Shared Flashcard Set

Details

Pharmacology Exam 2
N/A
382
Veterinary Medicine
Professional
11/07/2011

Additional Veterinary Medicine Flashcards

 


 

Cards

Term
What is the neurotransmitter in cholinergic receptors?
Definition
Acetylcholine
Term
Where are muscarinic cholinergic receptors found?
Definition
Smooth muscle
Term
How many subtypes are there of muscarinic cholinergic receptors?
Definition
5 subtypes
Term
Where are nicotinic cholinergic receptors found?
Definition

1. Skeletal muscle

2. CNS

3. Presynaptic ganglion (ANS)

Term
How many subtypes of nicotinic cholinergic receptors are there?
Definition
2 subtypes
Term
What part of the extracellular nicotinic cholinergic receptor does Ach bind to?
Definition
Ach binds to the alpha subunits
Term
What are the three parts of the neuromuscular junction?
Definition

1. Pre-synaptic membrane

2. Synaptic cleft

3. Post-synaptic membrane

Term
What increases surface area at the post-synaptic membrane?
Definition
The undulations of the post-synaptic membrane increases surface area
Term
What mineral is essential for release of Ach from the presynaptic terminal?
Definition
Calcium
Term
What causes muscle contraction?
Definition
Bound Ach at the post-synaptic terminal
Term
True/False - chemicals are not an important part of vesicle binding to the presynaptic membrane.
Definition
FALSE! There are many chemicals and all are needed to allow binding
Term
About how many vesicles are there to a presynaptic membrane?
Definition
Usually about 60 vesicles to a membrane
Term
What is the important enzyme that facilitates the reuptake of Ach?
Definition
Acetylcholinesterase
Term
What two enzymes is Ach broken down into?
Definition

1. Acetate

2. Choline

Term
What is the end plate potential?
Definition
Depolarization of muscle membrane causes contraction
Term
What is the safety factor of the end plate potential?
Definition

There are 15 to 40 million ACh receptor

There are 60 ACh vesicles

Each vesicle contains 10,000 ACh

Term
What is blocked in Nondepolarizing blockades of the endplate?
Definition
It is a postsynaptic blockade that prevents access of neurotransmitter to its receptor and thus prevents depolarization
Term
What type of things cause nondepolarizing blockades?
Definition

1. Pharmacologic drugs

2. Disease

3. Toxins

Term
What happens in depolarizing blockade of endplate?
Definition
There is sustained depolarization of muscle membrane
Term
What happens in depolarizing blockades?
Definition
There is repetitive firing of the membrane, the membrane is unable to repolarize, this causes fibrillations and weakness
Term
What can be affected in neuromuscular junction disease at the pre-synaptic membrane?
Definition
Disease can either increase or decrease presynaptic ACh synthesis, release, or reuptake
Term
What can be affected in neuromuscular junction disease at the synaptic cleft?
Definition
Disease can alter [ACh] by altering removal
Term
What can be affected in neuromuscular junction disease at the post-synaptic membrane?
Definition
Disease can affect ACh interaction w/receptor organ
Term
What will you see with weakness at the neuromuscular junction?
Definition

1. Episodic exercise intolerance

2. Weakness - stilted gait, varies in severity, usually more prominent in hind limbs vs. thoracic limbs (b/c longer pathway for neurotransmission)

3. +/- tremors

4. Normal muscle tone, flaccidity

5. Normal postural reactions

6. Normal, decrease to absent spinal reflexes

7. +/- cranial nerve signs (megaesophagus)

8. Normal sensation

Term
What is another name for Myasthenia Gravis?
Definition
Grave Muscle Disease
Term
What do you see in Myasthenia Gravis?
Definition

Muscle weakness

It is a postsynaptic disease

Term
What are the causes of Myasthenia Gravis?
Definition

Congenital

Acquired - Immune

Term
What breeds is congenital Myasthenia Gravis seen in?
Definition

1. Jack Russell terrier

2. Dachshund

3. Springer spaniel

Term
What is seen in congenital Myasthenia Gravis?
Definition
Low-density ACh Receptors
Term
What breeds is acquired Myasthenia Gravis seen in?
Definition

1. Golden retriever

2. German shepherd

3. Labrador retriever

4. Newfoundland

5. Pure bred cats

Term
What is the signalment of acquired Myasthenia Gravis?
Definition
Bimodal distribution
Term
True/False: acquired myasthenia gravis is immune caused.
Definition
TRUE
Term
What do the antibodies in immune-caused myasthenia gravis attack?
Definition
The antibodies are against the receptor - they attack the alpha subunit and bind there to prevent ACh from binding
Term
What two things occur in the immune-mediated disease of myasthenia gravis?
Definition

1. Ab-mediated ACh receptor destruction

2. Complement mediated damage to endplate

Term
What is affected in the neuromuscular junction of myasthenia gravis?
Definition
The undulation of the post-synaptic membrane is greatly decreased
Term
What are the three types of clinical signs of myasthenia gravis?
Definition

1. Focal

2. Generalized

3. Acute Fulminating

Term
What is seen with focal clinical signs?
Definition

1. Double vision/blurry vision

2. Dilated esophagus - megaesophagus

3. Cranial nerve signs

4. Pharyngeal/laryngeal weakness

 

26-43% of MG cases present this way

Term
What is seen with generalized clinical signs of MG?
Definition

1. Appendicular muscle weakness, predominately pelvic limb weakness, some involve thoracic limb

2. Megaesophagus - in 75% of cases

Term
What is seen with Acute Fulminating clinical signs of MG?
Definition

1. Patients are showing flaccid weakness

2. Can't support limbs

3. Lose ability to breathe

4. Severe appendicular muscle weakness

5. Cranial nerve involvement

6. Respiratory distress

7. Acute onset and rapid progression

 

B/c patients come in w/acute respiratory distress it can be mistaken for toxicity

Term
What four things causes morbidity in MG?
Definition

1. Skeletal muscle weakness

2. Pharyngeal muscle weakness

3. Megaesophagus

4. Aspiration pneumonia

 

Term
What three other acquired diagnoses are seen with MG?
Definition

1. Thymoma (5% in dogs, 25% in cats)

2. Paraneoplastic disease

3. Hypothyroidism

Term
What is the gold standard for diagnosis of MG?
Definition

Measure serum ACh receptor antibodies

 

Radioimmunoassay (>0.6 nmol/L in dogs; >0.3 nmol/L in cats)

Term
What is another test to diagnose MG?
Definition
The Edrophonium Challenge test
Term
What is edrophonium?
Definition
Edrophonium chloride (Tensilon) is an ultra-short-acting anticholinesterase agent.
Term
What does edrophonium do?
Definition
The drug enables more ACh molecules to be available and interact with the remaining ACh receptors.
Term
What does it mean when a patient responds positively to IV injection of 0.1 to 0.2 mg/kg of edrophonium?
Definition
An obvious improvement in muscle strength shortly administration is considered a positive response and allows you to make the presumptive diagnosis of aquired MG
Term
What receptors are affected with edrophonium?
Definition
Both muscarinic and nicotinic receptors because anticholinesterase agents are non-specific
Term
What must an animal be pre-treated with if you want to avoid the muscarinic effects associated with edrophonium?
Definition
Pre-treat with IM or SC atropine (0.02-0.04 mg/kg) and you also should have it available for immediate IV administration to counteract undesirable muscarinic side effects?
Term
What is the acronym for muscarinic effects that can be associated with edrophonium and what does it stand for?
Definition

DUMBELS

Diarrhea

Urination

Miosis

Bronchospasm

Emesis

Lacrimation

Salivation

Term
What is the first line therapy for MG treatment?
Definition

Cholinesterase inhibitors

 

Term
What are the two long-acting cholinesterase inhibitors used in MG treatment?
Definition

1. Pyridostigmine (PO)

2. Neostigmine (IM)

Term
What are two other possible MG treatments?
Definition

1. Immunomodulation

2. Thymectomy

Need to make sure that you have supportive care

 

Term
What is the terminal half-life of Pyridostigmine Bromide?
Definition
T1/2 = 8.3 hours +/- 2.1 SD
Term
True/False:  Pyridostigmine Bromide does not have high affinity for peripheral tissues.
Definition
FALSE! - does have high affinity for peripheral tissues
Term
True/False:  Pyridostigmine bromide does easily cross BBB
Definition
FALSE - does not easily cross BBB
Term
What is the dosing range for dogs with pyridostigmine bromide?
Definition
0.5 - 3.0 mg/kg PO q 12 hr, q 8 hr
Term
What is the dosing range for cats with pyridostigmine bromide?
Definition

0.25 mg/kg/day

They are sensitive to anticholinesterase agents

Term
What is the dosing range for Neostigmine?
Definition
0.04 mg/kg IM q 6 hr
Term
True/False:  You want to titrate the dose of pyridostigmine  bromide to the patient.
Definition

True - start with a low starting dose and dose to effect.  Want to see a reduction of clinical signs with the fewest unacceptable side effects.

