibit anti-nociceptive effects of nicotine
--inhibit nicotine-induced motor impairment
Factors affecting Local Anesthetic Pharmacokinetics
-Proximity of injection to nerve
-Anesthetic flow around neural tissue
--flow of blood and lymphatics take local anesthetic away from intended site, decreases concentration
-Diffusion across barriers into tissue
--depends on pH, pKa, concentration, and lipid solubility
-Binding of local anesthetic to non-neural sites
-Absorption into vascular and lymphatic systems
Local Anesthetic Absorption
-Even locally, will end up being absorbed and will end up acting systemically
-Systemic system serves as depot for drug
-Different tissues have more or less absorption
--muscle and intercostal speces have most sytemic absorption
Lidocaine in the Lung
-Pharmacokinetics are altered by the lung
-Lung absorbs anesthetic and drops the plasma levels of the drugs
-Helpful to decrease systemic absorption
-Venous blood goes to lung, leads up uptake by the lung
Lidocaine Elimination
-Metabolized in liver via aromatic hydroxylation, N-dealkylation, and amide hydrolysis
-high extraction ratio: more than 70% of drug is cleared from plasma in one circulation through the liver
Extraction Ratio
-Amount of drug that is removed from plasma circulation by the liver
-Liver metabolism depends on blood flow more than enzymes
-Liver function does not really affect metabolism of local anesthetics
-Drugs with high-extraction ratio depend on blood flow, are not affected by liver function
--generally safer
-Drugs with low extraction ratio depends on cytochromes and enzymes, are affected by liver function
Local Anesthetic Toxicity
-Depends on rate of absorption and metabolism
-Toxicity is additive
--multiple drugs can have additive effect, need to be careful
-Allergic reactions, mostly with ester local anesthetics
-Neurotoxicity
-Methemoglobinemia
-CNS toxicity
--can lead to unconsciousness, seizures, and coma
--only an issue with certain local anesthetics
Cardiovascular Toxicity of Local Anesthetics
-Associated with local anesthetic potency
-Acidosis, hypoxia, pregnancy, K, addition of epinephrine
-Dose related, decreases cardiac contractility
-Can constrict or dilate vasculature
Hypercarbia and local anesthetics
-Animals under anesthesia do not breathe well
-With respiratory distress comes respiratory acidosis
-Toxicity of drug is higher
-Want to keep animal away from an acidic state
Bupivicaine IV
-NEVER USE IV!
-Can kill a healthy young animal with 1 dose
-Related to potency
Bupivicaine use
-4x more potent nerve block
--lasts much longer, 4-6 hours
-70x more potent cardiovascular toxicity
-Delays conduction
Toxicity of Local Anesthetics in Awake Dogs
-Determined by a study on awake dogs that increased dosages and looked at resulting signs
-When dog is anesthetized, can give more local anesthetic because anesthetic agents work as muscle relaxant and decrease muscle tremors
-Lidocaine has less toxicity that bupivicaine
Toxicity of Local Anesthetics in Anesthetized Dogs
-Toxic dose is MUCH higher in anesthetized dogs
-In awake dogs, CNS effects will be more dramatic before cardiovascular effects are seen
Local Anesthetic Toxic effects
-CNS effects in awake animals
-Cardiovascular effects in anesthetized animals
-Different doses used for awake vs. anesthetized animals
Local Anesthetic Toxicity in Cats
-Need to be very careful in cats
-Small V1 compartment
-Toxic effects are very dependent on plasma levels
-Have different metabolism and different compartments leading to different distribution
--different pharmacokinetics
-Will often see side-effects if not careful
-Usually only give lidocaine to treat arrhythmias, and even then cats do not respond very well
Cardiovascular Toxicity of Local Anesthetics
-Lidocaine is used IV very often
-As doses increase, difference in mean arterial blood pressure is not that evident
-Therapeutic/clinical doses of lidocaine are cardiovascular sparing
-At lower doses, CO actually increases
-Safe drug to use in cardiovascularly unstable patients as long as you stay away from toxic doses
Treatment of Local Anesthetic Toxicity
-Use lipid emulsions
-Lipid acts as a sink, local anesthetic is trapped in the lipid
-Very effective, especially with cardiac arrest
Esters vs. Amides
-Esters have more allergic reactions
-Amides have more CNS and cardiovascular toxicity at high doses
-Esters are hydrolyzed by plasma cholinesterase
--produces allergic metabolite
-Amides are degraded in the liver
Lidocaine
-Very old drug, 2nd local anesthetic amide to be synthesized
-Short onset and long duration
-Hypoallergenic
-Main local anesthetic used
-Only downfall is short duration, only reason you would use something else
-has vasodilatory effects
Special Effects of Lidocaine
-Analgesia and MAC sparing-effects
--Can decrease concentration of inhalant anesthetics used at the same time
-Cardiovascular sparing
-Anti-arrhythmic effects
--similar to procainamide
-Useful in neuropathic pain
-Anti-endotoxic
-Prokinetic, can be beneficial for GI system
--used pre-surgical or post-surgical in colic situations
Lidocaine as anti-endotoxic
-Beneficial for re-perfusion injury
-Na enters cells, but with no ATP to pump Na out will switch and pump Ca out
--causes necrosis
-Giving lidocaine blocks Na channel and decreases amount of Na that goes into cell
-Will not have a lot of Ca flux
Functions of the GI tract
-Continual supply of water, elecrtolytes, and nutrients
-Highly regulated movement
-Most drugs are absorbed in the small intestine, NOT stomach
--need to be stable to get through the stomach
Stomach Anatomy
-Fundus
--cardia
--esophagus
-Corpus/Body
--rugae increase surface area
-Antrum
--pyloric antrum
--pylorus
Storage of the Stomach
-Muscle plasticity
--stretches to accommodate load
-Vagal reflex inhibits muscle activity during filling
-Competitive eaters control vagal reflex
Mixing action of the Stomach
-Regulated by vagal nerve
-Slow constrictor waves in the body of the stomach
-Peristaltic constrictor waves in the antrum
-Regulation of waves by vagal nerve activity
Emptying of Stomach
-Coordinated action
-Pyloric pump and musculature in the antrum
-Solids must be less than 2mm to leave the stomach and enter duodenum
-Pyloric sphincter
-Regulated by food in the stomach, gastrin, enterogastric reflexes, and hormonal feedback
Cell Types in the Stomach
-Superficial epithelial cells produce mucin and HCO3
-Mucous neck cells produce mucin and pepsinogens
-Stem cells/regenerative cells
-Parietal cells produce HCl and intrinsic factor
-Chief Cell produces pepsin
-Endocrine cells
Intrinsic Factors
-glycoprotein that allows for vitamin B12 absorption in ileum
Gastric Acid Functions
-Protein digestion
--pepsin preferentially cleaves proteins at carboxylic groups of aromatic AAs
-Iron absorption
-Inactivation of bacteria, viruses, and parasites
Pepsin production
-Pepsinogen is cleaved at low pH (les than 3.5)
-unfolds, clips part of self, and becomes active
-Activated pepsin can covert pepsinogen into active pepsin
-Increasing concentration of pepsin will increase pH, and pepsinogen will no longer be activated
-Pepsin preferentially cleaves at carboxylic groups
Regulation of Stomach Acid Secretion
-H/K ATPase proton pump is key factor
-Puts H into intestinal lumen
-brings K into cell cytoplasm
-H comes from H2O
Transmitter Systems involved in Regulation of Stomach Acid
-Gastrin: acts on CCKb receptor, increases acid secretion
-Histamine: Acts on H2 receptor, increases acid secretion
-ACh: acts on muscarinic M3 receptors, increases acid secretion
-Prostaglandin: acts on EP2 receptors, decreases acid secretion
-Somatostatin: acts on SST receptors, decreases acid secretion
Gastrin and Stomach Acid Secretion
-Peptide
-Made in G-cells in antrum of stomach
-Acts on CCKb receptor to increase acid secretion
--GPCR to activate IP3 and Ca release
-Activates H/K ATPase pump to increase secretion
-Direct stimulation via ACh and CCkb receptors
-Indirect stimulation as well
Histamine and Stomach Acid Secretion
-Released from enter-chromaffin cells
-Binds to histamine receptors (H2 receptor)
--activates Gs protein to up-regulate cAMP
--leads to increased action of PKA, stimulates H/K proton pump
-Increases acid secretion
ACh and Stomach Acid Secretion
-NT released from vagal nerve
-Binds to muscarinic M3 receptor
-Increases acid secretion
Prostaglandin and Stomach Acid Secretion
-Acts on Gi protein, inhibitory
Somatostatin and Stomach Acid Secretion
-Acts on Gi protein, inhibitory
Direct stimulation for Gastric Acid Secretion
-Vagal nerve
--releases ACh directly onto parietal cells
-Gastrin binds to CCK receptor on parietal cells
Indirect Stimulation for Gastric Acid Secretion
-Vagal neve releasing ACh onto enterochromaffin-like cells
--releases histamine
-Gastrin binds to CCK receptor on ECL cell
Cephalic Phase of Gastric Acid Secretion
-Automated conditioned reflexes
-Comes from the brain
-based on smell, taste, chewing, swallowing
-Also hypoglycemia acts as a trigger
-Acid is released before food is even in stomach
Gastric Phase of Gastric Acid Secretion
-Distention sends signal up to brain
-Brain send signal back to increase acid secretion
-Once food is present, AA and peptides activate
Gastric Ulcers
-Go through mucosa into submucosa
Helicobacter pylori
-Gram- bacillus bacteria, colonizes antral mucosa of the stomach
-Can cause gastric ulcers
--60% of gastric ulcers
--90% of duodenal ulcers
-Antral inflammation may decrease somatostatin release from antral D-cells
--Decreased somatostatin results in increased gastrin release, increases acid production
Treatment of gastric Ulcers
-Amoxicillin
-Clarithromycin
-Proton pump inhibitor

One week therapy in adults
NSAIDs and gastric ulcers
-NSAIDs are non-selective COX-1 and COX-2 inhibitors
-Prevent formation of PGE and Arachadonic acid
-PGE play pivotal role in maintaining gastric mucosal integrity
-Blocking prostaglandin synthesis can lead to gastric ulcers
PGE and Gastric Mucosal integrity
-Pivotal role in maintaining gastric mucosal integrity
-Inhibit gastric acid secretion
-Stimulate HCO3- secretion
-Stimulate mucous secretion
-INcrease mucosal blood flow
-Modify local inflammatory response caused by acid
Rofecoxib
-Vioxx
-Selective COX-2 inhibitor
-Withdrawn in 2004 due to increased risk for heart attack and stroke
COX-1
-Mediates synthesis of PGEs responsible for protection of the stomach lining
