• 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

1/347

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;

347 Cards in this Set

  • Front
  • Back
in pharmacology terms, what is a 1% solution?
1 g/100 mL
what is the conversion factor between kg and lbs?
1 kg = 2.2 lbs
what is the conversion factor between fl oz and mL?
1 fl oz ~ 30 mL
what is the conversion between pints and fl oz?
1 pint = 16 fl oz
what is the conversion between pints and quarts?
2 pints = 1 quart
how many pints are in one gallon?
8 pints = 1 gallon
what information must be on a veterinary prescription for a CIII - CV drug?
- Clinic/DVM name, address, phone number
- date
- Patient ID (name/animal number)
- species
- client name and address
- drug name & strength
- number
- directions for use
- refill instructions
- DEA registration number
- DVM signature
- DVM printed name
what are the four main uses for a drug?
1. prevent disease
2. cure disease
3. treat disease
4. diagnose disease
what is an "Approved Drug?"
drug that meets FDA requirements for safety and effacacy
what is an "Unapproved Drug?"
drug with health claims but no FDA approval
how are food supplements / nutraceuticals treated by the FDA?
there is no FDA oversight; however, they can make no health claims
how are food additives regulated by the FDA?
they must meet the FDA requirement for safety
what are the three basic processes studied by pharmacokinetics?
1. absorption (into bloodstream)
2. distribution (from blood to tissue)
3. elimination (metabolism to inactive drug & excretion)
comment on if there is a species relation to:
- pharmacodynamics
- pharmacokinetics
pharmacodynamics: no species differences

pharmacokinetics: species differences
the study of the mechanism of drug action on the body
pharmacodynamics
the study of what the body does to a drug, in terms of absorption, distribution, and elimination
pharmacokinetics
applied use of drugs in disease states
therapeutics
individualized drug therapy
clinical pharmacology
use of a specific drug for a specific disease
chemotherapy
the study of adverse effects of drugs
toxicology
standardization and dispensing of drugs
pharmacy
science of formulating drugs
biopharmaceutics
study of drugs from plant sources
pharmacognosy
what are the four basic ways in which drugs are classified?
1. chemical structure
2. mechanism of action
3. indication
4. system the drug affects
what are the four basic goals of rational therapeutics?
understanding
1. the disease process and its cause
2. how the drug produces its pharmacodynamic/chemotherapeutic properties
3. pharmacokinetics of the drug, especially how the disease process impacts their fate in the body
4. drug interactions (beneficial and detrimental)
term used to describe how the chemical makeup of a drug affects its pharmacodynamic properties
structure-activity relationship
what four major pharmacodynamic properties does chemical structure affect?
1. effectiveness
2. potency
3. dosage
4. toxicity
what four major chemical properties of a drug affect its pharmacokinetics?
1. molecular weight
2. acid/base properties
3. lipophilicity/hydrophilicity
4. functional groups
what are four general physiological and pathological factors that can alter drug effectiveness?
1. the disease itself influencing drug action

the use of multiple drugs can result in...
2. addition
3. synergism/potentiation
4. antagonism
... of drug effects
for a properly prescribed drug, what is the veterinarian's foremost concern to achieve the desired effect?
owner compliance
how do youth and old age influence drug effectiveness and why?
both groups tend to be more sensitive to a drug.

- youth: unable to metabolize a drug as fast as an adult

- old: decreased renal clearance
what are six major medical-ethical responsibilities of a veterinarian prescribing drugs?
1. proper Dx
2. knowledge of drug actions (efficacy, mechanisms/kinetics, safety, interactions) so they are ordered correctly
3. a VCPR
4. proper supervision
5. consideration of the client for recommended regimen and cost
6. adverse drug reaction reporting
tendency for a drug to bind to a receptor
affinity
what does an agonist do?
initiates the propagation of a message in a cell, thereby causing a biological response
what does an antagonist do?
it has affinity for a site, but does not initiate a biological response
what two pharmacodynamic terms have to do with a drug's binding to a site, and therefore its affect on the body?
affinity and specificity
what are the three basic sites of drug action?
1. outside the cells, not involving receptors (e.g. laxatives, antacids, osmotic diuretics)
2. in or on cells without specific molecular receptors, but rather cause a whole-organelle or whole-cell effect
3. drug action involving receptors
how are target specificity and side-effects related?
increased target specificity tends to decrease side effects
what are the four general pharmacodynamic properties of receptors?
1. specific or not
2. bind to normal body components
3. may have subtypes that can be exploited in therapeutics
4. can be desensitized
what are the two types of dose-response curves and what do they represent?
1. graded (quantitative) - represents the efficacy of a drug with respect to dosage amount
2. quantal - represents the number of individuals that will exhibit a response as a function of dosage
graded (quantitative) dose-response curve:

- axes
- shape of curve
- points on the curve that represent pharmacodynamic properties
ability to bind to a receptor
affinity
the property of effecting a response when a drug is bound to a receptor
- or -
magnitude of response obtained from drug-receptor binding
efficacy or intrinsic activity
a drug that does not cause a maximal response
partial agonist
concentration of drug at a receptor needed to initiate a response
threshold
comparative term to describe the position of two or more agonists on a dose-response graph
potency
what is surmountable antagonism and how does it change the dose-response curve?
when two drugs bind to the same receptor the more potent drug requires a higher dosage to have its effect. This will shift the curve to the right (including threshold), but not change its shape.
what is insurmountable antagonism and how does it change the dose-response curve?
1. when one drug binds to a receptor, but not at the target site, it changes the shape of the target site, thus making the other drug less potent.
- or -
2. when the drug has no agonist activity, but decreases the pool of receptors
- or -
3. with a partial agonist competitively binding

This will change the shape of the curve, lower the maximum efficacy, and possibly raise the threshold.
what are the axes of the two types of quantal dose-response curves?
1. number responding versus log dose (bell-shaped curve) - less common
2. cumulative number responding versus log dose (integral of #1) - most common
how is the therapeutic index calculated? What do greater values mean? What is required of the dose-response curves for this data to be meaningful?
LD50/ED50 or TD50/ED50

LD50 = lethal dose 50%, ED50 = effective dose 50%, TD50 = toxic dose 50%

greater numbers mean a safer drug. The curves must be parallel in shape
how is the margin of safety calculated? What do the values mean?
TD01/ED99

TD01 = toxic dose 1%, ED99 = effective dose 99%

It represents how much one can overdose a patient before toxic effects set in. It must be greater than 1 to be considered safe.
how do the following effects affect the ability of drug translocation