 

Also monitor for overdose and side effects

Term
What are the cholinesterase inhibitors side effects - for both muscarinic and nicotinic receptors?
Definition

Muscarinic - Cholinergic crisis (DUMBELS)

 

Nicotinic - paradoxical muscle weakness (depolarizing neuromuscular junction blockade)

Term
What do you do to treat a myasthenic crisis or cholinergic crisis while treating MG with an anticholinesterase therapy?
Definition

Myasthenic crisis - reduce dosage

Cholinergic crisis - reduce dosage and reduce frequency

Term
What drugs do you use in immunomodulation treatment of MG?
Definition
Corticosteroids - such as prednisone
Term
What is the treatment plan for immunomodulation with MG?
Definition

You want to SLOWLY titrate to immunosuppression

1. initiated 0.25 to 0.5 mg/kg q 24 hr (SID)

2. increase to 2.0 mg/kg SID/BID for immunosuppressive effects

3. After remission slow taper every 2 to 4 weeks to alternate day therapy

Term
How can you tell remission has been achieved in immunomodulation of MG?
Definition
ACh receptor Ab titers
Term
What are the six possible side effects with immunomodulation treatment of MG?
Definition

1. Iatrogenic Cushings

2. Muscle weakness

3. Wasting

4. PU/PD

5. Polyphagia

6. Personality Changes

Term
What are four other immunosuppressants that can help to decrease the amount of prednisone needed?
Definition

1. Azathioprine

2. Mycophenolate

3. Cyclosporine

4. Plasmapharesis

Term
Why do you not want to start immunomodulation therapy dose at immunosuppressive levels?
Definition
Could send the patient into a myasthenic crisis
Term
About how many cases of MG spontaneously go into remission?
Definition
About 40% of these cases
Term
What drugs are contraindicated for MG?
Definition

Antibiotics - penicillin, aminoglycosides, fluoroquinolones, tetracyclines, quinolones, and beta-blockers

Antiarrhythmics

Phenothiazines

Term
What is the prognosis of MG?
Definition

Guarded in early stages

If there is a poor response to therapy w/in 2 weeks - 50% mortality

1 - year mortality 40%

 

Term
What can help guide the course of therapy of MG?
Definition
Periodic testing of ACh receptor antibody titers
Term
What does organophosphate toxicity cause?
Definition

Inhibits action of acetylcholinesterase (irreversible inhibition, "aging")

Depolarizing neuromuscular junction blockade

Term
What causes organophosphate toxicity?
Definition
Insecticides - organophosphate, carbamate (reversible)
Term
What are the muscarinic and nicotinic clinical signs of organophosphate toxicity?
Definition

Muscarinic signs - hypersalivation, lacrimation, urination, diarrhea, miosis, bronchoconstriction, increased GI motility, cyanosis and incontinence

Nicotinic signs - muscle weakness (usually generalized) - depolarizing NMJ blockage

Term
True/False - organophosphate toxicity leads to increased CNS sensitivity.
Definition
True - it crosses the BBB and you see hyperactivity, seizures, and anorexia
Term
How can you diagnose organophosphate toxicity?
Definition
Reduced cholinesterase activity (<50%) in blood is supportive of exposure in dogs.  It is not a prognostic test in cats b/c feline blood is composed of pseudocholinesterase that is very sensitive to inhibition by organophosphate
Term
What are the two ways to treat organophosphate toxicity?
Definition

1. Symptomatic therapy

2. Enzyme reactivators

Term
What is used in symptomatic treatment of organophosphate toxicity?
Definition

Atropine (muscarinic cholinergic blocker)

Diphenhydramine (nicotinic cholinergic blocker)

Term
What is used in enzyme reactivators treatment of organophosphate toxicity and how does it work?
Definition

2-PAM, pralidoxime chloride is used.

It acts specifically on the organophosphate-enzyme complex and freeing the enzyme from the organophosphate compound.  Should not be used in carbamate toxicity.

Term
What species of ticks lead to tick paralysis in the US?
Definition

1. Dermacentor andersoni

2. Ambylomma americanum

3. Dermacentor variabilis

Term
What species of tick leads to tick paralysis in Australia?
Definition
Ixodes holocyclus
Term
What is tick paralysis?
Definition
Blockade of ACh release from the presynaptic membrane of the NMJ.
Term
About how long does it take to develop tick paralysis?
Definition
Develops within 7 to 9 days and then have a rapid progressive flaccid paralysis
Term
What causes death in tick paralysis?
Definition
Death is by respiratory paralysis
Term
True/False:  Cranial nerve involvement in tick paralysis is common.
Definition
False - it is rare
Term
What is the diagnosis and treatment of tick paralysis?
Definition

Diagnosis - diagnosed by rapid improvement after tick removal

Treatment - removal of ticks making sure to remove the head as the toxin is secreted from the salivary gland; should also be treated with an ectoparasiticide therapy

Prognosis is good

Term
What is involved in botulism?
Definition

Causative agent - Clostridium botulinum

Botulinum toxin C and D

Term
How does one aquire botulism and when do clinical signs show up?
Definition

Toxin is absorbed from the GI tract

Clinical signs show up w/in 12 hours to 6 days

Term
What is the mechanism of botulism?
Definition
Toxin prevents the presynaptic release of ACh at NMJ and autonomic synapses
Term
What are the clinical signs of botulism?
Definition

Progressive, symmetric, generalized lower motor neuron disease.

Severity varies w/amount of toxin ingested - range from mild generalized weakness to tetraplegia w/respiratory failure.

Cranial and Spinal nerves are affected

Term
What chemical is affected in botulism that helps ACh bind to the presynaptic terminal?
Definition
Syntaxin is prevented which stops binding of ACh to the presynaptic terminal
Term
What are the cranial nerve clinical signs associated with botulism?
Definition

1. Facial nerve paralysis

2. Megaesophagus

3. Decreased gag reflex

4. Decreased jaw tone

Term
What are the autonomic clinical signs associated with botulism?
Definition

1. Heart rate variation

2. Mydriasis

3. Urinary retention

Term
What is the treatment for botulism?
Definition

Mainly supportive care - make sure to prevent aspiration pneumonia; monitor bladder

Antitoxin for Type C1 is made, but hard to obtain and is only effective if given before binding to NMJ which is not usually possible

Antibiotics are usually contraindicated

Term
What affects the synaptic cleft?
Definition
Organophosphate toxicity
Term
What two things affect the presynaptic terminal?
Definition

1. Tick paralysis

2. Botulism

Term
What affects the post-synaptic membrane?
Definition
Myasthenia gravis
Term
In the US color coding & packaging standards of ocular drugs what does it mean when the it is red?
Definition

The drug is a mydriatics/cycloplegics drug

Don't want to put in your own eyes! (wash hands after using)

Dispensed in uveitis

Term
What does green mean in ocular drug color coding & packaging standards of the US?
Definition
The drug is a miotics drug
Term
What does orange mean according to the color coding & packaging standards of US ocular drugs?
Definition
It is a CAIs drug
Term
What does it mean when a drug is yellow/blue in ocular drugs in color coding & packaging standards in the US?
Definition
The drug is a beta blockers drug
Term
What does the color grey mean in color coding & packaging standards of ocular drugs in the US?
Definition
The drug is an NSAIDs drug
Term
What does mydriatic mean?
Definition
Dilated pupil
Term
What does miosis mean?
Definition
Constricted pupil
Term
What two nerves provide parasympathetic innervation to the orbit?
Definition

1. Cranial nerve III - oculomotor nerve

2. Cranial nerve VII - facial nerve

Term
Where does the oculomotor nerve synapse?
Definition
Synapses in ciliary ganglion
Term
Where does the facial nerve synapse?
Definition
Synapses in pterygopalatine ganglion
Term
What three things occur after synapses in the ciliary ganglion?
Definition

1. Iris sphincter muscle contracts --> miosis (this opens the ciliary cleft which reduces IOP)

2. Ciliary muscle --> accomodation

3. Uveal vascular smooth muscle --> vasodilation

Term
What happens after the synapse at the pterygopalatine ganglion?
Definition
Lacrimal glands --> lacrimation
Term
What is the sympathetic innervation to the orbit?
Definition

Preganglionic fibers - T1-T3(or T4) to sympathetic trunk

Autonomic ganglion - cranial cervical ganglion

Postganglionic fibers - distributed to orbital structures via branches of CN V

Term
What are the effects of sympathetic innervation to the orbit?
Definition

Iris dilator muscle constricts --> mydriasis

Trabecular meshwork and the ciliary body epithelium help to reduce IOP

Uveal vascular smooth muscle --> vasoconstriction

Term
For cholinergic pharmacology of the eye what type of cholinergic antagonists are used?
Definition
Parasympatholytics
Term
What types of parasympatholytics are used in cholinergic antagonists of ocular pharmacology?
Definition

Direct acting - reversible, competitive muscarinic antagonists

Atropine

Tropicamide

Term
What are the two indications to use parasympatholytics as cholinergic antagonists in ocular pharmacology?
Definition

1. Treat uveitis

2. Diagnostic mydriasis

Term
What is the mechanism of action of cholinergic antagonists and what does this cause in ocular pharmacology?
Definition

Block muscarinic receptors on post-synaptic terminal of neuroeffector junction

Causes mydriasis and cycloplegia

Term
What is cycloplegia?
Definition

Paralysis of the ciliary muscle in the eye; paralysis in accomodation

Usually accompanied by dilation of the pupil

Term
What are mydriatic/cycloplegic agents used for?
Definition
For the treatment of uveitis
Term
What do mydriatic/cycloplegic agents do?
Definition

1. Eliminates ciliary spasm and decreases pain

2. Dilates pupil and prevents synechia (pupil getting stuck where we don't want it to get stuck)

3. Stabilizes blood aqueous barrier (anti-inflammatory effect)

 