-Do not want to inhibit
COX-2
-mediates the synthesis of PGEs responsible for pain and inflammation
-Want to inhibit
-COX-2 specific inhibition may minimize gastric ulcers
Deracoxib
-Deramaxx
-Selective COX-2 inhibitor
-Pain and inflammation after operations
-Used long-term for pain and inflammation associated with osteoarthritis
-Large doses can cause COX-1 inhibition and gastric issues
Gastric Mucosal Defense Systems
1. Mucosal cell renewal
2. Prostaglandins
3. Mucosal Blood Flow
4. Mucus Secretion
5. HCO3- secretion
Normal gastric Epithelium
-Mucus acts as a barrier to H and pepsins
-traps alkaline solution
-Acid generally tends to stay in the lumen
-Keeps mucosa buffered and safe from pepsin digestion
Damaged mucosal barrier
-Mast cell degranulation and release of histamine is bad news
--inflammatory response
Therapy for Gastric Erosion and gastric Ulcer
1. Dietary management
2. Antacids
3. Chemical diffusion barriers
4. ACh receptor antagonists
5. Histamine receptor antagonists
6. Gastrin receptor antagonists
7. H, K, ATPase agonists
8. Prostaglandins
Dietary Management and Gastric Ulcers
-Provide adequate nutrition
-Maintain continuous neutralization of gastric acid with food
-Minimize acid secretions
-Reduce mechanical, thermal, and chemical irritation to the gastric mucosa
Antacids and gastric Ulcers
-Neutralize acid in the gastric lumen
-Rapid onset and short duration
--starts in 30 min, lasts 2-3 hours
-Mainly reduce pain associated with acid secretion
--prevents HCl from reaching nerves and illicit pain
-Binds HCl in lumen, neutralize
Sodium Bicarbonate Antacid
-Alka-Seltzer
-Water soluble
-Can be absorbed systemically
--leads to alkalosis and potentially kidney stones
-Rapid generation of CO2, can lead to gastric distention and discomfort
Calcium Carbonate Antacids
-Tums
-Can neutralize 2 molecules of HCl
-Increases Ca load in the urine
--can lead to development of kidney stones
--issue with renal patients
-Antacids with Ca can cause constipation
Magnesium Hydroxide
-Milk of Magnesia
-Magnesium containing antacids
-Cause diarrhea, acts as laxative
-up to 20% of Mg can be absorbed systemically
--can cause hypermagnesia in renal patients
-Mg can accumulate in patients with renal disease, causes hypermagnesia
-Ion imbalances leads to cardiac problems and CNS depression
Acid Rebound
-If stomach pH is increased (above pH4) stomach is stimulated to secrete acid to maintain low pH
--compensatory HCl secretion by stomach
-Aluminum salts do not raise gastric pH, do not stimulate compensatory HCl secretion
-Mg salts raise gastric pH, can stimulate compensatory HCl secretion
-If stop taking Mg salt antacids, can cause acid rebound
--compensatory HCl secretion
Aluminum Hydroxide Antacid
-Maalox
-Aluminum-containing antacid
-Constipation is a side-effect
-Al can form insoluble Al-Phosphate complexes
--Can lead to hypophosphatemia with chronic use
Magaldrate Antacid
-Riopan
-Antacid contains mix of Al and Mg
-Constipation and laxative effects are off-set
Chemical Diffusion Barriers
-Coat irritated area of the stomach, forms physical barrier against stomach acid
-Mixture of sucrose octasulfate and AlOH
-Sucrose octsulfate polymerizes and forms a paste
--binds to damaged gastric epithelial cells
-Binds and activates bile salts and pepsin
-Stimulates prostaglandin synthesis
-Increases mucosal blood blow by increasing vasodilatory NO production
--increased NO leads to vasodilation
Bismuth Subsalicylate
-Pepto-bismol
--cleaved to bismuth and salicylate in intestine
-Bismuth coats ulcerated mucosal surfaces, absorbs toxins, and has mild antibacterial action
-Salicylate has anti-inflammatory action
-Do not use in cats due to salicylate and inefficient hepatic metabolism in cats
--leads to toxicosis
Montmorillonite
-Diarsanyl
-Contains montmorillonite, smectite clay
-Combines with mucus to form protective layer on intestinal mucosa to help reduce irritation and fluid loss
-High capacity to abosorb toxins, bacteria, viruses, enzymes, free radicals
--more absorbent than kaolinite
-Given as paste containing dextrose and glycerol
ACh receptor Antagonists and Stomach Acids
-Pirenzepine
-M1-selective antagonist, decreases antimuscarinic-mediated side effects
-Exact location of M1 receptors are not know, but work
-M3 receptors on parietal cells
Anti-Histamines and Stomach Acids
-Histamine Receptor Antagonists
-H2 antagonist
H1 location and action
-Smooth muscle and endothelium
-In CNS
-Promotes vasodilation, bronchoconstriction
-Smooth muscle activation
-CNS activation
H2 location and action
-Parietal cells
-Regulates histamine-mediated gastric acid secretion
H3 location and action
-Inhibitory autoreceptor
-Regulates histamine release
Cimetidine
-Cimetidine (Tagamet)
-Prototypical H2-selective antagonist
-Suppresses gastric acid secretion without H1 side-effects
--no drowsiness
-Inhibits many CYP450 enzymes
-Numerous drug interactions can occur due to CYP inhibition
--Coumadin, benzodiazepines, oral contraceptives
-Decreases hepatic blood flow
-Can decrease clearance of flow-limited drugs
--propranolol, lidocaine
Ranitidine
-Zantac
-2nd generation H2-selective antagonist
-H2-selective antagonist
-Much less CYP450 inhibition than cimetidine
-Longer lasting
-10x more activity
Famotidine
-Pepcid
-3rd generation H2-selective antagonist
-Poor bioavailability, 37%
-30x more potent than cemitidine, more potent than ranitidine
-No CYP450 inhibition
-Pro-motility effects on the stomach
Pepsid Complete
-Famotidine, CaCO3 antacid, and MgOH antacid
-Take effect immediately due to CaCO3 and MgOH, famotidine has long-lasting effect
-Ca antacids have constipation effect and Mg antacids have laxative effects
Nizatidine
-4th generation H2-selective
-No CYP450 inhibition
-Excreted unchanged in the urine
-Has pro-motility effects on stomach
Gastrin Receptor Antagonists
-Experimental CPDs
-High affinity selective antagonists to CCK receptor
-Effective in animal models
-Inhibits gastrin-stimulated gastric acid secretion
-Decreased ethanol-induced damage to gastric mucosa
-Adjunct therapy?
Proton Pump Inhibtors
-Omeprazole (Prilosec)
-Lansoprazole (Prevacid)
-Esomeprazole (Nexium)
-Rabeprazole (Aciphex)
Omeprazole
-Prilosec
-Weak base, unstable at low pH
--needs to be coated to survive stomach
-Absorbed in alkaline pH of duodenum, low pH
-Once inside low pH environment of parietal cell, becomes protonated
-Protonated form covalently and irreversibly binds to and inhibits proton pump
-One dose lasts 2-3 days due to concentration of drug and irreversible inhibition of the pump
-Decreases pump numbers, decreases total acid produced
Prolonged use of Omeprazole
-Prolonged decrease in gastric acid production
-Will increase stomach pH (more alkaline)
-Allows GI bacterial growth
--risk of pneumonia in certain patients
-Sustained HCl reduction can lead to increased gastric levels
--hypergastrinemia
--need to co-administed CCKb antagonist?
-Has caused parietal cell hyperplasia in animals
Prostaglandins and Gastric Ulcers
-Pivotal role in maintain gastric mucosal integrity
-Inhibits gastric acid secretion
-Stimulates HCO secretion
-Stimulates mucus secretion
-Increases mucosal blood flow
-Modified local inflammatory response caused by acid
Misoprostol
-Cytotec
-Binds to prostaglandin receptor on parietal cells
-Decreases intracellular cAMP levels, leads to decrease in proton pump activity
-Increases mucous production in gastric ulcers
-CO-administered with NSAID to prevent ulceration
-Off label uses:
--labor induction, smooth muscle contraction
--abortion
--erectile dysfunction
Renin-Angiotensin System Function
-Major role in cardiovascular and body fluid homeostasis
-Boosts blood pressure when blood volume has been challenged
-Contributes to central hypertension
-Works through actions of circulating angiotensin II
--angiotensin II acts on target organs
-targeted pharmacologically to treat hypertension
Renin Angiotensin System Discovery
-Extract of the kidney
-Realized that it was an enzyme, not a substance of the kidney itself
-Angiotensin was found to trigger aldosterone
Angiotensin Synthesis
1. Angiotensinogen precursor in blood (biologically inactive)
-Renin cleaves angiotensinogen into angiotensin I
-Angiotensin I is also biologically inactive
-Rate limiting step in synthesis
2. Angiotensin I is converted into Angiotensin II by Angiotensin Converting Enzyme (ACE)
-provides active peptide
3. Angiotensin III is also formed and may have a small effect
Antiogensin I
-Biologically inactive
-Formed from Angiotensinogen by renin
--renin is enzyme the cleaves
-Formation is rate-limiting step in formation of Angiotensin II
-Needs to be cleaved by ACE to become Angiotensin II
Renin
-In Kidney
-Plasma concentration is rate limiting factor in Angiotensin II formation
--converts angiotensinogen into angiotensin I
-Secretion is controlled by reduced renal perfusion pressure, reduction of Na concentration in DCT, or sympathetic activation of b-adrenergic receptors
Renin Secretion
-Determines activity of Renin-Angiotensin-Aldosterone system
--Other components are always present, but not activated until Renin is released
1. Reduced renal perfusion is sensed by intrarenal stretch receptor
--juxtaglomerular apparatus
2. Reduced Na concentration in DCT triggers renin release
3. Sympathetic activation of b-adrenergic receptors stimualtes renin release
--Epi, NorEpi

Redundant system to ensure that when BP drops, body secretes renin
Juxtaglomerular Cells
-Produce renin, secrete renin
-In afferent arterioles of renal glomerulus
-Act as intra-renal pressure sensor
--have stretch sensors, monitor how much pressure is on arteriole
-Low pressure results in secretion of renin
--increases systemic blood pressure
Angiotensinogen
-Abundant protein in plasma in healthy people
-Cleaved from N-terminus by renin to form angiotensin I
-Made by the liver, also found elsewhere
-Biologically inert, needs renin to be converted to become active
Angiotensin Converting Enzyme
-Always present
-Really a more non-specific enzyme, can also cleave other peptides
-Cuts 2 AA from carboxy end of a peptide
-Found in lumen of endothelial cells, including lung
--highest concentration is in lung
-Prefers substrates that do not have proline as dominant AA at carobxy terminus
--will not cleave angiotensin II due to presence of proline
--Does not cleave active peptide
Angiotensin II
-Most active form
-Most potent vasoconstrictor in the body!
