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

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/328

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

328 Cards in this Set

  • Front
  • Back

What are the two components of the extracellular buffer system

bicarbonate buffer system & proteins

What are the two components of the Intracellular buffer system

phosphate buffer system & proteins

Define Hydrophilic

water loving

Define Hydrophobic
water–fearing
Define Amphipathic
molecules having both phobic and philic portions (cell membranes)
Predict the movement of water across membranes with a Hypotonic extracellular fluid
Higher water to solute ratio than the cell, floods the cell. The Cell will expand and eventually rupture
Predict the movement of water across membranes with a Hypertonic extracellular fluid
lower water to solute ratio than cell, transfer H2O from cell into solution causing shrinkage
Predict the movement of water across membranes with a Isotonic extracellular fluid
same water to solute ratio as cell, no net change = neutral situation
Describe Henderson–Hasselbalch equation for acids
pH – pKA = log of [ionized & unprotonated acids] / [protonated & unionized acids]
Describe the Henderson–Hasselbalch situation where a acid’s pKa is higher than the pH
the equilibrium will force more of the substance towards the fully protonated, unionized right side of the equation. A–+H+ > AH
Describe the Henderson–Hasselbalch situation where a acid’s pKa is lower than the pH
the equilibrium will force more of the substance towards the unprotonated, ionized left side of the equation. A–+H+ > AH
Describe with the Henderson–Hasselbalch: the differences in ionization/protonation between acids and bases
pH – pKA = log of [unionized base] / [protonated & ionized acids]
Name the steps in protein expression [general]
DNA located in the nucleus are transcribed into mRNA which are transported to the Endoplasmic Reticulum where they are translated into polypeptides
Describe the important pieces of the Amino Acid Structure [general]
Amino Acid – H3N+
Carboxyl Group – COO–
alpha–Carbon – center carbon attached to the Carboxyl group
Functional Group – R–group
Explain how the properties of amino acids determine protein structure/function
Folding provides the overall shape which determines the affinity/fitting of the receptor sites
Describe the characteristics of polar proteins
typically hydrophilic, water–soluble, and require active transport across membranes
Describe the characteristics of non–polar proteins

typically hydrophobic, lipid–soluble, and utilize passive diffusion across membranes

Describe the characteristics of the Primary protein structure
Sequence of the Amino Acid chain
Describe the characteristics of the Secondary protein structure
relationship of H–Bonding between the R–groups that cause alpha–helices and beta–sheets
Describe the importance of the cysteine amino acid
the cysteine amino acid contains an SH on the R–group that creates Disulfide (covalent) bonds with other cysteine molecules that are very strong.
Describe the characteristics of the Tertiary protein structure
overall/3–D structure of a single protein
Describe the bonding associated with the Tertiary protein structure
h–bonding, ionic bonding, disulfide bonds, Hydrophobic reactions
Describe the bonding associated with the Quaternary protein structure
h–bonding, ionic bonding, disulfide bonds, Hydrophobic reactions
Describe the characteristics of the Quaternary protein structure
combination of multiple protein monomers together
Define protein: Homodimer
2+ polypeptide chains, identical order, number, & kind of A.A. Residues
Define protein: Heterodimer
2+ polypeptide chains, differ in order, number, & kind of A.A. Residues
Define term: Isoform with regards to HbF & HbA
different structures, same function, slight alteration of specificity, activity, binding efficiency. Fetal Hemoglobin has 2–alpha globins + 2 gamma–globins while the Adult hemoglobin has 2–alpha globins + 2–beta–globins
Define polymorphic
Genetic Diversity, slight variations in a population
Describe various ways [general] that a protein structure can be disrupted
alterations in folding
temperature, pH, ionic detergents, heavy metal ions, organic solvents such as alcohol
molecules that alter bonding
post–translational modifications [specific] – phosphorylation and glycosylation
Glycation [non–specific]
Proteases
Define the function of proteases
break the amino acid backbone, a part of the protein half–life regulatory system
Describe the Proteasome
protein chopping factor in the cell, maintains cellular homeostasis through removing unwanted or dangerous proteins
Describe Ubiquitination

enzymatic addition of lysine residue to proteins that are targeted for proteolysis by the proteasome

Describe the equilibrium dissociation constant Kd
Kd is a measurement of the binding Strength/Affinity between a protein and a ligand
Describe the result of a low Kd value
lower Kd values reflect a high affinity or a more tightly bond between the ligand and the protein
Describe the result of a high Kd value

Higher Kd values reflect a low affinity or a loose bond between the ligand and the protein

Define the term Saturable in regards to Protein–Ligand Data

Saturable refers to the percent of the substrate that is bound to a ligand. When 100% of the substrate is bound, any excess ligand is extraneous and ineffective

Describe Allosteric behavior

change in protein activity by binding of effector|Enzyme NOT at the active site

Describe Cooperativity

assists in reaction rates based on changes within the molecule. Binding of first O2 on Hgb increases likelihood of #2 binding etc. [unloading also cooperative]

What are the two CoEnzymes involved in Redox Reactions
FADH – flavin adenine dinucleotide
NADH2 – nicotinamide adenine dinucleotide
Define the mnemonic OIL RIG
Oxidizing is Losing e–, Reducing is Gaining e–
Define Reducing Agent

The reducing agent is the molecule that is donating/losing the electron. The reducing agent is ultimately oxidized (lost electron)

Define Oxidizing Agent

The oxidizing agent is the molecule that is accepting/gaining the electron. The oxidizing agent is ultimately reduced (gained electron)