Term
What are the contraindications of mydriatic/cycloplegic agents?
Definition

1. Ocular hypertension/glaucoma

2. Decreased tear production

Term
What are the cholinergic agonists?
Definition
Parasympathomimetics
Term
What are the two types of parasympathomimetics and the drugs used in each category of cholinergic agonists of ocular pharmacology?
Definition

Direct-acting parasympathomimetics - pilocarpine, carbachol, and acetylcholine

Indirect-acting parasympathomimetics - organophosphates (echothiophate), carbamates (demecarium bromide)

Term
What do direct acting parasympathomimetics do?
Definition
Agonize ACh receptor
Term
What do indirect-acting parasympathomimetics do?
Definition
Inhibit ACh-esterase
Term
What are the mechanisms/effects of cholinergic agonists on ocular pharmacology?
Definition

1. Induce miosis

2. Increase aqueous outflow

3. Stimulate lacrimation

4. Enhance uveitis

Term
What are the indications for treating with cholinergic agonists in ocular pharmacology?
Definition
Treatment for glaucoma and dry eye
Term
What are alpha and beta adrenergic receptor agonists in ocular pharmacology?
Definition

1. Epinephrine

2. Dipivefrin

Term
What are alpha adrenergic receptor agonists used in ocular pharmacology?
Definition

1. Phenylephrine (mixed alpha receptor agonist)

2. Apraclonidine (alpha-2 agonist)

3. Brimonidine (alpha-2 agonist)

Term
What are the indications/effects of adrenergic receptor agonists in ocular pharmacology?
Definition

1. Vasoconstriction (want to constrict pupil to open up the drain and reduce IOP)

2. Hemostasis

3. Reduce IOP

4. Mydriasis w/o cycloplegia (iris dilator muscle not ciliary muscle is affected)

5. Diagnosis and treatment of autonomic disorders

Term
What is Horner's syndrome?
Definition
Sympathetic denervation to the eye
Term
What are the four classic signals of sympathetic denervation to the eye?
Definition

1. Ptosis - Muller's muscle (droopy upper eyelid)

2. Miosis - Iris dilator muscle

3. Enopthalmos - Orbitalis muscle

4. Elevation of the third eyelid - Passive, due to enophthalmos

Term
True/False:  the localization of the lesion in Horner's syndrome can be in the central neuron, preganglionic neuron, or postganglionic neuron.
Definition
True - usually pre- or post-ganglionic
Term
What do we do when we create miosis?
Definition
We open up the drain
Term
Where can adrenergic receptros be found in the eye?
Definition

1. Muller's muscle (eyelid)

2. Iris dilator muscle

3. Trabecular meshwork

4. Ciliary epithelium

5. Uveal vascular smooth muscle

Term
What is denervation hypersensitivity?
Definition

In the post ganglionic neuron if something is wrong with it, what happens is that the iris is wanting epinephrine so it upregulates the number of receptors looking for epinephrine.  If you give it enough time to upregulate (think it's about 10 days) and you expose it to epinephrine or a similar drug it is going to respond quicker than the other eye.

Resolution of clinical signs if post-ganglionic lesion

Term
What it is the test for Horner's syndrome?
Definition

Denervation hypersensitivity

 

Take dilute phenylephrine (0.1 - 1%) which is an adrenergic agonist

It's diluted to the [] that has no effect on normal eye

Give one drop to each eye so that what you give is equal amounts

Check every five minutes

W/in 15-20 minutes if it is a post-ganglionic lesion - affected eye will be widely dilated and third eyelid will be down

Term
What are the adrenergic receptor antagonists used in ocular pharmacology?
Definition

Beta-blockers!

Beta-1 & beta-2 non-selective:  timolol, metipranolol, carteolol, levobunolol

Beta-1 selective:  betaxolol

Term
What are the indications/effects of adrenergic receptor antagonists in ocular pharmacology?
Definition
Decrease aqueous humor production --> reduce IOP
Term
True/False:  Fluid production w/in the eye = tears.
Definition
FALSE! - fluid production w/in the eyedoes not equal tear production which is fluid production outside the eye
Term
What do you want aqueous production to equal?
Definition
Aqueous outflow
Term
What is glaucoma?
Definition
A problem in the drain - decreased aqueous outflow or obstruction
Term
What is aqueous humor produced by?
Definition
Ciliary body
Term
Why is aqueous humor important?
Definition
Main component of intraocular pressure
Term
What are the two ways that aqueous humor leaves the eye?
Definition

1. Conventional route - iridocorneal angle

2. Unconventional route - uveoscleral outflow

Term
What is the pathway for conventional outflow of aqueous humor?
Definition

1. Ciliary body epithelium

2. Posterior chamber

3. Anterior chamber

4. Iridocorneal angle

5. Uveal trabeculae meshwork

6. Angular aqueous plexus

7. Out the Scleral venous plexus

Term
True/False:  Percentage of drainage via unconventional outflow of aqueous humor varies by species.
Definition

TRUE!

Cats - 3%

Dogs - 15%

Horses - >15%

Term
What is the pathway for unconventional outflow of aqueous humor?
Definition

1. Aqueous humor percolates thru the ciliary muscle

2. To suprachoroidal space, through sclera

3. To extraorbital tissue/fat

4. To orbital lymphatics

Term
What is one of the leading causes of blindness in patients?
Definition
Glaucoma
Term
What is the goal in treatment of glaucoma?
Definition
To decrease intraocular pressure
Term
What are the two ways we can decrease intraocular pressure in glaucoma?
Definition

1. Decrease production of aqueous humor

2. Increase outflow of aqueous humor

Term
What are the two broad categories of drugs used to treat glaucoma?
Definition

1. Autonomic agents

2. Non-autonomic agents

Term
What are the three types of autonomic agents used to treat glaucoma?
Definition

1. Cholinergic agonists

2. Adrenergic agonists

3. Adrenergic antagonists

Term
What cholinergic agonists are used to treat glaucoma?
Definition

1. Pilocarpine

2. Demecarium

3. Echothiopate

Term
What are the adrenergic agonists used in treatment of glaucoma?
Definition

1. Dipivefrin

2. Apraclonidine

3. Brimonidine

Term
What adrenergic antagonists are used in treatment of glaucoma?
Definition
Beta-blockers
Term
What are the three non-autonomic agents used to treat glaucoma?
Definition

1. Osmotic diuretics

2. Carbonic anhydrase inhibitors

3. Prostaglandins

Term
What are the osmotic diuretics used to treat glaucoma?
Definition

1. Mannitol

2. Glycerin

Term
What are the carbonic anhydrase inhibitors used to treat glaucoma?
Definition

1. Methazolamide

2. Dorzolamide

Term
What prostaglandin is used to treat glaucoma?
Definition
Iatanoprost
Term
What drugs decrease aqueous production?
Definition

1. Osmotics

2. CAIs

3. Beta-blockers

4. Adrenergic agonists

Term
What drugs increase aqueous outflow?
Definition

1. Cholinergic agonists

2. Prostaglandins

3. Adrenergic agonists

Term
How do cholinergic agonists work to treat glaucoma?
Definition

Cause miosis

Increase aqueous outflow by conventional outflow

Term
How do beta-adrenergic antagonists work to treat glaucoma?
Definition
Decrease aqueous production
Term
How do adrenergic agonists work to treat glaucoma?
Definition

Decrease aqueous production

Increase aqueous outflow by conventional outflow

Term
What are the three ways of aqueous humor production?
Definition

1. Active secretion (70%)

2. Ultrafiltration

3. Diffusion

Term
What is active secretion of aqueous humor production?
Definition
Requires energy and enzymes to move solutes against a concentration gradient
Term
What is ultrafiltration of aqueous humor production driven by?
Definition
Starling's forces
Term
What is diffusion of aqueous humor production?
Definition
Passive movement of solutes down a concentration gradient
Term
What agents will help to counteract diffusion of aqueous humor production?
Definition
Osmotic agents
Term
What agents will help to counteract ultrafiltration of aqueous humor production?
Definition

1. Adrenergic agonists - vasoconstriction

2. Osmotic agents

Term
What agents will help to counteract active secretion of aqueous humor production?
Definition

1. Alpha-2 agonists

2. Beta-blockers

3. Carbonic anhydrase inhibitors

Term
What are the two osmotic diuretic hypotensive agents used in vet med and what is their method of ingestion?
Definition

1. Mannitol - intravenous

2. Glycerin - oral

Term
True/False: Mannitol and Glycerin are hyperosmotic agents.
Definition
TRUE!!
Term
What are osmotic diuretics used for, and what is their method of action?
Definition

1. Short term use for emergency reduction of intraocular pressure

2. Causes rapid increase in serum osmolarity, dehydration of vitreous, and decreases aqueous humor production

Term
True/False - Hypotensive agents are used more and more in vet med.
Definition
FALSE! - used less and less in vet med
Term
What are some problems associated with glycerin in ocular pharmacology?
Definition

1. May induce nausea and vomiting

2. Metabolized to sugars --> hyperglycemia and glucosuria

Term
What is glycerin contraindicated for?
Definition

1. Diabetes mellitus

2. Cardiac disease - can precipitate pulmonary edema

3. Severe renal disease

4. Dehydration

Term
What are the limitations of mannitol?
Definition

1. Must be warmed to dissolve crystals

2. Filter in IV line

3. NPO for 4 hours

Term
What are the positives for mannitol use?
Definition

Not metabolized, excreted in urine

Can be administered to diabetics

Term
What are the contraindications of mannitol use in ocular pharmacology?
Definition