-Most of information for specific activity is in C-terminal
--last 7 AA are really important
-Agonist activity depends on phenylalanine in 8 position on carboxy terminus
-Substitution at position 1 and 8 creates a compound that is less susceptible to degradation by peptidases
Angiotensin III
-Potent in some systems, important for aldosterone release
-Not useful for boosting blood pressure
Angiotensin II metabolism
-Non-specific enzymatic breakdown
--Enzymes are very common, and angiotensin II does not last for long in the blood
--15-60 seconds
-Mostly destroyed by single pass through pulmonary vascular bed
-Rapid removal by "angiotensinases" from circulation
--aminopeptidases
--endopeptidases
Steps in Activation of RAA system
1. Stimulus that causes reduced blood pressure
-decreases perfusion of juxtaglomerular apparatus
2. Renin released by JG apparatus
3. Angiotensin II formation
--most potent vasoconstrictor in the body
--Vasoconstriction leads to increased BP
--Aldosterone secretion from adrenal cortex prevents Na release

Process increases blood volume (Na retention) and blood pressure (vasoconstriction) to increase renal perfusion
Aldosterone
-Steroid hormone
-Made in adrenal cortex zona glomerulosa
-Regulates blood Na and K balance
--allows reabsorption of Na and excretion of K
-Synthesis is stimulated by increased angiotensin II, ACTH, or K levels relative to Na
-Can affect acid excretion in kidney, can be stimulated by acidosis
-Can also be stimulated by atrial stretch receptors
Mechanism of Aldosterone Secretion
1. Low blood pressure causes stretch receptors to stimulate adrenal gland to release aldosterone
2. Aldosterone increases Na resorption from urine, sweat, and gut
-causes increased osmolarity of extracellular fluid
3. Water is retained and blood pressure goes back to normal
Renin-Angiotensin System in Adrenal Cortex
-Stimulates synthesis and release of aldosterone from zona glomerulosa cells
-Augmented by hyponatremia and hyperkalemia
-Allows for Na conservation
Renin-Angiotensin System in Kidneys
-Antidiuresis and antinatriuresis
-Promotes Na reabsorption by kidney
-Reduces glomerular filtration rate
--slows filtering of fluids, might lose fluids
Renin-Angiotensin System in Cardiovascular System
-Very potent vasoconstrictor
-Direct vasoconstriction action on vascular smooth muscles
--augmented by Sympathetic nervous system
-Increases cardiac contractility with little change in HR
-Major role in pathogenesis of hypertension, even when plasma renin activity is not elevated
--involved in pathological hypertension
Renin-Angiotensin System in Peripheral Autonomous Nervous System
-Activates sympathetic nervous system via central effects in the brain
-Sympathetic post-ganglionic neurons are releaseing NorEpi
-target organs increase sensitivity to NorEpi, results in more response
-Adrenal medulla is activated and released catecholamines into the blood
Renin-Angiotensin system in the Central Nervous System
-Circulating angiotensin II will increase sympathetic nervous system drive
-Angiotensin II can get access to brain areas that do not have BBB or weak BBB
--activates receptors in these areas
-Central pressor response activates sympathetic nervous system
-Acts on endocrine system to release ADH and ACTH
--ADH conserves H2O in kidney
--activates cortisol response, boosts sympathetic activity and responsiveness
-Changes behavior to increase thirst and appetite for Na
Renin-Angiotensin System in Cell Growth
-Increases mitosis in vascular and cardiac cells
-May contribute to cardiovascular hypertrophy
Angiotensin Receptor
AT1
-Mediates most known functions of angiotensin
--Most cardiovascular effects
-GPCR
-Found on vascular smooth muscle
-Activates phospholipase C and liberates IP3 and intracellular Ca
--causes smooth muscle contraction
-Activation leads to vasoconstriction
-Blocking will prevent vasoconstriction
Angiotensin Receptor
AT2
-Similar affinity for angiotensin as AT1
-GPCR
-Present in high density in fetal tissues
--contributes to normal growth and development early in life
-Found in some brain areas
-Functions via serine and tyrosine phosphatases
-Up-regulated in pathological conditions
--heart failure, myocardial infarction
-Ideally do not want to block AT2, has protective effects
Inhibition of Renin-Angiotensin System
-Lots of steps in production, many ways to block action of system
1. Block renin secretion
2. Inhibit renin enzymatic activity
3. Block Convert angiotensin I to angiotensin II
4. Block angiotensin II receptors
Block Renin secretion
-block sympathetic nerves
-Reduces angiotensin II levels
Inhibition of renin enzymatic activity
-mostly experimental drugs
Block conversion of angiotensin I to angiotensin II
-ACE inhibitors
-Very convenient way to prevent effects of angiotensin II
ACE inhibitors
-Decrease systemic vascular resistance without increasing HR
-Promote natriuresis, excretion of Na (and H2O)
-Effective treatment of hypertension
--decrease mortality and morbidity in heart failure and left ventricular dysfunction
-Delays diabetic nephropathy
Adverse Side effects of ACE inhibitors
-ACE is not specific to Angiotensin I, also converts other peptides
-Change breakdown of other peptides
-Inhibits degradation of bradykinin
--common side effect is coughing
Captopril
-ACE inhibitor
-Highly specific blocker of angiotensin I to angiotensin II
-Very effective in treatment of hypertension
-Most useful in combination with diuretic
-Useful for congestive heart failure
--reduces pre-load and afterload
--improves CO and decreases congestion
Enalapril
-ACE inhibitor
-Prodrug ethyl ester that is hydrolyzed in the body
-More potent than captopril
-Lasts longer in the body
--only need 1 dose per day
Angiotensin Receptor Antagonists
Peptide Congeners
-Saralasin
-Antagonists with substitutions at positions 1 and 8 of angiotensin II
-Bind to receptors but do not stimulate receptors
-Anti-hypertensive effect
-Only effective if administered IV
Angiotensin Receptor Antagonists
Non-peptide antagonists
-Losartan, Valsartan
-Anti-hypertensive drugs with lower side-effects than ACE inhibitors
--do not influence bradykinin system
-Interact with AT1 receptor, not AT2 receptor
--do not remove beneficial effects of angiotensin II
Kinins
-Potent vasodilator proteins
-Formed by cleavage of protein substrates (kininogens)
-Kallikreins break down precursors into active forms
-Lots of similarities to Renin-Angiotensin system
Kallikreins
-Glycoproteins
Kininogens
-Precursors of kinins and substrates of kallikreins
-Present in plasma, lymph, interstitial fluid
-High molecular weight form: 15-25%
--in blood stream
-Low Molecular weight form: act in tissues, can cross capillary walls
Kinins in Mammals
1. Bradykinin
2. Iysylbradykinin
3. Methoionyllyslbradykinin
Bradykinin
-Slows down HR
-Local inflammatory effects
-Reduces BP
-Very short half-life in blood
-Designed to have local effects, not global effects
Kininase
-Breaks down bradykinin into an inert peptide
-Kininase=ACE!
