Pharmacology Quiz: Drug Actions and Bioavailability
56 Questions
0 Views

Choose a study mode

Play Quiz
Study Flashcards
Spaced Repetition
Chat to Lesson

Podcast

Play an AI-generated podcast conversation about this lesson

Questions and Answers

What are the principal pharmacokinetic factors, and how do they determine bioavailability?

The principal pharmacokinetic factors—absorption, distribution, metabolism, and excretion—determine bioavailability by influencing the concentration of a drug at its site of action.

Compare and contrast different drug administration methods, their advantages, and disadvantages.

Drug administration methods vary in terms of onset, bioavailability, and convenience, each having specific advantages and disadvantages.

Explain how lipid solubility and ionization affect drug absorption.

Increased lipid solubility enhances drug absorption, while ionization typically decreases it, as ionized drugs are less able to cross lipid membranes.

What is the blood-brain barrier, and why is it important for psychopharmacology?

<p>The blood-brain barrier is a protective layer of cells that restricts the passage of substances from the bloodstream into the brain, which is crucial in psychopharmacology because it affects drug access to the brain.</p> Signup and view all the answers

Define depot binding and its impact on drug action duration and intensity.

<p>Depot binding is the attachment of drugs to inactive sites in the body, reducing the amount of drug available to reach the target site, which affects drug action duration and intensity.</p> Signup and view all the answers

Explain the difference between synthetic and non-synthetic drug metabolism.

<p>Synthetic drug metabolism involves phase II reactions like glucuronidation, which conjugate drugs with polar molecules to enhance excretion, while non-synthetic (phase I) metabolism involves oxidation, reduction, or hydrolysis to modify the drug's structure.</p> Signup and view all the answers

Describe factors that influence drug metabolism and elimination.

<p>Factors influencing drug metabolism and elimination include genetics, liver function, age, and interactions with other drugs, which can alter enzyme activity.</p> Signup and view all the answers

Define agonist, antagonist, partial agonist, and inverse agonist in drug-receptor interactions.

<p>An <em>agonist</em> activates a receptor; an <em>antagonist</em> blocks a receptor; a <em>partial agonist</em> partially activates a receptor; and an <em>inverse agonist</em> causes an opposite effect to that of an agonist.</p> Signup and view all the answers

What is a dose-response curve? How does it illustrate ED50 and maximum response?

<p>A dose-response curve shows the relationship between drug dose and effect, illustrating ED50 as the dose producing 50% of the maximum effect and the maximum response as the greatest effect a drug can achieve.</p> Signup and view all the answers

How do potency and efficacy differ, and how are they shown graphically?

<p><strong>Potency</strong> refers to the amount of drug needed to produce an effect, while <strong>efficacy</strong> is the maximum effect a drug can produce. Graphically, potency is shown by the position of the curve along the x-axis, and efficacy is shown by the height of the curve.</p> Signup and view all the answers

Explain the importance of the therapeutic index in drug safety evaluation.

<p>The therapeutic index (TI) is crucial in drug safety evaluation, as it indicates the margin between effective and toxic doses, helping to determine the safety of a drug.</p> Signup and view all the answers

How does a competitive antagonist affect drug potency and efficacy?

<p>A competitive antagonist reduces drug potency by requiring a higher concentration of the agonist to achieve the same effect, but it does not affect the drug's efficacy if sufficient agonist is present.</p> Signup and view all the answers

Define drug tolerance and describe the three major types of tolerance.

<p>Drug tolerance is the decreased response to a drug after repeated use. The three major types are metabolic tolerance, pharmacodynamic tolerance, and behavioral tolerance.</p> Signup and view all the answers

What is pharmacogenetics, and how does it contribute to personalized medicine? Provide an example.

<p>Pharmacogenetics is the study of how genes affect a person's response to drugs, contributing to personalized medicine by predicting drug efficacy and adverse effects. An example is testing for CYP2C19 variants before prescribing clopidogrel.</p> Signup and view all the answers

Describe the structure of a typical axodendritic synapse, including both presynaptic and postsynaptic elements. How do axosomatic synapses, axoaxonic synapses, and neuromuscular junctions differ from axodendritic synapses?

<p>A typical axodendritic synapse includes a presynaptic axon terminal and a postsynaptic dendrite. Axosomatic synapses connect axons to cell bodies, axoaxonic synapses connect axons to other axons, and neuromuscular junctions connect motor neurons to muscle fibers, differing structurally and functionally from axodendritic synapses.</p> Signup and view all the answers

List the criteria required for a substance to be verified as a neurotransmitter. Of these, which might be considered most important?