What vitamin is NAD derived from
niacin
What vitamin is FAD derived from
riboflavin
What vitamin is TPP derived from
Thiamine (B1)
What vitamin is CoA derived from
Pantothenate (B5)
What vitamin is Tetrahydrofolate derived from
Folic Acid
What vitamin is Pyridoxal phosphate derived from
Pyridoxine (B6)
Define Michaelis–Menton’s Km
kM is the substrate concentration that is required for the reaction to occur at ½ of the maximum (Vmax) reaction rate
Define Michaelis–Menton’s Vmax

Vmax is the maximum reaction rate

What does a low Michaelis–Menton Km mean
lower Km values show a higher affinity for the substrate, requires less [s] to achieve Vmax
Describe the graph of a Lineweaver–Burk plot with regards to the changes in the graph caused by a competitive inhibitor

The Km of the graph changes which changes the slope of the line and the point where it intercepts the X–Axis while the Y–Intercept stays the same

Describe the graph of a Lineweaver–Burk plot with regards to the changes in the graph caused by a noncompetitive inhibitor
The Vmax of the graph changes which Y–intercept point but the slope of the line and the X–intercept remain relatively unchanged.
With regards to the Lineweaver–Burk plot, what does a larger or smaller X–Intercept value mean
a larger X–intercept means a higher Km and a lower affinity for the substrate while a lower X–intercept means a lower Km and a stronger affinity for the substrate
Which of the inhibitors can be overcome by increasing the concentration of substrate
The competitive inhibitors
Define Allosteric inhibitors
binds at somewhere other than the active site to alter function
Define Reversible inhibitors

bind non–covalently to the enzyme, do not change it, and are able to be separated

Define Irreversible inhibitors
bind covalently to the enzyme, change it chemically, permanently
Explain the difference between a receptor agonist and a receptor antagonist
A receptor agonist binds & fully activates the receptor while the receptor antagonist binds but does not activate the receptor
Describe the methods by which enzyme activity are regulated
non–specific inhibition of enzymes such as overall temperature and pH, any proteolytic behavior
Specific inhibition of enzymes such as intentional substrates, products, drugs etc.
Phosphorylation and dephosphorylation
post–translational modifications such as the addition of objects to the protein structure or specific folding sequences
protein–protein interactions
expression level – what is needed at that point in the cellular life and function
proteolysis – ubiquitination of unneeded or harmful proteins
define accuracy
closeness to the known standard
Define Precision

reproducibility

Define Sensitivity
the smallest amount of detectable analyte
Define Specificity | Selectivity
ability of a method to measure one analyte vs another. How well a ligand will bind to Substrate A vs Substrate B
Describe the general method and clinical uses for electrophoresis
electrophoresis is the process of separating proteins based on size and molecular weight. Clinical Significance is the ability to detect amounts of expressed proteins.
Describe the clinical uses for Enzyme Assays
The examination of expressed enzymes and proteins such as the glucose oxidase test that measures the light–scattering effect of light shone through a sample of a sample
Describe the clinical uses for Mass Spectrometry

able to tell the amount and sequence of proteins

Describe the clinical uses for Immunoassays
utilizes detection antibodies labelled with radioactive, fluorescent, or colorimetric parts that allow the examination of expressed mRNA and proteins. This is useful with cancer patients. A subset of this is the ELISA test.
Describe the general method and clinical uses for DNA sequencing
allows for the sequencing by synthesis of DNA by tagging the bases in various methods, able to explore a patient’s genome
Describe the general method and clinical uses for DNA fingerprinting
DNA fingerprinting allows for the examination by fragment length, variations in the genome due to RFLP, STR, and VNTR alterations.
Define RFLP with regards to genetic structure
Restriction fragment length polymorphism, enzymatic digestion of the genome at known points resulting in varying fragment lengths due to polymorphisms or small sequential differences in the populous
Define STR with regards to the genetic structure

Short Tandem Repeats, alterations in the genetic code 2–7 base pairs long

Define VNTR with regards to the genetic structure

Variable Number Tandem Repeats – alterations in the genetic code > 8 base pairs long

Define SNP with regards to the genetic structure

Single Nucleotide Polymorphisms – alterations in the genetic code only 1 base pair long

Describe the PCR
PCR is polymerase chain reaction and is the process of generating large amounts of DNA from a small initial sample. This allows specific regions of DNA to be examined and tested
Explain the difference between Northern, Southern, and Western Blotting
Northern – studies RNA, the North is Really Cold
Southern – studies DNA, the Dukes of Hazard were in the South
Western – studies proteins, the Pacific ocean is on the West Coast
What information can Proteomic studies provide

proteomics refer to the investigation of all the proteins expressed by the cell. This is not examining the genome itself but rather the proteins expressed by it which are susceptible to many alteration

What information can DNA microarray studies provide
DNA Microarray or DNA Chip detects specific genetic mutations and determines which mRNAs are being expressed in the patient
Define Physical Diagnosis
determine nature of disease by physical measures that include inspection, palpation, percussion and auscultation
History provides what percent of the diagnosis?
90%
What are the 4 intellectual processes involved in arriving at a diagnosis?
Pattern Recognition
Sampling the Universe
Algorithms
Hypothesis
Define using pattern recognition to arrive at a diagnosis:

If it look likes a Duck, quacks like a duck.....

Define using Sampling of the Universe to arrive at a diagnosis:
Sampling the Universe involves an exhaustive method of performing copious tests in order to slowly limit the probability of the disease process. Useful in situations where limited information is available (ER/Trauma)
Define using Algorithms to arrive at a diagnosis:
Algorithms provide a distinct, tried, tested pathway of following s/sx to a diagnosis and subsequent treatment plan
Define using hypotheses to arrive at a diagnosis:
Gather information
Assessment of the information
Form hypothesis most likely to fit assessment
Assess the probability and utility of various ways to test the hypothesis
Define Baye's Theorem
use sensitivity and specificity of disease in the patient's population to determine if a certain course of action should be taken. Probability and utility enter the process to allow the physician to decide how probable a patient is to have a disease and the utility determines whether certain procedures have worth to the patient.
Define Rapport

Seriously.... treat your patient like he was your Mom, or... rather, how you should have treated your Mom.