-inhibition results in Bradykinin increase, slower HR
Purpose of Inflammation
-Bring fluid, proteins, and cells from blood into damaged tissues
-Tissue response to damage
-Facilitate healing process, repair damage
Cardinal signs of Inflammation
1. Heat: localized increased vasodilation
2. Redness
3. Swelling
4. Pain
5. Loss of Function
Acute Inflammation
-Initial response to tissue injury
-Release of autacoids
Chronic Inflammation
-Long-term inflammation
-No resolution of the underlying injurious process
-Deposition of fibrous tissue
Mediators of Acute Inflammation
1. Histamine: vasodilation, increased vascular permeabiluty
2. Serotonin: increased vascular permeability
--may or may not cause vasodilation
3. Bradykinin: vasodilation, increased vascular permeability, Pain
4. Prostaglandins: Vasodilation, vascular permeability, chemotaxis, pain
5. Leukotrienes: vascular permeability, chemotaxis
Prostaglandins and leukotrienes
-Major contributors to pain involved with inflammation
-Reducing prostaglandins and leukotrienes will reduce pain
-Both are products of arachadonic acid, derived from fatty acids in lipid membranes
Mediators of Chronic Inflammation
1. Interleukins 1,2,3
2. GM-CSF
3. TNF-a
4. Interferons
5. PDGF3 (platelet-derived growth factor)
PDGF3
-Stimulates fibroboastic migration into area of inflammation
-Causes fibrous material to be deposited in an area
-leads to fibrosis
Outline of Inflammatory Response
1. Cell damage causes:
--amplification of arachadonic acid cascade and production of leukotrienes and prostaglandins
--Activation of inflammatory cells
--Production of Oxygen derived Free radicals from neutrophil membrane, interact with fatty acids
Activation of Inflammatory cells in response to Tissue Damage
-Release of kinins, neuropeptides, histamines, and complement components
--stimulate some cells to produce arachadonic acids, leukotrienes, prostaglandins
-Releases autosomal lysosomes
Oxygen derived Free Radicals
-Released during tissue damage from neutrophil membrane
-Activate immune cells in area
-Interact with fatty acids and initiate arachadonic acid cascade
Arachidonic Acid Cascade
-Synthesis of Arachadonic acid from membrane phospholipids
--rate limiting step
-Once arachadonic acids are formed, cell quickly makes leukotrienes and prostaglandins
-Synthesis of eicosanoids (Prostaglandins and leukotrioenes) from arachadonic acid
Arachadonic Acid Cascade Steps
1. Tissue damage and disturbance of cell membranes
2. Phospholipase 2 activated, cleaves fatty acid chains from phospholipids in membrane
--produce arachadonic acid
3. Arachadonic acid is converted into prostaglandin or leukotriene depending on cell type
--specific type depends on cell
--Lipoxygenase enzyme forms leukotrienes
--cyclooxygenase enzyme forms prostaglandins
4. Eicosanoids mediate bulk of the inflammatory response
Cyclo-oxygenase
-Converts arachadonic acid into prostaglandins, thromboxane, and prostacyclin
-Target of many anti-inflammatory drugs
Lipoxygenase
-Enzyme that converts arachadonic acid into leukotrienes
Leukotrienes
-Important role in airway inflammation and asthma
-Cause bronchoconstriction in airways
-Potent chemotactic factor for macrophages, eosinophils, neutrophils
-Potent vasoconstrictors
-Secreted during asthma attacks and anaphylaxis
-Stimulate T-cell expansion
Eicosanoids
-Prostaglandins, leukotrienes, thromboxanes
-Product of arachadonic acid metabolism
-Have very short half-life
-Bind membrane receptors on target cells
-Induce changes in intracellular Ca levels
-Stimulate inflammation response
-MANY prostaglandin receptors
Factors governing which eicosanoids are produced
-Species
-Cell type
-Phenotype of cell
-Manner in which cell is activated
Prostaglandin Receptors
-LOTS of different receptors
-Some are inhibitory, some are contractile, some are relaxants
-Could be inhibited, but inhibiting the receptors are not as effective as targeting something up-stream
Prostaglandin other functions
-Have homeostatic functions, are not only involved in inflammation
-Role in vascular smooth muscle contraction
-Role in platelet formation and aggreagation
-Role in thermoregulation
-Gastric protection, stimulate bicarbonate production
Prostaglandin Types
-PGD, PGE, PGF
--responsible for edema and local swelling
--regulate renal vasculature
-PGE1 and PGE2: vasodilation
--induce fever when present in cerebral ventricles
Inflammation-associated fever
-Infection leads to production of bacterial endotoxins
-Macrophages release IL-1 (pyrogen)
-Has effect in hypothalamus to produce PGE
-PGE elevate temperature set point, results in a fever

-Fever is caused by PGE
Prostacyclin
PGI2
-Synthesized by vascular endothelial cells
-Inhibits platelet aggregation
--allows fluid accumulation in region of tissue damage
-Inhibits smooth muscle contraction, vasodilation
Thromboxane
TXA2
-Synthesized by platelets and macrophages
-Promotes platelet aggregation
--antagonist for prostacyclin
-Causes smooth muscle contraction, vasoconstriction
Pain associated with Inflammation
-Pain due to swelling and nociceptors becoming sensitized by prostaglandins and other inflammatory mediators
-INhibiting prostaglandin biosynthesis will dampen pain sensation
Strokes and NSAIDS
-NSAID disturbs the balance between prostacyclin and thormboxane
-Favors more inhibitory prostacyclin synthesis and results in strokes
Phospholipase A2
-Stimulated by cellular damage
-Cleaves some of fatty acid chain from cell membranes
--converts to arachadonic acid
-Inhibited by corticosteroids
5-lipoxygenase
-Present in lung, platelets, white cells
--neutrophils, mast cells, monocytes, macrophages, others
-Converts arachadonic acid to leukotriene A4
-Following enzymatic steps produce bioactive leukotrienes
PGH Synthase
Cyco-oxygenase
-Converts arachadonic acid to PGH2 in 2 steps
-Cyclooxygenase converts arachadonic acid into PGG2
-Peroxidase converts PGG2 into PGH2
-Biochemically adds 2 O2 molecules to arachadonic acid
--peroxidase removes oxygen free radical
Cyclo-oxygenases
-PGH synthase-1 (COX-1)
-PGH Synthase-2 (COX-2)
COX-1 (PGH Synthase-1)
-Widely distributed "housekeeping" enzyme
-Constitutively expressed in platelets
-present in most tissues
-Has role in inflammation, larger role in homeostasis and gastric cytoprotection
--Stimulates bicarbonate and mucous secretions in gastric mucosa
-Exists as a dimer
-Closely associated with membranes
COX-2 (PGH Synthase-2)
-Inducible in inflammatory and immune cells
-Induced 10-20x by growth factors and cytokines
--IL-1 and TNF-a, activate NF-kB transcriotion factor
-Want to inhibit during inflammatory reaction
NF-kB
-Transcription factor that is activated during inflammation
-Expression is elevated in many disease states
-Enhanced in cancerous cells
--plays some role in promoting cancer propagation
-Stimulates production of many cytokines and COX-2
NSAIDS
-Function by inhibiting Cyclo-oxygenase
-Prevent synthesis of prostagpandins and throboxanes
-DO NOT inhibit synthesis of leukotrienes
Anti-inflammatory drugs
-Dampen inflammation pathways
-Do not eliminate cause of inflammation
--underlying injury or autoimmune condition
-Palliative effect
-Have side effects
Pharmacological Targets of Arachadonic Acid Cascade
-Anything in the pathway could be inhibited
-Main inhibition occurs at Phospholipase A2 and cyclooxygenase stages
-Phispholipase A2
-Cyclo-oxygenase
-Lipoxygenase
-Prostaglandin synthesis
-Autacoid receptors
Glucocorticoids as Anti-inflammatories
-Steroid hormone derived from cholesterol
-Affect gene expression
-Inhibit Phospholipase A2
--early step
-Inhibit COX-2 induction
--disrupt NF-kB function
-Potent anti-inflammatory activity
-Many side effects
--interfere with adrenal function
Adrenal Glands
-Provide animal with capacity to adjust to environmental and metabolic stresses
-Produce glucocorticoids
Hormones of the adrenal cortex
-Glucocorticoids
--cortisol
--corticosterone
-Mineralocorticoids
--aldosterone
-Androgens and estrogens
Glucocorticoid Function
-Regulates blood sugar levels
-Regulates protein, fat, carbohydrate metabolism
--regulates starvation stress
-Suppresses inflammatory response
-Regulated by ACTH from pituitary gland
Mineralocorticoids
-Aldosterone
-Increases Na reabsorption from glomerular filtrate in kidney
-Increases K excretion
-Increases fluid volume
-Restores electrolyte balance
-Raises blood pressure
Hypothalamus-Pituitary-Adrenal Axis
-Natural glucocorticoids in the blood negatively feed back in hypothalamus and anterior pituitary
--inhibit release of corticotropin-releasing factor from the hypothalamus
-No ACTH stimulation for release in the pituitary
-Shut down synthesis of ACTH and glucocorticoids
-Stimulated by neural mechanisms physical or psychological stress, and diurnal fluctuations
-Arenal cortex starts to atrophy if not stimulated by ACTH
--causes addisonian crisis
Hydrocortisone
-Cortisol
-Predominates in man, dog, horse, pig
-80-95% protein bound in the blood
--corticoid binding globin produced in liver
--patients with liver issues are more sensitive to glucocorticoid drugs
-20% is free or loosely bound to albumin
-Short plasma half-life, 60-90 minutes
-Synthesized from cholesterol
-
Corticosterone
-Cortisone
-Predominates in rodents
-less protein bound that cortisol, shorter half-life
Corticoid Binding Globin
-produced in the liver
-Binds cortisol in the blood
--80% of cortisol is bound
-Synthesis increases during pregnancy
-When saturated, free cortisol levels rise rapidly
-Patients with liver issues are more sensitive to glucocorticoid drugs
Cortisol Metabolism
-1% excreted unchanged in the liver
-20% converted to cortisone in the kidney and other tissues
-Inactivated and metabolized in the liver
--conjugated with glucuronic acid
-Excreted in the urine
Glucocorticoid Mechanism of Action
-Bound to globin in blood, not biologically active
-Free form enters target cells
-Binds to intracellular receptor
--glucocorticoid receptor
-Receptor dimerizes and translocates into the nucleus
--binds specific DNA sequences
--Glucocorticoid response elements
-Affects gene transcription
--can activate or repress gene expression
Induced Glucocorticoid Genes
-Lipocortin1/Annexin 1
--inhibitor of Phospholipase A2, negative feedback
-MAP kinase phosphatase
-IkB, NF-kB inhibitor
Repressed Glucocorticoid Genes
-POMC/ACTH
-IL-1
-IL-2
Metabolic Effects of Glucocorticoids
-Maintain adequate glucose supply to the brain
-Stimulate gluconeogenesis during gasting
-Increases glucose production from proteins
--proteolysis of muscle, bone, skin, lymphoid etc.
-Increase liver glycogen deposition
-Increased glucose stimulates insulin secretion
-Decreases cellular uptake of glucose
-INhibits glucose uptake by peripheral fat cells
-Increased fat deposition around the abdomen
Anti-Inflammatory effects of Glucocorticoids
-Block all known pathways of eicosinoid synthesis
-Indirectly inhibit phospholipase A2 synthesis
--may interfere with phospholipid binding
-Stimulate synthesis of lipocortin I/Annexin I
--directly inhibits Phospholipase A2
-Inhibits NF-kB dependednt COX-2 induction
Mechanisms by which Glucocorticoids dampen inflammation
1. INhibit COX-2 expression
--interferes with NF-kB
2. Stimulates lipocortin/annexin expression
--inhibits phospholipase A2
3. Inhibits phospholipase A2 by inducing MAP kinase phosphatase I expression
Lipocortin 1
-Annexin 1
-Production is stimulated by....