<p>Criteria for a neurotransmitter include synthesis within the neuron, release upon stimulation, receptor binding, and a mechanism for inactivation. Release upon stimulation is arguably the most important.</p> Signup and view all the answers

Neurotransmitters can be classified based on the chemical category to which they belong. Name these categories and give at least one example of a member of each category.

<p>Neurotransmitter categories include amino acids (e.g., glutamate), monoamines (e.g., dopamine), peptides (e.g., endorphins), and others (e.g., acetylcholine).</p> Signup and view all the answers

Describe how the synthesis of neuropeptides differs from that of other types of neurotransmitters.

<p>Neuropeptides are synthesized in the cell body and transported to the nerve terminal, unlike other neurotransmitters, which are often synthesized directly in the nerve terminal.</p> Signup and view all the answers

What is the difference between a neurotransmitter and a neuromodulator? How are neuromodulators related to the concept of volume transmission?

<p>A neurotransmitter acts directly on postsynaptic receptors at a synapse, while a neuromodulator affects neuronal activity more diffusely. Neuromodulators are linked to volume transmission as they can affect multiple neurons over a wider area.</p> Signup and view all the answers

What is exocytosis and what is its role in neurotransmitter release?

<p>Exocytosis is the process by which vesicles fuse with the presynaptic membrane to release neurotransmitters into the synaptic cleft.</p> Signup and view all the answers

Describe the process of vesicle recycling and the various models that have been proposed to explain the recycling process. How do these models differ with respect to factors such as speed of recycling, area of the nerve terminal where vesicle membrane retrieval occurs, and the involvement of the protein clathrin and of endosomes?

<p>Vesicle recycling involves the retrieval of vesicle membranes from the presynaptic terminal after exocytosis. Models vary in speed, area of retrieval (directly from the membrane or via endosomes), and clathrin involvement. Some propose rapid 'kiss-and-run' recycling, while others involve endocytosis and clathrin-mediated retrieval.</p> Signup and view all the answers

Discuss the concept of a retrograde messenger and how this concept applies to lipid and gaseous transmitters.

<p>A retrograde messenger is released by the postsynaptic neuron to affect the presynaptic neuron. Lipid (e.g., endocannabinoids) and gaseous (e.g., nitric oxide) transmitters can act as retrograde messengers, influencing presynaptic neurotransmitter release.</p> Signup and view all the answers

What is the difference between somatodendritic and terminal autoreceptors? How do these receptors control the rate of neurotransmitter release?

<p>Somatodendritic autoreceptors are located on the cell body and dendrites, while terminal autoreceptors are found on the axon terminal. Both control neurotransmitter release by inhibiting further release when activated by the neurotransmitter itself.</p> Signup and view all the answers

Discuss the mechanisms by which neurotransmitters are inactivated.

<p>Neurotransmitters are inactivated by reuptake into the presynaptic neuron, enzymatic degradation in the synaptic cleft, or diffusion away from the synapse.</p> Signup and view all the answers

Which neurotransmitters make up the category called catecholamines? What are the distinguishing chemical features of this category?

<p>Catecholamines include dopamine, norepinephrine, and epinephrine. They are characterized by a catechol nucleus (benzene ring with two hydroxyl groups) and an amine group.</p> Signup and view all the answers

Describe the steps involved in the biosynthesis of dopamine and norepinephrine. Name the enzyme that catalyzes each biochemical reaction, and indicate which reaction is the rate-limiting step in catecholamine synthesis.

<p>Dopamine synthesis involves tyrosine being converted to L-DOPA by tyrosine hydroxylase (the rate-limiting step), then to dopamine by DOPA decarboxylase. Norepinephrine synthesis involves dopamine being converted to norepinephrine by dopamine beta-hydroxylase.</p> Signup and view all the answers

Discuss the factors that regulate the rate of catecholamine synthesis.

<p>The rate of catecholamine synthesis is regulated by the availability of precursor amino acids (tyrosine), the activity of tyrosine hydroxylase, and feedback inhibition from dopamine and norepinephrine.</p> Signup and view all the answers

List the names of the proteins that transport catecholamines in synaptic vesicles. Which of these proteins is expressed in the brain, and which is expressed in the adrenal medulla?

<p>The vesicular monoamine transporters (VMATs) transport catecholamines into synaptic vesicles. VMAT2 is expressed in the brain, and VMAT1 is primarily expressed in the adrenal medulla.</p> Signup and view all the answers

What is meant by single-spiking versus burst firing mode as applied to the firing patterns of midbrain dopaminergic neurons? How do these different firing patterns influence dopamine release at the nerve terminal?