Professional appearance, attitude, caring, confidentiality, communication, and preservation of dignity.

Describe the aspects of history taking
history taking includes the Chief Complaint, HPI, PMH, Social History, the Review of Systems
Define HPI and the associated mnemonic
OLD CARTS: Onset, Location, Duration, Character, Associated & Aggravating factors, Relieving Factors, Temporal factors, Severity of Symptoms
Define PMH & the associated mnemonic

MIIFMASH: Medical Illness, Injuries, Immunization, Family History, Medications, Allergies, Surgeries, Hospitalization

Describe when you would include first and second degree relatives in family history

Always include first degree relatives but only include second degree relatives when situationally appropriate

Detail the mnemonic used for history taking–

history taking includes the Chief Complaint, HPI, PMH, Social History, the Review of Systems

Explain when to do a complete history
On every new patient
Explain when a focused history is appropriate

on returning patients or emergent/trauma patients

What are the cardinal principles of the physical exam

Inspection
Auscultation
Percussion
Palpation

Physical Exam: provide details regarding

Inspection
yields the most # of diagnoses, generalized, localized
Physical Exam: provide details regarding

Auscultation

is performed from the moment you walk in the door

ex: patients breathing pattern with and without the stethoscope

Physical Exam: provide details regarding

Percussion
helps to locate organs and map out sizes, tests for density in tissue & air/fluid filled cavities
What three aspects are important re: Percussion
Tympany
Resonance
Flatness
Define Tympany
air filled stomach
Define Resonance
air filled lungs
Define Flatness
solid muscle e.g. thigh
Physical Exam: provide details regarding

Palpation
Tenderness
Texture
Temperature
Tone
Masses
Consistency
Location
Mobility
Pulsation
What is the most important part of palpation during the physical exam?
Keep the patient at ease
Define Ballottement
solid structure suspended in fluid e.g. patella
What is important regarding the sequencing of the various parts of the physical exam?
the sequence should be divided by system but adjusted to minimize the amount of changing positions the patient will have to do.
Describe parts of the physical exam to be performed while the patient is in the seated position
general appearance
vital signs
extremities
parts of the neurological & musculoskeletal systems
Describe subjective vs. objective information
Subjective – information given to you, what the patient tells you.

Objective – measurable information such as vital signs & head to toe exam
Describe the difference between Autocrine, Paracrine, and Endocrine action

Autocrine – target sites are on the same cell that produces the messenger

Paracrine – Secreted into interstitial fluid targeting adjacent cells

Endocrine – Secreted into the blood

Describe the general characteristics of signal transduction systems

signal/ligand released due to stimulus
ligand travels to site of action
Recognition & Binding of ligand by receptors
Binding results in a change in the receptor that creates a response in the cell

Signal is terminated

Regarding signal transduction, describe small messenger molecules
steroids – cortisol, estrogen, testosterone

lipid–soluble, able to diffuse across membranes
Regarding signal transduction, describe peptide messenger molecules
proteins
cytokines
growth factors
Hormones

require active/facilitated transport
List the main types of plasma membrane receptors

Ligand–gated ion channels
Receptor kinases
Receptors that act through accessory kinases
G–protein coupled receptors

Describe intracellular receptors
act primarily as transcription factors which regulate gene expression in the response signals being released
Describe Protein Kinases

Protein Kinases transfer a phosphate group from ATP to the –OH group of a specific Amino Acid side chain on a protein

Describe Protein Phosphatases

Protein Phosphatases catalyze the dephosphorylation & modulate the phosphorylation cascade process

Describe Co–Repressors / Co–Activators
modulate signaling through intracellular receptors that are transcription factors
Explain the general mechanisms through which signal termination & desensitization occur
Uncoupling – GPCRs uncoupling from G–proteins after the ligand binds
Endocytosis – internalization and degradation of protein receptors
Modifications by other proteins decreasing signal activity
Altered expression of the proteins
Describe the process by which a Tyrosine Kinase Receptor connects to an adaptor protein in general terms:
Ligand binding
Dimerization
Autophosphorylation
Connection to Adaptor protein on activated receptor site
Describe the process by which Tyrosine Kinase Receptors activate the Ras pathway
Ligand binding
Dimerization
Autophosphorylation
binding of Adapter protein
binding of GEF protein
attachment & activation of Ras to Ras–GTP
Ras–GTP activates Raf
Raf continues on into the MAP–K pathway
What type of receptor initiates the MAP–K pathway through Ras
Tyrosine–Kinase Receptors
Define GEF proteins
Guanine Nucleotide Exchange Factors
Define GAP proteins
GTPase Activating Protein
Define the function of GTPase
replace GTP with GDP
What is the domain on the Adaptor Protein that binds to the phosphorylated region of the Tyrosine Kinase Receptor
SH2 domain
What is the domain on the Adaptor Protein that binds to the GEF protein
SH3
What is the function of the GEF protein?
Catalyzes the exchange of GDP to GTP on Ras
In Signal Transduction:

Define PI
Phosphotidylinositol
Define the importance of phosphatidylinositol
membrane bound structure that through phosphorylation becomes PIP2 and PIP3
What processes work on PIP2 leading to the release of Calcium intracellularly
Phospholipase C + PIP2 creates IP3 and DAG

IP3 stimulates the release of Calcium from the Endoplasmic Reticulum
What molecule is needed to change PIP2 to PIP3?