-Lipocortin 1 binds to ... and keeps in inactive
-May share therapeutic activities of glucocorticoids with fewer side effects
--not practical for therapeutic uses
Anti-inflammatory effects of Glucocorticoids
Peripheral Leukocytes
-Suppresses immune system
-Decreases number of neutrophils at site of inflammation
--decreased migration from vessels
--endothelial cell adhesion molecules
-Increases overall number of neutrophils in blood
--influx into blood from bone marrow
-Decreases lymphocytes, eosinophils, basophils, and monocytes
--movement of cells from vascular compartment to lymphoid tissue
Anti-inflammatory effects of Glucocorticoids
-Inhibit ability of lymphocytes to respond to antigens
-Inhibit histamine release
-Reduce cytokine function
-Inhibit macrophage phagocytosis
-Decrease vascular permeability
--inhibits autacoid release
--inhibits induction of an endothelial enzyme that increases NO
Therapeutic uses of ACTH
-Not practical as an Anti-inflammatory drug
--to many side-effects
--stimulates production of androgens and mineralocorticoids
-Can be used to diagnose adrenal insufficiency (Addison's disease)
Addison's Disease
-Chronic Adrenal insufficiency
-Inability to maintain glucose levels during fasting
-Hyperpigmentation, weakness, fatigue, weight loss, electrolyte imbalance
-Minor trauma or infection may produce acute adrenal insufficiency and lead to shock or death
-Treat with glucocorticoids and mineralocorticoid
Cushing's Disease
-Adrenalcortical hyperfunction
-Usually caused by pituitary adenoma that produces excess ACTH
-Leads to excessive glucocorticoid production
--hyperglycemia, hypertenions, diabetes
-Will have face and trunk obesity
-Muscle wasting, thinning of skin
-Poor wound healing
-Osteoporosis
-Tx: drug therapies or removal or tumor
--follow with hormone replacement therapy
Cortisol/Hydrocortisone
-glucocorticoid
-Standard for steroid anti-inflammatory drugs
-Has many side-effects
Metabolic effects of Glucocorticoids
-Suppress ACTH release
--leads to atrophy of the adrenal cortex
-Cause weight gain via hyperglycemia and increased insulin requirements
-Electrolyte imbalances
--increased Na and increased body volume
Other effects of Glucocorticoids
-Weakness
-Increased gastric acid and pepsin secretion
-increased number of platelets and RBCs
-Effects on fetal development
--higher abortion rates in cattle
Immunosuppression and glucocorticoids
-Slow wound repair
-Large doses can suppress antibody production
-Impaired hypersensitivity reactions
-Infections may persist
Fluid and electrolyte disturbances with glucocorticoids
-Mineralocorticoid effect
Glucocorticoids and glaucoma
-Increased intraocular pressure leads to cataracts and glaucoma
Synthetic Glucocorticoids
-Actions are similar to cortisol
--bind same receptor
-Can be short, intermediate, or long-acting
-Oral, topical, IV, and IM forms
Cortisol an Mineralocorticoid activity
-It has it
-Can make chemical modifications to cortisol to engineer out mineralocorticoid activity
-Change glucocorticoid to mineralocorticoid ratio
--add a double bond
--forms prednisolone
--less water retention with prednisolone than with cortisol and increases half-life
Categories of Action for Synthetic Glucocorticoids
-Short acting: 1-24 hours
-Intermediate acting: 24-48 hours
-Long acting: more than 48 hours
-Ultra long acting: several weeks
-Addition of fluoride increases time of elimination, increases halflife
Modifications to Ester groups in Glucocorticoids
-In general, less soluble esters increase glucocorticoid half-life
-Drug is not active until esterase cleaves groups
Solubility of groups on synthetic glucocorticoids
1. Water Soluble
--phosphate
--succinate esters of a drug
2. Poorly water soluble
--acetate, diacetate
3. Non-water soluble drugs
--Acetonide, diproprionate
Hydrocotisone
-Low anti-inflammatory activity
-Has some mineralocorticoid activity
--retains water
-Oral, injectable, and topical forms available
Prednisone
-More anti-inflammatory activity that cortisol
-Less mineralocorticoid activity
Prednisolone
-Short-acting
-Double bond between carbons 1 and 2
--gives less mineralocorticoid activity, 70% less
-More anti-inflammaotry activity than cortisone, 4x
-Water soluble
-Orally administered
--also topical or injectable
Methylprednisolone
-Methylated derivative of prednisolone
-short-acting
-4-5x anti-inflammatory activity of cortisol
-Very little mineralocorticoid activity
-Oral, topical, and injectable forms exist
Triamcinolone
-Intermediate-acting synthetic glucocorticoid
-Very similar to dexamethasone, has substitution instead of methyl group
-Oral and topical forms
--oral is 5x activity
--topical is 5-100x activity, acetonide increases surface activity
-Very little mineralocorticoid activity
-Similar to dexamethasone
Dexamethasone
-Long-acting, fairly slow onset of action (6 hours)
-Lower doses needed
-30x anti-inflammatory activity of cortisol
-Very little mineralocorticoid activity
-oral, topical, and injectable forms
Most common Side Efects of Short-term Glucocorticoid Therapy
-Increased susceptibility to infection
-Increased urination and thirst (PU/PD)
-Increased appetite
-Behavioral mood changes
-Diarrhea
-Development of pancreatitis
-gastric ulceration
-Dogs get hepatomegaly
-Long-term leads to suppression of HPA axis and cushing's syndrome
Hyperadrenocorticism Symptoms
-Looks like Cushing’s disease
-Suppression of the immune system:
--decreased lymphocyte proliferation
--increased incidence of infections in skin, urinary tract, and respiratory tract
-Poor wound healing
-Tendon, ligament, joint abnormalities
-Dull/dry hair coat or hair loss
-Dry, thin skin that bruises easily
-Decreased muscle mass and tone
--weak abdominal muscles give pendulous abdomen
--Muscle pain, stiff gait, rigid limbs
Glucocorticoid Therapy
-Short-term therapy is faster acting
-Good for acute inflammation
-Sooner patient is treated with glucocorticoids, the less inflammation-associated damage there is
-Taper off the drug
--if withdrawn abruptly adrenal gland may not recover
-Prolonged adrenal suppression may cause irreversible damage
Glucocorticoid Alternate Day Therapy
-For long-term therapy
-Short half-life and water soluble
--drug should be gone from system by day 2
-Ensures less suppression of the adrenal gland endogenous cortisol and ACTH
Glucocorticoid Repositol Therapy
-Good for cats, not good for dogs
--Cats are less sensitive to side effects
-Methylprednisolone acetate, triamcinolone acetonide
-Long-lasting (weeks to months)
-Give when oral medication is impossible
Topical Therapy of Glucocorticoids
-Ointments, creams, lotions, etc.
-readily absorbed through the skin
-May suppress HPA axis
NSAIDs
-Competitively inhibit cyclo-oxygenase from interacting with arachadonic acid to convert into prostaglandins
Cyclooxygenase Inhibitors
-Chemically diverse
-Weak acids
-Most inhibit both COX-1 and COX-2
-Unwanted side effects are due to inhibition of COX-1
-Do not inhibit tissue damage caused by release of lysosomal enzymes and toxic oxygen radicals
Aspirin and Salicylates
-Similar to Sodium Salicylates
-Absorbed in stomach and upper small intestine
-Acid medium in stomach promotes absorption
--keeps salicylate in non-ionized form
--poorly absorbed in horse and cows
-Salicylate binds albumin in serum
--higher dosages result in higher concentration of un-bound drug
Salicylate
-Aspirin is converted to salicylate by esterases in tissues and blood
-Competitive inhibitor of arachadonic acid
Irreversible COX inhibitor
-Aspirin
-Only NSAID to covalently modify COX
-Acetylates COD, prevents enzyme binding to arachadonic acid
-More effective COX-1 inhibition than COX-2
-Peroxidase activity is not affected
-Scavenges oxygen free radicals
--contributes to inhibition of prostaglandin synthesis
Other effects of Aspirin
-Analgesic: reduces pain by inhibiting pain stimuli and reducing inflammation
-Antipyrogenic: blocks pyrogen-induced prostaglandin synthesis
-Platelet effects: inhibits COX-1, inhibits thromboxane synthesis in platelets, inhibits platelet aggregation
--irreversible until new platelets are formed
--Platelets do not have nuclei, no way to up or down-regulate production
--no place for glucocorticoids to have effect
Aspirin Inactivation
-Inactivated by conjugation to glucuronic Acid
-Different species have different amounts of glucuronidation activity
--lower glucuronidation activity leads to longer plasma half-life for aspirin
--T1/2 is variable
-Salicylate conjugates are cleared via kidney
--excretion is faster in more alkaline urine
-Conjugates to glutathione when glucuronic acid is absent
Adverse effects of Aspirin
-gastric Ulceration and GI disturbances
-COX-1 inhibition of gastric cytoprotection
--prostaglandins stimulate bicarbonate and mucous secretions in gastric mucosa
-Gastric irritation can be decreased by raising gastric pH to 3.5 or higher
-Can administed misoprostol, Prostaglandin analog
Adverse effects of Aspirin at High Doses
-Salicylism
--dizziness, deafness, uncoupled oxidative phosphorylation, increased O2 consumption
-Fever
-Dehydration
-Renal damage
-Generalized hemorrhage
-Convulsions or coma
Para-Aminophenols
-Acetaminophen: tylenol, paracetamol
-Phenacetin: breaks down to acetaminophen
-Reversible inhibition of COX, COX-3??