<p>Single-spiking refers to regular, isolated action potentials, while burst firing involves clusters of action potentials. Burst firing results in greater dopamine release at the nerve terminal.</p> Signup and view all the answers

Describe how catecholamine release is regulated by autoreceptors, including differences in the location and mechanism of action of terminal versus somatodendritic autoreceptors. What adrenergic subtypes function as noradrenergic autoreceptors? Name an adrenergic autoreceptor agonist and an antagonist and indicate what effects these drugs have on nor-adrenergic cell firing.

<p>Catecholamine release is regulated by autoreceptors, which inhibit further release when activated. Terminal autoreceptors are on axon terminals and reduce exocytosis, while somatodendritic autoreceptors on cell bodies slow firing. Alpha-2 adrenergic receptors function as noradrenergic autoreceptors. Clonidine is an agonist (reduces cell firing), and yohimbine is an antagonist (increases cell firing).</p> Signup and view all the answers

What are the two basic mechanisms by which catecholamine transmission is terminated?

<p>Catecholamine transmission is terminated by reuptake into the presynaptic neuron via transporters and enzymatic degradation by enzymes like MAO and COMT.</p> Signup and view all the answers

Name the major metabolites of DA and NE.

<p>The major metabolites of dopamine (DA) are DOPAC and HVA. The major metabolites of norepinephrine (NE) are MHPG and VMA.</p> Signup and view all the answers

Name and discuss the clinical uses of drugs that alter either catecholamine reuptake or catecholamine metabolism.

<p>Drugs that alter catecholamine reuptake (e.g., cocaine, amphetamine) are used to treat ADHD and narcolepsy by increasing DA and NE levels. Drugs that alter catecholamine metabolism (e.g., MAO inhibitors) are used to treat depression by preventing the breakdown of DA, NE, and 5-HT.</p> Signup and view all the answers

Describe the two major dopaminergic path-ways that originate in the midbrain and project to forebrain structures. Include in your answer the derivation of each pathway's name.

<p>The two major dopaminergic pathways are the mesolimbic pathway (VTA to nucleus accumbens, involved in reward) and the mesocortical pathway (VTA to prefrontal cortex, involved in cognition). 'Meso' refers to the midbrain origin, 'limbic' to the limbic system target, and 'cortical' to the cortical target.</p> Signup and view all the answers

The midbrain dopaminergic cell groups have been shown to play important roles in motor function, motivation, and cognition. Much of this information has been obtained using either neurotoxins to damage/kill the cells or genetic engineering methods to produce a biochemical DA deficiency (i.e., DD mice). Compare and contrast these methodological approaches, and then discuss the behavioral characteristics of laboratory animals that have been generated using one or the other technique.

<p>Neurotoxins (like 6-OHDA) cause cell death, leading to rapid DA depletion, but can affect other cell types. Genetic engineering (like DD mice) creates specific DA deficiencies, allowing for chronic study but can activate compensatory mechanisms. Neurotoxin-lesioned animals show severe motor deficits; DD mice might show more subtle cognitive or motivational impairments.</p> Signup and view all the answers

Describe (a) the behavioral functions of the mesolimbic versus the mesocortical dopaminergic pathways, and (b) the involvement of different subsets of VTA dopaminergic neurons in responding to rewarding versus aversive stimuli.

<p>(a) The mesolimbic pathway is involved in reward and motivation, while the mesocortical pathway is involved in cognition and executive functions. (b) Different VTA DA neuron subsets respond to rewarding stimuli (increasing firing) or aversive stimuli (decreasing firing).</p> Signup and view all the answers

How many different subtypes of DA receptors exist? How are these subtypes grouped into families? Discuss the differences between D1 and D2 receptors with respect to signaling mechanisms and affinity for DA.

<p>There are five subtypes of dopamine (DA) receptors which are grouped into two families: D1-like (D1 and D5) and D2-like (D2, D3, and D4). D1 receptors activate adenylyl cyclase and increase cAMP, while D2 receptors inhibit adenylyl cyclase and decrease cAMP. The D1 receptors typically have a lower affinity for DA than D2 receptors.</p> Signup and view all the answers

What is behavioral supersensitivity? In the case of D2 receptor supersensitivity, how is this phenomenon produced pharmacologically, and what is the hypothesized mechanism?

