• 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

1/960

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;

960 Cards in this Set

  • Front
  • Back

[OPP Structural Screening]

What does a static structural screening exam provide details for?

the presence of a somatic dysfunction

does not identify the dysfunction, just that one is present
[OPP Structural Screening]

What are the lateral anatomical aspects of the optimal gravitational line?
Anterior aspect of the lateral malleolus
middle of the tibial plateau
greater trochanter
body of L3 (center of the body mass)
middle of the humeral head
external auditory meatus
[OPP Structural Screening]

What are the posterior anatomical aspects of the optimal gravitational line?
Popliteal creases
greater trochanter
iliac crests
inferior angles of the scapula
acromion processes
mastoid processes
[OPP Structural Screening]


Interpret positive hip drop test
–ask the standing patient to bend one knee
–this causes the hip on that side to drop toward the floor
–this induces the lumbar spine to bend in the opposite direction to keep the eyes level
–the hip usually drops >=25 degrees
–if less drop = positive test
positive hip drop test = restricted lumbar side–bending on OPPOSITE SIDE

[OPP Structural Screening]


Interpret positive hip drop test

–stand behind the patient & stabilize the head & shoulder with one arm
–use the other arm to push the shoulder inferiorly & medially
–this induces sidebending in the thoracic spine
–comparing the sides = the side w more resistance indicates restricted thoracic sidebending

[OPP Structural Screening]


Discus the treatment of a patient with short leg syndrome (unlevel sacral base)

OMT
Exercise
PT
heel–lift on side w depressed sacral base

[OPP Structural Screening]

Properly name a scoliotic curve
Major double scoliosis
single scoliosis
junctional scholiosis
[OPP Structural Screening]

List some risk factors associated with the presence & severity of scoliotic curves
Females: 3–5 times more likely
Bone Maturity
Initial curve magnitude
– smaller curves in mature patients = lower risk of progression
– > 45 deg curves in growing patients or >50 deg curves in mature will progress over time
[OPP Structural Screening]

What are the causes of scoliosis?

Idiopathic! (70–90% nfc)

[OPP Structural Screening]

Differentiate between structural & functional scholiosis
If patient sidebends towards convexity (toward rib hump)

functional – hump diminishes/disappears
Structural – does not reduce
[OPP Structural Screening]

Describe the utilization of the Adam's Test to screen for scholiosis
method: patient bends forward = presents a rib hump
Rib hump on right, rotation to right
Rib hump on left, rotation to left
[OPP Structural Screening]

Classify scoliosis based on the degree of deviation found by the Cobb method of measuring scoliotic curves
mild = 10–15 degrees

moderate = 20–45 degrees

severe > 50 degrees
[OPP Structural Screening]

Describe at what scoliotic angle you will encounter impaired respiratory function
> 50 degrees deviation
[OPP Structural Screening]

Describe at what scoliotic angle you will encounter impaired cardiovascular function
> 75 degrees deviations

[OPP Structural Screening]

What are the three etiological classifications of scoliosis

Congenital
Neuromuscular
Idiopathic
[OPP Structural Screening]


Describe the Congenital classification of scoliosis

Failure of formation

failure of segmenetation

mixed

[OPP Structural Screening]


Describe the Neuromuscular classification of scoliosis
Neuropathic

Myopathic

mixed
[OPP Structural Screening]


Describe the Idiopathic classification of scoliosis
Infantile – birth to 3y

Juvenile – 4–9y

Adolescent – >10y
[OPP Structural Screening]


Detail the heel lift protocol
flexible pt = 1/8" lift, no more than 1/16" q2wk

fragile pt = 1/16" lift, no more than 1/16" q2wk

injured – full amount immediately
[OPP Structural Screening]


Define kyphosis
excessive outward curvature of the spine, causing hunching of the back.
[OPP Structural Screening]


Define Lordosis
a posture assumed by some female mammals during mating, in which the back is arched downward.
[intro to epidemiology]

Describe the useful aspects of epidemiology

–to study the history of the disease
–community diagnoses
–determine risk for individuals, groups, populations
–assessment, evaluations, research
–Identification of syndromes
–Determine causes of disease, sources of disease, & effective tx for disease

[intro to epidemiology]

Define ENDEMIC
A disease or pathogen present or usually Prevalent in a given population or geographic region at all times.
[intro to epidemiology]

Define EPIDEMIC
A disease occurring suddenly in numbers far exceeding those attributable to ENDEMIC disease, occurring suddenly in numbers clearly in excess of normal expectancy.
[intro to epidemiology]

Define PANDEMIC
A widespread epidemic distributed or occurring widely throughout a region, country, continent or globally.
[intro to epidemiology]

Define INCIDENCE
The rate of occurrence of an event, the number of NEW cases of disease occurring over a specified period of time; may be expressed per a known population size.
[intro to epidemiology]

Define PREVALENCE
The number of cases of disease occurring within a population at any one given point in time.
[intro to epidemiology]

Define Primary Prevention
involves halting any occurrence of a disease or disorder before it happens.
(1) Immunizations
(2) Smoking Cessation
(3) Weight loss
(4) Physical fitness activities
(5) Nutrition
(6) Sun exposure limitation
[intro to epidemiology]

Define Secondary Prevention
Health Screening and Detection activities.
(1) Breast self exam, Mammography
(2) Prostate exam, PSA
(3) Stool Hemocult, Colonoscopy
(4) Blood Pressure Screening
(5) Periodic blood tests – glucose, lipids, others
[intro to epidemiology]

Define Tertiary Prevention
preventing complications in those who have developed disease/illness and have been diagnosed.
(1) CVA (stroke rehabilitation)
(2) Cardiac rehabilitation, following MI
(3) Post knee/hip replacement Physical Therapy
(4) Diabetic foot exams
[intro to epidemiology]

Describe the types of studies useful to epidemiology
Cohort study
Case Control Study
Occupational Epidemiology study
Cross–sectional study
Clinical trial study
[intro to epidemiology]

Describe the Clinical trial study...
(a) Study pointed at development of new preventive strategies and new treatments for disease.
(b) Also called Randomized Clinical trials.
[DS – Intro to Immunology]

Describe the non–specific or innate defense system
–is always prepared, responding immediately to protect the body from all foreign substances
–First–line of defense within nonspecific immune system includes the body membranes (skin & mucosae) that prevent entry of MOs
–Second–line of defense within the nonspecific immune system is the chemical signals released when the external defenses are penetrated, using antimicrobial proteins as well as phagocytes & other defense cells to inhibit the spread of foreign bodies
[DS – Intro to Immunology]

Describe the specific (or adaptive) defense system
performs the attack against foreign substances & provides a 3rd line of defense for the body.
– the adaptive defense system is a functional system that is made of individual immune cells (lymphocytes), which inhabit lymphatic tissues & circulate in the body fluids, & a diverse array of molecules.
–the immune system protects the body from most infectious MOs, cancer cells, & transplanted organs/grafts. The immune system will directly attack foreign bodies & indirectly attack by releasing mobilizing chemicals & protective antibody molecules. Immunity is a high resistance to disease.
[DS – Intro to Immunology]

Describe the major features & characteristics of adaptive immune systems
– It is antigen specific: recognizes and is directed against particular antigens, against pathogens or foreign substances that stimulate the immune response
– It is systemic: Immunity is not restricted to the initial infection site
– It has memory: After an initial exposure, it recognizes and mounts even stronger attacks on the previously encountered pathogens
[DS – Intro to Immunology]

Compare & contrast active immunity & passive immunity
– Active Immunity: Naturally acquired active immunity occurs when the person is exposed to a live pathogen, develops the disease, and becomes immune as a result of the primary immune response.
– Artificially acquired active immunity can be induced by a vaccine, a substance that contains the antigen.
Passive Immunity: –Artificially acquired passive immunity is a short–term immunization by the injection of antibodies. Naturally acquired passive immunity occurs during pregnancy, in which certain antibodies are passed from the maternal into the fetal bloodstream.
[DS – Intro to Immunology]

Describe the primary immune response
– constitutes cellular proliferation and differentiation, occurs on the first exposure to a particular antigen.
i)
[DS – Intro to Immunology]

Describe the secondary immune response
occurs during a re–exposed to the same antigen; short lag phase, increased immune response
[DS – Intro to Immunology]

Describe the major functions of the B–Cells
lymphocytes that oversee humoral immunity; B–cells can be distinguished from other lymphocytes, such as T–cells and natural killer cells (NK cells), by the presence of a protein on the B–cell's outer surface known as a B–cell receptor (BCR). This specialized receptor protein allows a B–cell to bind to a specific antigen.
[DS – Intro to Immunology]

Describe the major functions of the T–Cells
a type of lymphocyte (itself a type of white blood cell) that play a central role in cell–mediated immunity. They can be distinguished from other lymphocytes, such as B cells and natural killer cells (NK cells), by the presence of a T–cell receptor (TCR) on the cell surface. They are called T–cells because they mature in the thymus (although some also mature in the tonsils).[1][2] There are several subsets of T cells: Helper, Cytotoxic, Memory, Regulatory, Natural Killer, and Mucosal Associated Invariant T Cells.
[DS – Intro to Immunology]

List the major Antigen–Presenting Cells
Dendritic Cells

Macrophages

B–Lymphocytes
[DS – Intro to Immunology]

Describe the APC aspect of the dendritic cell
process antigen material and present it on the cell surface to the T cells of the immune system.
They act as messengers between the innate and the adaptive immune systems.
Dendritic cells are present in those tissues that are in contact with the external environment, such as the skin
Once activated, they migrate to the lymph nodes where they interact with T cells & B cells to initiate and shape the adaptive immune response.
[DS – Intro to Immunology]

Describe the APC aspect of the Macrophage

are a type of WBC that engulf & digest cellular debris, foreign substances, microbes, and cancer cells in phagocytosis

[DS – Intro to Immunology]

Describe the APC aspect of the B–lymphocyte

principal functions of B–cells are to make antibodies against antigens, to perform the role of antigen–presenting cells (APCs), and to develop into memory B–cells after activation by antigen interaction. B–cells also release cytokines (proteins), which are used for signaling immune regulatory functions.

[DS – Intro to Immunology]

Explain the role of Naive Lymphocytes
– A naive T–cell is considered mature and unlike activated T cells or memory T cells it has not encountered its cognate antigen within the periphery.
– A naive B–cell has not been exposed to an antigen. Once exposed, it either becomes a memory B cell or a plasma cell that secretes antibodies specific to the antigen that was originally bound. Plasma cells do not last long in the circulation, this is in contrast to memory cells that last for very long periods of time. Memory cells do not secrete antibody until activated by their specific antigen.
[DS – Intro to Immunology]

Explain the role of Effector Lymphocytes
– Effector cells are the relatively short–lived activated cells that defend the body in an immune response.
– Effector B cells are called plasma cells and secrete antibodies
– Activated T cells include cytotoxic T–cells & helper T–cells, which carry out cell–mediated responses.
[DS – Intro to Immunology]

Explain the role of Memory Lymphocytes
– Memory T cells have become "experienced" by having encountered antigen during a prior infection, cancer, or vaccination. At a second encounter with the invader, memory T–cells can reproduce to mount a faster & stronger immune response than the first time the immune system responded to the invader.
– Memory B cells are a B–cell sub–type that are formed within germinal centers following primary infection and are important in generating an accelerated and more robust antibody–mediated immune response in the case of re–infection
[DS – Intro to Immunology]

List the primary immunologic organs & tissues
Thymus gland

Bone Marrow
[DS – Intro to Immunology]

List the secondary immunologic organs & tissues
Spleen
Lymph Nodes
Lymphoid Tissues
[DS – Intro to Immunology]

Contrast Cell–Mediated Immunity with Humoral Immunity
– Cell–Mediated Immunity does not involve antibodies, but rather involves the activation of phagocytes, antigen–specific cytotoxic T–lymphocytes, and the release of various cytokines in response to an antigen.
– Humoral Immunity: called the antibody–mediated immune system, is the aspect of immunity that is mediated by macromolecules (as opposed to cell–mediated immunity) found in extracellular fluids such as secreted antibodies, complement proteins and certain antimicrobial peptides. Humoral immunity is so named because it involves substances found in the humours, or body fluids.
[Intro to Immunology]

Compare & contrast the innate defenses & the adaptive/acquired immune response
Innate: body's natural barrier/defenses, non–specific, cells use pattern–recognition receptors to recognize pathogen–associated molecular patterns
Adaptive: antigen–specific, clonal response, Memory, Lymphocytes use antigen–specific receptors that recognize specific epitopes
[Intro to Immunology]

What are PRRs?
Pattern–recognition Receptors
[Intro to Immunology]

What are PAMPs?
Pathogen–associated Molecular Patterns
[Intro to Immunology]

What are the Mechanical, Chemical, and Microbiological mechanisms of defense in the

SKIN
– Epithelial cells joined by tight junctions
– Flow of fluid, perspiration, sloughing off of skin
– Sebum (fatty acids, lactic acid, lysozyme)
– Antimicrobial peptides (defensins)
– Normal flora
[Intro to Immunology]