-Competes with arachadonic acid binding
-Effective analgesic and antipyrogenic, poor anti-inflammatory
-Not very useful as anti-inflammatory agents in vet med
-Rapidly absorbed in the GI tract
-80-90% is conjugated to glucuronic acid
-High doses cause accumulation of toxic by-product in liver
Adverse effects of Acetaminophen
-HIGHLY toxic to cats
-Oxidized in liver to toxic metabolite that depletes glutathione
-Causes hepatic necrosis and methemoglobin formation
-Hemolytic anemia
-Denaturation of RBC membranes causes hypoxia
Phenylproprionic Acids
-Ibuprophen
-Naproxen
-Carprophen
-Competitive reversible inhibitors of COX
--interfere with arachidonic acid binding
-Compete with aspirin for plasma protein binding sites
Ibuprophen
-Moderate COX inhibitor
-More selective for COX-2
-Less toxic to has
-Toxic to dogs
--causes ulcers and kidney failure
Naproxen
-Approved for use in horses and dogs
-Works equally on COX-1 and COX-2
Adverse effects of Phenylproprionic Acids
-Possible nephrotoxicity
-Nausea
-GI toxicities
-Peripheral edema
COXIBs
-COX-2 selective drugs
-Carprophen (Rimadyl)
--least selective
-Etodolac (Etogesic)
-Deracoxib (Deramaxx)
-Firocoxib (Previcox)
-Robenacoxib (Onsior, for CATS)
COX-2 selectivity
-Make molecule a little larger so it can only fit into COX-2 binding site
-Will only inhibit COX-2
Homeostasis of Platelet Aggregation
-Some coxibs can disrupt balance
-Shift balance towards platelet aggregation
-Occurs when only COX-2 is inhibitied
-has not been demonstrated to occur in animals, only in humans
Carprophen
-Rimadyl
-Only officially approved for dogs
-Pain relief, joint pain, post-surgical inflammation
-Proprionic acid
-More selective for COX-2, inhibits COX-1 only slightly
-Not approved for use in cats
-Insoluble in water
-Metabolized in the liver and excreted in the feces
-T1/2 in dogs is 8 hours
Adverse Effects of Carprophen
-Gastric Ulceration
-Nausea, appetite loss, vomiting, diarrhea
-Hepatopathy, liver toxicity
--occurs within first 3 weeks
--If ignored, can be life-threatening and irreversible damage
--3-4x high concentration of liver enzymes in the blood
-Labrador Retrieveres are over-represented
-Make sure animal does not have liver damage before giving
Renal side effects fo Carprophen
-Due to COX-1 inhibition
-Excess water consumption and urination (PU/PD)
-Loss of balance, hyperactivity, depression, and aggression
Carprophen Conraindications
-Pre-existing liver, kidney disease or GI ulcerations
-Should not be used with other NSAID or corticosteroid drugs
-Not recommended for pregnant or nursing females
--has not been sufficiently tested
Etodolac
-Approved for dogs
-T1/2 is 8-14 hours
-Metabolized in the liver and excreted in feces
-Extensive enterohepatic recycling
-Side effects of GI ulceration
Deracoxib
-Approved for use in dogs
-Control post-operative pain and inflammation
-Relatively short half-life with low doses
-Larger doses have longer half-life
Firocoxib
-Previcox, Equioxx
-Approved for dogs and horses
-T1/2:
--8 hours in dogs
--30-40 hours in horses
-Hepatic clearance
Selectivity Assays
-IC50 ratio: give idea for how selective a drug can be
-Concentration of a drug needed to inhibit processes by 50%
-high ratio is more COX-2 selective
-Ratio does not correlate to efficacy
--drugs with large COX-1 action will also have COX-2 activity
-Deracoxib has highest ratio, least COX-1 inhibition
-Aspirin has lowest ratio, affects both COX-1 and COX-2
Most Common NSAID side effect
-Gastric Intolerance and Ulceration
-Especially in Dogs
NSAIDs for Cats
-Not commonly used
-Cats do not tolerate any NSAID well
-Robenacoxib and Meloxicam are only 2 approved
Robenacoxib
-NSAID in cats
-New COX-2 selective inhibitor
-Tablet and injectable forms
-27 hour halflife
-Metabolized in liver and excreted in feces
-Adverse reactions in digestive tract reported
Meloxicam
-NSAID in cats, relatively safe in dogs
-Considered COX-2 selective at low doses
-97% is bound to plasma proteins
-Metabolized in the liver
-Eliminated in feces
-Only approved for a single injection to relieve post-operative pain
-Repeated use is associated with renal failure and death
Flunixin meglumine
-Banamine
-Irreversible, non-covalent COX inhibitor
-Possibile weak inhibitor of 5-lipoxygenase
-Powerful analgesic
-Long duration, 24-36 hours
-2 hours onset of action, maximum action at 12 hours
-Administered IV or IM
-Used mostly in horses and cattle
-DO NOT use in cats
Flunixin meglumine Advantages
-More potent analgesic than other available inhibitors of prostaglandin synthesis
-Rapid onset and long duration of action
--will have effect within 2 hours of administration, lasts 24-36 hours
-Useful for relieving pain associated with musculoskeletal disorders, surgery, GI spasm, and endotoxic shock
--helpful for colics
Flunixin meglumine Adverse effects
-Ulceration of tongue, lips, gums, palate, stomach
-Anorexia
-CNS depression
-May cause acute renal failure in dogs
--unknown mechanism
--need to ensure adequate hydration, avoid decreases in BP and blood volume
Phenylbutasone
-Irreversibly binds COX
--non-covalent
-Metabolized to oxyphenobutasone active metabolite
-Can stick around in the body for a long period of time
-NOT approved for food animals
-99% of drug binds to plasma proteins
-Approved for dogs and horses
--most popular NSAID in horses
Bute Duration of Action
-LONG duration of action
--24-72 hours
-New COX has to be synthesized in order to synthesize more prostaglandins
-Oxyphenbutazine metabolite persists in the body
-Plasma bound drug can penetrate inflamed tissues
-Excreted via kidneys
Bute administration
-Orally or IV in dogs and horses
-Can cause local irritation and phlebitis if injected perivascularly
Bute Adverse Effects
-Oral and GI tract erosions, diarrhea, depression
-Death (if enteropathy leads to decreased blood volume, hypovolemic shock, circulatory collapse)
-Do not use if patient has hematocytologic disorders
--cardiac, renal, or hepatic dysfunction also no-go
-Can cause permanent kidney damage if animal is dehydrated
Aspirin Unique Features
-Irreversible COX inhibitor
-Increased toxicity in Cats
Phenylbutazone Unique Features
-Most popular NSAID in horses
-Induces hepatic CYP450 enzymes
-Idiosyncratic bone marrow toxicity
Acetominophen Unique Features
-Does not inhibit platelet function
-TOXIC in cats
Ibuprophen unique feature
-Enhanced ulcer formation in dogs
Naxproxen unique features
-Enhanced ulcer formation in dogs
Carprophen unique feature
-Idiosyncratic hepatotoxin
Flunixin meglumine Unique features
-Potent analgesic agent for horses
Tepoxalin
-Zubrin
-Cox inhibitor
-Dual LOX/COX inhibitor?
--inhibits 5-lipoxygenase
-Approved for drugs
-Lipophilic
-Converted to an active carboxylated metabolite
-Hepatic clearance
-T1/2 in dogs is 12-14 hours
Dimethy Sulfoxide
DMSO
-Non-COX inhibitor
-Clear, odorless liquid
-Solvent for Aromatic and Unsaturated hydrocarbons, organic nitrogen cpds, and inorganic salts
-Rapidly penetrates the skin
-Breaks down to DMS and other metabolites
--excreted via kidney
-Some is expelled in breath
--halitosis is common side effect, smells like garlic/oysters
DMSO therapeutic Features
-Anti-inflammatory
-Analgesic
-Anti-microbial
-Anti-fungal
-Anti-diuretic
-Anti-cholinesterase
DMSO Anti-inflammatory effects
-Not well understood
-Traps cytotoxic free radicals released from leukotrienes during injury and inflammation
DMSO Analgesic
-Thermal activity may relieve muscle and joint pain
-Appears to slow conductance of non-myelinated nerve fibers
-Has anti-microbial, anti-fungal, anti-diuretic, and anti-cholinesterase function with unknown mechanism
DMSO Administration
-Topical
-Readily penetrates the skin
-Causes hemolysis when injected IV in concentrations greater than 40%
-Readily penetrates the skin, rapidly distributed to all parts of the body
DMSO Adverse Effects
-Can introduce external toxins into the body
--Wear gloves when administering
-Can induce degranulation of mast cells
-May cause cataracts in dogs
-May be teratogenic
Enteric Nervous System
-Branch of Autonomic nervous System
-Receives input from SNS and PNS
-Can act semi-autonomously, thought of as a 3rd division of the ANS
-Myenteric plexus: GI motility
-Submucosal plexus: secretion and absorption
Electrical Activity of the GI system
-GI smooth muscle activity is controlled on 3 levels
--Extrinsic Autonomic Nervous System
--Intrinsic Nervous System
--Receptors mediated by neuropeptides
-ACh used for intrinsic and extrinsic systems
--adrenergic stimulation decreases motility
-Vasoactive intestinal Peptide, substance P, gastrin, motilin, and ghrelin modulate gasric and intestinal motility
-Slow waves during resting
-Spikes during activity, leads to depolarization
Laxatives and Cathartics
-Promote defecation
-Cleanse bowels for radiographic procedures, surgery, colonoscopy
-Eliminate toxins during poisonings
-Reduce fecal impaction, non-dietary constipation
Laxatives
-Elimination of soft formed stools
Cathartics
-Elimination of more liquid stools
Bristol Stool Chart
Type 1: separate hard lumps, like nuts
--hard to pass
Type 2: sausage-shaped but lumpy
Type 3: Like a sausage but with cracks on surface
Type 4: like a sausage or snake, smooth and soft
Type 5: Soft blobs with clear-cut edges
Type 6: Fluffy pieces with ragged edges, mushy stool
Type 7: Watery, no solid pieces, entirely liquid
Types of Laxatives and cathartics
-Stimulants
-Surfactant laxatives
-Lubricants
-Bulk forming laxatives
-Osmotic Agents
Stimulants
-Promote accumulation of water and electrolytes in lumen of the colon
-Stimulate intestinal motility, muscle contraction
-Slow onset of action (6 hours)
-Ex: Bisacodyl
--intestinal and bacterial enzymes convert into active metabolite
Bisacodyl
-Diphenthylmethane derivative
-Slow onset of action, works in colon
-Rapidly converted by intestinal and bacterial enzymes into active desacetyl metabolite
Docusate
-Anionic surfactants that have detergent-like action
-Acts as stool-wetting and stool-softening agent
-Allows mixing of water, lipids, and fecal material
-Stimulates intestinal water and electrolyte secretion to soften stools
Surfactant laxatives
-Hydrophilic end and long tail
-Anionic
-Detergent-like action
-Stool-wetting and stool softening agent
-Allows mixing of water, lipids, and fecal material
-Stimulates intestinal water and electrolyte secretion to soften stools
Lubricants
-Soften and lubricate fecal material to facilitate expulsion
-Mineral oil
Mineral Oil
-Lubricant
-Aliphatic hydrocarbon, obtained from petroleum
-Indigestible and not absorbed by intestine
-Needs to be taken for 2-3 days for effect
-Can cause malabsorption of fat-soluble vitamins and drugs
Bulk Forming Laxatives
-Dietary fiber
-Indigestible plant cell wall material
-Not digested in intestine, but absorb water and expands, increases bulk
-Expansion stimulates stretch reflex, stimulates expulsion
-Day onset of action
-Flatulence is side-effect
-Need to stay hydrated, material absorbs water out of lumen
Osmotic Agents
Hypertonic Agents
-Cause osmotic movement of water into gut lumen
-Hypertonic agents cause osmotic movement of water into the gut lumen
--Mg, Tartarate Na salts, lactulose, glycerine, sorbitol, mannitol, polyethylene glycol
--Movement of water into lumen increases stool volume
--Stimulates mechanoreceptors and results in increase in peristaltic activity
-Potential for dehydration exists
Osmosis
-Movement of water across a semi-permeable membrane from areas of high concentration to areas of low concentration
-Moves from Low concentration of solute to high concentration of solute
-Seeks equilibrium of concentration
Osmotic Agents
Isotonic Agents
-PEG electrolyte solution
-Isotonic water in, water out
-results in copious watery stools
-No movement of fluid into intestine
-No dehydration, no differential osmotic pressure
Magnesium Hydroxide
“Milk of Magnesia”
-Has laxative action
-Cause diarrhea
-Draw fluid into the lumen of intestine
-Distention of bowl due to increased fluid in intestine induces peristalsis
Anti-diarrheal agents
-Oral rehydration
-Opioid agonists
-Bismuth salicylates
-Kaolin-pectin suspensions
Oral Rehydration for Diarrhea
-Main risk in acute diarrhea is dehydration
-Need to replace fluids and electrolytes lost
Opioid Agonist for diarrhea
-Act on Mu and Delta receptors in myenteric plexus
-Decreases motility of large intestine
-Allows more time for water resorption
-Activate Mu receptors and increase sphincter tone
-Imodium, Lomotil
--structurally similar to demerol, potentially addictive
--sold in combination with atropine to induce unpleasant side-effects and decrease abuse potential
Bismuth Subsalicylate
Anti-diarrheal
-treats mild to moderate diarrhea
-Mechanism is not understood
-Absorption of toxins by bismuth?