<p>Behavioral supersensitivity is an enhanced response to a drug after repeated exposure or withdrawal. D2 receptor supersensitivity can be induced by chronic antagonist treatment; the proposed mechanism involves receptor upregulation or increased receptor sensitivity.</p> Signup and view all the answers

Discuss the major sources of NE in the forebrain and in the peripheral nervous system. What is the "fight-or-flight” response, and how do EPI and NE mediate this response?

<p>In the forebrain, NE is mainly from the locus coeruleus. In the peripheral nervous system, NE is from sympathetic neurons and the adrenal medulla. The &quot;fight-or-flight” response is mediated by EPI and NE, leading to increased heart rate, blood pressure, and alertness in response to stress.</p> Signup and view all the answers

Describe the adrenergic receptor subtypes and their signaling mechanisms.

<p>Adrenergic receptors are divided into alpha (α1, α2) and beta (β1, β2, β3) subtypes. α1 receptors increase intracellular calcium, α2 receptors inhibit adenylyl cyclase, and beta receptors stimulate adenylyl cyclase.</p> Signup and view all the answers

Discuss the involvement of the central noradrenergic system in arousal and cognition. Include in your answer information derived from pharmacological manipulations of this system and the role of specific adrenergic receptor subtypes.

<p>The central noradrenergic system is involved in arousal, attention, and cognition. Pharmacological activation (e.g., amphetamine) enhances alertness, while blockade (e.g., clonidine) reduces it. α2 agonists can improve attention, while β-blockers can impair cognitive functions.</p> Signup and view all the answers

What is the evidence that peripheral EPI plays a role in the consolidation of emotional memories? What are the hypothesized mechanisms underlying this effect of EPI?

<p>Peripheral EPI enhances consolidation of emotional memories; evidence comes from studies where EPI administration after a learning experience improves later recall. Mechanisms involve activation of peripheral β-adrenergic receptors, leading to increased amygdala activity via vagal nerve stimulation.</p> Signup and view all the answers

Describe the uses of specific medications that work by either stimulating or blocking peripheral adrenergic receptors.

<p>Medications stimulating peripheral adrenergic receptors (e.g., pseudoephedrine) are used as decongestants. Medications blocking peripheral adrenergic receptors (e.g., beta-blockers) are used to treat hypertension and anxiety.</p> Signup and view all the answers

List the steps involved in 5-HT synthesis, including the name of the enzyme catalyzing each step. Which is the rate-limiting step in the synthetic pathway?

<p>Serotonin (5-HT) synthesis involves tryptophan being converted to 5-hydroxytryptophan by tryptophan hydroxylase (the rate-limiting step), then to 5-HT by aromatic L-amino acid decarboxylase.</p> Signup and view all the answers

Describe the pharmacological and dietary methods used either to increase or to decrease brain 5-HT levels.

<p>Pharmacological methods to increase 5-HT include SSRIs (selective serotonin reuptake inhibitors) which prevent the reuptake of serotonin, and MAO inhibitors, which prevent serotonin breakdown. Dietary methods include increasing tryptophan intake by eating protein-rich foods, which can modestly boost serotonin synthesis but is less reliable.</p> Signup and view all the answers

Discuss the effects of increasing or decreasing brain 5-HT on mood and cognition in humans.

<p>Increasing brain 5-HT (e.g., via SSRIs) generally improves mood and reduces anxiety. Decreasing brain 5-HT can worsen mood and impair cognitive functions such as attention and memory.</p> Signup and view all the answers

Describe the processes involved in 5-HT storage, release, and inactivation. Name the drugs mentioned in the text that influence these processes, including the effect of each drug on serotonergic transmission.

<p>5-HT is stored in vesicles, released via exocytosis, and inactivated by reuptake (via SERT) or enzymatic degradation (by MAO). SSRIs block reuptake, increasing 5-HT; MAO inhibitors block degradation also increasing 5-HT; MDMA triggers release.</p> Signup and view all the answers

What is the name given to the group of cells that synthesize 5-HT in the brain? Name the two specific cell groups that are responsible for most of the serotonergic projections to the forebrain and list the major forebrain areas that receive these projections.

<p>Serotonergic neurons synthesize 5-HT in the brain. The two specific cell groups that are responsible for most of the serotonergic projections to the forebrain are the dorsal raphe nucleus and the median raphe nucleus. Major forebrain areas receiving these projections include the cortex, hippocampus, amygdala, and hypothalamus.</p> Signup and view all the answers

Discuss how the firing of dorsal raphe serotonergic neurons varies with behavioral state and in response to rewards and punishments.