1. Cardiac disease

2. Severe renal disease

3. Dehydration

Term
What is another class of hypotensive agent?
Definition
Carbonic Anhydrase Inhibitors (also diuretics)
Term
What are some CAIs?
Definition

Topical - Dorzolamide

Oral - Methazolamide, Acetozolamide

Term
What is Carbonic Anhydrase and where is it made?
Definition

An enzyme that is made in non-pigmented ciliary body epithelium

Enzyme catalyzes this reaction:  CO2 + H2O --> HCO3- + H+

Aids in the formation of aqueous humor

Term
What do CAIs do?
Definition
Decrease aqueous humor production
Term
What are the CAI side effects?
Definition

Oral medication:

CA is present in other places other than eye like:

Renal PCT epithelium - diuresis, hypokalemia, metabolic acidosis, compensatory respiratory alkalosis

GI - anorexia, vomiting, diarrhea

Higher prevalence of side effects w/acetozolamide than methazolamide

 

Topical medication:

Rare

(dorzolamide)

Term
What prostaglandin is used in vet med for ocular pharmacology?
Definition

Latanaprost

A PGF2alpha analogue

Term
What is the way that latanaprost works?
Definition

Increases unconventional (uveoscleral) outflow of aqueous humor

Think there may also be an effect on conventional as well

Also causes intense miosis in the dog and cat

Term
What drug does Dr. G recommend just having in your clinic for glaucoma treatment?
Definition

Latanaprost!

Says it is essentially replacing mannitol

And now is in a generic form so it isn't as expensive

Term
When I say aqueous fluid production, what am I talking about?
Definition

TEARS!!!!!!!

not aqueous humor (very important to not mix these up)

Term
What are the three components of the precorneal tear film and where do they come from?
Definition

1. Lipid - Meibomian glands

2. Aqueous - Lacrimal glands (Gland of 3rd eyelid, Lacrimal gland)

3. Mucin - Conjunctival goblet cells

Term
What is the term for dry eye?
Definition
Keratoconjunctivitis sicca
Term
What are the two types of treatment for dry eye?
Definition

1. Lacrostimulants (trying to make more tears)

2. Lacrimomimetics (tear substitutes)

Term
What are some lacrostimulants used in ocular pharmacology?
Definition

1. Cholinergic agents

2. Immunosuppressive agents like cyclosporine A

Term
What are the three types of lacrimomimetics used in ocular pharmacology?
Definition

1. Aqueous replacement

2. Mucinomimetics

3. Lipid replacement

Term
If a dog presents with a super dry eye, and an ipsilateral super dry nostril what does it most likely have?
Definition
Neurogenic KCS
Term
What is the leading cause of dry eye in dogs?
Definition
Autoimmune - immune system attacks lacrimal gland
Term
What does pilocarpine do and how do you know when you've given too much of it?
Definition

1. Parasympathetic innervation to the lacrimal glands

Topical solution is given orally

2. See a systemic increase in parasympathetic activity - know we've given too much and have to back down a dose when the dog starts vomiting

Term
What is Cyclosporine A?
Definition

A drug given to transplant patients - noticed that it made women who were on it didn't have dry eyes

 

ONE OF THE FEW DRUGS APPROVED FOR USE IN VET MED BEFORE GOING TO PHYSICIANS

Term
What is the mechanism of action of Cyclosporine A?
Definition

Inhibits T cell activation, thereby decreasing inflammation w/in the lacrimal gland - decreases IL-2 release and IL-2 receptor expression

Stimulates tear production

Non-inhibitory to corneal healing so can use w/ulcer

Vacillates btwn commercially available and not available - usually compound it if it is available

Term
What is a new immunomodulating drug in ocular pharmacology?
Definition
Tacrolimus
Term
What are the four principles of drug therapy of CNS pharmacology?
Definition

1. Define therapeutic goals

2. Select appropriate drug

3. Monitor progress towards goal

4. Know potential adverse effects

Term
What is a seizure?
Definition

Abnormal electrical activity in the brain (on EEG) with one or more of the following:

1. Loss or deranged consciousness

2. Altered tone & movement

3. Autonomic disturbances

4. Abormal behavior (sensation)

Term
What is epilepsy?
Definition
Repeated seizures over time
Term
What is status epilepticus?
Definition
Continuous seizures
Term
What are the two ways seizures are caused?
Definition

1. Altered resting membrane potential

2. Altered balance of excitation and inhibition

Term
What are the four mechanisms of action of anticonvulsant drugs?
Definition

1. Enhance inhibition thru the GABA-chloride ion channel complex

2. Pre-synaptic mechanism

3. Topiramate

4. Decrease action potential generation thru the sodium channels

Term
What is the mechanism of action used by most traditional anticonvulsants?
Definition
Enhance inhibition thru the GABA-chloride ion channel complex
Term
What four drugs enhance inhibition thru the GABA-chloride ion channel complex?
Definition

1. Pregabalin (Lyrica)

2. Bromide

3. Benzodiazepines

4. Barbiturates

Term
What is Pregabalin (lyrica)?
Definition
GABA analog
Term
How does bromide work to enhance inhibition thru the GABA-chloride ion channel complex?
Definition

Displaces chloride and diffuses thru channel more readily.  Thus more inhibition per channel opening

Used as a potassium salt orally

Term
How doe benzodiazepines work to enhance inhibition thru GABA-chloride ion channel complex?
Definition

Affect GABA binding and channel kinetics

1. Diazepam used primarily in status epilepticus

2. Clonzepam or clorazepate occasionally used as short-term anticonvulsant

Term
How do barbiturates work to enhance inhibition thru the GABA-chloride ion channel complex?
Definition

Affect channel kinetics

1. Phenobarbital used intravenously to treat status epilepticus and used orally as maintenance anticonvulsant

2. Pentobarbital occasionally used intravenously for treating status epilepticus

3. Primidone - metabolized to phenobarbital but more hepatotoxic, only drug approved for use in epilepsy in dogs

Term
What drug(s) use the pre-synaptic mechanism of action in anticonvulsant drugs and how does it work?
Definition

1. Levetiracetam (Keppra) - newer anticonvulsant available as a generic

Very low toxicity

2. Mechanism not clear but appears to alter neurotransmission thru an effect on presynaptic vesicle binding

May decrease excitation thru excitatory AA neurotransmitters and calcium channels

Term
What is topiramate and how does it work?
Definition

1. Type of mechanism of action of anticonvulsant drugs

Short serum terminal half-life but may have longer brain levels

2. Multiple mechanisms - sodium channel inactivation, GABA prolongation, Glutamate antagonism (AMPA receptor)

Term
What drugs decrease action potential generation thru the sodium channels in CNS pharmacology and how do they work?
Definition

1. Phenytoin & carbamazepine (also valproate) - limits high frequency opening of sodium channels at axon hillock by enhancing channel inactivation, commonly used in ppl but half-life is too short in dogs

2. Zonisamide - new drug that is seeing increased use in dogs, reduces calcium currents and increases sodium channel inactivation

Term
What is the oldest antiepileptic drug?
Definition
Potassium  bromide
Term
What are some actions of Diazepam (valium)?
Definition

1. Anti-anxiety

2. Anticonvulsant

3. Muscle relaxant

4. Appetite stimulant

 

Term
What are the four pharmacologic considerations of CNS pharmacology?
Definition

1. Route of administration

2. Delivery to target tissue

3. Half-life

4. Core principles of therapy

Term
What are the differences in route of administration in CNS pharmacology?
Definition

1. Oral - easy to give, but slower to action.  Bioavailability may be a concern for some drugs

2. Intravenous - rapid action, but difficult to give, more likely to have side effects

3. Per rectum (intranasal, transdermal) - can get rapid blood levels w/o having to give IV if highly lipid soluble

Term
What is the importance of half-life in CNS pharmacology?
Definition

1. Important in maintaining therapeutic levels

2. Influences time needed to reach steady state blood levels - loading dose can achieve rapid levels, but more risk of side effect

Term
What is the goal for treatment of status epilepticus?
Definition
Stop seizures quickly b4 permanent brain damage or death
Term
What are the four problems with drug delivery to the CNS in status epilepticus treatment?
Definition

1. The BBB:  tight junctions and lack of fenestration in endothelial cells inhibit diffusion of most substances into brain.  High lipid solubility necessary to adequately cross

2. Blood supply:  brain receives a relatively high proportion of total blood flow due to its high metabolic demand

3. Intravenous bolus will lead to high blood levels initially

4. Highly lipid soluble drugs given IV will reach high brain concentrations due to the high blood levels, high blood flow and large lipid pool (the brain).  However, they cross back into the bloodstream just as quickly and will then get redistributed into the body fat.  Thus the effective half-life following an IV bolus is much shorter than the elimination half-life.  With repeated doses body fat gets saturated and equilibrium established

Term
What is the drug of choice for status epilepticus?
Definition
Diazepam
Term
What are the three reasons that diazepam is the drug of choice for status epilepticus?
Definition

1. High lipid solubility - very rapid action, but short effective half-life due to redistribution; can be given per rectum if necessary

2. Good safety margin - sedation (rare paradoxical excitation/aggression); hypotension; propylene glycol/alcohol base can cause arrhythmia