-Antiinflammatory effect of salicylate?
-Used in dogs, horses, cattle
-NOT in cats
Kaolin-pectin suspensions
-Anti-diarrheal
-Colloid suspension with kaolinite
--Absorbs water
-Decreases fluidity of fecal material
-need to be sure to stay hydrated
-Give orally every 4-6 hours
-Used in dogs, cats, horses, cattle, swine, sheep, birds
Vomiting
-Protective mechanism to remove toxins from the body
-Can be humorally mediated or neurally mediated
Humorally-mediated emesis
-Kidney failure
-Liver disease
-Endotoxemia
-Digoxin toxicity
-Apomorphine
Neurally-mediated emesis
-GI infection
-GI toxicity
-GI inflammation
-Malignancy
Physiology of events during vomiting
-contraction of abdominal muscle
-Crainal movement of diaphragm
-Contraction of pylorus and antrum
-Relaxation of gastroesophageal sphincter
-Relaxation of the cricopharyngeus
-Closure of the glottis
-Respiratory inhibition
-Closure of the nasopharynx
-Main concern is aspiration of vomitus
Physiology of Emesis
-Highly regulated protective reflex
-Coordinated in emetic center in lateral reticular formation in medulla
--Lower order process
-Afferent inputs:
--chemotrigger zone
--Vestibular system
--peripheral sensory receptors in stomach and intestine
--higher CNS nervous system centers
Signals promoting vomiting
1. Chemotrigger zone
2. Ear vestibular nucleus
3. Gut sensory receptors
4. Gut irritant receptors
Metabolic consequences of Vomiting
-Dehydration
-Electrolyte imbalances
--Hypokalemia
--hypochloremia
--hyponatremia
--Alkalosis, increased blood pH due to loss of acids
Drugs controlling emesis
-A2 Adrenergic antagonists
-Dopamine antagonists
-5-HT3 serotonin antagonists
-Opioid antagonists
-Antihistamines
-Anticholinergics
-Neurokinin-1 antagonists
-Cannabinoids
A2 adrenergic antagonists and Emesis
-Compounds are Antagonists for multiple receptors
-Chlorpromazine
-Prochlorperazine
-Weak dopamine receptor antagonism
-Weak histamine receptor antagonism
-Weak cholinergic receptor antagonism
-Blocks vomiting reflex at emetic center at CRTZ
-Used mainly in dogs and cats as antiemetic
Side effects of A2 adrenergic Antagonists as anti-emetics
-mild sedation due to histamine receptor blockade
-Hypotension due to A1 receptor blockade
-Contraindicated in horses due to ataxia
Dopamine Antagonists and Emesis
-Anti-emetic action due to blocking D2
--Receptors in CRTZ
-At high doses blocks 5-HT3 serotonin receptors
--can contribute to anti-emetic properties
-Prokinetic, increases peristalsis in jejunum and duodenum
--due to muscarinic activity, D2 antagonism, and 5-HT4 receptor agonism
-Side effects include hyperactivity, tremors, and constipation
Serotonin Antagonists and emesis
-Ondansteron and Granisteron
-Anti-emetic action due to blocking serotonin receptors in CRTZ and GI tract
-Oral or IV dosage used to control nausea due to radiation and chemotherapy
-Side effects include transient headache, constipation, and dizziness
Opioid Antagonists and emesis
-Butorphanol (torbugesic)
-Synthetic opioid receptor antagonist
-Acts on opioid receptors in emetic center and higher CNS centers, blocks receptors for anti-emetic effect
-Sedation is common side-effect
-Used to prevent emesis after chemotherapy
Anti-histamines and Emesis
-Dimenhydrinate (Gravol, Dramamine)
-Mainstay therapy for nausea caused by motion sickness
-Anti-emetic activity due to blockade of H1 histamine receptors in CRTZ
-Sedation is common side-effect (CNS H1 receptors)
-Not effective in cats
-Used to prevent vomiting due to motion sickness
CRT zone in Cats
-Signal from vestibular nucleus in ear goes directly into emetic center in cats
-Does not go to CRT zone, impulse is not mediated by CRT zone
-In dogs, vestibular nucleus impulses pass through CRT zone to emetic center
Meclizine
-Dramamine II
-Therapy for nausea caused by motion sickness
-Anti-emetic activity due to blockade of central H1 histamine recptors
-Sedation is a less common side effect
-Used to treat vertigo due to inner ear infections
Anti-cholinergics and emesis blocking
-Scopolamine
-Topane alkaloid with antimuscarininc activity
-Blocks muscarinic receptor mediated emetic impulses from vagal affferent pathways to emetic center
-Effective for travel sickness
Propantheline, Isopropamide
-Anti-cholinergics used in dogs and cats to treat vomiting
-Not as effective alone as when combined with other medicines
-Side effects include tachycardia, loss of visual accommodation, urine retention, constipation, and xerostomia
Neurokinin1 Receptor Antagnoists and Emesis
-Maropitant
-Neuropeptide Substance-P is cognate ligant for NK-1
-Activation of NK-1 in CRTZ induces vomiting
-Used for motion sickness and comiting
Aprepitant
-"Emend"
-Neurokinin-1 antagonist used to treat vomiting
-Chemotherapy-induced nausea and vomiting
-Prevents post-operative nausea and vomiting in humans
Cannabinoids and Emesis
-Binds to CB1 and CB2 GPCR receptors in higher centers in the brain
-CB1 receptors are pre-synaptic modulatory receptors
-Marinol has been approved for treatment of refractory nausea and vomiting in chemotherapy patients
Emetics
-Cause vomiting
-Expel non-corrosive poisons from stomach prior to induction of general anesthesia
--80% of stomach contents are expelled
-Centrally acting emesis
-Peripherally acting emetics
Centrally acting Emetics
-Apomorphine:
--Non-sepective dopamine agonist related to morphine
--Can be given IV, IM, SC
--Stimulates CRTZ
--Can cause paradoxical excitement in cats
--contraindicated in cats
-Xylazine:
--A2-adrenergic agonist
--Induces vomiting in cats, then mild sedation
--dose for emesis is lower than dose for sedation
Peripherally acting Emetics
-Local irritants
-NaCl: saturated solution of table salt, induces vomiting in dogs and cats
-H2O2: induces vomiting in cats
-Syrup of Ipecac: induces vomiting in 15-30 minutes
--peripheral irritant, has some central actions
--contrainidcated for use in cats
Ayahuasca
-Psychoactive infusion from vine in S. America
-Used in shamanic rituals for religious ceremonies
-Also has purgative properties
Sites of Action of Inhaled Anesthetics
CNS
1. Spinal cord and Brainstem
--Inhibition of sensory processing
--inhibition of nociceptive signaling
--Inhibition of motor response to noxious stimulation, immobility
2. Brain
--hypnosis and amnesia
--analgesia
3. Interrupts communication between thalamus and cerebral cortex
--patients cannot perceive pain
Lipid Theory of General Anesthetic Action
-Interactions with lipid matrix of the membrane
-Expands volume of lipid membrane by interacting with it
--puts pressure on ionic channels, inhibits ion channels and prevent AP generation
-Changes membrane physical state, change in consistency
--prevents ion channels from functioning appropriately
Ion Channel Mechanism of Action
-Trans-membrane ion channels
-Gates are closed, inhibits movement of ions across cell membrane
-When a ligand binds to receptor, gates open and ion can move down-gradient across cell membrane
-"Resting Membrane": closed ion channel
-"Activated Membrane": open ion channel
Effects of Anesthetics on Membrane Integrity
-Membrane area expansion
--increased pressure on ion channels, prevents opening
-Fluidization of phospholipid bilayer
-Inhibition of conversion into space-saving gel phase
Protein Theory of Anesthetic Mechanism of Action
Excitation
-Anesthetics act on proteins in lipid bilayer
-Direct interactions with hydrophobic sites on specific membrane protein channels
--lipophilic anesthetics interact with hydrophobic sites more
-Binding Causes conformational changes in ion channel
-Can stabilize ion channels in closed state
--closed ion channel, no ions move into cell
-Ex: Central nicotinic ACh receptor, NMDA receptor
Protein Theory of Anesthetic Mechanism of Action
Inhibitory
-Enhance function of inhibitory amino acid receptors or ion channels
-Increases influx via GABAa and Glycine receptors
--make membrane potential more negative
-Decreases excitability of the CNS
-Potentiation of inhibitory currents via GABAa receptor and Glycine receptor
Protein theory of Molecular Action and Effects on Nociceptive Signaling
-Inhibits glutamatergic synaptic transmission in dorsal horn of the spinal cord
-No nociceptive inputs to the dorsal horn of the spinal cord
-Inhibits nociceptive signaling
Nociceptive Signaling of Pain
-1st order neuron from free sensory nerve endings into dorsal horn of spinal cord
-2nd order neuron from spinal cord up to brain
-3rd order neuron with brain
Inhibition of Nociceptive Signaling
-Isoflurane administration decreases excitatory currents in neurons
-decreases activity of neurons responsible for transmitting sense of pain
GABAa receptor
-Inhibitory NT, causes membrane hyperpolarization
-Inhbits excitability, hyperpolarizes membrane
-Allows Cl into the cell, increases Cl permeability
-Decreased excitability at the brain level
-Sedation, amnesia, muscle relaxation, anticonvulsion
Glycine Receptor
-Inhibitory NT, causes membrane hyperpolarization
-Increases Cl permeability
-Has effect on spinal reflexes and startle responses
-Major inhibitory receptor in spinal cord
Neuronal Nicotinic ACh receptor
-Excitatory NT
-Highly permeable to monovalent cations and Ca
-Modulates NT release
-Association with memory, nociception, and autonomic functions
Serotonin Receptor
-increases excitability by inhibiting K leaking out of the cell in the resting state
-Roles in arousal and possibly emesis
Glutamate Receptors
-NMDA subtype
-AMPA/Kainate subtype
-Fast excitatory neurotransmission
-Cation conductance for Ca and Mg
-Inhibition prevents perception, learning, memory
--No perception and memory
Group 1a compounds
-Provide amnesia and hypnosis
-No major role in pain transmission or loss of motor functions
--minimal analgesia and immobility
-Etomidate, propofol, Benzodiazepines
Group 1b Compounds
-Provide amnesia, hypnosis, and immobility
-Not much analgesic effect
-Isoflurane, Sevoflurane, barbituates
-Affect GABAa, Glycine, Glutamate, Nicotinic ACh receptots, and K-channels
Group 2 Componds
-Ketamine, NO
-Not very specific
-Provide analgesia
-Provides amnesia?