<p>Dorsal raphe serotonergic neurons firing varies with behavioral state, showing increased activity during active wakefulness and decreased activity during sleep. Firing rates also tend to decrease in response to both rewards and punishments, often signaling changes in reinforcement value rather than pure reward or aversion.</p> Signup and view all the answers

List the names of all the serotonergic receptor subtypes. Which of these receptors are metabotropic, and which are ionotropic?

<p>Serotonergic receptor subtypes include 5-HT1A, 5-HT1B, 5-HT1D, 5-HT1E, 5-HT1F, 5-HT2A, 5-HT2B, 5-HT2C, 5-HT3, 5-HT4, 5-HT5A, 5-HT5B, 5-HT6, and 5-HT7. All are metabotropic (GPCRs) except 5-HT3, which is ionotropic (ligand-gated ion channel).</p> Signup and view all the answers

Describe the signaling mechanisms of the 5-HT1A and 5-HT2A receptor subtypes.

<p>5-HT1A receptors primarily signal via Gi/o proteins, inhibiting adenylyl cyclase and reducing cAMP levels, which hyperpolarizes the cells. 5-HT2A receptors signal via Gq proteins, activating phospholipase C and increasing IP3 and DAG, which depolarizes the cells.</p> Signup and view all the answers

Some of the important functions mediated by brain 5-HT have been studied using Tph2-knockout mice along with mice that have developed without central serotonergic neurons. Discuss the various ways in which these mice differ from normal mice behaviorally and physiologically.

<p>Tph2-knockout mice (lacking brain serotonin synthesis) and mice without central serotonergic neurons show increased aggression, anxiety, and altered feeding behaviors. Physiologically, they may exhibit altered sleep patterns, reduced stress responses, and changes in pain sensitivity.</p> Signup and view all the answers

Several medications used clinically to treat anxiety disorders exert their primary actions on the serotonergic system. Which serotonergic receptor subtypes have been implicated in the control of anxiety and anxiety-related behaviors? Provide relevant experimental findings to support your answer.

<p>The 5-HT1A receptor subtype has been strongly implicated in anxiety control. Drugs that act as 5-HT1A receptor agonists, like buspirone, have been shown to reduce anxiety in both animal models and clinical trials. Decreased 5-HT1A receptor binding in certain brain regions is associated with increased anxiety.</p> Signup and view all the answers

Discuss the involvement of 5-HT in pain regulation and the use of serotonergic medications to treat pain-related disorders.

<p>5-HT is involved in pain regulation through descending pathways that modulate nociceptive transmission in the spinal cord. Serotonergic medications, particularly SNRIs (serotonin-norepinephrine reuptake inhibitors), can be used to treat chronic pain conditions like fibromyalgia and neuropathic pain by enhancing the activity of these descending inhibitory pathways.</p> Signup and view all the answers

5-HT1A, 5-HT4, and 5-HT6 receptors have all been implicated in processes of learning and memory. Describe the experimental findings obtained from laboratory animals that implicate each receptor subtype, including the brain area(s) of receptor expression thought to be important for each subtype.

<p>5-HT1A receptors in the hippocampus and prefrontal cortex are involved in spatial and working memory; activation of these receptors can improve performance in memory tasks. 5-HT4 receptors in the hippocampus enhance long-term potentiation (LTP) and improve memory consolidation. 5-HT6 receptors, also in the hippocampus and cortex, modulate neurotransmitter release and synaptic plasticity, and their blockade can enhance cognitive function.</p> Signup and view all the answers

What are the sources of 5-HT in the GI tract, and why are pharmacologists interested in 5-HT from these sources? Include in your answer a discussion of the clinical relevance of gut 5-HT, including serotonergic medications that have been developed to treat GI disorders.

<p>The primary source of 5-HT in the GI tract is enterochromaffin cells. Pharmacologists are interested in gut 5-HT because it regulates gut motility, secretion, and sensation. Serotonergic medications such as 5-HT3 receptor antagonists (e.g., ondansetron) are used to treat nausea and vomiting, while 5-HT4 receptor agonists (e.g., prucalopride) are used to treat constipation.</p> Signup and view all the answers

Flashcards

Pharmacokinetics

The study of how drugs move through the body affecting their bioavailability.

Drug Administration Methods

Different ways drugs can be given to patients, each with pros and cons.

Lipid Solubility

A measure of how well a drug can pass through cell membranes, impacting absorption.

Blood-Brain Barrier

A selective barrier that protects the brain by blocking harmful substances.