3. High efficacy

Term
What is given if diazepam doesn't work to treat status epilepticus and what are the differences btwn them?
Definition

IV phenobarbital or pentobarbital

1. shower to equilibrate across BBB

2. Narrow margin of safety; may end up having to intubate and support

3. Longer half-life

Term
What are the six therapeutic goals of long term treatment of epilepsy?
Definition

1. Reduce frequency of seizures by at least 50%

2. Reduce severity of seizures

3. Prevent cluster seizures

4. Reduce postictal behavior changes

5. Ensure client compliance

6. Minimize side effects

Term
What is the therapeutic range of drugs?
Definition
Range of blood levels of drug that usually works (efficacious) in most cases
Term
What is the loading dose (in CNS pharmacology)?
Definition
Blood level desired x volume distribution
Term
What are some problems that can be associated with metabolism in CNS drugs?
Definition

1. For some drugs, the metabolite is more active than the parent compound.

2. Some can have different metabolism in different species

3. Lipid solubility = non-polar = hepatic metabolism necessary for excretion for many drugs

4. If hepatic problems exists, need a drug that is not metabolized by the liver or toxic to it

Term
True/False: Some anticonvulsants have half-lifes too short to be useful in dogs, and some have very long half-lifes which create worries about toxicity.
Definition
TRUE
Term
What are four things that can cause anticonvulsants to lose efficacy over time?
Definition

1. Tolerance (benzodiazepines for chronic epilespy in dogs)

2. Enzyme induction - (phenobarbital)

3. Liver failure

4. Cimetidine inhibits P450 enzymes

Term
Do you want to do therapeutic drug monitoring of anticonvulsant drugs?
Definition
Yes - want to make sure that levels not too low or high
Term
What are the drugs most commonly used in CNS pharmacology?
Definition

1. Phenobarbital - Dr. O'Brien's drug of choice b/c inexpensive, effective in 70% of epileptics, side effects not too bad

2. Bromide - add on if phenobarbital is ineffective.  Can use as sole anticonvulsant esp. if hepatic disease is present

3. Diazepam used per rectum in cases which tend to have clusters of seizures.  Not used much for chronic maintainence since develop tolerance

Term
What are the four main side effects of anticonvulsants?
Definition

1. CNS effects

2. Hepatoxicity

3. Possible pancreatitis w/bromide

4. Allergic reactions

Term
What are the CNS side effects for anticonvulsants?
Definition

1. Sedation, ataxia

2. Rarelly excitability, irritability (disinhibition)

3. Usually develop tolerance to these after a few days unles levels are too high

4. Polyphagia, polydipsia (secondary polyuria)

5. Most are addictive and sudden withdrawal can precipitate seizures in normal animals

6. The side effects are usually dose dependent

Term
What area of the brain controls motivation, attention and arousal?
Definition

Reticular activating system - pons, midbrain & diencephalon

 

Oversimplification of action - projects to cortex & regulates consciousness

Term
What are the major neurotransmitters of the RAS?
Definition

Catecholamines:

Norepinephrine

Dopamine

Serotonin (5-HT)

Term
What is the ideal action of tranquiler drugs?
Definition

1. Abolish or enhance specific behaviors

2. Tranquilization w/o excess lethargy

3. Sedation w/o side effects

4. +/- analgesia

5. +/- amnesia

Term
True/False:  ideal tranquilizer drugs already exist.
Definition
FALSE! - don't exist yet
Term
What are neuroleptics and how do they act?
Definition

1. Major tranzuilizers

2. Act by blocking catecholamine receptors (antagonists)

Term
What do phenothiazines neuroleptics do and what are some examples?
Definition

1. Block multiple receptor types

2. Acepromazine, chlorpromazine, azaparone

Term
What do buterophenones neuroleptics do and what are some examples?
Definition

1. More selective for dopamine receptors

2. Droperidol, haloperidol

Term
True/False:  neuroleptics don't have analgesic properties and are often used in conjunction with opiate for minor procedures and pre-anesthetics
Definition
TRUE
Term
What are the side effects of neuroleptics?
Definition

1. Hypotension and other cardiovascular effects

2. Hypothermia

3. May lower seizure threshold.  (want to avoid this w/epileptics)

4. Extrapyrimidal signs - tremors & weakness (rare)

Term
What does stimulating the alpha-2 receptor do?
Definition
Blocks norepinephrine release
Term
True/False:  Alpha-2 adrenergic agonists are also neuroleptics.
Definition
TRUE
Term
What are some examples of alpha-2 adrenergic agonists in CNS pharmacology?
Definition

Xylazine

Dexmedetomidine

Term
What are some side effects of alpha-2 adrenergic receptors in CNS pharmacology?
Definition

1. Often induces emesis

2. In addition to tranquilizer, also has analgesic properties

3. Cardiovascular side effects

4. Is reversible by alpha-2 antagonists

Term
What are some examples of anxiolytics?
Definition

1. Benzodiazepines

2. Buspirone

Term
What is unique about buspirone?
Definition
Antianxiety w/o the muscle relaxation, anticonvulsant & sedative effects of benzodiazepines
Term
What are some problems with anxiolytics?
Definition

1. Slight risk of irritability & aggression (release from inhibition)

2. Rare acute hepatic necrosis reported in cats following diazepam

Term
What are two appetite stimulants?
Definition

1. Benzodiazepines - very potent in cats

2. Cyproheptadine - histamine and serotonin antagonist

Term
What are some caveats with psychoactive drugs?
Definition

1. Should always be used in conjunction w/behavior modification

2. Few are approved for use in animals

3. Most recommendations based on anecdotal reports; very little hard science

4. Effects could be unpredictable

5. Need to consider liability if human is injured

Term
What are antidepressants used for?
Definition
Compulsive behavior, separation anxiety, phobias, urine marking, aggression?
Term
What are two kinds of antidepressants?
Definition

1. Tricyclics

2. Fluoxetine (prozac)

Term
What do tricyclics antidepressants do?
Definition

1. Block catecholamine reuptake

2. Cardiovascular side effects

3. Clomipramine now approved for use in separation anxiety in dogs - needs to be combined w/behavior modification

Term
What do fluoxetine antidepressants do?
Definition

1. Selective serotonin re-uptake inhibitor

2. Approved for use in dogs

3. Aggression and urine marking

Term
What drug has been promoted for treating senility and how does it work?
Definition

1. L-deprenyl (selegiline)

2. Inhibits monoamine oxidase B, thus increasing catecholamines

Term
What are appetite stimulants used for and what are some problems associated with specific stimulants?
Definition

1. Used especially in cats to prevent hepatic lipidosis

2. Diazepam - worry about precipitating acute hepatic necrosis (rare)

Cyproheptadine - antihistamine and serotonin antagonist

Term
What are the five functions of the kidneys?
Definition

1. Regulation of water and inorganic-ion balance

2. Removal of metabolic waste products from the blood (urea, uric acid, and creatinine)

3. Removal of foreign chemicals from the blood and their excretion in the urine

4. Gluconeogenesis (during prolonged fasting)

5. Secretion of hormones - erythropoietin (controls erythrocyte production); renin (enzyme) (controls formation of angiotensin, which influences blood pressure and sodium balance); 1,25-Dihydroxyvitamin D3 (influences calcium balance)

 

Those in bold are affected by diuretics - number 1 is most important

Term
What cells make up the Juxtaglomerular apparatus?
Definition

1. Macula densa:  specialized cells sense Na+ load

2. Juxtaglomerular cells:  secrete renin (modulated by sympathetic nervous system)

Term
What is the major process affected by diuretics?
Definition
Reabsorption! by diffusion or mediated transport
Term
What is excreted = filtered (what things)?
Definition

Inulin or creatinine

~ GFR

Term
If a thing is excreted < filtered what is happening?
Definition
It is partially reabsorbed (conserved)
Term
If something is filtered but not excreted what is it?
Definition

Completely reabsorbed like many nutrients

Specifically glucose

Term
If something is excreted >> filtered what is it?
Definition

Filtered and secreted to the extent that all of it passes thru the kidney to the urine

Ex:  PAH ~ renal blood flow

Term
What are the four principles for reabsorption of Na+ and water?
Definition

1. Reabsorption of Na+ is by diffusion and/or mediated transport

2. Water reabsorption is by diffusion - dependent on Na+ reabsorption (in general)

3. Reabsorption of water and ions is regulated. (nutrient reabsorption is not regulated)

4. Reabsorption of Na+ is an active process occurring in all tubular segments except descending limb of loop of Henle

Term
What do diuretics do?
Definition

Increase the volume of urine (also can change composition of urine)

Predominant effect:  Na+ and H2O reabsorption

Term
What does the glomerulus do?
Definition
Formation of an ultrafiltrate of plasma (free of cells & protein)
Term
What is the primary role of the proximal tubule?
Definition
Reabsorption of filtered water & solutes
Term
What are two important functions of the proximal tubule?
Definition

1. Active Na+ transport (major site for it)

2. Major site for solute secretion (EXCEPT for K+)

Term
How is Na+ reabsorbed at the tubular lumen?
Definition

1. Carbonic acid is formed by enzyme, carbonic anhydrase

2. Bicarbonate is reabsorbed

3. H+ is secreted into tubular lumen in exchange for Na+

4. Electrogenic Na+/K+ ATPase maintains low intracellular Na+

Term
What is the function of the loop of Henle?
Definition
Reabsorbed major ions (to a lesser extent also reabsorbs water)
Term
What is important about the Loop of Henle?
Definition

Creates a hyperosmotic interstial fluid:  countercurrent multiplier system

Have a hyperosmotic medullary interstitium

Term
What occurs in the descending thin limb of the Loop of Henle?
Definition

Highly permeable to H2O (diffuses out - down the conc. gradient)

NO NA+ REABSORPTION

Increased solute conc. in lumen

Term
What occurs in the ascending thin limb of the Loop of Henle?
Definition

IMPERMEABLE TO H2O!