What are the Mechanical, Chemical, and Microbiological mechanisms of defense in the

GI tract
– Epithelial cells joined by tight junctions
– Flow of fluid, mucus, food, saliva
– acidity, enzymes (proteases)
– Antimicrobial peptides (defensins)
– Normal flora
–vomit, diarrhea
[Intro to Immunology]

What are the Mechanical, Chemical, and Microbiological mechanisms of defense in the

Respiratory Tract
– Epithelial cells joined by tight junctions
– Flow of fluid, mucus, (cilia, air flow)
– lysozyme in nasal secretions
– Antimicrobial peptides (defensins)
– Normal flora
– cough, sneeze
[Intro to Immunology]

What are the Mechanical, Chemical, and Microbiological mechanisms of defense in the

Urogenital Tract
– Epithelial cells joined by tight junctions
– Flow of fluid, urine, mucus, sperm
– Acidity in vaginal secretions, spermine & zinc in semen
– Antimicrobial peptides (defensins)
– Normal flora
[Intro to Immunology]

What are the Mechanical, Chemical, and Microbiological mechanisms of defense in the

Eyes
– Epithelial cells joined by tight junctions
– Flow of fluid, tears
– lysozyme in tears
– Antimicrobial peptides (defensins)
– Normal flora
[Intro to Immunology]

Compare & contrast the innate immunity & adaptive immunity

[think: summary table of differences]
Innate: recognition, rapid response (hours), fixed spectrum, limited # of specificities, constant during response

Adaptive: recognition, slow response (days to wks), variable spectrum, numerous highly selective specificities, improves during response
[Intro to Immunology]

List the basic principles of the clonal selection mechanism of lymphocyte activations
1. Lymphocytes clone with diverse receptors arise in generative lymphoid organs
2. Clones of mature lymphocytes specific for many antigens enter lymphoid tissues
3. antigen–specific clones are activated/selected by antigens
4. antigen specific immune responses occur
[Intro to Immunology]

Given, clonal selection, why do we still have infections?
– The initial attack that bypasses the primary defense will trigger the adaptive the second defenses.
– These need to initially learn to identify the foreign substance which take time (1–3 wks).
– Subsequent invasions will trigger a 2–3 day response instead due to memory cells
[Intro to Immunology]

Define immunologic memory
long–lived lymphocyte clones that derive from contact with the antigen
virus inhaled/inoculated > innate/barrier defenses > virus attaches to cell > innate cellular defenses > "foreign" antigen presented to immune system > activation of naive lymphocyte > expansion & differentiation of lymphocytes > lymphocyte effector function > Memory
[Intro to Immunology]

Define specificities
ensures that distinct antigens elicit responses that target those antigens
[Intro to Immunology]

Define Diversity
enables immune systems to respond to a large variety of antigens
[Intro to Immunology]

Define Memory
leads to rapid & enhanced responses to repeated exposures to the same antigens
[Intro to Immunology]

Define Clonal Expansion
increases the # of antigen–specific lymphocytes to keep pace with microbes
[Intro to Immunology]

Define Specialization
generates responses that are optimal for defense against different types of microbes
[Intro to Immunology]

Define Contraction & Homeostasis
allows immune systems to respond to newly encountered antigens
[Intro to Immunology]

Define Non–reactivity to self
prevents injury to the host during response to foreign antigens
[Intro to Immunology]

Describe the difference between (graph) the primary & secondary immunologic response (think vaccination)
//fce–study.netdna–ssl.com/2/images/upload–flashcards/32/03/12/6320312_m.jpg
[Intro to Immunology]

Describe the purpose of a vaccine

[think memory cells]
vaccination is the induction of a primary response to an organism in order to allow the creation of Memory B cells, so that when you encounter the actual disease, it induces the faster, secondary response.
[Intro to Immunology]

Compare & contrast antigen & antigenic determinant (epitope)
– an antigen is an agent of infection/disease

– the epitope is the piece of the antigen that the lymphocytes' receptors recognize
[Components of the Immune System]

What are the primary immune organs
where immune cells arise & mature

Thymus

Bone Marrow
[Components of the Immune System]

What are the secondary immune organs
Where immune cells get together to initiate adaptive immunity / released in an immune response

Lymph nodes, spleen, Waldeyer's Ring, lymphoid tissue, Peyer's Patch, MALT, Appendix
[Components of the Immune System]

Describe the thymus' involvement with T–Cells
Cortex – residence of immature T–cells

Medulla – residence of mature T–cells with forming/displayed receptors. ~5% survive & are released
[Components of the Immune System]

Describe the Bone marrow's involvement with lymphocytes
Formation of lymphocytes from stem cells

B–cells > create & mature in bone marrow into granulocytes & monocytes

T–cells > created & relocated to the thymus (thymocytes)
[Components of the Immune System]

What are the two parts to the Waldeyer's Ring?
Tonsils & adenoids
[Components of the Immune System]

Describe the shape & function of the lymph nodes
– bean–shaped, encapsulated, vascularized, secondary lymphoid organs clustered in groups around lymphatic channels.
– B–cells located mostly in the primary lymphoid follicles
– T–cells located mostly in the paracortical area
[Components of the Immune System]

Describe the Spleen in regards to the lymph system
–secondary lymphoid organ

–white–pulp surround arteries & in follicles; blood is filtered & antigens are presented to the lymphocytes (mostly B–cells)
[Components of the Immune System]

Describe the MALT
Mucosal–Associated Lymphoid Tissue (MALT)
– lymphoid cells & follicles in mucus membranes
– Respiratory, GI, urogenital
[Components of the Immune System]

Compare & contrast primary & secondary lymphoid Follicles
– A lymph follicle is a dense collection of lymphocytes, the number, size and configuration of which change in accordance with the functional state of the lymph node.
– primary – spherical aggregates of B–lymphocytes, both virgin & memory B–cells in the process of entering or leaving the node
– secondary – spherical aggregates of B–lymphocytes, both virgin & memory B–cells in the process of entering/leaving the node, Form only in response to an antigenic challenge. Germinal Centers – pale staining center where memory B cells & plasma cells are formed
[Components of the Immune System]

List the four classes of cells of the immune system
Lymphocytes
Antigen–presenting cells
Inflammatory Cells
Bridge Innate & Adaptive
[Components of the Immune System]

Describe the general function of the lymphocytes

[under 4 classes of cells associated with the immune system]
T–lymphocytes – regulators, helpers (for a proper B–cell antibody response, killers
B–lymphocytes – differentiate after encountering antigens into either a plasma cell or a memory cell
[Components of the Immune System]

Describe MHCs
Major Histocompatibility Complex – display peptides for T–cell activation, are responsible for self / non–self distinctions
[Components of the Immune System]

Describe CD molecules
a nomenclature system for cell surface molecules

CD4
CD8
CD21
[Components of the Immune System]

What is the primary function of CD4, CD8, & CD21
CD4 – Helper T–cell – Adhesion
CD8 – Killer T–Cell – Activation
CD21 – B–Cell – Activation
[Components of the Immune System]

What are the antigen–presenting cells & why are they needed

[under 4 classes of cells associated with the immune system]
Dendritic Cells, Macrophages, B–Lymphocytes

T–cells need antigens presented on surface of another cell
[Components of the Immune System]

What are the types of Inflammatory Cells & their purpose

[under 4 classes of cells associated with the immune system]
Neutrophils – 40–60% of blood leukocytes –> phagocytize
Eosinophils – 1–3%, combat parasitic worms
Basophils – 0–1%, mediate allergic rxs
Mast Cells – ––– mediate allergic reactions
others: epithelial, endothelial, platelets, lymphocytes, monocytes
[Components of the Immune System]

What are the cell types that bridge the innate & adaptive systems

[under 4 classes of cells associated with the immune system]
NK – Natural killer cell – kills tumors & virus infected cells
NKT – natural killer T–cell – recognize glycolipids
gama–delta T–cells – recognize a range of molecular patterns
[Components of the Immune System]

Describe the general role of the MHC/HLA genes & proteins in the immune system

displays peptides for the T–cell activation & are responsible for self / non–self distinctions

HLA = Human version of MHC

[Components of the Immune System]

Describe the term complement with regards to an immune response
a system of plasma proteins & surface molecules that facilitate inflammation & can destroy pathogens
[Components of the Immune System]

Describe cytokines with regards to an immune response
Intracellular communication molecules
[Components of the Immune System]

Describe Immunoglobulin with regards to an immune response
Immunoglobulins = antibody – antigen specific receptors on B–cell surface or secreted
–antigen binding, neutralization of microbial toxins, inhibiting attachment of microbes, opsonization, activate the protein–complement systems, protection of developing fetus

[Components of the Immune System]

Define opsonization

facilitating phagocytosis = bind to surface of MOs, help phagocytic cell to phagocytize

[Fates of a Cell]

1. Define Pathology & its significance in medicine

Understanding the changes in cells helps us to recognize the disease process, to explain why patients present with certain symptoms to accurately diagnose normal & abnormal conditions in patients, & to adequately care for patients on a daily basis

[Fates of a Cell]

2. Describe cellular adaptations & their relations to disease processes
cellular adaptations adjust to new conditions/demands for optimal functioning, reversible

affect size/number/phenotype/metabolic activity and/or functions of cells
[Fates of a Cell]

2. Define Hypertrophy
increase in the size of the cells

cause an increase in organ size
[Fates of a Cell]

2. Define Hyperplasia
Increased # of cells in tissue & organs

cause an increase in organ size

uncontrolled hyperplasia ~ cancer
[Fates of a Cell]

2. Define Atrophy
decrease in cell size = decreased organ size
[Fates of a Cell]

2. Define Ischemic Atrophy
loss of oxygenated blood supply

always accompanies Hypoxia
[Fates of a Cell]

2. Define Hypoxia
lack of oxygen
[Fates of a Cell]

2. Define Nutritional Atrophy
– less available nutrients than metabolically necessary = cell shrinkage/death

causes inflammation disease & cancer because of cytokine TNF (tumor necrosis factor)
[Fates of a Cell]

2. Define Metaplasia
one differentiated cell type is replaced by another

Squamous > columnar
Columnar > squamous
[Fates of a Cell]

2. Define Dysplasia
disordered growth
sometimes in metaplastic epithelium
[Fates of a Cell]

2. Carcinoma in Situ means?
dysplasia but the basement membrane is intact = non–malignant
[Fates of a Cell]

3.What are the 5 types of cellular adaptions

(list)
Hypertrophy
Hyperplasia
Atrophy
Metaplasia
Dysplasia
[Fates of a Cell]

3.What are the typical causes of cellular injury?
Oxygen deprivation
Physical Agents
Chemical Agents & drugs
Infectious Agents
Immunologic reactions
Genetic derangements
Nutritional Imbalances
[Fates of a Cell]

3.Cell Injury: describe oxygen deprivation
–ischemia
–cardiopulmonary failure
–Anemia / Carbon Monoxide / cyanide poisoning
–Severe blood loss
[Fates of a Cell]

3.Cell Injury: Describe Chemical Agents & drugs
–electrolyte imbalance
–oxygen toxicity
–toxins, industrial chemicals
–alcohol, recreational drugs, therapeutic drugs
[Fates of a Cell]

4. Describe cell death & the consequences
Necrosis is denaturing of intracellular proteins, enzymatic digestion of lethally injured cells, no membrane integrity
– contents leak out, inflammation results

Apoptosis = programmed cell death, plasma membrane remains intact
[Fates of a Cell]

What are the goals of cellular adaptation that reflect the dynamic ability of the cell to alter.
Gene transcription & translation (g1,g2)
Synthesis of new DNA/chromosomes (S)
Proliferation (M)
[Fates of a Cell]

What are the types of cells that undergo hypertrophy?
* usually in non–dividing cells, no new cells, only increase in the size & # of intracellular components

Cardiac muscle, Skeletal Muscle, Pregnant Uterus
[Fates of a Cell]

Fluff: What are the two biochemical pathways by which a cell will undergo hypertrophy
physiological = Phosphoinositide 3–kinase / Akt

pathological = signaling downstream of GPCRs
[Fates of a Cell]


What are the two stimuli causing cellular hypertophy
increased workload = muscles
increased hormonal stimulation = pregnancy
[Fates of a Cell]

What are the two mechanism/stimuli for hyperplasia?
increased growth factors

increased production of cells from tissue stem cells
[Fates of a Cell]

What are the types of pathologic hyperplasia?
– hormonal = increase the functional capacity (pregnancy)
– compensatory = increase tissue/organ mass after partial resection/damage (liver)
[Fates of a Cell]

What are the types of pathological
hyperplasia?
– excessive hormones & growth factors
endometrial hyperplasia & BPH
– viral infections = HPV
[Fates of a Cell]

What are the two types of hyperplasia
Controlled – no gene mutations, regression of hyperplasia if stimulation is removed

Uncontrolled – genetic aberrations occur, unrestrained proliferation, can become malignant
[Fates of a Cell]

Give examples of physiologic atrophy
embryonic structures – notochord

postpartum uterus
[Fates of a Cell]