Phosphatidylinositol 3–Kinase

Define DAG
Diacylglycerol
What is the function of DAG
DAG interacts with Protein Kinase C
Describe the Jak–STAT pathway
Receptor binds cytokines, dimerizes, and binds 2 Jaks which phosphorylate each other and the receptor. The receptor binds & phosphorylates 2 STATs which dissociate from the receptor, dimerize, and translocate to the nucleus
Describe the Serine–Threonine Kinase Pathway
TGF–beta binds to Type 2 receptor which phosphorylates the type 1 receptor.

The activated Type 1 receptor phosphorylates R–Smad.

R–Smad complexes with Co–Smad & migrates to nucleus
Define the term GPCR
G–Protein coupled Receptor
Describe the G–protein structure [general]
Subunits – alpha, beta, gamma

Alpha subunit is activated with the exchange of GDP to GTP, leaves the beta/gamma to go off intracellularly to create a cell response
Describe the three types of G–alpha sub proteins that are relative to [general] signal transduction as covered in the directed study
G–alpha–S
G–alpha–i
G–alpha–q
What is the general function of G–alpha–S protein

G–alpha–S is attached to GTP, detaches from gamma & beta components of the G–protein.
It targets the Adenylyl Cyclase which hydrolyzes GTP, G–alpha–S dissociates & returns to g/b portions of the G–protein.
Adenylyl Cyclase converts ATP to cAMP
cAMP activates Protein Kinase A which initiates the phosphorylation cascade
cAMP is degraded by cAMP phosphodiesterase

Describe the function of G–alpha–I protein
G–alpha–i protein inhibits Adenylyl Cyclase and thus the production of cAMP and the phosphorylation cascade
What structure degrades cAMP
cAMP phosphodiesterase
What is the general function of G–alpha–Q protein
G–Alpha–Q activates phospholipase C which ultimately stimulates DAG and IP3 release
State the function of GAPs
GTPase activating proteins – terminate reactions that utilize GTP
What [DS] signal transduction reactions are inhibited by GAPs
G–alpha–S
G–alpha–Q
Ras
State the function of cAMP phosphodiesterase

turns off cAMP which goes on to stimulate the phosphorylation cascade through Protein Kinase A

What [DS] signal transduction reactions are inhibited by cAMP phosphodiesterase

G–alpha–S due to the reliance on Adenylyl cyclase which catalyzes ATP to cAMP which travels on–wards to stimulate PKA and the phosphorylation cascade.

What [DS] signal transduction reactions are inhibited by protein phosphodiesterase

all phosphorylations

kinases–TK, JakSTAT, PI3–K

State the function of protein phosphatase

dephosphorylates proteins

Describe receptor internalization and degradation and the effect
tendency of cells to internalize structures and recycle the components. The reduction in the number of receptors bound to the membrane can decrease the overall sensitivity of the cell to outside messaging unless it maintains spare receptors and the fluctuation is negligible.
Describe chemical chirality
tendency for substances to have a combination right & left handed enantiomers

e.g. Mirror Images
Describe chirality and the effect on pharmaceutical action
chemical chirality influences pharmaceutical action because different isomers may be +/– effective or inactive
Describe chirality and the effect on pharmaceutical toxicity
chemical chirality influences toxicity because one isomer may have a therapeutic effect while the other may be toxic

ex: lamivudine. L–form tx HIV, R–form toxic
Describe chirality and the effect on pharmaceutical metabolism
chemical chirality influences drug–metabolism because if a dose is 50/50 racemic mixture of R–L handed structures and only half is absorbed via the GI or if one undergoes faster processing via the Renal/Liver systems, the overall effective dose is lessened than a purely 100% functional isomer
Describe Drug Selectivity
ability to affect one tissue, cell type, or organ while not others
Describe determinants of drug selectivity
Drug Dose
Drug Distribution
Receptor Distribution
Receptor Specificity & Selectivity
What is the significance of a drugs selectivity
a drug is given to stimulate/inhibit an effect in a specific target tissue, the drug's selectivity determines whether that drug will inhibit/stimulate ancillary tissues/processes causing side–effects not beneficial to the original cause of the administration.
Describe the term pharmaceutical Agonist
a drug that has an affinity to the receptor and also has intrinsic activity
Define Intrinsic activity [pharm]
able to produce a measurable effect
Define the term pharmaceutical Antagonist
a drug that has an affinity for a receptor but does not have intrinsic activity

receptor is activated but no measurable effect is produced
Define the term pharmaceutical partial agonist
a drug that interacts with the same receptors but cannot produce the same maximal effect as a full agonist
Define the term Affinity [pharm]
how strong an attraction that the drug has for the receptor.

measure of binding
Define the term Potency [pharm]
related to the drug binding affinity (Kd)

related to the amount of drug needed to produce a given intensity of effect

Higher binding reduces the overall drug needed to produce a specific effect
Define the term Efficacy [pharm]
the largest response or maximal effect – Emax
that a drug can produce
Describe the [pharm] term Intrinsic Activity
extent to which a bound Ligand activates a receptor

Agonist: IA=1
Antagonist: IA=0
Describe the graph of two drugs, an agonist & a partial agonist
The agonist will have a E–max of 100 while the partial agonist will partial agonist will have a reduced E–max.