Na+ diffuses out - down the conc. gradient

Term
What occurs in the thick ascending limb of the Loop of Henle?
Definition

IMPERMEABLE TO H20 & UREA

Active Na+ transport (Na+K+/2Cl- symport) - reabsorbs 25% filtered Na+

Lumenal fluid - becomes dilute

Term
What happens in the late distal tubule & cortical collecting duct?
Definition

K+ secretion & hormonal regulation

Active Na+ reabsorption in exchange for K+

Aldosterone promotes Na+, K+ exchange

K+ gets into the urine and you lose K+

Term
What is unique about the cortical collecting duct?
Definition
Lumenal membrane has K+ channel - unique to this part of the renal tubule so that K+ that enters cells is secreted into tubular fluid
Term
What type of system is the Renin-Angiotensin-Aldosterone System?
Definition
Negative feedback system
Term
What do the macula densa cells sense and what does this cause?
Definition

1. Sense decreased Na+ load

2. Makes JG cells secrete renin

Term
What is the pathogenesis of increasing renin?
Definition
Increased renin --> increased Angiotensin --> increased aldosterone --> increased Na+ reabsorption --> decreased Na+ excretion
Term
What does aldosterone do?
Definition

Also controlled by K+

Increased K+ in extracellular fluid of adrenal cortex leads to

Increased aldosterone secretion

Causes:

Increased K+ excretion by kidney instead of saving Na+

Term
What is important about the medullary collecting duct?
Definition

Major site for hormonal control of Na+ and H2O reabsorption

Vasopression (ADH) is required for H2O reabsorption

Some H+ excretion - carbonic anhydrase dependent

Term
Where do osmotic diuretics work?
Definition
Proximal tubule & Descending loop of Henle
Term
What are the four characterastics that you want osmotic diuretics to have?
Definition

1. Freely filtered

2. Limited reabsorption

3. Pharmacologically inert

4. Resistant to metabolism

Term
What is the site/mechanism of action of osmotic diuretics?
Definition

1. Proximal tubule and loop of Henle:  osmotic effect - hold water in tubular lumen

2. Reduced Na+ reabsorption - reduced medullary tonicity - reduced water reabsorption along loop of Henle

Term
What are the three effects of osmotic diuretics?
Definition

1. Initial expansion of intravascular fluid volume, increased renal blood flow - sometimes a good thing, but not usually

2. Increased excretion of urinary electrolytes - isosmotic water loss due to disturbance w/countercurrent balance

3. Maintain diuretic effect during hypovolemia or shock - trauma

Term
What is the preferred osmotic diuretic?
Definition

Mannitol!!!

 

Term

How should you give mannitol and what are the indications and contraindications of mannitol?

 

Definition

1. Poor GIT absorption - give parenterally

2. Indications - cerebral edema, glaucoma, prophylaxis of acute renal failure (questionable value if given after renal damage), promote excretion of certain toxins

3. Contraindications - congestive heart failure (especially w/pulmonary edema); anuric renal failure that does not respond to a test dose

Term
What are the site/mechanism of action for carbonic anhydrase inhibitors diuretics?
Definition

Primarily proximal tubule (inhibits carbonic anhydrase - duh!)

Decrease H+ secretion, decreases Na+ and HCO3- reabsorption

Minor effects at collecting duct - more Na+ in lumenal fluid, more Na+ exchanged for K+ - increased K+ secretion

Term
What are the effects of carbonic anhydrase inhibitors?
Definition

1. Increased excretion of HCO3-, Na+, K+ and water (transient)

2. Metabolic acidosis and decreased excretion of ammonia

3. Decreased production of aqueous humor (b/c dependent on CA)

Term
What is the preferred CAI diuretic?
Definition
Methazolamide (neptazane)
Term
What are the indications of using CAI diuretics?
Definition

1. Major use if for treatment of glaucoma (to lower IOP)

2. Can be used to produce alkaline urine (transient effect)

3. Also have been used for udder edema in cattle (extralabel use)

Term
What are the contraindications for CAI diuretics?
Definition

1. Allergy to sulfonamides (these are related compounds)

2. History of renal or urinary calculi that form in alkaline urine

3. Presence of Na+ or K+ depletion

4. Presence of metabolic or respiratory acidosis

Term
What is another name for loop diuretics?
Definition
High ceiling diuretics
Term
What is the site/mechanism of action for loop diuretics?
Definition

1. Thick ascending limb of loop of Henle

2. Inhibit Na+/K+/2Cl- symport (why used so much in vet med)

3. Inhibit paracellular diffusion of Na+, K+, Mg++, Ca++ (decreased electrical gradient)

4. Minor effect - some inhibition of Na+ and Cl- reabsorption in proximal and distal tubules

 

Term
What are the characteristics of loop diuretics?
Definition

1. Highly protein bound in plasma, but actively secreted by proximal tubule

2. Profound diuresis - most effective for edematous conditions - where the name "high ceiling diuretic" comes from

Term
What are the effects of Loop diuretics?
Definition

1. Profound increase in Na+ excretion (up to 25% of filtered load) - also increase excretion of Cl-, H2O, K+, H+, Ca++, and Mg++ (most profound effects on Na+ and Ca++ excretion)

2. Mild systemic venodilator (acutely increase renal blood flow)

3. Bronchodilator in humans, horses and guinea pigs

4. Increased renin secretion (due to decreased plasma Na+)

5. Metabolic alkalosis - rarely clinically significant

Term
True/False:  Improvement of a cough with furosemide means that the animal was in heart failure with pulmonary edema.
Definition
FALSE - does not necessarily mean this - but can be in DDx
Term
What is the preferred loop diuretic and what is its pharmacokinetics?
Definition

FUROSEMIDE

Onset of action - IV = 5 min; oral = 1 hr

Peak effect:  IV = 30 min; oral = 1-2 hrs

Duration: IV ~ 2-3 hrs; oral 4-6 hrs

Oral absorption may be decreased in right heart failure

 

Term
What are the indications of loop diuretics?
Definition

1. Congestive heart failure:  ascited, pleural effusion, & pulmonary edema (i.e. non-inflammatory edema)

2. Hypercalcemia

3. Prophylaxis for epistaxis (exercise induced pulmonary hemorrhage, EIPH) in the equine

4. Udder edema (food animal withdrawal time = 48 hrs)

5. Cerebral edema (if haven't had significant blood loss; if you do have that use mannitol)

6. Only diuretic w/significant effect in patients w/impaired renal function

Term
What are the contraindications/cautions with loop diuretics?
Definition

1. Anuria/progressive renal disease

2. Hypokalemia (b/c causes K loss)

3. Allergy to sulfonamides

4. Possible ototoxicity (ear loss) w/rapid IV administration (rare)

5. Ca++ wasting in cows may precipitate milk fever

6. Beware of profound dehydration (esp. if stops drinking - duh)

7. NSAIDS attenuate natriuretic effects of furosemide (might have to increase dose)

Term
What is another name for Thiazide diuretics?
Definition
Rescue diuretics - b/c not going to be first choice and not going to be used by themselves but can cause diuretic effect
Term
What is the site/mechanism of action of thiazide diuretics?
Definition

1. Early distal tubule (BEFORE Na+/K+ exchange)

2. Inhibit Na+/Cl- symport

3. Also minor inhibition of CA in proximal tubule

Term
What are the characteristics of thiazide diuretics?
Definition

1. Secreted into proximal tubule

2. Decreased Na+ reabsorption and increased K+ secretion

(increased Na+ load in tubular fluid, promotes Na+/K+ exchange)

Term
What are the effects of thiazide diuretics?
Definition

1. Moderate diuresis - rarely used as first line diuretic

2. Increased excretion of Na+, Cl-, and K+

3. Decreased excretion of Ca++

Term
What is the preferred thiazide diuretic and its pharmacokinetics?
Definition

HYDROCHLOROTHIAZIDE (HydroDiuril)

Onset of action (oral): 2 hrs for hydrochorothiazide

Peak effect at 4 hrs

Duration ~ 12 hrs

Term
What are the indications for thiazide diuretics?
Definition

1. Not first-line for mono-therapy

2. Refractory heart failure (resistance to furosemide) - rescue w/thiazide - canine (diuretic effect on own + restores diuresis to furosemide)

3. Ascites due to liver disease

4. Diabetes insipidus (paradoxically decreases urine flow)

5. Udder edema (extra-label use)

Term
What are the contraindications/cautions with thiazide diurectics?
Definition

1. Hypokalemia

2. Diabetes mellitus (due to diabetogenic effect)

3. Use w/caution in CHF (high aldosterone levels) - may already have K+ loss

4. If using as "rescue", decrease dose of furosemide by 50% (may need K+ replacement)

Term
What are the site and two mechanisms of action of K+ Sparing diuretics?
Definition