Give examples of pathologic atrophy
Atrophy of disuse (use it or lose it)
Denervation atrophy (spinal cord injury)
Ischemic atrophy ( loss of blood)
nutritional atrophy (protein–calorie malnut.)
Endocrine atrophy (menopause)
Pressure atrophy (tourniquet)
[Fates of a Cell]

Define Marasmus & Cachexia
marasmus – profound protein–calorie malnutrition

Cachexia – marked muscle wasting
[Fates of a Cell]

Which atrophy is due to an overproduction of cytokine TNF
nutritional atrophy – cachexia
[Fates of a Cell]

Define cytokine TNF
TNF = tumor necrosis factor
[Fates of a Cell]

What are the mechanisms of atrophy
–Decreased protein synthesis
–increased protein degradation
–increased autophagy
[Fates of a Cell]

Describe autophagy
starved cells eat their own components

–autophagic vacuoles or residual bodies
[Fates of a Cell]

What is known as brown atrophy?
Lipofuscin granules
[Fates of a Cell]

Metaplasia: What is the cell type change in the respiratory tract due to smoking?
Columnar change to Squamous cells to resist physical damage from smoking
[Fates of a Cell]

Metaplasia: What is the cell type change in the esophagus due to GERD
squamous epithelial cells change to columnar in the esophagus to resist chemical damage

[Fates of a Cell]

What is barrets esophagus?

the change in the esophagus from squamous cell to columnar to resist chemical damage

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

[ANS Pharmacology]

Review the Neuroanatomy of the sympathetic neurons
spinal cord
pre–ganglionic neuron
ACh to N–receptor @ synapse/ganglia
post–ganglionic neuron
target tissue
*sympathetic ganglion is "typically" farther from the target tissue than the parasympathetic ganglia
[ANS Pharmacology]

Tyrosine is changed by Tyrosine Hydroxylase to becomes what drug inside the Nerve Terminal
Dopa
[ANS Pharmacology]

Dopa is changed by what to becomes Dopamine inside the Nerve Terminal
Dopa decarboxylase
[ANS Pharmacology]

Dopamine is changed to Epinephrine via what enzyme?
Dopamine Beta–Hydroxylase
[ANS Pharmacology]

Norepinephrine is changed to Epinephrine by what enzyme?
phenylethanolamine–N–methyltransferase
[ANS Pharmacology]

Describe the release & signalling of neurotransmitter from cholinergic & adrenergic neurons
– an action potential causes the release of Ca2+ from the axon bulb so that vesicles with the neurotransmitter can go through exocytosis
–the exocytosis causes the release of the neurotransmitter into the synaptic cleft & then the neurotransmitter binds to receptor sites on the post–synaptic area
[ANS Pharmacology]

Differentiate between the direct–acting & indirect–acting cholinomimetics
–Direct–acting are Choline Esters that stimulate the same receptors that ACh itself would without any intermediates (RISK: will stimulate both PSNS & SNS)
–Indirect–acting are either reversible or irreversible cholinomimetics that inhibit acetylcholinesterase
[ANS Pharmacology]

Explain the aging process of organophosphates
– organophosphates covalently bind via phosphate group to the serine–OH group @ active site
–permanently inactivates enzyme
–restoration requires new enzyme molecules
–slow release of ethyl group (aging) makes it impossible for 2–PAM to break the bond of drug–enzyme
[ANS Pharmacology]

Explain the effect after Muscarinic & Nicotinic Receptor activation & blockade for:

Secretory
stimulation of the thermo–regulatory sweat
lacrimal & nasopharyngeal glands

Blockade – suppressed thermo–regulatory sweating "atropine fever"
[ANS Pharmacology]

Explain the effect after Muscarinic & Nicotinic Receptor activation & blockade for:

Gastrointestinal
activation – increased secretory & motor activity, salivary & gastric glands are stimulated, peristaltic activity is increased, sphincters are relaxed

Blockade – reduced motility & secretion
[ANS Pharmacology]

Explain the effect after Muscarinic & Nicotinic Receptor activation & blockade for:

Genitourinary
activation – stimulation of detrusor muscle & relaxation of the trigone & sphincter muscles of the bladder

Blockade – relaxes smooth muscle of the ureters & bladder walls, slow voiding
[ANS Pharmacology]

Explain the effect after Muscarinic & Nicotinic Receptor activation & blockade for:

Respiratory
activation – contracts the smooth muscle of the bronchial tree,

Blockade – ?
[ANS Pharmacology]

Explain the effect after Muscarinic & Nicotinic Receptor activation & blockade for:

Cardiovascular
activation – reduction in peripheral vascular resistance & change in the heart rate & contractility

blockade – SA & AV node sensitive to blockade, vasodilation blocked in the blood vessels
[ANS Pharmacology]

Explain the effect after Muscarinic & Nicotinic Receptor activation & blockade for:

Central Nervous
activation – regulation of the neurotransmitter release

blockade – sedative effects
[ANS Pharmacology]

Explain the effect after Muscarinic & Nicotinic Receptor activation & blockade for:

Peripheral Nervous
activation – initial response is simultaneous activation of PANS & SANS

blockade – ?
[ANS Pharmacology]

Explain the effect after Muscarinic & Nicotinic Receptor activation & blockade for:

Neuromuscular Junction
activation – contraction

blockade –
[ANS Pharmacology]

Identify the major adverse effects of cholinergic agents
excessive activation of the cholinergic system

DUMBBELSS: Diarrhea, Urination, Miosis, Bronchoconstriction, Bradycardia, Emesis, Lacrimation, Salivation, Sweating
[ANS Pharmacology]

Explain why anti–nicotinics are rarely used
blocking N–receptors will block the entire ANS
[ANS Pharmacology]

List the common side effects of Anti–Muscarinic Agents (Anti–DUMBBELSS)
Opposite of DUMBBELSS

Constipation, No Urination, Mydriasis, Bronchodilation, Tachycardia, No Emesis, No Tearing, Dry Mouth, No Sweat
[ANS Pharmacology]

Describe the major effects of alpha–1 receptors
–Vasoconstriction
–Increased peripheral resistance
–Increased blood pressure
–mydriasis
–increased closure of internal sphincter of the bladder
[ANS Pharmacology]

Describe the major effects of alpha–2 receptors
–inhibition of norepinephrine release
–inhibition of ACh release
–inhibition of insulin release
[ANS Pharmacology]

Describe the major effects of Beta–1 receptors
–Tachycardia
–Increased lipolysis
–increased myocardial contractility
[ANS Pharmacology]

Describe the major effects of Beta–2 receptors
–vasodilation
–slightly decreased peripheral resistance
–bronchodilation
–increased muscle & liver glycogenolysis
–increased release of glucagon
–relaxed uterine smooth muscle
[ANS Pharmacology]

Identify the effects of the sympathomimetic agonists & antagonists for:

Genitourinary System
alpha agonists can promote urinary continence
[ANS Pharmacology]

Identify the effects of the sympathomimetic agonists & antagonists for:

Cardiovascular System
a1 receptor – activation leads to arterial & venoconstriction
a2 receptor – activation leads to vasoconstriction
B receptors – Heart (B1)–stimulation increases cardiac output, Vascular Beds (B2) decrease peripheral resistance = vasodilation
[ANS Pharmacology]

Identify the effects of the sympathomimetic agonists & antagonists for:

Respiratory System
B2 receptors in bronchiole smooth muscle causes bronchodilation
[ANS Pharmacology]

Identify the effects of the sympathomimetic agonists & antagonists for:

Central Nervous System
depends on ability to cross BBB

effects vary from mild alerting to full block psychosis
[ANS Pharmacology]

Identify the effects of the sympathomimetic agonists & antagonists for:

Apocrine Sweat System
adrenoceptor stimulation leads to increased sweat production
[ANS Pharmacology]

Identify the effects of the sympathomimetic agonists & antagonists for:

Eye System
alpha agonists can reduce intraocular pressure & cause mydriasis
[ANS Pharmacology]

Identify the effects of the sympathomimetic agonists & antagonists for:

Metabolism System
activation of beta receptors can lead to lipolysis

can enhance glycogenolysis in the liver

regulation of insulin secretion
[ANS Pharmacology]

Identify the adverse drug reactions of alpha & beta receptor blockade
postural hypotension

reflex tachycardia

beta receptor blockade
[ANS Pharmacology]

Describe the mechanism of postural hypotension via a/b receptor blockade
due to antagonism of sympathetic nervous system stimulation of alpha–1 receptors in venous smooth muscle that causes less contraction = vasodilation = lower blood pressure

(often seen after the first dose of medication)
[ANS Pharmacology]

Describe the mechanism of Reflex Tachycardia via a/b receptor blockade
result of exposure to nonselective alpha antagonists when blood pressure is lower than normal

in the heart, the release of norepinephrine activates primarily Beta receptors as a result of agents that block
[ANS Pharmacology]

Describe the mechanism of Beta Receptor Blockade via a/b receptor blockade
depresses myocardial contractility & excitability, causes cardiac decompensation, severe hypotension, bradycardia, heart failure, fatigue/exercise intolerance
[Autonomics of Select Organs]

What sympathetic receptor type causes smooth muscle contraction in the Eye
alpha–1 adrenergic receptor
[Autonomics of Select Organs]

What sympathetic receptor type causes smooth muscle relaxation in the eye?
Beta–2 adrenergic receptor
[Autonomics of Select Organs]

What parasympathetic receptor type causes smooth muscle contraction
M Receptor
[Autonomics of Select Organs]

Describe how the action of the Ciliary Muscle affects the lens
Relaxed = taut suspensory ligaments = flat lens = far vision

Contracted = loosened suspensory ligaments = rounded lens = near vision
[Autonomics of Select Organs]

What nerve, receptor, transmitter is responsible for relaxing the Ciliary muscle in order to enhance far vision
Sympathetic

Beta–2 Receptor

Epinephrine
[Autonomics of Select Organs]

What nerve, receptor, transmitter is responsible for contracting dilator pupillae muscle and thus dilating the pupil in order to enhance far vision
Sympathetic

alpha–1 Receptor

Norepinephrine
[Autonomics of Select Organs]

What nerve, receptor, transmitter is responsible for contracting the sphincter pupillae muscle and thus constricting the pupil
Parasympathetic

M Receptor

Acetylcholine
[Autonomics of Select Organs]

Describe the s/sx of Horner's syndrome
pupil constricts in one eye
superior eyelid droops
dilation of ipsolateral blood vessels
No sweating on ipsolateral side
[Autonomics of Select Organs]

Horner's Syndrome

Explain why sympathetic nerve damage causes the pupil in one eye to constrict
lack of the sympathetic dilation mechanism via the b–1 adrenergic receptor on the dilator pupillae smooth muscle
[Autonomics of Select Organs]

Horner's Syndrome

Explain why sympathetic nerve damage causes the dilation of ipsolateral blood vessels
the sympathetic nerves are responsible for the vasoconstriction via alpha–adrenergic receptors
[Autonomics of Select Organs]

Horner's Syndrome

Explain why there is a lack of sweating on the ipsolateral side
The sympathetic nerves control sweat glands (other than the palms) via M–Receptors & Acetylcholine
[Autonomics of Select Organs]

Horner's Syndrome

Explain why the ipsolateral eyelid droops
the sympathetic system controls the positioning of the eyelid via contraction

(alpha–1 receptors)
[Autonomics of Select Organs]

Describe the sequence of nerve stimulation that results in bronchodilation
Bronchioles with Beta–2 receptors

Albuterol | Epinephrine–like agonist

Bronchial dilation
[Autonomics of Select Organs]

Describe the sequence of nerve stimulation that results in bronchoconstriction
Bronchial M–Receptors

Parasympathetic Nerve + Ach neurotransmitter

Bronchoconstriction
[Autonomics of Select Organs]

Explain one of the risks of administering too much B2 receptor agonist
B2 receptors will hide if exposed to too much agonist
[Autonomics of Select Organs]

What are the basic roles of the SNS and PSNS in urinary tract function
SNS– helps with filling, PSNS– helps with emptying
[Autonomics of Select Organs]

List the target tissues, receptors and actions involved in filling and emptying the bladder
SNS relax the detrusor muscle which is activated by B2 receptors, contract the trigone and internal sphincter which is activated by A1 receptors. PSNS contract the detrusor muscle which activate M receptors, and relax the trigone and internal sphincter which activate M receptors.
[Autonomics of Select Organs]

How do we prevent or initiate micturition
Spinal reflex– stretch receptors in the urinary bladder send afferent message to the spinal cord at sacral level, parasympathetic NS sends efferent message to contract the detrusor muscle, and relax the trigone and internal sphincter. Spinal reflex with superimposed higher brain center efferents use the Cortex and Brainstem to determine when it is socially acceptable to urinate.
[Autonomics of Select Organs]

Define the role of the SNS for sweating
The SNS stimulates the release of acetylcholine via sympathetic cholinergic neurons which bind to M receptors and cause all other sweat glands other than those in the palms of the hands to produce sweat.
[Autonomics of Select Organs]