The ED–50 value will be identical between the two.
Define the [pharm] term E50

The drug dose needed to reach 50% of the maximal drug effect (Emax)

Describe the [pharm] term clinical efficacy
depends on maximal efficacy & the drugs ability to reach the relevant receptors
[pharm] Describe the relationship between ED–50, potency, binding affinity, and Kd

The higher the binding affinity(lower Kd), the lower the drug concentration needed to reach 1/2 Emax (ED–50)

low ED50 = high affinity & potency, Low Kd



higher ED50 = low affinity & potency, HIGH Kd

Describe the graded log–dose response curve

the curve looks like a vertically stretched S with the maximum point being the Emax of the drug with the lowest point being the threshold dose.

[pharm] Define threshold dose
the lowest drug dose that will produce a therapeutic effect or an effect first appears
[pharm] Describe the change to a graded log–dose response curve graph of agonists that differ in potency
potency = drug concentration vs. effect

graph will shift R or L

there will be no change in E–Max

[pharm] Describe the change to a graded log–dose response curve graph of agonists that differ in efficacy

E–max will have a vertical shift (up/down)
Describe [pharm] graphically the effect of a competitive antagonist on an agonist drug's action
the antagonist binds to the same receptor site as the agonist, preventing it from binding while producing no intrinsic activity.
potency of drug is reduced, graph shifts right because the antagonist can be overcome with more a higher agonist concentration

No change to E–Max
Describe [pharm] graphically the effect of a non–competitive antagonist on an agonist drug's action
the antagonist binds to a non–active site on the receptor causing a conformational change in the receptor shape, eliminating the agonists ability to bind. IA=0.

increasing the agonist conc. has zero effect

Graph will show a downward shift in E–max
Describe [pharm] graphically the effect of an irreversible antagonist on an agonist drug's action
the antagonist covalently binds to receptor first, eliminating the agonists ability to bind. IA=0.

increasing the agonist conc. has zero effect

Graph will show a downward shift in E–max
What are the advantages & disadvantages to irreversible inhibitors?
adv – a bound inhibitor stays bound until replaced. Eliminates the need to repeatedly dose a patient to maintain therapy

dis–the body must replace the receptor before any other action can be taken (tetanus)
Describe the concept of Spare receptors
A cell maintains more receptors than strictly necessary to produce the maximal internal response. This increases cellular sensitivity to messengers such as hormones that are typically in low serum concentrations

effect: lower dose to provide a result
Detail how spare receptors will interact with agonist and antagonistic drugs in a graph
The graph of a drugs response will shift to the right without changing E–max if there are spare receptors present as there is no initial competition for sites between the agonist–antagonists.

Once all receptors are occupied, the antagonist will force the E–max downwards as they fight for an increased % of total receptors.
[Pharm] describe chemical antagonists
chemical interaction of two substances where a receptor is NOT involved

overall effect of the active substance is lost

IA = 0
Name examples of chemical antagonists
Metal chelators (EDTA) vs. toxic metals (lead)

Antiseptics
[Pharm] describe pharmacologic antagonists
a drug that has an intrinsic activity of 0

prevents action from happening
[Pharm] describe physiological antagonists
when there are opposing effects by 2 agonists
Biceps vs. Triceps = no movement of the arm
Define ED50, LD50, TD50
ED50 – Effective Dose in 50% of the populous

LD50 – lethal dose in 50% of the populous

TD50 – Toxic dose in 50% of the populous
Define therapeutic index
the ratio of Lethal dose (50%) to Effective dose (50%) that provides a measure of the safety of a drug

higher value = safer drug
Describe the purpose of the cellular membrane
structure/rigidity
transportation of substances in/out
Describe why substrate transport across the cell membrane is necessary for cellular function
cellular homeostasis

metabolism

cellular signaling
Describe [bio] selectively permeable
the ability of the cellular membrane to allow only desired substances to pass
Cellular Membrane, describe the movement of Gases, Water, Ions, metabolic substrates, and proteins across the cellular membrane
Gases & water(maybe) – simple diffusion

Ions & metabolic substrates – facilitated diff

proteins – facilitated diffusion if not necessary for cellular function
Name the factors that determine substances membrane permeability
size & charge – smaller = better

larger area = larger diffusion rate
Describe the two forces in molecular movement in physiological systems
Diffusion – randomized movement, movement from a higher to lower gradient

Bulk Flow – concerted manner, driving pressure
List two systems that utilize bulk flow to function
Circulation

Respiration
Describe the term Concentration Gradient
the concentration/charge/forces on a molecule on one side of a membrane versus those applied to the other side of the membrane.

Movement always from higher conc. gradients to lower if possible
Describe the difference between simple & facilitated diffusion
Simple diffusion – transfer of a molecule across a membrane without help

Facilitated diffusion – carrier molecules (transport proteins or ion channels) are needed for the molecule to move across the membrane
Name the two types of proteins that facilitate diffusion
transport proteins

ion–channel proteins
list the factors that determine the rate of diffusion across the membrane

concentration gradient
area of a membrane
membrane permeability of molecule


lipid solubility, size, charge

Describe the graphic relationship between the rate of diffusion & concentration gradient across a membrane for a freely permeable molecule

straight line with slope dependent on permeability coefficient vs concentration gradient
higher slope = higher permeability coeff.

Describe the effect of saturation on the graph of the rate of molecular facilitated diffusion & concentration gradient across a membrane

the graph will appear roughly straight with a slope depending on the permeability coefficient and concentration gradient until all of the facilitating proteins are occupied at 100% capacity where the graph will level off to a maximum rate

Explain the role and selectivity of gating channels in controlling membrane permeability to ions
Channels assist with membrane permeability in allowing specific ions to pass. Channels maintain different shapes.