- higher molecular weight?
- bound to a plasma protein?
- lipophilicity?
- charge state (degree of ionization)?
- body pH?
- drug pKa?
- higher molecular weight: lower
- bound to a plasma protein: lower
- lipophilicity: the more lipophilic, the easier it is to cross the membrane
- charge state (degree of ionization): charged molecules don't cross; uncharged molecules can cross
- body pH: lower pH will increase the ability of weak acids to cross; higher pH will increase the ability of weak bases to cross. ***This is assuming that the conjugate base of the weak acid is an anion and that the conjugate acid of the weak base is a cation.
- drug pKa: lower pKa favors lower pH; higher pKa favors higher pH
what is the main "goal" of the body metabolizing a drug?
to make it more water soluble for elimination
a drug designed to become more potent after it has been metabolized
prodrug
what properties of a drug are changed by biotransformation?
- water solubility
- activation/inactivation
- potency
- affinity and specificity
- duration of drug action
- side effects and/or toxicity
the passage of a drug through a cellular membrane
translocation
in what organs does drug biotransformation occur?
- LIVER (main site)
- intestine
- kidney
- plasma
- others
in what organelles does drug biotransformation occur?
- microsomes (SER) - major
- mitochondria
- cytosol
why do neonates and elderly animals have less efficient biotransformation of drugs?
because they have lower liver function
generally, what organic functional groups are most susceptible to biotransformation?
hydroxyl, carboxyl, amino
what are the phases of drug metabolism and what is each phase's general purpose?
Phase I: transformation
Phase II: conjugation
what are the three most common Phase I biotransformation reactions in order of prominence?
1. oxidation
2. reduction
3. hydrolysis
which Phase I biotransformation is most rapid and what are the most common organic functional groups associated with this reaction?
hydrolysis is fastest.

- esters, amides, glucuronides, glycosides
why is oxidation a more prominent biotransformation pathway than reduction?
because reduction requires a relatively oxygen-free environment
which phase of drug metabolism has the most species differences and why?
Phase II, because different species have different amounts of the transferase enzymes required for the conjugation reactions.
what general type of reaction is in a Phase II biotransformation, and what is required to catalyze these reactions?
these are conjugation reactions and require transferase enzymes
what are the five predominant Phase II biotransformation reactions, in order of most important to least important? note species differences.
1. Glucuronic acid conjugation (cats and neonates are deficient in the transferase enzyme)
2. Sulfate conjugation (pigs have limited capability)
3. Mercapturic acid conjugation (primates and guinea pigs are deficient in the transferase enzyme)
4. Acetylation (dogs are deficient)
5. Methylation
which species is/are deficient in the Phase II biotransformation enzyme for

- glucuronic acid conjugation?
- sulfate conjugation?
- mercapturic acid conjugation?
- acetylation?
- methylation?
- glucuronic acid conjugation: cats and neonates
- sulfate conjugation: pigs
- mercapturic acid conjugation: primates and guinea pigs
- acetylation: dogs
- methylation: none noted
glucuronic acid conjugation:

- organ and organelle
- reversibility
- product
- functional groups
- mechanism
- species specific comments
- organ and organelle: this is a steroid hormone pathway found in the liver that is affected by age and health; microsome (SER)
- reversibility: reversible (GI flora can do this)
- product: organic acid for tubular secretion
- functional groups: hydroxyl, carboxyl, amino, also sulfhydryl
- mechanism: UDPGA + drug + transferase --> product
- species specific comments: cats and neonates are deficient in the transferase enzyme
sulfate conjugation

- organ and organelle
- reversibility
- product
- functional groups
- mechanism
- species specific comments
- organ and organelle: takes place primarily in liver; cytosol
- reversibility: irreversible
- product: sulfate ester
- functional groups: aromatic hydroxyl, aromatic amino, sometimes aromatic carboxyl
- mechanism: PAPS + drug + transferase enzyme --> conjugate;
- species specific comments: pigs are deficient in the transferase enzyme
mercapturic acid formation

- organelle
- reversibility
- product
- functional groups
- mechanism
- species specific comments
- organelle: cytosol; a reserve pathway for many of the same functional groups that under glucuronidation/sulfation
- reversibility: irreversible
- product: a mercapturic acid
- functional groups: carboxyl, hydroxyl, amino, halogen, sulfhydryl
- mechanism: drug + glutathione (GSH) + 4 enzymes --> product
- species specific comments: primate and guinea pig are limited
acetylation in a Phase II biotransformation:

- organelle
- reversibility
- product
- functional groups
- mechanism
- species specific comments
- organelle: cytosol
- reversibility: irreversible
- product: amide
- functional groups: amino group only (including sulfa drugs)
- mechanism: AcCoA + drug + transferase enzyme
- species specific comments: dogs are deficient
methylation in a Phase II biotransformation

- organelle
- reversibility
- product
- functional groups
- mechanism
- species specific comments
- organelle: cytosol
- reversibility: reversible
- product: methylated product
- functional groups: endogenous alcohols, amino, sulfhydryl
- mechanism: SAM + drug
- species specific comments: none noted
how many Phase II biotransformation reactions are reversible and which ones are they?
two: glucuronidation and methylation
what are the main irreversible Phase II biotransformation reactions?
- sulfation
- metcapturic acid formation (glutathione)
- acetylation
which biotransformation reactions take place in microsomes (SER)?
only two: oxidation and glucuronidation
what is the main organ in which microsomal biotransformation takes place?
liver
what are traits and conditions that affect microsomal enzyme activity?
- age
- sex
- disease state
- genetics (e.g. breed)
- environment
- nutritional status
testing of biotransformation enzyme isoforms in the DNA for pharmacokinetic properties such as safety and efficacy
pharmacogenetics
what is the predominant enzyme involved with microsomal oxidative biotransformation? What type of molecule is it?
Cytochrome P450, which is an iron-containing protein
what is induction, with regards to pharmacokinetics? what are its pharmacological effects?
- up-regulation of the production of microsomal CP450 due to increased demand for drug oxidation (e.g. increased amount to drug intake)

- can result in shorter duration and/or lower drug concentrations
what is the major reason that the body will decrease protein synthesis associated with inhibition of microsomal enzymes?
disease
with regards to pharmacokinetics, what is inhibition, what are the two ways in which this can occur, and how does this change the body's response to a drug?
- decreased activity of microsomal enzymes due to

1. decreased protein synthesis (delayed effect)
2. competitive inhibition (immediate effect)

this can cause hypersensitivity to a drug
in what four major ways can the veterinarian apply the knowledge of drug chemical structure, reactions they may undergo, and species uniqueness?
1. increase drug effectiveness (e.g. species, age)
2. help establish dosages and regimens, especially in extralabel drug use
3. avoid drug toxicities
4. consider special situations (such as sensitivity of cats and Greyhounds, aged animals, animals with liver disease)
what probable biotransformation pathway(s) exist for esters and amides?
hydrolysis
what probable biotransformation pathway(s) exist for aromatic rings (the ring itself, not its functional groups)?
oxidation (hydroxylation)
what probable biotransformation pathway(s) exist for aliphatic hydroxyl groups?
glucuronidation > sulfate conjugation
what probable biotransformation pathway(s) exist for aromatic hydroxyl groups?
glucuronic acid conjugation, sulfation, mercapturic acid (glutathione) conjugation, methylation
what probable biotransformation pathway(s) exist for aliphatic carboxyl groups?
glucuronidation >> sulfation, mercapturic acid (glutathione) conjugation
what probable biotransformation pathway(s) exist for aromatic carboxyl groups?
glucuronidation >> sulfation, mercapturic acid (gultathione) conjugation
what probable biotransformation pathway(s) exist for aliphatic amines?
oxidation (deamination)
what probable biotransformation pathway(s) exist for aromatic amines?
glucuronidation, sulfation, mercapturic acid (glutathione) conjugation, acetylation