List the ANS function, transmitter and receptors for the rate and contractility of the heart
The sympathetic system will stimulate the SA Node, AV Node, and Atria and ventricles, causing an increase in heart rate, conduction through the node, and contractility, respectfully. The sympathetic receptor for the heart is the β1, the transmitter NE
[Autonomics of Select Organs]

Describe how the ANS alters blood flow during exercise
Blood pressure is a combination of cardiac output and peripheral vascular resistance. Essentially no parasympathetic innervation of blood vessels. Sympathetics include α1 receptors for vasoconstriction (rest of blood vessels) and β2 receptors for vasodilation (blood vessels to liver/skeletal muscle/coronary/cerebral).
[Autonomics of Select Organs]

List the ANS receptor sites and resulting function on GI sphincters and smooth muscle along the intestine
Parasympathetic relax sphincter and increase tone and motility by the binding of Acetylcholine to M receptors. Sympathetic contract sphincter and decrease tone and motility by the binding of Norephinerine to A1, A2 and B2
[OPP]

Describe the osteopathic approach to patient care
The osteopathic approach has a patient–centered focus, and it optimized health using standard and unique skills, including hands–on manual diagnosis and treatment. OMT is indicated when a physician makes the diagnosis of somatic dysfunction AND relates it to a patient’s problem. It may also include pharmacological agents, exercise, nutritional counseling, surgical procedures, or lifestyle modifications.
[OPP]

List the 5 components of the 5 Model approach
Biomechanical
Respiratory–Circulatory
Metabolic
Neurologic
Behavioral
[OPP]

Describe the Bioimechanical approach & goals
views the patient from a structural or mechanical perspective. The objective is to optimize the patient’s adaptive potential through restoration of structural integrity and function. The goal of this model is biomechanical adjustment and mobilization of joints.
[OPP]

Describe the Respiratory–Circulatory approach & goals
focused on the respiratory and circulatory components of the homeostatic response in pathophysiological processes. This model concerns itself with the maintenance of extra– and intracellular environments through the unimpeded delivery of oxygen and nutrients and the removal of cellular waste products. The goal of this model is to improve all of the diaphragm restrictions in the body.
Think: arthritic knee, no direct OMT, instead, improve R–C flow
[OPP]

Describe the Metabolic approach & goals
focused on energy conservation and efficiency of metabolic functions. The goal is to enhance the self–regulatory and self–healing mechanisms while fostering energy conservation by balancing the body’s energy expenditure and enhance immune system, endocrine, and organ functions
[OPP]

Describe the Neurologic approach & goals
This model views the patient’s problems in terms of aberrancies or impairments of neural function. The goal is to attain autonomic balance and address neural reflex activity, remove facilitated segments, decrease afferent nerve signals, and relieve pain
[OPP]

Describe the Behavioral approach & goals
This model recognizes that the assessment of a patient’s health includes assessing his or her mental, emotional, and spiritual state of being as well as person lifestyle choices.
[OPP]

Explain how these 5 coordinated body functions are interrelated & interdependent
Case study: Otitis Media
Biomechanical: blocked eustachian tube
R–C: decreased lymphatic & vascular flow to inflamed area
Metabolic: infected middle ear = impaired immune system
Neurologic: Pain, pressure on nerves
Behavioral: altered mental state due to OM
[OPP]

Describe the relationship of the musculoskeletal system to the 5 models
provide the framework within which all other systems reside

represents the entry–point for both dx & tx

often reflects or gives clues as to what is occurring internally
[OPP]

Explain how the osteopathic manipulative treatment affects physiological function & restores health:

Biomechanical
OMT can positively affect physiological functions such as coordination and symmetry of musculoskeletal and myofascial structures in posture and motion, and works to improve quality and quantity of motion
[OPP]

Explain how the osteopathic manipulative treatment affects physiological function & restores health:

Respiratory–Circulatory
OMT works to increase/improve respiration, circulation, venous, and lymphatic drainage
[OPP]

Explain how the osteopathic manipulative treatment affects physiological function & restores health:

Metabolic
Tx (although not manipulative in nature) works to maintain metabolic processes, homeostasis, energy balance, regulatory processes, immunology, digestion, waste removal, etc.

e.g. nutritional aspects: zinc, selenium, Vit D, iron deficiencies, formula vs. breast milk
[OPP]

Explain how the osteopathic manipulative treatment affects physiological function & restores health:

Neurological
Treatment affects focuses on the reduction of mechanical stresses, balance of neural inputs, and the elimination of nociceptive drive
[OPP] Define Nociceptive Drive
of, relating to, or denoting pain arising from the stimulation of nerve cells (often as distinct from that arising from damage or disease in the nerves themselves).
[OPP]

Explain how the osteopathic manipulative treatment affects physiological function & restores health:

Behavioral
OMT is employed within this model with the goal of improving the body’s ability to effectively manage, compensate, or adapt to certain stressors.
[OPP]

Describe the anatomical regions most commonly associated with:

Biomechanical
bones, muscles, postural muscles, spine, extremities, etc
[OPP]

Describe the anatomical regions most commonly associated with:

Respiratory–Circulatory
thoracic inlet, thoracic and pelvic diaphragms, Tenotium cerebelli, costal cage
[OPP]

Describe the anatomical regions most commonly associated with:

Metabolic
internal organs, endocrine glands
[OPP]

Describe the anatomical regions most commonly associated with:

Neurological
head, brain, spinal cord, autonomic nervous system, peripheral nerves
[OPP]

Describe the anatomical regions most commonly associated with:

Behavioral
Brain
[ANS]

Name the two components of the ANS
Sympathetic

Parasympathetic
[ANS]

Describe the Sympathetic Nervous System's purpose
Fight or Flight

increase or dilate
control peripheral blood vessels
glycogenolysis
[ANS]

Describe the Parasympathetic Nervous System's purpose
Rest & Digest

decrease or constrictg
[ANS]

List the names of the neurons & synapses that make up the ANS
preganglionic | presynaptic
postganglionic | postsynaptic

Synapse | ganglion

Target tissue
[ANS]

List the function of each of the Sympathetic target tissues
Smooth Muscle of all organs
Cardiac Muscle
Glandular cells [sweat, salivary, digestive,
[ANS]

List the function of each of the Parasmpathetic target tissues
Smooth Muscle
Glands – GI & Excretory Organs, genitalia, lungs,
Atria
tear & salivary glands
eye muscles
[ANS]

Describe the anatomical Origin & route of the parasympathetic innervation to each target tissue
long presynaptic neuron in which there is a short postsynaptic neuron. The synapse is within or very close to the target organ
[ANS]

Describe the anatomical Origin & route of the sympathetic innervation to each target tissue
short presynaptic neuron from the spinal cord then to the sympathetic chain out to the body
[ANS–2]

Describe the anatomical organization of the spinal nerve & Sympathetic Nervous System
Anterior Root & Posterior Root for a specific segment of the body

Spinal cord gray matter (cell bodies) consist of a:
Posterior horn to which the sensory neuron enter
Anterior Horn to which the motor neuron originate

Sympathetic nerves enter sympathetic chain for synapse/ganglia
[ANS–2]

What is segmentation in the nervous system
Each spinal nerve innervates a different section of the body
[ANS–2]

Sympathetic Somatic Nervous System Segmentation
Each segment consists:
Myotomes: of a group of muscles
Schlerotomes: a group of connective tissues
Dermatomes: a patch of skin
[ANS–2]

What vertebral region enervates:

cardiovascular
T1–5
[ANS–2]

What vertebral region enervates:

Respiratory
T2–7

lungs extend slightly lower than the Heart
[ANS–2]

What vertebral region enervates:

Stomach, Liver, Gall Bladder
T5–9
[ANS–2]

What vertebral region enervates:

Pancreas
T5–11
[ANS–2]

What vertebral region enervates:

Small Intestines
T9–11
[ANS–2]

What vertebral region enervates:

Ovaries & Testicles
T9–11
[ANS–2]

What vertebral region enervates:

Kidneys & Ureters
T10–L1
[ANS–2]

What vertebral region enervates:

Urinary Bladder
T10–L1
[ANS–2]

What vertebral region enervates:

Large Intestines, Rectum
T8–L2
[ANS–2]

What vertebral region enervates:

Uterus
T10–L1
[ANS–2]

What vertebral region enervates:

Prostate
L1–2
[ANS–2]

Describe the Somato–Somatic Spinal Reflex
Somatic sensory nerve is stimulated, afferent signal enters spinal cord, stimulates interneuron, interneuron stimulates presynaptic motor neuron, efferent signal exits spinal cord, reaches synapse, signals postsynaptic motor neuron, postsynaptic motor neuron signals effect in somatic tissue.
[ANS–2]

Describe the Viscero–Visceral Spinal Reflex
Visceral sensory nerve is stimulated, afferent signal enters spinal cord, stimulates interneuron, interneuron stimulates presynaptic motor neuron, efferent signal exits spinal cord, reaches synapse, signals postsynaptic motor neuron, postsynaptic motor neuron signals effect in visceral tissue.
[ANS–2]

What is the role of the Interneuron
//fce–study.netdna–ssl.com/2/images/upload–flashcards/19/24/13/6192413_m.png
[ANS–2]

Describe spinal facilitation's relationship to somatic dysfunction
Abnormal Reflexes:
Afferent Drive: Problem in tissue causes increased sensory input to spinal cord, which causes Facilitated Spinal Cord Segments: the properties of the other neurons in that segment change, accepting that signal as if it was meant for them, causing abnormal reflexes (somatic –> visceral or visceral –> somatic), which causes somatic dysfunction.
[ANS–2]

What are the clinical aspects of facilitated segments
Knowing which segments of nerves control which types of tissues allows a physician to trace the somatic dysfunction to its origin, and allow them to know which other body systems will be affected by an ailment and/or injury.
Define pharmacokinetics
What the body does to the drug.

e.g.
Distribution to peripheral tissues
excretion
site of action
Define pharmacodynamics
What the drug does to the body:
interaction with receptor
stimulus
effect
[Microanatomy]
List the common Histological Dyes
Hematoxylin
Eosin
Orcein
Silver
PAS – periodic acid–Schiff
[Microanatomy]

What is the color will Hematoxylin show up as?
blue <––> violet
[Microanatomy]

What is the color will Eosin show up as?
red <––> Pink
[Microanatomy]
Define PAS
Periodic acid–Schiff
[Microanatomy]

What color does PAS show as?
Magenta/Purple
[Microanatomy]

What structures are dyed by Hematoxylin
Nucleus
Basophilic regions of cytoplasm
Cartilage Matrix
[Microanatomy]

What will Hematoxylin dye?

Acid or Base
Basic structures
[Microanatomy]

What will Eosin dye?

Acid or Base
Acidic structures
[microanatomy]

What is the target structure of Orcein's elastic stain?
Brown: Elastic fibers
[microanatomy]

slide: describe the appearance of the nucleus
H&E will appear blue
[microanatomy]

slide: describe the appearance of the nucleolus
H&E will appear blue
appears w rRNA – protein formation
dark spot at center of the nucleus
[microanatomy]

slide: describe the appearance of the nucleolus with golgi
H&E will appear blue
dark spot at center of the nucleus
golgi: shipping new proteins out of the cell
[microanatomy]

slide: describe the appearance of the nucleolus without golgi
H&E will appear blue
dark spot at center of the nucleus
No golgi: proteins being made for use inside the cell
[microanatomy]

slide: describe the appearance of the Golgi Apparatus
H&E will appear as a clearing in the slide next to the nucleus
[microanatomy]

slide: describe the appearance of the secretory vesicles
H&E can be blue or pink

(only when very large)
[microanatomy]

slide: describe the appearance of the cytoskeletal elements
H&E will appear pink
[microanatomy]

slide: describe the appearance of the Lipid Droplets
H&E will appear as an empty hole
[microanatomy]

List the various levels of structural organization in the human body
chemical
cellular
tissue
organ
organ system
organism
[microanatomy]

Describe the chemical structural level & provide examples
histochemistry, cytochemistry

lipids, proteins
[microanatomy]

Describe the cellular structural level & provide examples
micro–anatomy
smallest living structure

fibrinoblast, keratinocyte, osteocyte, motor neuron, myocyte
[microanatomy]

Describe the tissue structural level & provide examples
microanatomy

Connective
Epithelial
Muscular
Nervous
[microanatomy]

Describe the Organ structural level & provide examples
gross anatomy

multiple tissue types working together
[microanatomy]

Describe the Organ system structural level & provide examples
gross anatomy

multiple organs

Digestive, skeletal, muscular, nervous, etc
[microanatomy]

Describe the organism structural level & provide examples
gross anatomy

multiple systems working together (osteopathic medicine)
[microanatomy]

Describe the characteristics of the Epithelium tissue type [5 items]
derived from all three germ layers
densely packed w little extracellular materials
always sits on a basement membrane
forms sheets that cover/line surfaces
form secretory glands
[microanatomy]

Describe the characteristics of the Connective tissue type
derived from mesoderm
mainly composed of extra–cellular elements, limited # of cells
[microanatomy]

Describe the differences between Embryonic, Adult, and specialized connective tissue
Embryonic: mesenchymal, mucous

Adult: connective tissue proper

Specialized: cartilage, bone, blood
[microanatomy]

Describe the characteristics of the Muscle tissue type
derived from mesoderm
contain contractile elements – myofilaments
possess many mitochondria – energy prod.
visibly longer than wide
[microanatomy]

Describe the characteristics of the Nervous tissue type
derived from ectoderm
[microanatomy]

What is the name of the supporting cell for Neurons?