Site:  late distal tubule and collecting duct

1. Antagonize aldosterone (spironolactone) - blocks active Na+/K+ exchange

2. Block sodium channels (triamterine) in tubular cells of the late distal tubule and collecting duct

Term
What are the characeristics of K+ sparing diuretics?
Definition
Actively secreted by the renal tubules
Term
What are the effects of K+ sparing diuretics?
Definition

1. Increased excretion of NaCl

2. Decreased excretion of K+, H+, Ca++ and Mg++

Term
What are the pharmacokinets of spironolactone, a K+ sparing diuretic?
Definition

1. Protein bound, but actively secreted by renal tubules

2. Gradual onset and offset of action:  peak effect 2-3 days after therapy is started

Term
What are the indications of spironolactone, a K+ sparing diuretic?
Definition

1. Used in small animals - mainly cats & dogs

2. Refractory heart failure - used mainly in combination w/other diuretic agents (primarily furosemdie - small additional diuretic effect & decreases K+ excretion); spironolactone + ACE inhibitor - be aware of risk of hyperkalemia (both are K+ sparing) unless furosemide is also given

3. Ascites caused by RHF, hepatic disease, or nephrotic syndrome

Term
What are the contraindications/cautions with spironolactone, a K+ sparing diuretic?
Definition

1. Hyperkalemia

2. Use caution administering w/other drugs that increase blood K+ (ACE inhibitors & NSAIDS)

3. Not advisable to combine w/ACE inhibitor, unless also giving furosemide

4. Induces hepatic microsomal enzymes (watch for drug interactions)

Term
List the properties of an ideal inhalation anesthetic.
Definition

1. Stable w/o preservatives

2. Compatible w/existing equipment

3. Non-flammable

4. Easily vaporized under ambient conditions

5. Non-irritating to airways

6. Low blood solubility to allow rapid changes of anesthetic depth

7. Very potent

8. No cardiovascular depression

9. Compatible w/catecholamines and other vasoactive drugs

10. Produces good skeletal muscle relaxation

11. Resists degradation in the body

12. Inexpensive

13. Non-toxic

Term
What is the Meyer-Overton theory of the mechanism of action of local anesthetics?
Definition
Volatile anesthetics dissolve in cell membranes and cause a distortion which decreases conductance of Na thru the cell membrane
Term
What is the protein receptor hypothesis of the mechanism of action of local anesthetics?
Definition
Volatile anesthetics interact w/an as yet unidentified receptor in the CNS
Term
What is the alterations in neurotransmitter availability theory of the mechanism of action of local anesthetics?
Definition
Volatile anesthetics could interfere w/the formation, release, or breakdown of neurotransmitters in the CNS
Term
Which organic inhalant anesthetic is not an ether?
Definition
Halothane
Term
What inorganic inhalant anesthetic is used today?
Definition
Nitrous oxide
Term
What is the difference btwn a gas and a vapor?
Definition

Gas = an agent that exists in its gaseous form at room temperature and sea level pressure

Vapor = the gaseous state of a substance that at room temperature and sea level pressure is a liquid

Term
Define saturated vapor pressure and why is the SVP of an anesthetic important?
Definition
SVP represents a maximum concentration of molecules in the vapor state that can exist for a given liquid at each temperature.  The SVP is important b/c anesthetic vaporizers are made based on SVP of the agent.
Term
Which anesthetic has a boiling point near room temperature and why is that important?
Definition

1. Desflurane

2. Important b/c a special vaporizer is needed in order to use it and it is very expensive

Term
Explain what a partition coefficient is and how it is clinically important when applied to inhalation anesthetics.
Definition

The partition coefficient is the ratio of the concentration of anesthetics in 2 phases, such as blood and gas.  It describes the affinity of an anesthetic for one solvent over another after equilibrium has been reached.

This is clinically important b/c this determines the solubility of an anesthetic

Term
When given the blood/gas partition coefficients for anesthetic agents be able to make comparisons about those agents based on the partition coefficients.
Definition
The blood/gas PC indicates the speed of anesthetic induction, recovery, and change of anesthetic depth.  The larger the blood/gas PC is, the greater the solubility of the agent.  An anesthetic agent w/a small blood/gas PC has a lower solubility, which allows more rapid changes to anesthetic depth than one w/a large blood/gas PC.
Term
Does the movement of inhalant anesthetic agents in the body occur due to concentration gradients or partial pressure gradients?
Definition
Partial pressure gradients
Term
How does increased solubility in the blood (larger blood/gas PC) affect anesthetic uptake from the alveoli?
Definition
Agent will be taken up in large amounts in the blood; b/c of their high solubility, a relatively large amount must be dissolved in blood b4 equilibrium is reached
Term
How does increased cardiac output affect anesthetic uptake from the alveoli?
Definition
Increases uptake in peripheral circulation, so there is a delay in the rise of the partial pressure of the anesthetic; blood flow to the brain is kept constant
Term
How does a decreased cardiac output affect how quickly an animal will go to sleep when being anesthetized w/an inhalant anesthetic?
Definition
With decreased CO, the partial pressure of the anesthetic in the brain increases faster (less is going out into the peripheral circulation) and the animal becomes unconscious faster
Term
Define minimum alveolar concentration (MAC).
Definition
MAC is the minimum alveolar concentration of an anesthetic that prevents gross purposeful movement in 50% of the subjects exposed to a noxious stimulus (similar to ED50); defined as the percentage of 1 atmosphere and represents an anesthetic partial pressure at the anesthetic site of action (Px = C/100 * Pbar)
Term
How can the MAC be useful clinically?
Definition
MAC is an indicator of potency (higher MAC value = less potent) of an agent and is useful in comparing different anesthetic agents against one another.  It also allows you to quantify analgesic properties of other drugs (if there is an analgesic effect, it affects the MAC).
Term
Can the MAC of an anesthetic be affected by any factors?
Definition
Yes, it is affected by changes in body temperature, age, and pregnancy.  For every 1 degree C drop in body temperature, the MAC drops 2-5%.  Increasing age and pregnancy also decrease MAC.
Term
What are the differences in the cardiovascular effects of the ether anesthetics and halothane?
Definition
They can cause dose-dependent hypotension and dysrhythmias.  All produce a dose-dependent decrease in CO due to a decrease in cardiac contractility, but halothane produces the most prominent effect.  Vasodilation also occurs, which is more prominent w/the ether anesthetics.  All inhalation anesthetics increase the automaticity of the myocardium, which may cause dysrythmias.  Halothane tends to sensitize the heart to catecholamines.
Term

Which anesthetic is the most metabolized?

1. Halothane

2. Methoxyflurane

3. Sevoflurane

4. Isoflurane

Definition
2. Methoxyflurane
Term

Which of the following anesthetics is metabolized the least?

1. Halothane

2. Methoxyflurane

3. Sevoflurane

4. Isoflurane

Definition
4. Isoflurane
Term
What is compound A?  How is it produced? Is it directly toxic?
Definition
Compound a is a vinyl ether that is produced from sevoflurane metabolism when it is exposed to the CO2 absorbents in the anesthetic machine.  It is inhaled and absorbed and undergoes metabolism in the kidney.  Compound A is not directly toxic; it is a metabolite of drug metabolism produced in the kidney, which is nephrotoxic.
Term
How do local anesthetics work?
Definition
Local anesthetics inhibit propagation (conduction) of nerve impulses by blocking sodium channels, which cause rapid depolarization by allowing rapid inward movement of Na.
Term
Briefly describe a differential blockade?
Definition
Differential blockade is when you block sensory fibers, but not motor fibers.  Sympathetic and pain fiber nerves typically block first, and the last to block are motor fibers.  However, the differential sensitivity of all nerve fibers is determined by fiber size, fiber type, nerve location, length of nerve exposed to local anesthetic, time for equilibration in tissue, and the agent used.
Term
In a basic environment are local anesthetics more ionized or non-ionized?  Why?
Definition
In a basic environment, they are more non-ionized, b/c most tertiary amines are weak bases and they have a pK above physiological pH.  More non-ionized drug means that more of the drug can interact w/the cell membrane and will have a faster onset and higher potency.
Term
What causes the local block to wear off after local anesthetic is injected near a nerve?
Definition
The drug is removed by systemic circulation.  Most local anesthetics cause vasodilation, so they tend to speed their own diffusion away from the site.
Term
Why are epinephrines sometimes added to lidocaine?
Definition
To produce vasoconstriction to prolong the local anesthetic's effect.
Term
What topically applied local anesthetic can cause hemoglobin damage, especially in cats?
Definition
Topical benzocaines
Term
What are the generic names of the 3 most commonly used local anesthetics in veterinary medicine?  Which is the longest acting and which is the shortest acting?
Definition

1. Lidocaine

2. Bupivicaine - longest

3. Procaine - shortest

Term

Which of the following is most cardiotoxic?