methylation (natural substrates)
what probable biotransformation pathway(s) exist for thiols?
glucuronidation and mercapturic acid (glutathione) conjugation
name three common types of reductive biotransformation reactions
1. azo reaction (azo --> amino)
2. nitro reaction (nitro --> amino)
3. alcohol (geminal diol --> primary alcohol)
name five common drug functional groups subject to hydrolysis.
1. ester
2. phosphate ester
3. amide
4. glucuronide conjugate
5. glycoside
where must a drug first go to be considered "in the body"?
bloodstream
what are advantages and disadvantages of oral administration?
advantages:
- convenient, cheap, no sterility needed, variety of dose forms
- can get the dose back if you act quickly
disadvantages:
- variability due to physiology, feeding, Dz, etc
- intractable patients
- first-pass effect (goes to liver first)
what patient and pharmaceutical differences increase variability of absorption from oral administration?
- pill compression, coatings, suspending agents, etc.
- GI transit time, inflammation, malabsorption syndromes
what aspects of the stomach (abomasum) can affect oral administration of drugs?
- mechanical preparation
- "flat" absorptive surface
- pH extremes
what aspects of the rumenoreticulum can affect oral administration of drugs?
- stratified squamous epithelium
- pH varies with diet
- metabolism by bacterial flora
- large amount of volume compared to body water
what aspects of the small intestine can affect oral administration of drugs?
- large absorptive surface
- specialized absorptive functions
- relatively neutral pH
what are advantages and disadvantages of IM administration of drug?
advantages:
- more consistent absorption (IM ~ IV)
- certainty of administration
- depot or sustained effect possible
- viable route for patients that can't be easily given other routes (e.g. unconscious, intractable, dehydrated)
disadvantages:
- more difficult for owners of small animals
- pain
- muscle damage
- cannot recover dose
what is the three-step process of drug absorption?
1. dissolve (lipid/suspension) into or mix (aqueous) with tissue fluid
2. diffuse into capillaries
3. carried to circulatory system
what does it mean when a drug "falls" out of solution?
the vehicle carrying the drug is absorbed more quickly than the drug, leaving a residue in the tissue
how does absorption of a spherical lipid bolus vary with time?
it slows (less surface area)
what are advantages and disadvantages of SQ administration?
advantages:
- can be given by owner
- vasoconstrictor can be added to prolong effect at site of interest
disadvantages:
- variability
how do heat, cold, and hydration status effect drug absorption from a SQ injection?
- heat: increases blood flow; more rapid absorption
- cold and dehydration: decreases blood flow; slower absorption
what are the two main pathways in which topical drugs enter the body?
1. diffusion through the stratified squamous epithelium
2. "passage" through adnexal structures
what are four patient and pharmaceutical factors affecting topical drug absorption?
1. lipid solubility and molecule size
2. skin hydration and abrasion
3. area of application
4. ambient and patient temperature
how does the vehicle affect topical drug absorption
- drugs in "like" vehicles are retained on skin surface (aqueous/water of lipid/lipid)
- drugs in "unlike" vehicles are absorbed (suspensions)
what are advantages and disadvantages of intraperitoneal drug administration?
advantages:
- larger absorptive surface than IM/SQ
disadvantages:
- drugs/vehicles may cause peritonitis
- damage to organs by needles
- injection into organs
what are advantages and disadvantages to intrathecal drug administration?
advantages:
- direct delivery to CSF
disadvantages:
- difficult to calculate because CSF volume is not proportional to body weight
- toxicity is likely and may be unusual
- risk of introducing infection
- very slow absorption because it is diffusion-limited
what comprises the vascular space?
plasma volume + RBC volume + WBC volume
what percentage of body weight is the vascular space?
7%
what three important equilibria exist in the vascular space?
1. solution and plasma proteins
2. ionized and unionized drug
3. plasma and cells
what comprises the tissue space
= the body space - vascular space - structural protein - bone
what three important equilibria exist in the tissue space?
1. water and tissue proteins
2. ionized and unionized drug
3. intracellular fluid and extracellular fluid
what comprises the extracellular space and what percentage of of the body weight is it?
plasma + extracellular tissue space
15-20% of body weight
how does extracellular space vary with age?
babies have more
what comprises the intracellular space and what percentage body weight is it?
intracellular vascular space + intracellular tissue space
35-45% of body weight
what two important equilibria exist in the extracellular space?
1. water and proteins
2. ionized and unionized drugs
what important equilibrium exists in the intracellular space?
ionized and unionized drug
how long after administration is drug distributed in the
- vascular space?
- tissue space?
- extracellular space?
intracellular space?
- vascular space: 10-30 minutes
- tissue space: hours/days/weeks
- extracellular space: 30-90 minutes
- intracellular space: 30 minutes to 12+ hours
what are the "reserved" spaces in the body, which have special barriers between plasma and tissue fluid?
1. CSF
2. aqueous humor
3. prostatic fluid
in what four ways do drugs move from extracellular vascular space to extracellular tissue space?
1. transcytotic
2. endothelial junctions (during inflammation)
3. diffusion through endothelial membranes
4. carried in cells or on proteins (very special circumstances)
in what two ways can drug move between extracellular space and intracellular space?
1. diffusion through lipid bylayer
2. active uptake (esp. WBCs)
the process of drug being reabsorbed from the bile in the intestines
enterohepatic circulation
what is the process of enterohepatic circulation? how does it affect volume of distribution? elimination rate? how can this circulation be interrupted clinically?
- drug or its Phase II conjugate excreted in bile
- drug reabsorbed or its conjugate cleaved by flora and reabsorbed
- increases Vz
- slows elimination rate
- can interrupt via activated charcoal or other substances that will prevent reabsorption or unconjugation
mammary excretion:
- how does it get into the milk?
- what properties of the drug affect this process?
- how does mastitis affect this process?
- why does drug stay in the milk?
- non-ionic diffusion
- lipid solubility and molecular size
- inflammation reduces barrier to penetration
- milk is lower pH, so it ion-traps the drug
why is mammary excretion important?
- may affect treatment of mastitis
- nursing animals may be exposed to drug
what are two reasons why salivary excretion can be important pharmacokinetically?
1. recycles certain drugs like enterohepatic circulation (Vz goes up)
2. trap in rumen based on pH
what are the two general processes of drug elimination?
1. biotransformation
2. excretion
what is the major route of elimination of a drug that is
- lipid soluble?
- aqueous?
- protein-bound?
- lipid soluble: metabolism
- aqueous: direct excretion
- protein-bound: metabolism
what four factors contribute to the rate of drug metabolism?
1. metabolic activity for a drug
2. blood flow to the metabolizing organ
3. health of the metabolizing organ
4. health of the circulatory system
what organ is most important for metabolizing autacoids?
lungs
what is secreted via active biliary excretion?
- drugs with MW > 300
- mostly conjugates
what is excreted by passive biliary excretion?
- drugs with MW < 300
- biliary concentrations similar to plasma water
what three factors sum up renal elimination?
renal elimination = glomerular filtration + tubular secretion - passive reabsorption
what happens to an unionized drug when concentrated in the nephron? why?
it gets reabsorbed because concentration in nephron > concentration in blood
how are protein-bound drugs eliminated by the kidney?
by active tubular secretion
what two things most commonly reduce renal passive reabsorption? how does this affect elimination rate of the drug?
1. disease (accidental)
2. therapy (intentional)
- increases the elimination rate of the drug
when does reducing the passive reabsorption of a drug not work?
- if reabsorption is not an important part of renal elimination
by what two ways can renal passive reabsorption be reduced therapeutically?
1. high urine production (less contact time with epithelium for drug to be reabsorbed)
2. urine pH (ionized drug will not be reabsorbed)
what ratio does the volume of distribution, Vz represent at any given time? How is it determined experimentally?
Vz = amount of drug in the body / plasma concentration