What is their function?
Neuroglial cells

assist with metabolism & support the neurons
[microanatomy]

What tissue type is derived from Ectoderm
Nervous Tissue
[microanatomy]

What tissue type is derived from mesoderm
Connective & Muscle
[microanatomy]

What tissue type has many mitochondria
Muscle tissue
[microanatomy]

Name the three germ layers
ectoderm, mesoderm, and endoderm
[microanatomy]

What tissue type is derived from all three germ layers?
Epithelium
[microanatomy]

How can you recognize a slide stained with Orcein?
Brown tint

stains Elastic fibers
[microanatomy]

What tissue does the Orcein's stain?
Elastic fibers
[microanatomy]

On slides with H&E, what component picks up the dark blue stain
epithelium tissue
[Action Potentials]

Describe the term Excitability
ability of the cell to generate an action potential

able to send information electrically
[Action Potentials]

list the types of cells that are excitable
nerve cells
cardiac myocytes
skeletal muscle cells
some smooth muscle cells (e.g. GI)
[Action Potentials]

Explain membrane potential involvement in driving insulin secretion
high glucose inside Beta–islet cells in pancreas force an increased production of ATP
High [ATP] causes the membrane K+ channels to be blocked
this reduction in membrane K+ permeability causes the Em to more away from the NP for K+ and become more positive.
This depolarization opens Ca2+ channels, allowing Ca2+ into the cell which triggers the release of insulin
[Action Potentials]

What channels are blocked and which are opened in the process of releasing insulin from the Beta–Islet cells in the pancreas
K+ channels blocked due to high [ATP]
Ca2+ channels opened due to increased + Em
[Action Potentials]

What type of gates close in the Beta Islet pancreatic cells when glucose concentration is low?
low [ATP]
K+ channels unblocked,
Em moves to NP for K+
Em less positive = Ca2+ voltage–gated ion channels close
No insulin secretion
[Action Potentials]

What types of channels control the release of Ca2+ in Beta–Islet pancreatic cells
Ca2+ voltage–gated ion channels
[Action Potentials]

Describe how the membrane potential changes during the action potential
starting Em: ~ –70 mV
ending Em: ~+30 mV

net change: ~100 mV
[Action Potentials]

Describe the 4 types of stimuli that can initiate an action potential
electrical – cardiac
mechanical – sensory receptors
thermal
chemical – neurotransmitter
[Action Potentials]

What are the stages of an action potential?
Depolarizing phase
Plateau (if cardiac)
Repolarizing phase
Hyperpolarizing phase
[Action Potentials]

Define Action potential
transient reversal of membrane potential (Em)
[Action Potentials]

Define threshold
minimum membrane potential required to produce an action potential
[Action Potentials]

Describe the depolarizing phase
rapid upstroke that is a response to a stimulus that is sufficient (–50mV) to pass the threshold causing a change in the membrane potential (Em) from –70mV to +30mV
[Action Potentials]

Describe the Action Potential Duration
length of time between reaching threshold to the return to Resting Membrane Potential
[Action Potentials]

Describe the repolarizing phase
process by which the cell membrane returns from +30 mV to the Resting Membrane potential at ~ –70 mV
[Action Potentials]

Describe the hyperpolarizing phase
the process by which the cell, during the process of returning to Resting Membrane Potential becomes more negative via the influx of extra K+ ions for a short period before it returns to ~ –70 mV
[Action Potentials]

Describe the differences in action potentials in various tissues
Neuronal action potentials: 1–2 ms
ventricular action potentials: 300 ms

~300 times longer
partly due to plateau phase
[Action Potentials]

What is one of the contributing factors in the differing lengths of repolarizations
ion permeability
[Action Potentials]

Define "all or none"
All: threshold Em met, full action potential produced.

None: threshold Em not met, no membrane response
[Action Potentials]

Describe the amplitude of the action potential
constant amplitude regardless of the strength of the stimulus
[Action Potentials]

Describe the behavior of the amplitude of an action potential as it is conducted along an excited cell membrane
the action potential is conducted along an excited cell with an amplitude that does not change with distance from the initial trigger
[Action Potentials]

Explain Refractoriness
refers to the period in which another action potential cannot be triggered during an action potential

cellular need to recover before further action
[Action Potentials]

Compare absolute vs. relative refractory periods
Absolute: action potential is not possible regardless of the strength of the stimulus

Relative: the period during repolarization where the cell can trigger another action potential if the stimulus is large enough
[Action Potentials]

Explain the membrane potential changes during an action potential
Stimulus
membrane more permeable to Na+
drives Em to E(Na) ––> more positive –> depol.
membrane permeable to K+ increases
Em moves to Ek ––> more negative –> Repol.
[Action Potentials]

explain the concept of driving force
difference between the Nernst potential for the ions involved & the membrane potential

NP = E(ion) – Em
far from NP for specific ion, the driving force is large
near NP(ion), driving force is low
[Action Potentials]

define conductance
flow of ions across a membrane

conductance = 1 / Resistance
[Action Potentials]

Explain the role of Na+ in the action potential
depolarization is brought about by a rapid activation of a Na+ current

As the Em approaches the NP(Na+), the driving force decreases, Na+ influx slows near the peak of the action potential then decreases as the gates are inactivated
[Action Potentials]

Explain the role of K+ in the action potential
the increasingly positive Em during depolarization causes an outward flow of K+ ions until at the peak of the action potential the Na+ conductance is equal to the K+ conductance
Repolarization is brought by a reversal where the K+ conductance increases & the Na+ conductance decreases
ultimately returning the state to the original [ion] and Resting membrane potential
[Action Potentials]

In which direction does Na+ flow during the action potential?
Na+ current is Inward: depolarization
[Action Potentials]

In which direction does K+ flow during the action potential?
K+ current is outward

repolarization
hyperpolarization
refractoriness [absolute | relative]
[Action Potentials]

Explain how the ion gradients are restored following an action potential
Na/K ATPase

pumps out Na+ in exchange for K+
[Action Potentials]

Describe how Na+ channels are opened during the action potential
stimulus: small depolarization
Critical Mass: enough Na+ channels must open to cause an influx of Na+ in order to steadily change the Em more positive until threshold is reached ~ 50mV

positive feedback process
[Action Potentials]

What is the Em needed to open Na+ channels
approx. –50 mV
[Action Potentials]

What determines the action potential threshold?
whether there is enough depolarization to overcome a resting K+ efflux
[Action Potentials]

Describe one of the causes of Hypercalcemia
hyperparathyroidism
[Action Potentials]

Describe the results of hypercalcemia on the action potential
hypercalcemia caused by hyperparathyroidism results in a high extracellular [Ca2+] that shields the membrane voltage receptors, increasing the apparent threshold value needed to stimulate a depolarization
[Action Potentials]

What physiological s/sx can show hypercalcemia
muscle weakness

higher threshold due to membrane voltage receptor shielding results in a more difficult to achieve Action Potential
[Action Potentials]

What is one of the causes for hypocalcemia?
hypoparathyroidism
[Action Potentials]

Describe the effect of hypocalcemia on the action potential
low extracellular Ca2+ reduces the shielding effect Ca2+ has on the membrane receptors resulting in less obstruction of the voltage sensors = lower threshold to depolarize
[Action Potentials]

What physiological s/sx can show hypocalcemia
muscle twitching

lower threshold due to less shielding of the membrane voltage receptor results in a quicker depolarization
[Action Potentials]

Explain what happens to the driving force for Na+ ions during the depolarization phase of the action potential
the driving force for sodium ions decreases towards the peak of the action potential as the E(m) gets closer to E(Na)

net driving force is difference between NP for that ion & the E(m), nearer the Em is to E(Na) during upstroke, the smaller the driving force
[Action Potentials]

Explain why the sodium ion current is brief during the action potential
the driving force of sodium ion influx lessens near the peak of the action potential
Sodium ion channel inactivation stops sustained ion influx during the action potential

inactivation is kinetic action only
[Action Potentials]


Describe what is meant by the Na+ channel inactivation
Inactivation gate (IG) begins to close @–55 mV
activation gate (AG) begins to open @–50 mV
IG begins to close at the threshold point while the AG is open fully during the depolarization stage but closes slowly as the Em approaches E(Na).
purely a kinetic action
[Action Potentials]

Be able to distinguish between Na+ inactivation, activation, closing
Activation: Na+ channels open for depolarization due to stimulus
Inactivation: begin to close once threshold is met, limiting the influx of Na+ ions
closing: the Na+ channels are fully closed and inoperable during Membrane Resting Potential without a stimullus
[Action Potentials]

Describe how Na+ channel activation & inactivation vary during the action potential
@ resting: closed
[Action Potentials]

Describe what causes Na+ channel inactivation & be able to predict hyperkalemia on Na+ channel inactivation
Resting Membrane Potential (RMP): –70 mV
Inactivation gates close @ –55 mV
Hyperkalemia: raises RMP to ~ –55 mV
inactivation gates close
No Na+ ions allowed in regardless of stimulus
total blocking of signal
[Action Potentials]

Explain how K+ channels are opened during the action potential
K+ channels open through depolarization allowing a K+ efflux during repolarization causing a return to Resting Membrane Potential
[Action Potentials]

Explain why the K+ current occurs after the Na+ current
K+ current occurs after the Na+ current because the K+ channels open more slowly than the Na+ channels

slight difference @ which the channels open with K after Na+
[Action Potential]

What are the two types of K+ channels that mediate K+ efflux
Voltage–gated ion channel
inward rectifier channels
[Action Potential]

Describe voltage–gated ion channels
open at a set membrane voltage
[Action Potential]

Describe inward rectifier channels
open @ – Em
close @+ Em

account for RMP @ –88 mV due the allowance of potassium membrane permeability at resting membrane potential.
potassium always goes out
[Action Potential]

Which gate is responsible for K+ permeability at rest?
inward rectifier channels
[Action Potential]

Which gate is responsible for repolarization
Voltage–gated channels
[Action Potential]

list the factors that determine action potential duration
the magnitude of K+ currents:
larger = faster repolarization
smaller = slower repolarization

determined also by membrane permeability
[Action Potential]

Explain the mechanisms of altered excitability
Altered Depolarizing Duration in the presence of other depolarizing currents

Calcium current is triggered after a Na current, can create the plateau seen in the action potential, delays repolarization
[Drug distribution & excretion]

describe the effects of ionization & lipid solubility on drug distribution
Ionization: non–ionized forms of the drugs can cross the lipophilic membrane while the ionized forms cannot

Lipid solubility – lipophilic drugs can easily pass lipid membranes (think blood brain barrier & propofol)

Water soluble – escape slower, limited solubility & diffusion, more dependent on hydrostatic pressure
[Drug distribution & excretion]

What percentage of blood flow is consumed by the brain?
20% of blood flow
[Drug distribution & excretion]


What percentage of blood–born energy is consumed by the brain?
25% of the energy
[Drug distribution & excretion]

Discuss the factors affecting the distribution of the drug into the CNS
glial wrapping – highly lipid–soluble can rapidly reach the brain
ABC transporter – rapidly perceived toxic drugs out of the CNS back into systemic circulation
Tight Junctions – limit drug movement out of the capillaries
Basement membrane – when intact presents a barrier to the brain but is lipophilic
[Drug distribution & excretion]

Describe the BBB – glial wrapping function
allows highly lipid–soluble drugs to rapidly reach the brain but presents a barrier to other types
[Drug distribution & excretion]

Describe the BBB – ABC transporters
recognize & evacuate toxic drugs out of the CNS back into the capillaries
[Drug distribution & excretion]

Describe the BBB – tight junctions
prevent drug movement out of the capillaries between the cells into the brain.

other tissue types are more lenient and allow freer movement of drugs into the tissues
[Drug distribution & excretion]

How does solubility determine a drugs entry into the CNS?
In order to pass through the BBB, the drug must be highly lipid soluble

If the drug is polar or highly water soluble, it might need to be directly dosed into the CNS, bypassing the BBB
[Drug distribution & excretion]

Describe possible drug related ramifications to disruptions in the integrity of the BBB
any inflammation or injury can render the BBB leaky, resulting in the ability of drugs to enter into the CNS that normally could not or in dose changes quicker than normally allowed by the strict controlling BBB
[Drug distribution & excretion]

Discuss the barrier characteristics of the placenta
exhibits all modes of transfer of molecules
passive diffusion due to lipid solubility
most drugs can easily cross
drugs metabolized by the fetus can accumulate because of a change in lipid solubility by the metabolite (ionization)
[Drug distribution & excretion]

What is the equation for the volume of distribution
Vd = D / Cpo

Vd =apparent volume of distribution
D = drug dose
Cpo = plasma conc. of drug @ time = 0
[Drug distribution & excretion]