Signup and view all the flashcards

Depot Binding

The process where drugs bind to inactive sites, affecting their duration and intensity.

Signup and view all the flashcards

Synthetic vs Non-Synthetic Metabolism

Synthetic drugs are made artificially, while non-synthetic are naturally occurring.

Signup and view all the flashcards

Drug Metabolism Factors

Factors like age, genetics, and health that influence how drugs are processed.

Signup and view all the flashcards

Agonist

Substances that activate receptors to produce a response.

Signup and view all the flashcards

Antagonist

Substances that block or dampen the action of agonists.

Signup and view all the flashcards

Dose-Response Curve

A graph that represents the relationship between drug dose and its pharmacological effect.

Signup and view all the flashcards

Potency vs Efficacy

Potency refers to the strength of a drug at a certain dose; efficacy is the maximum effect achievable.

Signup and view all the flashcards

Therapeutic Index

The ratio between the toxic and therapeutic doses of a drug, indicating safety.

Signup and view all the flashcards

Competitive Antagonist

Drugs that bind to the same receptor as the agonist but do not activate it, reducing agonist efficacy.

Signup and view all the flashcards

Drug Tolerance

The reduced reaction to a drug after repeated use, requiring higher doses.

Signup and view all the flashcards

Pharmacogenetics

The study of how genes affect individual responses to drugs, aiding personalized medicine.

Signup and view all the flashcards

Neurotransmitter Criteria

Conditions that a substance must fulfill to be classified as a neurotransmitter.

Signup and view all the flashcards

Neurotransmitter Categories

Groups based on chemical structure, e.g., amino acids and peptides.

Signup and view all the flashcards

Exocytosis

The process where neurotransmitters are released from vesicles into the synaptic cleft.

Signup and view all the flashcards

Vesicle Recycling

The process of reusing vesicles after neurotransmitter release.

Signup and view all the flashcards

Somatodendritic Autoreceptors

Receptors on the cell body that regulate neurotransmitter release.

Signup and view all the flashcards

Catecholamines

A group of neurotransmitters including dopamine, norepinephrine, and epinephrine.

Signup and view all the flashcards

Dopamine Synthesis Steps

The biochemical steps and enzymes required to produce dopamine.

Signup and view all the flashcards

Reuptake Mechanisms

Processes that terminate neurotransmitter action by absorbing them back into neurons.

Signup and view all the flashcards

Serotonin Synthesis Steps

The sequential reactions to produce serotonin, including rate-limiting steps.

Signup and view all the flashcards

5-HT Effect on Mood

Changes in serotonin levels influence mood and cognitions in individuals.

Signup and view all the flashcards

Cholinergic Synaptic Transmission

The process of how acetylcholine is synthesized, released, and broken down in the nervous system.

Signup and view all the flashcards

Parasympathomimetic Agents

Drugs that mimic the effects of the parasympathetic nervous system.

Signup and view all the flashcards

Study Notes

Pharmacokinetics and Bioavailability

  • Pharmacokinetic factors determine bioavailability, influencing how much drug reaches its target.
  • Principal pharmacokinetic factors affect drug absorption, distribution, metabolism, and excretion.

Drug Administration Methods

  • Different drug administration methods have varying advantages and disadvantages.
  • Comparing and contrasting various methods (e.g., oral, intravenous, inhalation) is crucial in understanding their suitability.

Drug Absorption

  • Lipid solubility and ionization affect drug absorption.
  • Lipid-soluble drugs generally absorb more readily.

Blood-Brain Barrier

  • The blood-brain barrier is crucial for psychopharmacology.
  • This barrier regulates passage of substances into the brain.

Depot Binding

  • Depot binding influences drug action duration and intensity.
  • It describes how drugs bind to tissues, altering their release rate and duration of action.

Synthetic vs. Non-Synthetic Drug Metabolism

  • Differences exist in how synthetic and non-synthetic drugs are metabolized.

Drug Metabolism and Elimination

  • Factors influence drug metabolism and elimination.
  • These factors include genetics, age, and physiological conditions.

Drug-Receptor Interactions

  • Key terms like agonist, antagonist, partial agonist, and inverse agonist define drug interactions with receptors.

Dose-Response Curves

  • Dose-response curves illustrate the relationship between drug dose and response.
  • ED50 (effective dose 50%) and maximum response are key parameters.

Potency and Efficacy

  • Potency and efficacy are distinct drug properties; their differences are explained.
  • Potency refers to the amount needed to produce an effect, while efficacy refers to the largest possible effect.