experimentally:
Vz = dose/Cp0, where Cp0 is the y-intercept of a plot of log(plasma conc) versus time
normally, how much of blood volume is comprised of plasma water?
55%
if clearance was 100% efficient, what would determine clearance rate?
blood flow
how is organ clearance calculated?
Clearance = Q x E

where Q is blood flow to the organ and E is efficiency of extraction
what are the three major components of total body clearance?
Cl_total = Cl_hepatic + Cl_renal + Cl_pulmonary
how is clearance measured experimentally?
Clearance = Vz x λz

where λz is the SLOPE of a plot of ln(drug plasma concentration) versus time
after how many half-lives has a drug been "effectively eliminated?" what percentage of drug is this?
5 half-lives, 97%
what is a loading dose?
an initial high dose of a drug given to shorten the time to reach steady-state concentrations
what is the absorption rate constant? How is this number applied clinically?
- ka describes the rate of drug movement to the site of administration to the circulatory system
- ka will determine the time required to reach a peak concentration of the drug
what is bioavailability?
the fraction of the dose of a drug that gets into the bloodstream
what are two major factors that affect bioequivalence between two drugs or dose forms?
1. bioavailability
2. rate of absorption
miosis:
- branch of ANS?
- receptors?
(pupil constrition)
- parasympathetic
- M3, M2
visual accommodation:
- branch of ANS?
- receptors?
- parasympathetic
- M3, M2
lacrimation:
- branch of ANS?
- receptors?
- parasympathetic
- M3, M2
pharyngeal mucus secretion:
- branch of ANS?
- receptors?
- parasympathetic
- M3, M2
salivation:
- branch of ANS?
- receptors?
- parasympathetic and sympathetic
- M3, M2, α1
bronchoconstriction:
- branch of ANS?
- receptors?
- parasympathetic
- M3 = M2
pulmonary mucus secretion:
- branch of ANS?
- receptors?
- parasympathetic
- M3, M2
lower heart rate:
- branch of ANS?
- receptors?
- parasympathetic
- M2
↓ atrioventricular conduction:
- branch of ANS?
- receptors?
- parasympathetic
- M2
higher gastric acid secretion:
- branch of ANS?
- receptors?
- parasympathetic
- M3, M2
increased GI motility:
- branch of ANS?
- receptors?
- parasympathetic
- M3, M2
micturition:
- branch of ANS?
- receptors?
- parasympathetic
- M3
erection (male):
- branch of ANS?
- receptors?
- parasympathetic
- M3
mydriasis:
- branch of ANS?
- receptors?
(pupil dilation)
- sympathetic
- α1
vasoconstriction:
- branch of ANS?
- receptors?
- sympathetic
- α1
sweating:
- branch of ANS?
- receptors?
- sympathetic
- M3, M2
bronchodilation:
- branch of ANS?
- receptors?
- sympathetic
- β2
increased heart rate:
- branch of ANS?
- receptors?
- sympathetic
- β1
increased atrioventricular conduction:
- branch of ANS?
- receptors?
- sympathetic
- β1
increased heart contractility:
- branch of ANS?
- receptors?
- sympathetic
- β1
decreased GI motility:
- branch of ANS?
- receptors?
- sympathetic
- α and β
renin secretion:
- branch of the ANS?
- receptors?
- sympathetic
- β1
increased hepatic glycogenolysis:
- branch of ANS?
- receptors?
- sympathetic
- β2
uterine relaxation:
- branch of ANS?
- receptors?
- sympathetic
- β1
urinary retention:
- branch of ANS?
- receptors?
- sympathetic
- α1
ejaculation:
- branch of ANS?
- receptors?
- sympathetic
- α1
for the parasympathetic and sympathetic branches of the ANS
- what is the preganglionic receptor?
- length of preganglionic fiber
- length of postganglionic fiber
PARASYMPATHETIC
- nicotinic preganglionic receptor
- long preganglionic fiber
- short postganglionic fiber
SYMPATHETIC
- nicotinic preganglionic receptor
- short preganaglionic fiber
- long postganglionic fiber
what are the postganglionic receptors for
- parasympathetic?
- sympathetic?
- somatic?
- parasympathetic: muscarinic
- sympathetic: α and β
- somatic: nicotinic (there is no ganglion)
what are the primary neurotransmitters that are agonists for the following receptors:
- nicotinic?
- muscarinic?
- α and β?
- nicotinic: acetylcholine
- muscarinic: acetylcholine
- α and β: norepinephrine (α), epinephrine (α and β)
which nerve endings are the primary targets of drugs?
post-ganglionic
what differentiates the different autonomic nervous system receptor sub-types?
the type of second-messenger
which enzyme
- produces acetylcholine in the neuron?
- breaks down acetylcholine in the synapse?
- choline acetyltransferase produces acetylcholine
- acetylcholinesterase breaks down AcCh in the synapse
which enzymes are involved in the breakdown of norepinephrine, epinephrine, and dopamine in the synapse?
- monoamine oxidase (MAO)
- catecholamine O-methyltransferase (COMT)
which enzyme is required to synthesize epinephrine from norepinephrine?
N-methyltransferase
what is the basic synthetic pathway from amino acid to epinephrine?
tyrosine --> DOPA --> dopamine --> norepinephrine --> epinephrine
what are major target organs for:
- α1
- α2
- β1
- β2
- dopamine
- α1: blood vessels, eye, uterus, urinary sphincter, CNS
- α2: CNS
- β1: heart
- β2: bronchi, muscle blood vessels, pancreas, bladder wall, uterus, liver
- dopamine: CNS, kidney, vasculature
what is the meaning of the suffixes "mimetic" and "lytic"?
- mimetic = agonist
- lytic = antagonist
what are the effects of the SNS and PNS on the eye?
- SNS dilates (α1)
- PNS constricts
what are the effects of the SNS and PNS on the heart?
- SNS increases hr (β1)
- PNS decreases hr
what are the effects of the SNS on blood vessels?
- α1 constricts blood vessels and raises bp
- β2 increases blood flow to skeletal muscles
what are the effects of the SNS on the bronchi?
- β2 dilates bronchi
what are the effects of the PNS on the GI tract and urination
- PNS increases peristalsis and urination
what are the effects of the PNS on exocrine glands?
- PNS increases secretions
how do endogenous substances of the autonomic nervous system tend to differ from synthetics?
endogenous substances tend to be more potent and shorter acting
what is the chemical structure of direct acting sympathomimetics?
phenylethylamines
if a sympathomimetic is modified at the alkyl group, how is it affected?
kinetics change; e.g. the drug lasts longer
if the nitrogen group on a sympathomimetic is modified, how does affect its action?
- affects pharmacodynamics
- preference of β over α receptors
if the substitution on the aromatic ring of a sympathomimetic is changed, how does that affect its action?
- dynamics and kinetics
- β > α
- affects duration of action