Describe the meaning of a low Vd value
a low Vd value relates to a low volume of distribution which relates to the concentration of the drug in the plasma at the time of the test. This means more of the drug is in the plasma vs. stored in the tissues
[Drug distribution & excretion]

Describe the meaning of a high Vd value
a high Vd value relates to a high volume of distribution which correlates to a low % of the drug in the plasma & a high % of the drug in the tissues or protein bound (e.g. somewhere else)
[Drug distribution & excretion]

What is the estimated value of Total Body Water (TBW)
~42 liters
[Drug distribution & excretion]

How much of the estimated TBW is in the plasma?
~3 liters
[Drug distribution & excretion]

How much of the estimated TBW is in the Extracellular fluid
~14 Liters
[Drug distribution & excretion]

How much of the estimated TBW is in the Intracellular fluid
~25 liters
[Drug distribution & excretion]

What does it mean if the Vd exceeds TBW?
that the drug is concentrated outside of the plasma
[Drug distribution & excretion]

Describe the factors that contribute to unequal drug distribution in the body
plasma binding
drug ionization
tissue perfusion
[Drug distribution & excretion]

What are the primary plasma proteins involved in drug binding?
Albumin

AAG – Alpha 1–acid glycoproteins
[Drug distribution & excretion]

Describe the characteristics of Albumin
binding is usually reversible

preference for acidic drugs
[Drug distribution & excretion]

Describe the characteristics of AAG
Alpha 1–acid glycoprotein

binding is usually reversible
preference for basic drugs
[Drug distribution & excretion]

Describe the potential effects of plasma binding on the concentration of free drug in the blood, Vd, & elimination
Increased protein binding will cause any bound drug to not be excreted except by active transport as well as
slow the drug movement out of the capillaries as the bound drug does not count towards the concentration gradient.
[Drug distribution & excretion]

How will the number of plasma protein binding sites influence drug competition
finite # of binding sites & amount of plasma proteins lead to a saturation & competition and potentially more free drug in the plasma, a higher gradient forcing more into the tissues
[Drug distribution & excretion]

Describe protein binding site competition with regards to drugs that have a low therapeutic index
if drug A with a low therapeutic index (higher dose = toxicity) is pushed off a protein in exchange for another then the plasma concentration of the drug A will increase, potentially risking toxicity
[Drug distribution & excretion]

Describe the potential effects of hypoalbuminemia
decreased plasma proteins will decrease the available protein binding sites potentially increasing the free drug, pushing more into the tissues, extending the duration of the drugs action
[Drug distribution & excretion]

describe the distribution characteristics of thiopental/propofol & the termination
these drugs pass quickly through the lipophilic BBB but also quickly into the Fat & muscle tissue. When the drug administration stops, the plasma concentration drops due to the increasing distribution to the muscles/fat, the drug leaves the brain. The drug concentration in the brain eventually reaches zero with the remaining amount in the muscles/fat but leaking out slow enough to stay subtherapeutic due to the speed of excretion
[Drug distribution & excretion]

describe elimination
removal of the drug from the body

metabolism + urinary excretion
[Drug distribution & excretion]

Describe (CL) clearance
quantitative measure of elimination (mL/min)
[Drug distribution & excretion]

Describe Excretion
process of drug elimination, primarily the kidney
[Drug distribution & excretion]

List the methods of drug excretion
Renal, biliary, salivary, mammary, skin, intestines, pulmonary excretion
[Drug distribution & excretion]

Describe enterohepatic circulation
drug metabolized once by the liver, returned to the GI tract, enzymes remove conjugate added by the liver, allowing the drug to return again into the blood
[Drug distribution & excretion]

What will enterohepatic circulation do to a drugs half–life?
potentially increase
[Drug distribution & excretion]

Explain renal secretion via active transport
active transporters (OAT, OCT) transport metabolites into the renal tubules from the peritubular capillaries

transporters are saturable

increase renal clearance
[Drug distribution & excretion]

Explain a drugs reabsorption during the renal excretion process
drug reabsorption greatly depends on urine pH (ionization/unionization)

pka > pH forces drug to unionized, lipophilic, reabsorbable.

decreases renal clearance
[Drug distribution & excretion]

What will pH change with tubular reabsorption of drugs
pKa > pH – unionized, less cleared, lower CL
pKa < pH – ionized, more cleared, higher CL
[Drug distribution & excretion]

What is the GFR for a normal kidney?
~120 mL/min
[Drug distribution & excretion]

give the equation that finds the ratio of drug concentration on each side of a membrane
Acid:
R1:2 = [1+10^(pH1–pKa)] / [1+10^(pH2–pKa)]

Base
R1:2 = [1+10^(pKa–pH1)] / [1+10^(pKa–pH2)]
[Drug distribution & excretion]

What factors may influence the renal excretion of drugs?

blood flow
pH / ionization
lipid–solubility
plasma protein binding
presence of other drugs
disease
age

[Phamacokinetics]

Define "therapeutic window"
a range of plasma concentrations between a minimal effective concentration (MEC) and a minimal toxic concentration (MTC).
[Phamacokinetics]

Define MEC
minimum effective concentration
[Phamacokinetics]

Define MTC
minimum toxic concentration
[Phamacokinetics]

Define therapeutic drug monitoring
Useful for: narrow therapeutic window drugs to ensure therapeutic effect & avoid toxicity
–evaluate pt compliance
–avoid irriversible toxicities
–evaluate changes in efectiveness r/t CL
–drugs that lack clearly defined endpoints
[Phamacokinetics]

Describe Pharmacokinetic parameters
Most important factors in dosing:

–Half–life
–Clearance
–Volume of distribution
[Phamacokinetics]

Explain the concept of half–life
Half–life = time required for 50% completion of a process
–index of: time–course for drug–elimination & drug–accumulation
[Phamacokinetics]

Describe the relationship between half–life & first–order processes
rate of elimination is directly PROPORTIONAL to drug CONCENTRATION

– %drug / minute
[Phamacokinetics]

Describe the relationship between half–life & zero–order processes
constant amount of drug is eliminated per unit of time regardless of drug concentration
– 10 mL / minute
[Phamacokinetics]

How many half–lives does it take to complete a process?
~5
[Phamacokinetics]

Recall the maximum clearance rates for a drug eliminated by hepatic clearance, renal tubular secretion, & glomerular
Liver blood flow (1500mL/min)
Renal blood flow (660mL/min)
Glomerular filtration rate (120–130mL/min)
[Phamacokinetics]

Factors Affecting Drug Half–Life
aging (decr. muscle mass, decr. distribution)

obesity (increase adipose mass, increase distribution)

Pathologic fluid (increase distribution)
[Phamacokinetics]

Explain the concept of clearance & its relationship to drug dose & steady–state drug plasma concentration
–apparent volume of fluid completely cleared of drug per unit of time (by metabolism and/or excretion)
–it’s an index of how well a drug is removed irreversibly from circulation
–quantitative measure of rate of removal of a substance from the body
CLsystemic = Renal + Liver + other
[Phamacokinetics]

What can change a drugs clearance?
P450 induction (increase CL)

P450 inhibition (decrease CL)
Cardiac failure (decrease CL)
Hepatic failure (decrease CL)
Renal failure (decrease CL)
[Phamacokinetics]
Explain the concept of a loading dose
a loading dose is the amount/concentration/dose of a drug that will force the drug into the tissues, quickly bringing the Vd into the therapeutic range.

Doseloading = Css * Vd
Doseloading = [Css * Vd ] / Foral
[Phamacokinetics]

Explain the relationship between half–life, volume of distribution and clearance. Employ these relationships to predict the pharmacokinetics of a drug.
t 1/2 = [ 0.693 * Vd ] / CL

(t1/2) is directly related to (VD)
(t1/2) is inversely related to clearance (CL)
(t1/2) is dependent on VD and CL
[Phamacokinetics]

Given the appropriate data, calculate a new dose or a new dose interval in an individual with decreasing renal or hepatic function.
New Dose = [CLdiseased / CLnormal] * Maint. Dose

New Dose Interval =
[Phamacokinetics]

Discuss First–Order Kinetics
constant fraction of drug in the body is eliminated per unit of time

–rate of elimination by metabolism or renal excretion is proportional to plasma drug concentration

–*half life, clearance, and volume of distribution are independent of dose, and clearance rate for any drug is constant
[Phamacokinetics]

Discuss Zero–Order Kinetics
–elimination rate is independent of drug concentration and constant per unit time
–a constant amount of drug is eliminated per unit time

–implies the clearance mechanism(s) is saturated or capacity limited (metabolism by enzymes or transporter
[Phamacokinetics]

Explain what is meant by dose‑dependent kinetics and explain the clinical implications. Name three drugs eliminated by dose–dependent kinetics.
Drug elimination is first order (fixed percentage of drug removed per unit time) at low concentrations; however, at high concentrations, enzymes for breaking down drug are saturated, and elimination becomes zero order (constant amount of drug removed per unit time)
–Clinical implications: A small increase in dosage can cause adverse effects.
–Phenytoin, Ethanol (reaches saturation quickly), aspirin
[Intro to Viruses]

What are the basic structural organization of human viruses?
– DNA or RNA genome (ss or ds)
– surrounding capsid composed of protein capsomeres
– naked or w lipid envelope
– shape: icosahedral, helical, complex
[Intro to Viruses]

Describe some of the general characteristics of viruses (general, extra)
– infrequently fatal
– majority of infections are asymptomatic
– obligate intracellular parasites
– no synthetic machinery
– only one type of genomic nucleic acid
– Not alive, complex mobile genetic elements
[Intro to Viruses]

List Common DNA viruses
HBV – Hepatitis B
HPV – human papilloma virus
Parvovirus B19
Adenovirus
Herpesviridae
Polyomaviruses
[Intro to Viruses]

List common RNA viruses
Influenza
RSV
Parainfluenza
Hepatitis A, C, D, E
Enteroviruses
Encephalitis viruses
Measles, Mumps, Rubella
Norwalk, Rotavirus
Virtually Everything else :P
[Cultural Awareness]

Identify populations at risk for certain disease processes & cultural barriers
Cancer screening & management Cardiovascular disease, diabetes, HIV infection/aids, Immunizations, Infant Mortality, Mental Health, Hepatitis, Syphilis, TB
[Cultural Awareness]

Describe the six focus areas in which racial & ethnic minorities experience serious disparities in health access & outcomes
Cancer screening & management
Cardiovascular Disease
Diabetes
HIV infection / Aids
Immunizations
[Cultural Awareness]

Describe how cultural beliefs & behaviors can affect assessment of the patient & adherence to your treatment plan
Communication
family relationships
diet & nutrition
health beliefs & practices
[Intro to Viruses]

Describe the classifications of viruses
By Nucleic Acid: RNA/DNA, segmented or non–segmented, linear or circular, SS or DS, +/– sense mRNA,
envelope
capsid
replication strategy
[Intro to Viruses]

Describe the nature of viral replication
–Attachment – viral attachment proteins connect w cellular receptors
–penetration/uncoating – retraction of receptors, capsid digestion
–biosynthesis – cellular synthetic processes redirected, become viral specific
–maturation – viral components assembled
–release – virus extracellular
[Intro to Viruses]

Mechanisms of viral pathogenesis, including alternatives to cell distruction
–cell destruction – lytic, inhibition of host DNA, RNA &/or protein synthesis; immune–mediated tissue damage
–transformation – cell transformation to oncogenic > cell division rate increases
–latent or persistent – ocult, virus becomes dormant, can later become re–activated
–cell fusion – to form multinucleated cells
–latent infection – chronic carrier, latent, or slow virus
[Intro to Viruses]

Epidemiological considerations with respect to the transmission & dissemination of viral diseases
Transmission, age, gender, ethnic background, country of origin, travel history, occupation, season, underlying medical conditions.
[Intro to Viruses]

the role of the immune system in viral disease symptomatology & recovery from infection
antigenic variation, some viruses encode receptors for various mediators of Immunity thus blocking their ability to interact with receptors on the virus, some viruses reduce expression of class I MHC proteins thus reducing ability of cytotoxic T–cells to kill the virus–infected cells, direct cell–to–cell propagation & attenuated virus
[Fungal]

How many species of fungi are pathogenic?

How many are pathogenic to humans?