Some require a ligand binding or specific voltage charge in order to allow passage to ions
Describe the difference between voltage & ligand gating
voltage – motivated by the net difference in charge across the channel

ligand – opens only when ligand–bound
Define Active transport & its purpose
Active transport refers to the process in which a solute is moved up a concentration gradient with an energy input (ATP)
Explain secondary active transport
utilizes the passage of another molecule (usually Na+) to facilitate the movement of a 2nd
What is the functional importance of Secondary Active Transport
move other substances into the cell by using the already existing process of sodium ion transport moving into the cell
List the two types of secondary active transport proteins
Symport – co–transport system, usually with glucose

Antiport – sodium in, another molecule out
Name the secondary active transport mechanism that prevents glucose from appearing in the urine under the normal glycemic conditions
glucose is reabsorbed via symports in the proximal renal tubules in conjunction with sodium ions
Explain why glucose might be present in urine
glucose will be found in the urine when the concentration of glucose exceeds the available symports, any excess glucose that is unable to be reabsorbed will be excreted in the urine (glycosuria)
Give the Professional definition of osteopathic medicine
Osteopathic medicine is a complete system of medical care practiced by physicians with an unlimited license that is represented by a philosophy that combines the needs of the patient with the current practices of medicine, surgery, and obstetrics. It emphasizes the interrelationship between structure and function, and that has an appreciation of the body's innate ability to heal itself.
Give the 4 Principles of Osteopathic Medicine
the body is a unit: body, mind & spirit

the body is capable of self–regulation, self–healing, and health maintenance

Structure & function are reciprocally interdependent

Rational treatment is based upon an understanding of these principles
Define somatic dysfunction
a somatic dysfunction is the impaired or altered function of related components of the somatic (body framework) system
Describe the diagnosis of a Somatic Dysfunction
1) Can treat with OMT

2) TART
Define the mnemonic TART
Tissue texture abnormalities
Asymmetry
Restriction of Motion
Tenderness
Describe the concept of Joint Play
movement of the synovial joint that is independent of & cannot be reproduced by a voluntary movement

essential for maximal pain free movement

can be restored by OMT
What are the absolute contraindications of OMT
The patient refuses to have OMT performed

Absence of a somatic dysfunction
What are the relative contraindications of OMT
When the potential benefits outweigh the risk of harm to the patient

Acute muscle spasms or muscle soreness or treatment reactions
Describe direct OMT techniques [general]
technique in which the restricted tissue is initially taken in the direction of the restriction of motion
Describe indirect OMT techniques [general]
techniques that initially position the restricted tissue toward the relative ease or freedom of motion

Less risk for the patient
Discuss the range of motion diagram

Depicts the RoM of any particular joint

Extremes:
Anatomical barrier: Passive RoM barrier

Physiological barrier: Active RoM barrier

Neutral: prone position

Define soft tissue techniques
a direct osteopathic technique that usually involves lateral & linear stretching, deep pressure, traction and/or separation of muscle origin & insertions while monitoring tissue response & muscle changes by palpation
Define the differences between OMT:

Traction

Traction – Longitudinal Muscle Stretch

think: tug of war on a rope

Define the differences between OMT:

Kneading

Kneading – Lateral Muscle Pressure

think: plucking a guitar string

Define the differences between OMT:

Effleurage

Effleurage – Stroking pressure to move fluid

Define the differences between OMT:

Petrissage

Petrissage – Squeezing pressure to move fluid

Define the differences between OMT:

Tapotement
Striking with the side of the hand
Define the differences between OMT:

Skin Rolling
Lifting the skin away from the deeper structures & rolling the skin fold along the body
List the physiological mechanisms of action & therapeutic effects of soft tissue techniques
Relax hypertonic muscles and reduce spasm
Stretch and increase the elasticity of shortened fascial structures
Enhance circulation to local myofascial structures
Improve local tissue nutrition, oxygenation, and removal of metabolic wastes
Improve abnormal neurological reflexes
Identify areas of restricted motion, tissue texture changes and sensitivity
Improve local systemic immune responses
Observe tissue response to the application of manipulative technique
Provide a general state of relaxation
Provide a general state of tonic stimulation
Optimize overall autonomic tone
What are indications for soft tissue techniques
any somatic dysfunction
What are contraindications of soft tissue techniques
local infection,
open wounds
lack of skin
soft tissue integrity
anti–coagulated patients with bruising
Define OMT Treatment Reaction
24–48 hours after OMT
muscle soreness relieved by rest, warm bath, mild anti–inflammatory/analgesic medication

Somatoemotional release
Define Somatoemotional release
if an emotion is connected in some way to a somatic dysfunction and this dysfunction later receives OMT has the risk of the unexpected recurrence of the emotion

Think: Doc's shoulder
Where and when does replication occur?
In the Nucleus

During S–Phase
What is the role of helicase in DNA replication?
separates the parental DNA strands

Hint: Zipper
What is the role of topoisomerase in DNA replication
relieves the supercoiling after helicase uncoils the strands

single–stranded binding proteins then keep the strands from binding back together

hint: Legs
What is the role of single–stranded binding proteins in DNA replication
prevents the parental strands from pairing back up after topoisomerase splits the strands and helicase uncoils the strand
Describe the role of primase in DNA replication
adds RNA primer once topoisomerase splits open the strands of DNA.