how does removing a hydroxyl group from a catechol based sympathomimetic affect its activity?
- increases stability because COMT enzyme no longer works on this substrate
- increases its α activity
how does substituting an OH group on the aliphatic carbons of a sympathomimetic affect its activity?
makes it more polar
how does substituting a methyl group on the aliphatic carbons of a sympathomimetic affect its activity?
increases stability because MAO will not work on this substrate
how does substitution of a methyl or larger group onto a sympathomimetic affect its activity?
increases its β activity
how does rearranging and/or substituting the OH groups on a sympathomimetic affect its activity?
makes it more β2 selective
what is the receptor selectivity of the following catecholamines:
- norepinephrine?
- epinephrine?
- isoproterenol?
- norepinepherine: α
- epinepherine: α and β
- isoproterenol: β
how are sympathomimetics used to affect the cardiovascular system?
- α1 drugs: decrease hemorrhage; nasal decongestant; localize drugs; increase BP; shock treatment
- α2 drugs: lower BP
- β1 drugs: raise HR and contractility
how are sympathomimetics used to affect the respiratory system?
β2 drugs are bronchodilators
how are sympathomimetics used to affect the eyes?
α1 drugs are mydriatics
how are sympathomimetics used to affect the urethra and uterus?
α1 drugs cause smooth muscle contraction
how are sympathomimetics used to affect the CNS?
- α1 and dopaminergic drugs are stimulants
- α2 drugs are tranquilizers and analgesics
what is tachyphylaxis?
diminished response to a drug as repeated doses are given
what are four general side-effects/toxic effects of sympathomimetics
1. action at non-selective sites (e.g. β2 pancreas receptors)
2. overstimulation (especially heart, BP, CNS; notable for α1 drugs)
3. tolerance and/or tachyphylaxis
4. drug interactions (e.g. arrhythmias with anesthetics)
α1 drugs:
- examples
- indications
- side effects
- epinephrine (more β than α), norepinephrine (more α than β)
- vasoconstrictors, mydriatics, increase smooth muscle tone (e.g. bladder, uterus)
α2 drugs:
- examples
- indications
- action
- side-effects
- xylazine, (detomidine, dexmedetomidine) - tranquilizers; (clondine) - antihypertensive (amitraz) - insecticide
- CNS effects: tranquilization, lower BP, analgesia
- decreases presynaptic outflow
β receptor agonists:
- examples
- indications
- isoproterenol (β2 bronchodilator; β1 cardiostimulatory effects)
- epinephrine (same effects, but also affects α)
what are the five types of sympathomimetics that act by indirect mechanisms?
1. phosphodiesterase inhibitors - especially methylxanthines (THEOPHYLLINE, caffeine, theobromine)
2. MAO inhibitors
3. monoamine reuptake inhibitors
4. CNS stimulants that increase the effects of norepinephrine or dopamine (e.g. amphetamine or methylphenidate)
5. COMT (catechol-O-methyltransferase) inhibitors
what is the basic reaction pathway of glucuronidation?
UDP-glucose --> UDP-glucuronic acid --(drug + transferase) --> drug conjugate
which drug conjugate is especially labile to intestinal flora?
glucuronic acid conjugates
why can certain substances such as acetaminophen, furosemide (Lasix), and bromobenzene, which undergo mercapturic acid conjugation, cause toxicities
because in high doses, the mercapturic acid pathway becomes saturated
in which type of conjugation is the moiety directly added to the drug?
acetylation
name three types of reductions
1. azo reaction
2. nitro reaction
3. alcohol reaction
what specifically is the reason for species differences in the ability to oxidize drugs?
the quantity and quality of the various Cytochrome P450 (CYP) enzymes
are Cytrochrome P450 enzymes required for all oxidation biotransformations? Why?
no, because CYP enzymes are only in the microsomes. Some oxidation can occur outside of the microsomes
what biochemicals must be present for microsomal drug oxidation?
NADPH, oxygen, Mg2+, flavoprotein, Cytrochrome P450
what is necessary for microsomal enzyme induction?
exposure to a chemical that increases protein synthesis, especially CYP450
what are the two major concerns arising as a result of enzyme induction?
1. drug tolerance
2. drug interactions
in which organelle is monoamine oxidase found?
mitochondria
list four areas outside the microsomes where drug metabolism can occur?
1. cytosol
2. blood
3. microflora
4. mitochondria
what are the three body compartments and their constituents?
1. central compartment - blood volume and organs of elimination
2. peripheral compartment - muscle, SQ, lung
3. deep compartment - fat, kidneys
how is elimination rate constant calculated?
λz = Clearance Time / Vz
what are four major uses of sympatholytics? Three common side-effects?
USES
1. lower BP
2. lower ocular pressure
3. depress CNS function
4. lower HR
SIDE-EFFECTS
1. increased peristalsis
2. decreased ejaculation
3. increased sodium retention
what is one major use for
- α1 blockade?
- α2 blockade?
- α1 blockade: lower blood pressure
- α2 blockade: antidotes to α2 agonists
what are three examples of uses of direct β antagonists?
1. arrhythmias
2. lower heart rate and cardiac output
3. lower ocular pressure (topical)
why should you not suddenly take a patient off of a β-antagonist that has been using the drug PO for a long time?
can cause cardiac arrhythmias and heart failure
what are two common side-effects of direct β antagonists
1. arrhythmia
2. increased airway resistance
why don't we use selective β2 blockers?
becuase they would cause severe side effects such as bronchoconstriction
give an example of a non-selective β blocker and its effects
propanolol
- lowers HR (competitive antagonist for β1 at heart)
- bronchoconstriction (competitive antagonist for β2 in bronchioles)
what are the three general mechanisms of action of indirect-acting sympatholytics?
1. antidopaminergic
2. reduces NE by synaptic action
3. decrease sympathetic outflow
an AGONIST of which receptor will cause indirect-acting sympathoLYTIC properties? What is a common drug that does this?
- α2 receptor
- xylazine
what are two examples of antidopaminergic indirect-acting sympatholytics
acepromazine and metoclopramide
what are the two main uses for antidopaminergic indirect-acting sympatholytics?
1. tranquilizer
2. antiemetics (DA causes emesis)
how do indirect-acting sympatholytics decrease available NE at the synapse? what are some of their therapeutic uses? what is a common side-effect?
- they are Calcium channel blockers (Ca influx into the neuron is required to release NE)