Therapeutic Index

  • The therapeutic index is essential for drug safety evaluation.
  • It describes the margin of safety for a given drug.

Competitive Antagonists

  • Competitive antagonists affect drug potency and efficacy.

Drug Tolerance

  • Drug tolerance involves the body's adaptability to a drug over time.
  • Tolerance can be categorized into different types (e.g., metabolic, cellular).

Pharmacogenetics

  • Pharmacogenetics relates to how genes affect drug response.
  • This concept is crucial for personalized medicine.

Synaptic Structure

  • Synapses, their presynaptic and postsynaptic components, and their variety are detailed.

Neurotransmitter Criteria

  • Criteria for identifying a substance as a neurotransmitter are discussed and one critical criteria is prioritized.
  • Key features needed to meet the criteria for neurotransmitters are explained.

Neurotransmitter Classification

  • Neurotransmitters are classified by their chemical makeup.
  • Examples of each category are identified.

Neuropeptide Synthesis

  • Neuropeptide synthesis differs from other neurotransmitter types.
  • This difference impacting their synthesis and function is detailed.

Neurotransmitter vs. Neuromodulator

  • The difference between neurotransmitters and neuromodulators, including their relationship to volume transmission.

Exocytosis

  • The function and role of exocytosis in neurotransmitter release are explained.

Vesicle Recycling

  • Various models of vesicle recycling are detailed, including the mechanisms involved and differences in factors like speed, area of retrieval, and the proteins involved.

Retrograde Messengers

  • The concept of retrograde messengers relates to lipid and gaseous transmitters.
  • Their function and how they are involved with retrograde signaling is detailed.

Autoreceptors

  • Somatodendritic and terminal autoreceptors control neurotransmitter release.
  • Difference in control mechanisms are provided.

Neurotransmitter Inactivation

  • The mechanisms by which neurotransmitters are inactivated are discussed.

Catecholamines

  • Catecholamines and their chemical features are detailed.
  • Synthesis, enzymes involved, rate-limiting step, and regulatory factors are explained.

Catecholamine Transport

  • Proteins that transport catecholamines in synaptic vesicles are listed.
  • The specific expression of these proteins in different brain regions (or other body parts) is detailed.

Dopaminergic Neuronal Firing

  • Single-spiking versus burst firing modes in certain dopamine neurons are described.
  • Influence on dopamine release at nerve terminals is discussed.

Catecholamine Release Regulation

  • Autroceptors regulate catecholamine release.
  • Specific differences in terminal and somatodendritic autoreceptors are elaborated.

Catecholamine Transmission Termination

  • Two basic mechanisms that terminate catecholamine transmission are outlined.

Catecholamine Metabolites

  • The major metabolic products of dopamine (DA) and norepinephrine (NE) are noted.

Clinical Uses of Catecholamines

  • The clinical usage of substances that affect catecholamine reuptake or metabolism is described.

Dopaminergic Pathways

  • Major dopaminergic pathways originating in the midbrain and projecting to forebrain structures are identified.
  • Each pathway's name and derivation are detailed.

Midbrain Dopaminergic Cell Groups Function

  • Methods for studying these cells, including neurotoxins and genetic engineering.
  • The behavioral characteristics of animal models used in research are provided.

Mesolimbic and Mesocortical Pathways

  • The roles these dopamine pathways play regarding rewards and aversive stimuli are explained.

Dopamine Receptor Subtypes

  • The number of dopamine receptor subtypes is given.
  • Differences between D1 and D2 receptors and signaling mechanisms are elaborated.

Behavioral Supersensitivity

  • Behavioral supersensitivity is examined.
  • Causes and mechanisms (specifically for D2 receptors) are detailed.

Norepinephrine (NE) in the Nervous System

  • Major sources of NE in the brain and peripheral nervous system.
  • The "fight-or-flight" response and the role of EPI and NE in mediating this response are shown.

Adrenergic Receptors

  • Adrenergic receptor subtypes and their signaling.

Noradrenergic System in Arousal and Cognition

  • The role of the noradrenergic system in arousal and cognition
  • Pharmacological manipulations and the role are elaborated.

Peripheral EPI Role

  • Evidence for peripheral epinephrine's role in emotional memory consolidation, and the hypothesized mechanisms.

Peripheral Adrenergic Receptors

  • Specific medication usage involving stimulation or blocking these receptors is described.

Serotonin Synthesis

  • Steps in the serotonin synthesis process are shown and the rate-limiting enzyme is identified.