this is followed by DNA polymerase adding in the daughter strands
What is the role of processivity factors in DNA replication?
helps hold DNA polymerase onto the substrate as it creates the daughter strands
Describe the role of RNAse in DNA replication?
RNAse removes the RNA primers

after RNAse: ends are then joined by the ligase
Which polymerase synthesizes the leading strand?
DNA Polymerase E (epsilon)
Which polymerase synthesizes the lagging strand?
DNA Polymerase d (delta)
Describe the function of Telomerase
Edits the end of the lagging strand in order to get a segment containing the hydroxyl group that a ligase can use to bind the Okazaki fragments together
What are Okazaki fragments?
the segments of the lagging strand that are written by DNA polymerase delta and are joined by the DNA ligase
What are the 3 types of RNA
rRNA
mRNA
tRNA
Describe the function of tRNA
transport RNA moves amino acids for use in protein translation
Describe the function of mRNA
messenger RNA that is transcripted from DNA and it transported out of the nucleus and translated into proteins
Describe the function of rRNA
ribosomal RNA is a component of Ribosomes which are used to translate mRNA into proteins
What is the importance of the TATA box?
TATA binding protein must find the TATA box to begin transcription
What is the role of the TATA binding protein
TATA binding proteins locate the TATA box
What is the role of transcription factors
transcription factors recruit RNA polymerase which is used to synthesize RNA
What is the function of RNA polymerase
RNA polymerase synthesizes RNA during transcription
What is the importance of the 5'–cap on RNA
the 5'–cap increases the half–life of the transcripted RNA & is important in binding the transcript to the ribosome
What is the part of RNA that increases the half–life & assists in binding the mRNA to the ribosome?
5'–cap
What is the polyA–tail and what does it do?
the polyA–tail protects the transcribed mRNA from degradation
Describe the composition of ribosomes in Eukaryotes vs. Prokaryotes
ribosomes = rRNA + ribosomal proteins

Eukaryotes: 60s + 40s = 80s

Prokaryotes: 50s + 30s = 70s
What are the two types of ribosomal subunits in Eukaryotes
60s + 40s

combine into a 80s
What are the two types of ribosomal subunits in prokaryotes
50s + 30s

combine into a 70s
Describe the process of transcription [general]
TATA binding proteins find TATA box

Transcription factors recruit RNA polymerase

RNA polymerase synthesizes new RNA into mRNA

mRNA travels to the ribosomes for processing
List the three steps of RNA translation [general]
Initiation
Elongation
Termination
Describe RNA translation Initiation
tRNA gets charged by Aminoacyl tRNA synthetase

initiation factors bind – initiate translation

charged tRNA finds 80s[40s/60s] ribosomal subunit

elongation begins
Describe RNA translation elongation
tRNA+Amino Acid are picked up by the elongation factors
complex enters ribosomal A site
Anticodon matched against mRNA at A–site
chain is shifted to the P–site where peptide bond formed between the amino acid in P–site with the incoming amino acid in A–site
proofreading via the elongation factors
What is the function of peptidyl transferase?
formation of the peptide bonds between the amino acids in P–site and A–site
What is the importance of chaperones & heat shock proteins?
these are required to overcome kinetic barriers

force the protein into the correct desired protein folding
Define Free ribosomes
ribosomes not attached to the endoplasmic reticulum
Compare the processing of proteins that are synthesized on free ribosomes vs those on the rough ER
free ribosomes – may stay in the cytoplasm or certain organelles

ER – enter golgi apparatus and are subjected to exocytosis
What is the translation function of aminoacyl tRNA synthetase
charges the tRNA before synthesis
What is the function of initiation factors in RNA translation
required to find the start codon & thus initiate translation
What is the role of histones?
chromatin remodeling is needed to unwind the chromatin that are tightly bound around the histone protein. Histones are proteins that wind DNA very tightly so as to compact it into a very small area. These combine into a 4–protein octamer within the nucleus.
What is the function of HATs
Histone acetyltransferase – responsible for turning ON gene expression. HATs transfer an acetyl group to the chromatin to neutralize the charge–attraction between the DNA strand and the histone resulting in the DNA unwinding

Recruited by activator proteins
What is the function of HDACs
Histone deacetylases turn OFF gene expression by removing the acetyl group & allowing the recondensing of the histone octamer.

Recruited by repressor proteins
Describe methylation and the effect on DNA
methylation of genes results in them being less readily transcribed. Problems are encountered when the STOP codon is methylated.
What is the function of methyltransferases
maintain methylation by adding methyl groups to the daughter strands and performing maintenance methylation
What is name for the process by which new methyl groups are added?
De Novo Methylation
By what mechanisms are transcription factors regulated?
their own expression level
expression of coactivators & corepressors
presence of inhibitors
phosphorylation
Describe alternative RNA processing & RNA editing
RNA editing – bases are altered chemically, using different exons

by alternative splicing to allow different protein sequencing from the same gene
Which stays with the protein: Intron or Exons?
Exons, they exit with the protein
Describe 3 mechanisms affecting RNA stability
regulatory proteins
5'–cap
poly–A tail
Describe the effect of MicroRNA on gene expression
induce transcript degradation
block translation
Define cell "resting membrane potential"
The resting membrane potential of a cell is the point at which there is zero net movement of ions across the cell membrane.

What are the two uses the resting membrane potential for a cell?

Excitability – ability to signal electrically

Absorb & Secrete solutes – Think Nernst Potential

What is the voltage of a cell at the typical resting membrane potential?