TREATS
- cardiovascular hypertrophy
- arrhythmias
- hypertension
SIDE-EFFECT: microsomial enzyme induction
what are four common uses for α2-agonists?
1. tranquilizers
2. antihypertensives
3. analgesics
4. emetics
examples of direct-acting parasympathomimetics?
1. acetylcholine
2. metoclopramide
3. pilocarpine
what are seven uses for parasympathomimetics
1. miotics
2. glaucoma
3. smooth muscle atony
4. urine retention
5. neuromuscular disease
6. anthelmintics and insecticides
7. antidotes
what are five contraindications for parasympathomimetics?
1. low heart rate
2. hypotension
3. asthma
4. hyperthyroidism
5. ulcers
what are the two binding sites of the acetylcholinesterase enzyme? what binds to each of these components?
1. anionic site - binds to quarternary ammonium ions
2. esteratic site - binds to esters and amides
what are the two common types of direct acting parasympathomimetics?
1. ester and amide acetylcholine-like structures
2. plant alkaloids
what is one example of a synthetic acetylcholine-like drug
metoclopramide
what is an example of a plant alkaolid that acts as a direct acting parasympathomimetnic?
pilocarpine
what is the major advantage of using a direct versus an indirect parasympathomimetic?
fewer side-effects
what is the mechanism of an indirect parasympathomimetic?
cholinesterase inhibitor
what are the two types of cholinesterase enzymes and where are they located?
- true: neural tissue, RBC
- pseudo: plasma, liver
which type of cholinesterase enzyme do indirect parasympathomimetics target?
"true" cholinesterase enzymes in neural tissue
how does cholinesterase inhibition cause a parasympathomimetic effect? is this a reversible or irreversible process? what is a specific example of a cholinesterase inhibitor?
- it increases the amount of AcCh in the synapse
- reversible
- neostigmine
what are short acting indirect cholinesterase inhibitors used for?
1. diagnostics
2. antidote for NM blocking agents (recovery from anesthesia)
what five examples of what long acting indirect cholinesterase inhibitors may used for?
1. neuromuscular diseases
2. miotics (glaucoma treatment)
3. myasthenia gravis
4. insecticides, antiprotozoal
5. Alzheimer's in people
what are the two chemical classes of cholinesterase inhibitors?
1. quarternary ammonium salts
2. carbamates
what is the only indication for therapeutic use of an irreversible cholinesterase inhibitor? where on cholinesterase do they bind? what type of drugs are they? what other non-therapeutic uses do this type of drug they have?
- glaucoma meds (miotics)
- they are organophosphates
- they bind to the esteratic site of cholinesterase
- also used as anthelminthics, antiprotozoals, insecticides
why are antidopaminergic compounds classified as indirect-acting sympatholytics?
because dopamine is the precursor to norepinephrine
what is the general chemistry profile of a parasympatholytic?
quarternary or tertiary amines, polar (many are esters), affinity for peripheral muscarinic sites, don't cross BBB well
name two examples of direct-acting parasympatholytics
1. atropine
2. acepromazine
name seven uses of parasympatholytics
1. cardiovascular: increase heart rate and cardiac output
2. lowers GI peristalsis
3. decrease exocrine secretions
4. mydriatics
5. decrease respiratory secretions; mild bronchodilation
6. CNS: motion sickness, antitremornergic, sedative, tricyclic antidepressants
7. antidotes, especially to carbamate and organophosphate poisoning
what is the common reason why a parasympatholytic has side-effects?
overdose
what are three common types of parasympatholytics?
1. Belladonna alkaloids (atropine)
2. Synthetics
3. Non-selective compounds (acepromazine)
what two sites in the body are the most common targets for agents acting at nicotinic sites?
1. autonomic ganglia
2. neuromuscular junction
what are the two classes of drugs that cause blockade at nicotinic receptors and what is their mechanism of action?
1. nondepolarizing blockade - surmountable antagonism
2. depolarizing blockade - stimulation, then block of receptor
what is an example of a non-specific nicotinic stimulator?
acetylcholine
in what circumstances would a direct ganglionic blocking agent be used?
- lower BP in emergencies or surgery
- paralyze muscles in surgery
heart rate:
- primary tone
- effects of stimulation
- effects of ganglionic blockade
- primary tone: PNS
- effects of stimulation: bradycardia
- effects of ganglionic blockade: tachycardia, arrhythmias
blood pressure:
- primary tone
- effects of stimulation
- effects of ganglionic blockade
- primary tone: SNS (α1)
- effects of stimulation: increased BP
- effects of ganglionic blockade: vasodilation, venous pooling, decreased venous return, decreased cardiac output, **orthostatic hypotension (hypotension when standing up)
bronchial smooth muscle:
- primary tone
- effects of stimulation
- effects of ganglionic blockade
- primary tone: SNS (β2)
- effects of stimulation: relaxation
- effects of ganglionic blockade: bronchoconstriction
urinary bladder:
- primary tone
- effects of stimulation
- effects of ganglionic blockade
- primary tone: PNS
- effects of stimulation: micturition
- effects of ganglionic blockade: urine retention
eye
- primary tone
- effects of stimulation
- effects of ganglionic blockade
- primary tone: PNS
- effects of ganglionic stimulation: miosis
- effects of blockade: mydriasis and cycloplegia (paralysis of the ciliary muscle of the eye)
salivary glands:
- primary tone
- effects of stimulation
- effects of ganglionic blockade
- primary tone: PNS
- effects of stimulation: secretions
- effects of ganglionic blockade: reduced secretion, dry mouth
what is the mechanism of neuromuscular blocking agents? what are they used for? what is a major adverse effect of overdose? what is an example of a drug?
- they block the pseudocholinesterases in the NM junctions
- they increase muscle relaxation, especially at surgery
- overdose can lead to respiratory muscles being paralyzed (without loss of consciousness)
- neostigmine
what are the four classes of CNS neurotransmitters of clinical importance and the specific natural neurotransmitters that are classified under these types?
1. ester: acetylcholine
2. monoamines: NE, DA, serotonin (5-HT)
3. amino acids: GABA, Glutamate
4. Neuropeptides: enkephalins (natural opoids)
CNS acetylcholine:
- putative role
- receptors
- degradative pathway(s)
- putative role: cognition, neural development
- receptors: muscarinic, nicotinic, other
- degradative pathway(s): acetylcholinesterase
CNS norepinephrine:
- putative role
- receptors
- degradative pathway(s)
- putative role: multipurpose
- receptors: α2 for tranquilization, stimulatory CNS receptors
- degradative pathway(s): reuptake, MAO, COMT
CNS dopamine:
- putative role
- receptors
- degradative pathway(s)
- putative role: multi-purpose (mustly stimulatory)
- receptors: D1, D1, D3, D4, D5
- degradative pathway(s): reuptake, MAO, COMT
CNS serotonin:
- putative role
- receptors
- degradative pathway(s)
- putative role: behavior
- receptors: 5-HT1, 5-HT2, etc.
- degradative pathway(s): reuptake, MAO; (note no COMT because serotonin has only one hydroxyl group)
CNS GABA:
- putative role
- receptors
- putative role: inhibitory NT
- receptors: GABA-A, GABA-B
CNS glutamate:
- putative role
- receptors
- putative role: excitatory NT
- receptors: NMDA, many others
CNS enkephalins:
- putative role
- receptors
- putative role: pain perception and pain tolerance
- receptors: mu, kappa, delta opoid receptors
what are the five main uses for CNS drugs in veterinary medicine?
1. tranquilizers (neuroleptics, anxiolytics, sedatives)
2. anticonvulsants
3. stimulants (antidepressants)
4. opoid analgesics
5. general anesthetics
what are the three most commonly used types of therapeutic GABA agonists?
1. benzodiazepine tranquilizers (diazepam)
2. barbiturates (thiopental, pentobarbital, phenobarbital)
3. general anesthetics
why can GABA-A agonists have an additive or synergistic effect?
because they bind to different sites on the GABA-A receptor
benzodiazepines:
- mechanism of action
- example of a generic
- uses
- analgesic potency
- advantages with respect to drug interaction
- legal classifications
- GABA agonist
- diazepam (Valium)
- tranquilizer, chemical restraint, anxiolytic, muscle relaxer, anticonvulsant
- no analgesic effect
- does not affect respiration, so it is good to use with general anesthesia
- C-IV drug
barbiturates:
- mechanism of action
- examples of generics
- uses
- legal classifications
- mechanism of action: GABA agonist
- examples of generics: thiopental, pentobarbital, phenobarbital
- uses: CNS, respiratory, and CV depressants; sedative-hypnotic-anesthetics; euthanasia; anticonvulsant
- legal classifications: C-II - C-IV
of the three common barbiturates that we are learning about, classify them according to duration of action and their most common usage in veterinary medicine
- thiopental: short-acting; anesthesia
- pentobarbital: medium-acting; euthanasia
- phenobarbital: long-acting; anticonvulsant
what are the two main mechanisms of action for the lipophilic general anesthetics such as isoflurane, propofol, etc?
- GABA agonists
- solvent-like to cell membranes, so they slow down cell function as a result
for barbiturates, what are the four dose-dependent effects?
sedation --> hypnosis --> anesthesia --> euthanasia
what are the major pharmacokinetic differences between inhalant and IV general anesthetics?
- inhalant: quick onset, quick recovery
- IV: needs to redistribute (longer onset), longer recovery
comment on antagonists/antidotes for the main classes of GABA agonists.
only the benzodiazepines have an antagoinst; barbiturates and general anesthetics do not
what are the three major uses for CNS GABA receptor inhibitors. Why aren't they more commonly used?
1. benzodiazepine antidote
2. pesticides
3. analeptics