Serotonin Levels Modification

  • Pharmacological and dietary approaches are described for increasing or decreasing brain serotonin levels.

Serotonin Effects on Mood and Cognition

  • Effects of increasing or decreasing brain serotonin on human mood and cognition are detailed.

Serotonin Storage, Release, and Inactivation

  • The process of serotonin storage, release, and inactivation is provided.
  • Drugs impacting these processes are included.

Serotonin Cell Groups

  • Major serotonin-synthesizing cell groups and their forebrain projections.

Dorsal Raphe Serotonergic Neurons

  • How firing patterns of these neurons relate to behavioral states and reactions to rewards and punishments.

Serotonin Receptor Subtypes

  • The names and types of all serotonin receptors are listed.
  • Types of receptors (metabotropic or ionotropic) are detailed.

5-HT1A and 5-HT2A Receptor Signaling

  • Mechanisms of signaling for these specific serotonin receptors are described.

5-HT Knockout Mice

  • How knock-out mice (Tph2 and those without central serotonergic neurons) are studied behaviorally and physiologically are provided.

Anxiety Treatment with Serotonergic Modifiers

  • How medications for treating anxiety disorders affect the serotonergic system.
  • Specific subtypes of serotonin receptors involved in those controls are detailed, with experimental findings provided.

5-HT in Pain Regulation

  • The role of serotonin in pain regulation and relevant serotonergic medications are shown.

5-HT Receptors for Learning and Memory

  • The roles of 5-HT1A, 5-HT4, and 5-HT6 receptors in learning and memory are elaborated.
  • Experimental findings and brain areas for these receptors are shown.

GI Tract Serotonin Sources

  • Sources of serotonin in the GI tract and the scientific interests in using it there are included.
  • The clinical relevance of the sources are elaborated, along with the development of related medications.

Acetylcholine (ACh) Synthesis and Breakdown

  • Chemical reactions and enzymes involved in synthesizing and breaking down Acetylcholine (ACh).
  • The use of these enzymes as markers for cholinergic neurons is detailed.

ACh Synthesis Regulation

  • Factors and current interventions that regulate ACh synthesis.

ACh Uptake and Storage

  • Mechanisms of ACh uptake and storage into vesicles.
  • Interference with ACh uptake by drug examples are included.

ACh Release Toxins

  • Effects of toxins that stimulate or inhibit ACh release, along with therapeutic applications.

ACh Recycling

  • Mechanisms for recycling ACh and how that impacts nervous system function.

ACh in the Peripheral Nervous System

  • Localization and functions of ACh in the peripheral nervous system.

Cholinergic Cell Groups (Brain)

  • Location of principle cholinergic cell groups in the brain.
  • The role of the BFCS and its association with cognitive function.

Nicotinic Cholinergic Receptors

  • Molecular structure and signaling mechanisms for nicotinic cholinergic receptors.

Nicotinic Receptor States

  • Different states (open, closed, desensitized) and their properties, including agonist binding and receptor channel states.

Muscarinic Cholinergic Receptors

  • Molecular structure, signaling mechanisms, and brain localization of muscarinic receptors.

M5 Muscarinic Receptors and Dopamine

  • Role of M5 muscarinic receptors in regulating dopaminergic firing and reward/reinforcement by abused drugs

Peripheral Muscarinic Receptors

  • Locations and functions of peripheral muscarinic receptors, including the "dry mouth" effect.

Pancreatic M3 Receptors, Insulin, and Antipsychotics

  • The role of pancreatic M3 muscarinic receptors in insulin secretion and regulation, along with insulin resistance issues related to some antipsychotic drugs.

Parasympathomimetics and Parasympatholytics

  • Definitions and example drugs (along with their physiological effects and medical uses) are provided.

Studying That Suits You

Use AI to generate personalized quizzes and flashcards to suit your learning preferences.

Quiz Team

Related Documents

Test #1 Neurophramacology PDF

Description

Test your knowledge on pharmacokinetics, drug administration methods, and factors influencing drug absorption and action. This quiz covers the essential principles of drug metabolism, the blood-brain barrier, and depot binding that are crucial for understanding pharmacology.

More Like This

Pharmacokinetics and Drug Absorption Quiz
5 questions
Gastrointestinal System Absorption Quiz
49 questions
Pharmacokinetics Overview Quiz
45 questions

Pharmacokinetics Overview Quiz

UndauntedHeliotrope500 avatar
UndauntedHeliotrope500
Use Quizgecko on...
Browser
Browser