-60 to -85 mV

Describe why the cellular resting membrane potential is maintained at a specific value?
the cell membrane overs around 60–85 mV because the membrane is fully permeable to K+ ions that have a Nernst potential of –88 mV but some Na+ ions (NP=+70) leak through forcing the membrane more positive
What is the extracellular membrane potential in mV?
always 0 mV
What are two types of cells that utilize membrane potential?
Nerve cells
muscle cells
What are the two forces that control movement of ions across the membrane
Concentration gradient
Electrical Gradient
Define concentration gradient
directs the flow of ions based on the concentration of solutes on the outside of the membrane versus those on the inside of the membrane. Solutes always move from high to low concentration if permitted
Define electrical gradient
difference in the electrical charges in vs outside the membrane. net flow will move to equalize the difference between negative & positive sides.
Define Driving Force [cell membrane]
force that dictates which direction the molecules move. Combines electrical & concentration gradients.
Define equilibrium in terms of membrane ion movement
No driving force for ion movement. It is established when the conc. & electrical gradient are equal but opposite
What is the Nernst equilibrium potential

is the membrane potential at which the concentration gradient + electrical gradient are equal but opposite e.g. balanced

What is the NP of Sodium?
+70 mV
What is the NP of Potassium?
–88 mV
What is the NP of Chlorine?
–47 mV
What is the NP of Calcium?
+122 mV
What are the concentrations of Sodium?
in: 10 mM
out: 140 mM
What are the concentrations of Potassium?
in: 140 mM
out: 5 mM
What are the concentrations of Chlorine?

in: 20 mM
out: 116 mM

What are the concentrations of Calcium?
in: 0.0001 mM
out: 1 mM
Describe the factors that make a membrane selectively permeable to an ion
membrane permeability to an ion depends on the # of open channels selective for that ion present in the membrane
Describe the graphic relationship between [K+]o, Ek, and the membrane potential
[K+]o is the concentration of K+ ions outside
Ek is the NP for potassium
Membrane potential is the membrane response to balance the gradients
If the membrane is solely permeable to Sodium ions, what is the membrane potential?
+70 mV
Explain why membrane potential is different from Ek at very negative membrane potentials
there will be slight movement of Na+ into the cell from Na+ leak channels
What is the ratio between Na+/K+ ions in a typical resting permeability
1 Na+ to 100 K+

in : out
10 : 140 Na
140 : 5 K
Describe the influence of Ca2+ & Cl– ions on the resting membrane potential in nerves and in the muscle cell

Calcium = zero influence on membrane potential

Chlorine influences skeletal muscle on most cell types.

What is the membrane potential for Skeletal muscle cells?
–85 mV
Define depolarization
a situation where the membrane or Nernst potential become less negative
Define hyperpolarization
Nernst or membrane potential becomes more negative
Describe the 7 basic elements of Healthcare communication
Open the discussion
Build a relationship
Gather information
Understanding the patients perspective
Sharing Information
Reaching agreement of problems/plans
Providing closure
List the methods drug administration
Oral
Describe the benefits & disadvantages of:

Oral administration
most common route, generally the safest, GI irritation, First–pass effect
Define First–Pass metabolism
metabolism of the drug by the GI bacterial enzymes, the GI tissue enzymes, and by the Liver enzymes prior to the drugs insertion into the systemic circulation
Describe the benefits & disadvantages of:

Rectal administration
potentially avoids first pass, absorption is slow and possibly erratic
Describe the benefits & disadvantages of:

transdermal administration
must be lipid–soluble, slow absorption, varying bioavailability
Describe the benefits & disadvantages of:

sublingual administration
direct absorption into the systemic circulation, higher bioavailability, rapid absorption
Describe the benefits & disadvantages of:

Buccal administration
direct absorption into the systemic circulation, higher bioavailability, rapid absorption
Describe the benefits & disadvantages of:

topical administration
no systemic exposure, localized effect

should be lipid–insoluble
Describe the benefits & disadvantages of:

subcutaneous administration
absorption is constant, complete, and slow
depends on blood flow for absorption rate
bypass First Pass effect
Describe the benefits & disadvantages of:

intramuscular administration

absorption by simple diffusion, generally rapidly, higher bioavailability
bypass First Pass effect

Describe the benefits & disadvantages of:

intravenous administration
avoids first–pass effect, fastest route, complete bioavailability, rapid effect, toxicity and occur with rapid drug administration
Describe the benefits & disadvantages of:

inhalation administration
local effect & systemic effect
absorption is rapid
avoids first–pass effect
Describe the benefits & disadvantages of:

Intrathecal administration
administration into the cerebrospinal fluid
used for local, rapid effect on the meninges & CS axis
avoids blood–brain barrier
Describe the graph of the IV drug concentration over Time

The IV dosage avoids the first pass effect and is placed directly into the systemic circulation at full concentration with subsequent elimination by the liver & kidneys

Describe the graph of a PO drug concentration over time

The PO dosage undergoes delivery through the GI system after undergoing First Pass metabolism prior to delivery into the systemic circulation where it subsequently undergoes elimination.
Rise from delivery to systemic concentration, then slow downgrade.

Describe primary process by which drugs cross the membrane
First Order process: rate is dependent on concentration (high to low)
Describe the importance of lipid–solubility on drugs membrane permeability

higher lipophilic = higher solubility = faster diffusion across the membrane

Ionization decreases the lipid–solubility.

pH controls %ionization and thus % absorbed

Describe the relationship between pH & ionization of a drug which is either a weak organic acid or base.

the drug must be administered so that the pH of the delivery system pushes the drug to unionized structure in order to pass through the membrane

Define Strong vs. Weak Acids & bases.

Strong Acid & Bases: fully ionized in all solutions

Weak Acids: only ionize at higher pH's
Weak Bases: only ionize at lower pH's

Define pKa
pH where the substance is 50% ionized & 50% unionized
Provide the equation for a weak acid


(think Log)
pH–pKa = log [ionized / unionized]
Provide the equation for a weak base


(think Log)
pH–pKa = log [unionized / ionized]
List the factors that affect the rate & extent of oral absorption

drug solubility
lipid solubility (nonionized form)
drug concentration @ absorption rate
surface area for absorption
blood flow @ absorption site
gastric emptying
food
metabolism of drug

What determines bio–availability

% of drug in systemic circulation after:

First pass effect
Rate of extraction