- they tend to cause convulsions, which is kind of a bad thing for a drug to do
comment on the usage of CNS glutamate agonists
they are not used because they cause convulsions, which have a tendency to make pet owners upset
give an example of a CNS glutamate antagonist and its uses and side-effects
ketamine
- dissociative anesthetic: catalepsy (immobilization), no respiratory side-effects
- causes amnesia and hallucinations (but animal can't move to "freak out" at hallucinations)
- restricted C-III due to it being a date rape drug
name the main CNS opioid receptors and the receptor function
- Mu (μ) - supraspinal analgesia, respiratory depression, euphoria, physical dependence
- Kappa (κ) - receptor for spinal analgesia, sedation, and miosis
Delta (δ) - unknown
what binding must an opioid have to be considered a full agonist? partial agonist?
full agonist: both μ and κ
partial agonist: either μ or κ
what are two examples of drugs that are full CNS opioid agonists?
morphine, codeine
what is an example of a drug that is a partial CNS opioid agonist?
butorphanol
what are six therapeutic uses for CNS opioid agonists? side-effects?
1. antitussive (codeine)
2. analgesic (morphine, codeine)
3. antidotes for opioid poisoning
4. emetics
5. sedation
6. miosis
side effects: respiratory depression, GI effects, euphoria, psychological dependence
what are the three main monoamine neurotransmitters in the CNS?
norepinephrine, dopamine, serotinin (5-hydroxytryptophan)
in general what three things are CNS MAO inhibitors used for in dogs?
1. behavioral modification (e.g. separation anxiety)
2. cognitive dysfunction
3. hyperadrenocorticism
what are the three types of drugs used in the CNS that have non-specific actions on monoamines?
1. MAO inhibitors
2. monoamine reuptake inhibitors
3. COMT inhibitors
what type of drug is a tricyclic antidepressant? what are they indicated for in dogs? what are side-effects? In general, why do some of these side-effects occur?
- monoamine reuptake inhibitor (SSRI)
- used for separation anxiety
- side-effects include confusion, twitching, and others
- side-effects occur because of non-specific action at other CNS sites and also SSRIs have cholinergic side-effects
in the CNS, what effects are seen with
- α1 agonists?
- α2 agonists?
- α1 agonists: stimulatory effects
- α2 agonists: tranquilizing; analgesic
what are MAO-B selective inhibitors indicated for in humans? dogs?
humans: Parkinson's
dogs: cognitive dysfunction; hyperadrenocorticism
which of the main CNS monoamine neurotransmitters will be affected by COMT inhibitors? unaffected? what are these drugs indicated for in humans?
- affected: norepinephrine and dopamine (they are catacholamines)
- not affected: serotonin (not a catechol)
- indicated for treatment of Parkinson's
what is an example of a CNS specific adrenergic drug? What are its pharmacodynamics? What are its indications?
- xylazine
- agonist of pre-synaptic α2 receptors, which blocks the release of norepinephrine
- indications: tranquilizer, analgesia, emesis
what type of drug would be used as a xylazine antidote
α2 blocker (e.g. yohimbine) because xylazine is an α2 agonist
why would IV dopamine administration have very limited affect in the CNS?
because it is too polar to cross the BBB
what are two effects of agonists of the CNS dopamine receptors D1 and D2?
1. emesis
2. lower prolactin secretion
what are two examples of dopamine antagonists that have antiemetic properties? which have tranquilizing effects and which do not?
1. acepromazine - tranquilizer
2. metoclopramide - not a tranquilizer
phenothiazine tranquilizers are what type of drug? what is an example of one? besides tranquilizing, what other effects does this drug have? side-effects?
- dopamine receptor blocker
- acepromazine
- antiemetic, antihistamine, antiadrenergic, anticholinergic
- seizures, Parkinson-like disorders with long-term use
why are *specific* CNS serotonin drugs necessary to be useful pharmaceuticals?
because there are so many receptors with widespread action that many side-effects would occur
what is the most common use for a specific serotonin agonist?
antianxiety
what are the three common serotinergic drug types?
1. direct agonists
2. specific serotonin reuptake inhibitors (SSRI)
3. 5-HT receptor blockers
in what ways are specific serotonin reuptake inhibitors (SSRIs) used in veterinary medicine?
- behavior modification (e.g. "OCD" like behaviors in dogs, like tail chasing)
- anxiety
- dominance aggression
for what purpose, in veterinary medicine, are 5-hydroxytryptamine (serotonin) receptor blockers used?
- antiemetics, especially for chemotherapy patients
what effect does acetylcholine stimulation of nicotinic and muscarinic receptors in the brain have?
stimulatory effects
why does neostigmine not commonly cause CNS side-effects?
because it does not cross the BBB
what are two uses of agents that block cholinergic receptors in the CNS?
1. antiemetics (acepromazine)
2. anesthetics
what is the second messenger at β receptors? what drug is a target for this system? how does it work? what are its uses
- cAMP is the second messenger
- theophylline (a methylxanthate)
- it is a phosphodiseterase inhibitor, which inhibits the breakdown of cAMP, thus having β sympathomimetic effects
- used as a stimulant and smooth muscle relaxor (e.g. bronchi)
acepromazine
- receptor action
- nervous system branches
- uses in veterinary medicine
- type of chemical
- side-effects
acepromazine
- receptor action: dopamine antagonist; antimuscarinic
- nervous system branches: PNS
- uses: tranquilizer, antiemetic
- type of chemical: synthetic
- side-effects: seizures, tremors (long-term use)
acetylcholine
- receptor action
- nervous system branches
- uses in veterinary medicine
- type of chemical
- side-effects
acetylcholine
- receptor action: nicotinic agonist; muscarininic agonist
- nervous system branches: SNS, PNS, neuro-muscular junction, CNS