How many are common?
over 100,000 species pathogenic

~175 cause disease in man

~30 common
[Fungal]

Describe saprophytic
lives on dead or decomposing matter, soil dwellers

[Fungal]

Describe general characteristic differences between Fungi & bacteria

Fungi vs. Bacteria


cell type - Eukaryotic vs. prokaryote


Cell interactions - uni/multicellular, able to differentiate vs. unicellular


Membrane - ergosterol, no cholesterol vs. no sterols


Cell Wall - peptidomanna, glycan, chitin, cellulose, chitosan vs. peptidoglycan, teichoic acid, lipopolysaccharide


Metabolism - heterotrophic only vs auto or heterotrophic


Oxygen Requirements - aerobic, anaerobic, facultative vs aerobic only


Reproduction - asexual (conidia), sexual (spores) vs asexual(fision)

[Fungal]

Describe the differences between molds, yeasts, & dimorphisms

molds: multicellular, long–tubelike extensions of the cell wall, grow naturally in soil environments @ 25 deg–C
Yeasts: unicellular, reproduce asexually by budding, grow @ body temperature ~37 deg–C
Dimorphisms: ability of a fungus to grow as a yeast or mold, depends on environmental condition
[Fungal]

Describe septation

crosswalls separating nuclei in a tube–like chain of fungi

[Fungal]

Describe the fungal characteristics that pertain to Adherence
some fungal species, particularly yeasts are able to colonized the mucosal surfaces of the oral, GI, & female genital tract

species of Candida that adhere best to epithelial cells are most often isolated from infections
– fungal "adhesins"
[Fungal]

Describe the fungal characteristics that pertain to Invasion
generally, non–invasive.

some proteases, lipases, ketatinases facilitate colonization, tissue invasion
[Fungal]

Describe the fungal characteristics that pertain to Resistance to phagocytosis
Most fungi are extremely susceptible to phagocytic killing. Pathogenic strains can often be shown to have increased anti–phagocytic properties

Histoplasma capsulatum in yeast form can multiply within macrophages
[Fungal]

Describe the fungal characteristics that pertain to Tissue Injury
No exotoxins, endotoxins. Fungal metabolic products do not injure tissues directly. Tissue injury is most often a result of inflammation & immune responses to fungal presence
[Fungal]

Describe the important methods available to aid in the diagnosis of fungal infections
KOH digestion & microscopy
10% KOH
Wood's lamp
culture, visualization, carbohydrate assimilation test, Ag/Ab detection, PCR
[Fungal]

Clinical classification of medically relevant fungi
Superficial – infections of the dead, superficial areas of the skin or hair shaft, no cellular response, cosmetic problems only
Cutaneous – living tissue is not invaded, organisms colonize the keratinized stratum corneum b/c of their keratinolytic ability > disease results from the host reaction to fungi metabolic products
subcutaneous – usually requires implantation, adaptation of organisms to tissue environment requires relatively long periods of time
systemic – agents that are inherently virulent and cause disease in healthy humans (acquired via respiratory tract) > requires significant exposure to fungal spores
opportunistic – in patients with impaired host defenses or alterations in normal bacterial flora
[Parasites]

Define Definitive Host
host in which the parasite reaches sexual maturity
[Parasites]

Define Intermediate Host
host in which asexual reproduction or larval development takes place
[Parasites]

Define Vector
transmitting agent, usually arthropod
[Parasites]

Explain the difference between trophozoite & a cyst
Trophozoite – metabolically active & motile stage of many protozoan parasites

Cyst – generally smaller & has an outer protective layer to enhance survival in the environment
[Parasites]

World–wide, how prevalent are infections caused by parasites
Toxoplasmosis = 1–2 billion
Ascariasis = 1 billion
Malaria = 200–300 million
Schistosomiasis = 200–300 million
Giardiasis = 200 million
Trypanosomiasis = 15–20 millions
[Parasites]

Describe the probable roles of IgE antibody & eosinophils in combating parasitic infections
IgE coats the parasite

Eosinophils will target, attack, & eat the parasite
[Parasites]

Describe clinical presentation & basic characteristics of Giardia infection
–most common cause of parasitic gastroenteritis
–infection from ingestion of cysts
–average incubation period of 10 days
–attaches to intestinal mucosa & causes shortening of the villi, inflammation of crypts & lamina propria
– diarrhea due to malabsorption
–immune response
–treatment – metronidazole
[Parasites]

Case Study!
–Your patient is a 5–year–old female accompanied by her mother.
–The chief complaint is nausea, vomiting, and foul–smelling diarrhea for several days.
–The mother states that the watery stool looks greasy. The child has had little appetite.
–The patient attends a pre–school this year, and had been in day care since she was 2–years–old.
–Physical exam reveals moderate epigastric tenderness. The child is slightly below normal weight.
[DS – Intro to Antimicrobial Pharm]

Describe bacteriostatic
inhibits/stops growth & reproduction of bacteria but do not kill them

works with the immune system to remove organisms from the body

*tetracycline
[DS – Intro to Antimicrobial Pharm]

Describe bactericidal
kill bacteria
– interference w a process essential for life

–useful with endocarditis, meningitis, & neutropenic cancer patients


*PCN
[DS – Intro to Antimicrobial Pharm]

Describe Time–dependent killing
killing effect is directly proportional to the amount of time the drug concentration at the site of infection is above the MIC of the organism
[DS – Intro to Antimicrobial Pharm]

Define MIC
minimum inhibitory concentration
[DS – Intro to Antimicrobial Pharm]

Are beta–lactam antibiotics time or concentration dependent?
time–dependent killers
[DS – Intro to Antimicrobial Pharm]

Describe concentration dependent antibiotics
bacterial kill increases with increasing levels of the drug

concentration is not important

Aminoglycosides & fluoroquinlones
[DS – Intro to Antimicrobial Pharm]

Describe selective toxicity
adversely affect only type of cell & spare normal human cells
[DS – Intro to Antimicrobial Pharm]

What are the 5 MoA of antibiotics
1. inhibition of cell wall synthesis
2. Disruption of cell membrane function
3. Inhibitors of protein synthesis
4. Inhibition of Nucleic Acid synthesis
5. Inhibition of synthesis of Essential Metabolites
[DS – Intro to Antimicrobial Pharm]

What antibiotics inhibit cell wall synthesis

Names & targets
b–lactam – targets TG
Vancomycin – targets TP
Bacitracin – stop dephosphorylation of bactoprenol
Cycloserine – stop the addition of d–ala
fosfomycin – inhibit nam
[DS – Intro to Antimicrobial Pharm]

Beta–lactam antibiotics: names
PCN
Cephalosporins
Carbapenem
Monobactams
[DS – Intro to Antimicrobial Pharm]

List the types of PCN
Natural PCN – PCN G
Extended–Spectrum PCN – amoxicillin
[DS – Intro to Antimicrobial Pharm]

List the generations & specific names of the Cephalosporins
1 – cephalexin
2 – cefoxitin
3 – ceftriaxone
4 – cefepime
[DS – Intro to Antimicrobial Pharm]

What are the benefits of the higher generation cephalosporins
– increased ability to cross the BBB
– increased resistance to b–lactamases
– increased activity vs. gram negative
[DS – Intro to Antimicrobial Pharm]

Carbapenem: names
Imipenem
[DS – Intro to Antimicrobial Pharm]

Monobactams: names
Aztreonam
[DS – Intro to Antimicrobial Pharm]

Describe the Beta–lactam ring, uses, makeup, & limits
–consists of b–lactam bonds
–can be cleaved by b–lactamases
– inhibits PBPs inducing cell wall bursting
[DS – Intro to Antimicrobial Pharm]

Names of antibiotics that disrupt cell membrane function
–Cidal & concentration–dependent

Amphotericin
Ketoconazole
Polymyxin
Daptomycin

Amy Keeps Pooping D***it
[DS – Intro to Antimicrobial Pharm]

Target & Names of the antibiotics that inhibit the synthesis of essential metabolites
Target: mostly folic acid

Trimethoprim
sulfonamides > sulfamethoxazole
[DS – Intro to Antimicrobial Pharm]

Target & names of antibiotics that affect nucleic acid metabolism
Bactericidal –
DNA Gyrase inhibitor
* Fluoroquinolones – Ciproflaxin
RNA Polymerase
* Rifampin
[DS – Intro to Antimicrobial Pharm]

Names of antibiotics that inhibit bacterial protein synthesis
Aminoglycosides – Gentamycin ******
Tetracyclines – Doxycyclin
Macrolides – Erythromycin
Chloramphenicol
Clindamycin
Mupirocin
Streptogramins
[DS – Intro to Antimicrobial Pharm]

Attributes of antibiotics that inhibit bacterial protein synthesis
Bacteriostatic (except AG=cidal)

target 30s/50s
[DS – Intro to Antimicrobial Pharm]

resistance factors for antibiotics that inhibit bacterial protein synthesis
Microbial Enzymes > AGs, alter enzyme, reduce binding
Ribosome protection > tetracyclines & Macrolides= compete for or alter binding site
Efflux pumps > Macrolides & Tetracyclines
Update reduced > Cephalosporins
Natural Selection –
[DS – Intro to Antimicrobial Pharm]

Define PBPs
peptidoglycan binding proteins
[DS – Microbial Genetics & Drug Res]

Discuss the misuses of antibiotics
–Given when they are not needed (according to CDC up to 50% of antimicrobial use in hospitals are inappropriate) (Outpatient prescriptions are often unnecessary particularly when treating URI)
–Continued when they are no longer necessary
–Given at the wrong dosage
–Wrong antibiotic is used to treat the infection
–In many countries antibiotics can be purchased over the counter which leads to even more indiscriminate use
– Antibiotic use and misuse has led to incr. in selective pressure
[DS – Microbial Genetics & Drug Res]

List the ways bacteria can resist antibiotics
–Degrade or alter antibiotic (splitting of the Beta–Lactam ring) (Modify aminoglycosides)
–Efflux Pump (Tetracycline) (Macrolides)
–Uptake reduced (Cephalosporins)
–Overproduction of target metabolic bypass (Sulfonamides) (Timethoprim) – produces enzymes that breakdown the drug
– Alteration of target (Penicillin binding proteins “transpeptidases”: Beta–Lactams) (50S ribosomal subunit modified so that it still functions but macrolide can no longer bind) (DNA gyrase and topoisomerase interferes with fluoroquinolones)
– Pathogens develop resistance through natural selection (When bacteria are exposed to an antibiotic, more susceptible organisms will be killed leaving behind the more resistant bacteria to grow and multiply)
[DS – Microbial Genetics & Drug Res]

Explain Spontaneous Mutations
Despite effective repair systems, mistakes in normal replication occur about 1 per 10^6 & 10^9 cells
[DS – Microbial Genetics & Drug Res]

Explain Induced Mutations
caused by mutagens
[DS – Microbial Genetics & Drug Res]

Explain Mutations resulting in antibiotic resistance
May arise prior to or in the absence of selective pressure; most antibiotic resistance occurs by point mutations
[DS – Microbial Genetics & Drug Res]

Explain point mutations:
substitution, deletion, or change of nucleotide base sequence
[DS – Microbial Genetics & Drug Res]

List three methods of intracellular DNA transfer
Transformation
Transduction
Conjugation
[DS – Microbial Genetics & Drug Res]

Explain Transformation
direct uptake & incorporation of exogenous genetic material (exogenous DNA) from its surrounding & taken up through the cell membranes
[DS – Microbial Genetics & Drug Res]

Explain Transduction
transfer of genetic material from one bacterium to another by means of a bacteriophage (virus)
[DS – Microbial Genetics & Drug Res]

Explain Conjugation

********
transfer of DNA between bacterial cells by direct cell to cell contact (sex pilus)

F+ to F– only = unidirectional
[DS – Microbial Genetics & Drug Res]

Explain the importance of R–plasmids
Conjugative Resistance R–plasmids

found in many gram–negative bacteria
Contain the "F" factor (replication & transfer Gene)

Contains resistant genes often coding for multiple resistances
[DS – Microbial Genetics & Drug Res]

Explain Transposons
Mobile Genetic Elements
–can move from place to place on the chromosomes & into/out of the plasmids
–carry both insertion sequences plus other genes often those coding for resistances
–if transposons insert into a functional gene, it will be destroyed = cell death
transposons are biological mutagens
[Drug Metabolism]

4. Discuss the major pathways of Drug metabolism:

Phase I only
–add group to molecule (OH, SH, COOH, NH2)
–alters drug to a more polar metabolite
– more water soluble
[Drug Metabolism]

4. Discuss the major pathways of Drug metabolism:

Phase II only
–conjugation: endogenous substrate added to the drug by acetylation, sulfonation, or glucuronidation
–usually forms a highly polar, inactive metabolite that is excreted via kidneys
–Phase 1 addition prep's the molecule for Phase 2 addition
– large change in structure, biological activity, LS, etc
[Drug Metabolism]

5. Recall the basic types of Phase I metabolism
Oxidations

Reductions

Hydrolysis

goal: add or change the molecule to make it more polar thus less LS & more water–soluble
[Drug Metabolism]

6. What are the top three CYP450s involved in drug metabolism in humans?
CYP3A4

CYP2d6

CYP2c9
[Drug Metabolism]

6. What food product inhibits intestinal CYP3A4
Grapefruit juice
[Drug Metabolism]

6. Which transporter works in cooperation with CYP450s to limit systemic exposure to drugs
P–glycoproteins
[Drug Metabolism]

7. Recall the different types of Phase II metabolisms & discuss its role in drug elimination & excretion
Glucuronidation, Acetylation, Sulfation
– attaches an endogenous substrate that usually increases the polarity, inactivates it, and makes it less LS
[Drug Metabolism]

8. Predict potential effects of a drug acting as an inducer or inhibitor of an enzyme that is responsible for the metabolism of another drug.
Induction: cause an increase in the expression of an enzyme, increasing the speed at which a SPECIFIC drug is metabolized and removed from the body.