1. Lidocaine

2. Bupivicaine

3. Procaine

Definition
2. Bupivicaine
Term
Briefly describe the differences btwn local infiltration of local anesthetic and a peripheral nerve block.
Definition
Local infiltration is when you are blocking several nerve fibers in an area by injecting the local anesthetic into a general area.  Usually the injection is made proximal to the area you want to block, and it requires less skill than doing a peripheral block.  Peripheral nerve block is when you are blocking a specific nerve (e.g. paralumbar block, dental block, etc.)
Term
2% lidocaine contains how many milligrams/milliliter?
Definition
20 mg/ml
Term
List the reasons indicated for the use of NMB anesthetic drugs.
Definition
Most common use in vet med is intraocular surgeries (for cyclopegia), but can also be used for thoracotomy, obese animals, improve surgical access, edoscopy/intubation, and balanced anesthesia
Term
Why is succinylcholine infrequently used as a NMB in veterinary medicine?
Definition
Succinylcholine is a depolarizing NMJ blocker, which cannot be reversed like non-depolarizing agents can.  Also, it has not analgesic properties and many side effects.
Term
What drugs enhance the effects of NMB?
Definition
MANY: inhalation anesthetics, aminoglycosides, diuretics, polymixins, tetracyclines, lincomycin, clindamycin, lithium, local anesthetics, barbiturates, propranolol, magnesium, calcium antagonists
Term
How is neuromuscular blockade monitored?
Definition
Blockade is monitored by setting up a peripheral nerve stimulator, which will stimulate motor nerves to assess the strength of contraction.  The tibial nerve, ulnar nerve, or facial nerve can be used for monitoring.  The twitch height is measured after stimulation (various modes of stimulation can be used; train of four and double burst are most common) and a fade in height/strength of twitch overtime corresponds to a good NMJ block.
Term
List the drugs used to reverse NMB.  What is their mechanism of action?
Definition

Acetylcholinesterase inhibitors are used including edrophonium, neostigmine, pyridostigmine.

They act by inhibiting the enzyme that breaks down ACh in nicotinic ACh receptor synapses, so that ACh stays around in the synapse longer.  They are usually administered along w/atropine or glycopyrollate to prevent bradycardia, lacrimation, etc. b/c of activation of muscarinic receptors.

4-Aminopyridine can also be used as a reversal, which acts by increasing the release of ACh at the NMJ

Term
What is GABA?
Definition
GABA is the major inhibitory neurotransmitter in the brain.  Most anesthetic drugs act at GABA receptors to enhance inhibitory effects and hyperpolarization.
Term
What is the chemical difference btwn thiopental and pentobarbital?
Definition
Thiopental has a sulfar atom connected to C2 of the barbituric acid moiety, and pentobarbital has an oxygen atom connected to C2 of the barbituric acid moiety.  Otherwise the structure is identical
Term
How do thiopental and pentobarbital differ, with respect to clinical use?
Definition
Thiopental is ultra short-acting, and pentobarbital is short-acting.  Thiopental is good for induction of anesthesia b/c it works very quickly, but pentobarbital is mainly used for anesthesia nowadays.  There are many side effects w/barbiturates, so other safer classes of drugs have largely replaced their use in anesthesia.
Term
Why does a patient wake up after a single injection of thiopental?  Why is recovery prolonged after multiple injections?
Definition
Thiopental is rapidly redistributed away from the central compartment (brain) to the muscle, which allows for rapid recovery.  However, thiopental tends to accumulate in the body, which prolongs the recovery.  This is because the redistribution sites become saturated, so the body must rely on metabolism to break down the excess, which happens very slowly.
Term
List the three drugs that have been used with guaifenesin to induce anesthesia in the horse.
Definition

1. Xylazine

2. Ketamine

3. Thiopental

Term
What are the major side effects associated w/Althesin administration?
Definition
It is associated w/allergic type reactions (decreased blood pressure, urticaria/erythema) due to its tendency to release histamine from mast cells.
Term
Name a major advantage of Althesin in cats.
Definition
It has a high therapeutic index, little cumulative effect, and rapid induction and recovery w/muscle relaxation.  Cats recover much better w/this than ketamine, so it may replace ketamine use in cats.
Term
How does ketamine affect cardiopulmonary parameters, intracranial pressure, and cerebral metabolic rate?  How does this compare to most other injectable agents?
Definition
Ketamine actually acts as an indirect cardiac stimulant, increasing heart rate, blood pressure, cardiac output, myocardial oxygen consumption, and decreases vascular resistance.  It can increase intracranial pressure and increases cerebral metabolic rate.  These are unique features, as most other injectable anesthetics depress the CV and don't affect cerebral blood flow
Term
What drugs are included in the drug combination marketed as Telezol?
Definition
Tiletamine and Zolazepam
Term
Etomide proveds for anesthetic induction w/minimal cardiopulmonary changes.  What are three DISADVANTAGES to etomidate use?
Definition

1. Inhibits steroidogenesis --> rise in cortisol blunted; stress response can be beneficial in helping animal heal

2. Can cause myoclonus, vocalizations, and pain on injection

3. Contains propylene glycol, so can cause hemolysis

Term
List the similarities btwn propofool and thiopental.
Definition
Minimal analgesia, sedative/inductive effects, enhance dysrhthmogenic effects of epinephrine (though thiopental does more)
Term
List the differences btwn propofol and thiopental.
Definition

Propofol is a phenol, thiopental is a Schedule III drug

Beware of stuff growing in propofol

Propfol metabolizes faster and does not have cumulative effects like thiopental can

Term
Why isn't urethane used in clinical veterinary medicine?
Definition
It is carcinogenic; it is long-acting, and it is only used in non-survival lab animal surgeries
Term
Which of the following is most likely following chronic opioid use:  Constipation/Diarrhea?
Definition
Constipation
Term
Describe the effects of opioids on respiratory function.  How do the effects of fentanyl differ from those of butorphanol?
Definition
Opioids depress respiratory function, due to a secondary increase in intracranial pressure and hypercapnia.  Butorphanol is only a partial agonist, so there is less respiratory depression w/it than fentanyl (a full agonist)
Term
Name the opioid receptors and the endogenous ligand for each.
Definition

Mu1/Mu2 = morphine, endorphins, endomorphins

Kappa = dynorphins, ketocyclazine

Delta = enkephalins

Sigma = SKF-10, 047

N/OFQ = buprenorphine

Term
What is an agonist (using an anesthetic drug as an example)?
Definition

Fentanyl is an example

Act at the receptor to have an effect

Determines magnitude of effect and potency (also depends on receptor density, efficiency, affinity and efficacy)

Term
What is a partial agonist (using an anesthetic drug as an example)?
Definition

Butorphanol is an example

Effect is less than what it would be if it was a full agonist

Has a ceiling effect (affords some safety from overdose)

Effects most at mu receptors

Term
What is an agonist-partial agonist (using an anesthetic drug as an example)?
Definition

Morphine-Butorphanol is an example

Partial agonist partially reverses effect of agonist

Term
What is an agonist-antagonist (using an anesthetic drug as an example)?
Definition

Morphine + Naloxone is an example --> decreases amount of analgesia (doesn't completely turn off agonist)

Act as antagonist at some receptors and agonist at others

Shifts curve to the right

Term
How is the extracellular signal initiated by an opioid transduced to the cytoplasm of the cell?
Definition
Via a seven transmembrane domain receptor, which can be receptor-dependent K channels or voltage-gated Ca channels; by inhibiting adenylate cyclase (decrease cAMP), phospholipase C, and MAP kinase.  This inhibits release of acetylcholine, glutamic acid, serotonin, substance P, and norepinephrine.
Term
Describe how opioids may be used in the perioperative period.
Definition

Central effects on supraspinal and spinal = can be given in CSF, alter ascending nocioceptive transmission, alter descending pain control circuits via midbrain to spinal cord dorsal horn

Sedation (dysphoria or excitement can also occur)

Vomiting = vomiting at low doses, inhibition at high doses

Increased sphincter tone

Term
What are the central nervous sytem effects of opioids?
Definition
Opioids alter ascending nocioceptive transmission, which means that the signal strength from the site of pain to the CNS decreases (spinal cord dorsal horn).  They also activate descending pain control circuits to further inhibit tranmission (midbrain to spinal cord dorsal horn)
Term
How do opioids affect body temperature?
Definition
They decrease body temperature (except maybe in cats w/full agonists - increase body temperature)
Term
Why is general anesthesia used for diagnostic or therapeutic procedures?
Definition
To facilitate humane restraint, efficiency, control convulsions, prevent recognition of pain, euthanasia, induce and maintain sedation; decrease stress (for both animal and personnel), protection of veterinary staff, allow immobilization of animal for procedure, decrease autonomic activity, optimize cardiovascular and respiratory function
Term
Name drugs that could be used for premedication of anesthesia.
Definition

Atropine

Glycopyrollate

Acepromazine

Xylazine

Diazepam

Morphine

Butorphanol

Carprofen

Meloxicam

Term
Name drugs that could be used for induction of anesthesia.
Definition

Ketamine/Xylazine

Thiopental

Propofol

Etomidate

Alphaxalone

Tiletamine/Zolazepam

Sevoflurane

Isoflurane

other Benzodiazepines

Term
Name drugs that could be used for maintainence of anesthesia.
Definition

Isoflurane

Sevoflurane

Ketamine

Tiletamine

Propofol

Etomidate

Alphaxalone

Inhalant + opioid (fentanyl)

Term
Name physiological parameters that may be monitored in anesthetized patients.
Definition

Blood pressure

Heart rate

Respiratory rate

Oxygen saturation

ECG

Temperature

Term
Name factors that may influence outcome from anesthesia.
Definition
Duration of anesthesia, types & dosages of drugs used before, during, and after anesthesia, pain management, drug interactions, species, breed, age, physical status, concurrent disease, surgical procedure, equipment available, skill of personnel
Supporting users have an ad free experience!