- uses: stimulation of SNS, PNS, CNS, NMJ
- type of chemical: amino-ester
- side-effects: lots of them
atropine
- receptor action
- nervous system branches
- uses in veterinary medicine
- type of chemical
- side-effects
atropine
- receptor action: antimuscarinic
- nervous system branches: PNS
- uses: increase HR and cardiac output, preanesthetic antisecretory
- type of chemical: alkaloid (Belladonna)
- side-effects: dry mouth, dry eye
butorphanol
- receptor action
- nervous system branches
- uses in veterinary medicine
- type of chemical
- side-effects
butorphanol
- receptor action: partial opioid agonist
- nervous system branches: CNS
- uses: emetic
- type of chemical: opioid
- side-effects: puking
codeine
- receptor action
- nervous system branches
- uses in veterinary medicine
- type of chemical
- side-effects
codeine
- receptor action: full opoid agonist
- nervous system branches: CNS
- uses: analgesic, antitussive
- type of chemical: opioid
- side-effects: respiratory depression, emesis, GI stasis
diazepam
- receptor action
- nervous system branches
- uses in veterinary medicine
- type of chemical
- side-effects
diazepam
- receptor action: GABA agonist
- nervous system branches: CNS
- uses: tranquilizer, anxiolytic, anticonvulsant, muscle relaxor
- type of chemical: benzodiazepine
- side-effects: generally safe
epinephrine
- receptor action
- nervous system branches
- uses in veterinary medicine
- type of chemical
- side-effects
epinephrine
- receptor action: α agonist; β agonist
- nervous system branches: SNS
- uses: increase HR and BP, bronchodilate
- type of chemical: catecholamine
- side-effects: tachycardia, arrhythmia, hypertension
isoproterenol
- receptor action
- nervous system branches
- uses in veterinary medicine
- type of chemical
- side-effects
isoproterenol
- receptor action: β agonist
- nervous system branches: SNS
- uses: increase HR, bronchodilate
- type of chemical: monoamine
- side-effects: tachycardia, arrhythmia
ketamine
- receptor action
- nervous system branches
- uses in veterinary medicine
- type of chemical
- side-effects
ketamine
- receptor action: glutamate receptor inhibitor
- nervous system branches: CNS
- uses: anesthetic, chemical restraint (catalepsy)
- type of chemical: dissociative anesthetic
- side-effects: amnesia, hallucinations
metoclopramide
- receptor action
- nervous system branches
- uses in veterinary medicine
- type of chemical
- side-effects
metoclopramide
- receptor action: dopamine antagonist; muscarinic agonist
- nervous system branches: PNS
- uses: antiemetic; GI stimulant (increases AcCh in GI tract)
- type of chemical: synthetic
- side-effects: none noted
morphine
- receptor action
- nervous system branches
- uses in veterinary medicine
- type of chemical
- side-effects
morphine
- receptor action: full opoid agonist
- nervous system branches: CNS
- uses: analgesic, antitussive
- type of chemical: opioid
- side-effects: respiratory depression, emesis, GI stasis
neostigmine
- receptor action
- nervous system branches
- uses in veterinary medicine
- type of chemical
- side-effects
neostigmine
- receptor action: cholinesterase inhibitor; non-depolarizing neuromuscular blocking agent
- nervous system branches: SNS, PNS, neuro-muscular junction
- uses: antidote to NM blockers
- type of chemical: carbamate neonicotinoid
- side-effects:overdose can lead to respiratory muscles being paralyzed (without loss of consciousness)
norepinephrine
- receptor action
- nervous system branches
- uses in veterinary medicine
- type of chemical
- side-effects
norepinephrine
- receptor action: α agonist
- nervous system branches: SNS
- uses: increase BP
- type of chemical: catecholamine
- side-effects: hypertension
pentobarbital
- receptor action
- nervous system branches
- uses in veterinary medicine
- type of chemical
- side-effects
pentobarbital
- receptor action: GABA agonist
- nervous system branches: CNS
- uses: euthanasia
- type of chemical: barbiturate
- side-effects: euthanasia
phenobarbital
- receptor action
- nervous system branches
- uses in veterinary medicine
- type of chemical
- side-effects
phenobarbital
- receptor action: GABA agonist
- nervous system branches: CNS
- uses: anticonvulsant
- type of chemical: barbiturate
- side-effects: sedation, respiratory depression, euthanasia
pilocarpine
- receptor action
- nervous system branches
- uses in veterinary medicine
- type of chemical
- side-effects
pilocarpine
- receptor action: muscarinic agonist
- nervous system branches: PNS
- uses: topical treatment of glaucoma
- type of chemical: alkaloid (plant)
- side-effects: none noted
propanolol
- receptor action
- nervous system branches
- uses in veterinary medicine
- type of chemical
- side-effects
propanolol
- receptor action: β blocker
- nervous system branches: SNS
- uses: slow heart rate, antiarrhythmetic, antihypertensive
- type of chemical: monoamine
- side-effects: heart failure (especially with sudden withdrawal or parenteral administration), bronchoconstriction
theophylline
- receptor action
- nervous system branches
- uses in veterinary medicine
- type of chemical
- side-effects
theophylline
- receptor action: phosphodiesterase inhibitor
- nervous system branches: SNS
- uses: increase BP, increase HR, bronchodilator, muscle relaxor (β2 action)
- type of chemical: methylxanthine
- side-effects: from OD
thiopental
- receptor action
- nervous system branches
- uses in veterinary medicine
- type of chemical
- side-effects
thiopental
- receptor action: GABA agonist
- nervous system branches: CNS
- uses: general anesthesia
- type of chemical: barbiturate
- side-effects: respiratory depression, euthanasia
xylazine
- receptor action
- nervous system branches
- uses in veterinary medicine
- type of chemical
- side-effects
xylazine
- receptor action: α2 agonist
- nervous system branches: SNS, CNS
- uses: tranquilizer, decrease BP, analgesic, antiarrhythmetic
- type of chemical: synthetic
- side-effects: emesis