-Do not provide hypnosis or immobility
Dosing Inhaled Anesthetics
-Determined by Minimum Alveolar Concentration (MAC)
Minimum Alveolar Concentration
-Concentration of anesthetic that provides 50% of patients with immobility when a supramaximal noxious stimulus is applied
-Will cause the given effect in 50% of the patients
-given as volume% or partial pressure
-MAC is agent-specific, depends on drug
-Can be species specific
-Correlates inversely to oil/gas partition coefficient
--More soluble anesthetics are more potent
--less soluble anesthetics are less potent, need higher concentration to reach same behavioral endpoint
-Give cushion to give more safety
--1.3-1.4x MAC will give ED95 (surgical MAC)
MAC value for different agents in Dog
Isoflurane: 1.28-1.5
Sevoflurane: 2.1-2.36
Desflurae: 7.2-10.3
N2O: 188-297
MAC value for agents in Cat
Isoflurane: 1.28-1.63
Sevoflurane: 2.58-3.41
Desflurane: 9.79-10.27
N2O: 255
MAC value for agents in Horse
Isoflurane: 1.31-1.64
Sevoflurane: 2.31-2.84
Desflurane: 7.02-8.06
N2O: 205
MAC value for agents in human
Isoflurane: 1.15
Sevoflurane: 1.58-2.05
Desflurane: 6.00
N2O: 104
Isoflurane MAC% in different species
Dog: 1.28-1.5
Cat: 1.28-1.63
Horse: 1.31-1.64
Human: 1.15
Sevoflurane MAC% in different species
-Dog: 2.1-2.36
-Cat: 2.58-3.41
-Horse: 2.31-2.84
-Human: 1.58-2.05
Desflurane MAC% in different species
Dog: 7.2-10.3
Cat: 9.79-10.27
Horse: 7.02-8.06
Human: 6.00
N2O MAC% in different spcies
-Not soluble AT ALL
-Need a hyperbaric chamber to get pure N2O concentrations to work
-Not potent on own, can be used as adjuvant
-Dog: 188-297
-Cat: 255
-Horse: 205
-Human: 104
Factors Affecting MAC value
Decreased inhalant needed
-Old age
-Pregnancy due to endogenous opioid release
-Hypothermia
-Hypotension due to poor perfusion
-Hypoxemia due to poor perfusion
-Hyponatremia
-CNS depressant drugs/analgesics
Factors Affecting MAC value
Increased inhalant needed
-Hyperthermia
-Hypernatremia
-CNS stimulants
Pharmacological Effects of Inhaled Anesthetics
-Gradual generalized depression of CNS
-Spinal cord goes 1st, then brain
-Iso is more potent than Sevo, more potent than Des
--N2O is much less potent
-In terms of analgesia, N2O is MUCH more potent that Iso, sevo, or des
Cerebral Effects of Isoflurane
-Cerebral metabolic rate decreases
-Seizure occurrence decreases
-Increased ICP
-Increased cerebral blood flow and increased preservation of cerebral blood flow autoregulation
-More responsive to CO2
Cerebral Effects of Sevoflurane
-Decreased cerebral metabolic rate
-Decreased seizures in most
--increased seizures in cats and children
-increased ICP
-Increased cerebral blood flow
-Slightly increased CO2 responsiveness
Cerebral Effects of Desflurane
-Decreases cerebral metabolic rate
-Increases ICP and cerebral blood flow
-Decreases seizures
Adverse effects of Inhaled Anesthetics on Respiratory System
-Cause respiratory depression
-Decrease ventilation, iso more than sevo and des
--increased PaCO2
-Decrease RR (more objective measurement)
-Causes Bronchodilation
-All cause dose-dependent respiratory depression
Adverse effects of Isoflurane on Cardiovascular System
-Significant decreases in myocardial contractility
--Less Ca available for contraction cross-bridging
-Decreased systemic vascular resistance
-Decreased BP
-Increases HR
Adverse Effects of Sevoflurane o Cardiovascular System
-Significantly decreases myocardial contractility
-Decreased systemic vascular resistance
-Decreased BP
-Decreases HR at lowed MAC, increases at MAC greater than 1.5
Adverse effects of Desflurane on Cardiovascular System
-Significantly decreases myocardial contractility
-Decreased systemic vascular resistance
-Decreased BP
Adverse effects of Inhaled Anesthetics on Kidneys
-Decreased renal blood flow due to decreased cardiovascular performance
-Decreased urine output
Adverse Effects of Inhaled Anesthetics on the Liver
-Decreases hepatic blood flow
--Desflurane decreases much more than iso and sevo
-Decreases injectable drug metabolism by 50-70%
--CYP450 system
-Can cause hepatotoxicity, usually negligible for iso, sevo, and des
Adverse effects of Inhaled Anesthetics on Skeletal Muscle
-Decrease skeletal muscle tone
--due to decreases in CNS
-Great muscle relaxants
-Augment neuromuscular blockade by NDNMB
Adverse effects of Inhaled Anesthetics on Fetuses
-Decreased uteroplacental blood flow
-Hypoxia to fetus!
-May me mutagenic? Cause changes in gene expression
--associated with malformations?
Clinical Highlights of Isoflurane
-Most used inhaled anesthetic
-CHEAP
-Useful for mask induction
--non irritating to airways
--Also pungent odor
-Minimal metabolism, no direct toxic effects
Clinical Highlights of Sevoflurane
-Effects are similar to isofluorane
-More expensive that Iso
-Less soluble in blood and tissues than iso
--can achieve changes in anesthetic depth faster
--faster induction and slightly faster recovery
-No pungent order, best choice for mask or inhalant inductions
-Potentially less hypotensive
-Causes less respiratory depression
Sevoflurane and Renal/Hepatic Toxicity
-Hepatic metabolism to inorganic Fluoride
-Has potential to cause renal and hepatic damage
-Renal toxicity has been demonstrated in rats
-No evidence of fluoride-induced renal or hepatic toxicity in humans, dogs, monkeys, horses
-not a major concern
Sevoflurane and Compound A
-Breakdown of Sevo forms vinyl-ether compound (Compound A)
-Causes renal damage in rats
-No evidence that Compound A causes toxicity in clinical vet med
-No large enough concentration of Compound A to cause kidney damage
Desflurane Clinical Highlights
-Least soluble inhalant anesthetic
--Very rapid induction and recovery
-Extremely expensive, prohibitively expensive
-Pharmacodynamic profile is similar to isoflurane
--less CO depressant
-Irritating to airways
-Need special vaporizer due to volatility of gas
-Needs time to warm up
-Basically no metabolism
N2O Clinical Highlights
-Gas at sea level
-Very weak anesthetic in animals
--Have to deliver a high concentrations to have an effect
-Has analgesic properties
-Limited respiratory and cardiovascular depression
-Stimulates sympathetic nervous system
--may improve CO and arterial blood pressure
-Has MAC sparing effects
N2O MAC Sparing effects
-When used with other inhalants, requires less concentration of other inhalant
-More N2O, less inhalant you have to give
-Inhalants cause respiratory and cardiovascular depression
--using N2O can reduce effects
N2O disadvantages
-Increased risk for arrhythmia
-Direct myocardial depression
--increases in sympathetic tone counteract and provide cardiovascular stimulation
-N2O diffuses into closed gas spaces
--into gut and distends gut
--into pleural space, causes pneumothorax
-Risk for hypoxemia in large animals
-Diffusion hypoxia
-Bone marrow depression and changes in blood composition
Balanced Anesthesia
-Combining injectable anesthetics, inhalation anesthetics, and regional anesthetics can provide for faster recovery
-Hypnosis and amnesia
-Analgesia
-Muscle relaxation
-Absence of somatic or autonomic responses to noxious stimuli
Balanced Anesthesia Protocols
-Injectible anesthetics with or without adjuncts
-Inhalation anesthetics
-Regional anesthesia
-Potent opioids, a2 agonists, ketamine, or lidocaine CRI
--has MAC sparing effects
-Neuromuscular blockers
-Can have a dose reduction of individual drugs
-Decreases adverse effects of any one drug
-Will get more rapid recovery