Inhibitor: cause the suppression of the enzymes that metabolize a SPECIFIC drug, increasing the time the drug stays in the body.
[Drug Metabolism]

consequences of drug metabolism
1. less lipid soluble, now kidney can excrete it
2. less stored in fat
3. more water soluble
4. more ionized at physiological pH
5. less bound to plasma proteins
6. less able to penetrate cell membranes
7. more readily excreted in urine by the kidneys
[Drug Metabolism]

general characteristics of drug metabolites
1. more water soluble
2. more easily excreted by kidneys
3. frequently inactivates a drug (inactivated by liver)
4. metabolism MAY activate a drug (bioactivation)
5. Prodrug concept à prodrug (precursor) is not active, metabolite is active may have no, <, =, or > pharmacological activity than parent drug can sometimes be toxic
[Drug Metabolism]

Explain the functional importance of drug metabolism as it relates to the elimination and excretion of drugs
1. Biotransformation makes drugs more lipid soluble, more water soluble, more ionized, less bound to plasma proteins, etc (see above). This allows the metabolites to be more easily eliminated and excreted through the kidneys.
2. Significance of metabolism: Termination of a drug's pharmacological effect(s) & enhancement of its excretion. This allows intermediates to become polar, non–toxic, metabolites which can then be excreted.
[Drug Metabolism]

Discuss the role of metabolism in acetaminophen toxicity
Metabolism allows acetaminophen to be turned into non–toxic metabolites. The major pathway (95%, phase 2) uses glucoronidation & sulfation to create inactive, non–toxic metabolites, which can then be excreted in the urine. The minor pathway (5%, phase creates reactive toxic intermediates, but these can be neutralized into non–toxic metabolites through glutathione conjugation.
[Drug Metabolism]

Describe the implications of an intermediary drug metabolite being more reactive than the parent compound
1. the reactive toxic intermediates are actually more reactive than the parent compound (acetaminophen) so the MUST be converted to non–toxic metabolites in order for them to be excreted
2. glutathione conjugation converts the toxic metabolites to the non–toxic form
3. happens when there is more drug present than the metabolic enzymes can handle
4. enzymes become saturated in the major pathway (phase 2) and so excess drug is metabolized by the minor pathway (CYP450 enzyme)
5. in this case the glutathione becomes depleted à drug cannot be converted to non–toxic metabolite so the toxic metabolic becomes reactive à hepatotoxicity cellular necrosis
[Drug Metabolism]

Discuss Phase I of drug metabolism
1. adds hydroxyl (OH) or some other group (SH, COOH, NH2)
2. more polar means more H2O soluble
3. if polar enough, may be excreted
4. many phase I products not eliminated rapidly
5. metabolites may be inactive
6. over ½ of all drugs are metabolized by a group of P450 enzymes
7. usually prepares a molecule for phase II
8. usually produces minor changes in chemical structure, LS, and biological activity
[Drug Metabolism]

Discuss Phase II of drug metabolism
1. conjugation an adds "something" to the drug; big water soluble drugs
2. glucuronic acid, sulfate, glutathione, acetyl groups
3. usually forms a highly polar, inactive metabolite that is renal excreted
4. Phase I metabolite may be needed as a substrate for phase II metabolism
5. Glucuronidation – hooks glucuronite groups; most important
6. Acetylation
7. Glutathione conjugation
8. Glycine conjugation
9. Methylation
10. produce large changes in structure, biological activity and LS (big decreases)
[Intro to Metabolism]

2. Describe the relationship between oxygen consumption & resting metabolic rate
–the amount of oxygen intake can be used to estimate metabolic rate
–measurement of the amount of energy needed to maintain normal function
–because energy prod. requires O2, oxygen consumption can be measured & used to estimate RMR/BMR
higher intake = higher Metabolic Rate
[Intro to Metabolism]

List the 3 phases in aerobic respiration
Phase 1: oxide fuels

Phase 2: Make ATP

Phase 3: Use ATP
[Intro to Metabolism]

Describe Phase 1 of aerobic respirations
turn everything into acetyl CoA
Oxidize acetyl CoA in the TCA
fuels oxidized to CO2
NAD & FAD reduced to NADH / FADH2
[Intro to Metabolism]

Describe Phase 2 of Aerobic Respiration
Oxidative Phosphorylation
– make ATP through the electron transport chain
– uses oxygen as the final electron acceptor
– FADH2 & NADH are re–oxidized back to
FAD / NAD
[Intro to Metabolism]

Describe Phase 3 of Aerobic Respiration
Using ATP will generate ADP

cycle starts over
[Intro to Metabolism]

1. Cellular carriers of Energy
ATP – Adenine Triphosphate
GTP – Guanine Triphosphate
CTP – Creatine Triphosphate
UTP – Uridine triphosphate
[Intro to Metabolism]

Glycolysis

Explain why the pathway exists & in which tissues it occurs
Pathway: glucose ––> pyruvate + 2 ATP

Where:
RBCs only use glucose for energy
– (no mitochondria)
Brain doesn't absorb lactate (unless forced) so will always use glucose & ketones
– (if forced w extended fasting)
[Intro to Metabolism]

TCA Cycle

Explain why the pathway exists & in which tissues it occurs
(aerobic respiration phase 1)

pathway: acetyl CoA converted to CO2, H2O, ATP
[Intro to Metabolism]

Oxidative phosphorylation

Explain why the pathway exists & in which tissues it occurs
(aerobic respiration phase 2)

pathway: makes ATP through the electron transport chain, FADH2 & NADH donate electrons to the process
– depends on oxygen

Where: inside the mitochondria
[Intro to Metabolism]

Fatty Acid synthesis

Explain why the pathway exists & in which tissues it occurs
pathway: acetyl CoA –––> Fatty acids
– NADPH oxidized to NADP
– pentose phosphate pathway reduces NADP to NADPH

Where: Liver
[Intro to Metabolism]

Triglyceride synthesis

Explain why the pathway exists & in which tissues it occurs
pathway: fatty acids ––> triglycerides

where: made in the liver, transported to the adipose via lipoproteins
– Dietary triglycerides are transported via
chylomicrons
[Intro to Metabolism]

Lipolysis

Explain why the pathway exists & in which tissues it occurs
pathway: triacylglycerols broken down into fatty acids & glycerols
– glycerol goes right into gluconeogenesis

Where: adipose tissues
[Intro to Metabolism]

Beta–oxidation

Explain why the pathway exists & in which tissues it occurs
pathway: fatty acids (oxidized) ––> acetyl CoA
– NADH & FADH2 are generated

where: muscle, liver, & "other tissues"
[Intro to Metabolism]

Gluconeogenesis

Explain why the pathway exists & in which tissues it occurs
pathway: converting lactate, certain amino acids, or glycerol into glucose
– glycerol comes straight from lypolysis
where:
– liver (during fasting state)
– kidneys (slight)
[Intro to Metabolism]


6. Explain the difference between aerobic & anaerobic metabolism
Aerobic metabolism: oxygen is available as the final electron acceptor
– acetyl CoA is used in the TCA cycle to produce ATP, CO2, H2O

Anaerobic metabolism: occurs where no O2 available. Pyruvate is converted to lactate
– cannot generate acetyl CoA, thus no TCA (no oxidative phosphorylation)
– primary metabolism of RBCs
[Intro to Metabolism]

6. Describe the uses of lactate in the fed & fasted states
Fasting: lactate is the product of glycolysis in RBC's & exercising muscles

Fed: RBCs primary metabolism is glycolysis whose result is lactate.
[Intro to Metabolism]

7. Indicate the metabolic process that will provide the cell with the energy during Fed & Fasted states.

Red Blood Cells
Fed: glycolysis

Fasted: glycolysis still
– only method available to RBCs
[Intro to Metabolism]

7. Indicate the metabolic process that will provide the cell with the energy during Fed & Fasted states.

Adipose Cells
Fed: glycolysis

Fasted: lipolysis
[Intro to Metabolism]

7. Indicate the metabolic process that will provide the cell with the energy during Fed & Fasted states.

Brain Cells
Fed: glycolysis, TCA, Oxydative Phosphorylation

Fasted: if extended: ketogenesis + whatever stores of glucose can be produced
[Intro to Metabolism]

7. Indicate the metabolic process that will provide the cell with the energy during Fed & Fasted states.

Muscle Cells
Fed: glycolysis (glucose = lactate)

Fasted: glycogenolysis + ketones
[Intro to Metabolism]

7. Indicate the metabolic process that will provide the cell with the energy during Fed & Fasted states.

Liver Cells
Fed: glycolysis

Fasted: B–oxidation, Amino Acids
[Intro to Metabolism]

8. Explain how the liver works with the other systems to maintain homeostatic blood glucose during a prolonged fast.
– continues to convert fatty acids to ketone bodies
– since the brain continues to use a limited amount of glucose, the liver needs to produce less glucose per hour during prolonged fasting than during shorter periods of fasting.
– because the stores of glycogen in the liver are depleted by ~30 hours of fasting, gluconeogenesis is the only process by which the liver can supply glucose to the blood
[Intro to Metabolism]

8. Explain how the Adipose Tissue works with the other systems to maintain homeostatic blood glucose during a prolonged fast.
– as blood insulin decreases or blood glucagon increases, adipose triacylglycerols are mobilized by lipolysis
– most fatty acids cannot provide carbon for gluconeogenesis ––> fatty acids serve as a fuel for muscle, kidney, & most other tissues and are oxidized to acetyl CoA & subsequently to CO2, H2O in the TCA cycle
– supplies the blood with its major source of fuel via the glycerol portion of the triglyceride
[Intro to Metabolism]

8. Explain how the RBCs works with the other systems to maintain homeostatic blood glucose during a prolonged fast.
transfer lactate to the blood so that it can be reconverted back to glucose
[Intro to Metabolism]

8. Explain how the Muscle works with the other systems to maintain homeostatic blood glucose during a prolonged fast.
– breakdown proteins to amino acids = transferred back to the liver to produce glucose
– acetyl CoA used in TCA cycle to generate ATP, CO2, H2O
– muscle use of ketones decreases
– brain use of ketones increases
– brain use of glucose decreases
– liver gluconeogenesis decreases
– muscle protein degradation decreases
– liver production of urea decreases
[DS – Metabolism]

1. What metabolic processes is insulin going to stimulate?
Insulin: increases the breakdown of glucose when blood glucose levels are high

Stimulates: glycolysis, TCA cycle, Oxidative Phosphorylation, Triglyceride Synthesis, Glycogenesis, Fatty Acid Synthesis
[DS – Metabolism]

1. What metabolic processes is glucagon going to stimulate?
Glucagon: increases during the fasted state, signals the liver to utilize shared carbohydrates to release glucose into circulation. Signals adipose to degrade triacylglycerols into fatty acids(energy via b–oxydation) & glycerol(gluconeogenesis)
Stimulates: TCA Cycle, Oxidative Phosphorylation, Lipolysis, Beta–oxidation, gluconeogenesis, glycogenolysis
[DS – Metabolism]

2. Name the primary regulator of insulin
Pancreas in response to a high–carbohydrate meal / serum glucose concentration
[DS – Metabolism]

2. Describe the mechanism through which the release of insulin occurs
–insulin is rapidly removed from circulation & degraded by the liver, so blood insulin levels decrease rapidly once the rate of secretion slows.
–neural signals from ANS help to coordinate insulin release with the secretory signals initiated by the ingestion of fuels
– certain amino acids can stimulate insulin secretion
– gut hormones in response to food intake can aid in onset of insulin release
–epinephrine decreases the release of insulin
[DS – Metabolism]

2. Name the primary regulator of glucagon
suppressed by insulin & glucose
[DS – Metabolism]

2. Describe the mechanism through which the release of glucagon occurs
– certain hormones stimulate glucagon secretion (cortisol & epinephrine)

– many amino acids also stimulate glucagon release
[DS – Metabolism]

4. Describe the effects of epinephrine on energy metabolism

[big picture]
–mobilize fuels during acute stress
–stimulate glucose production from glycogen
[DS – Metabolism]

4. Describe the effects of cortisol on energy metabolism

[big picture]
– provides for changing requirements over the long term
–stimulates glucagon secretion
–stimulates amino acid mobilization from muscle protein
–stimulates gluconeogensis
–stimulates fatty acid release from adipose tissues
[DS – Metabolism]

5. What class of receptor/signal transduction pathway does Glucagon exert its effect.
* binds to GPCRs to stimulate synthesis of cAMP

–activates glucose production from glycogen in the liver but not in the skeletal muscles
–glucagon–stimulated phosphorylation of enzymes simultaneously activates gl;ycogen degradation, inhibits glycogen synthesis, & inhibits glycolysis in the liver
[DS – Metabolism]

5. What class of receptor/signal transduction pathway does Insulin exert its effect.
* uses tyrosine–kinase receptors for signal transduction

–reverses glucagon–stimulated phosphorylation
–stimulates the phosphorylation of several enzymes
–induces & represses the synthesis of specific enzymes
–acts as a growth factor & has general stimulatory effects on protein synthesis
–stimulates glucose & amino acid transport into cells
[DS – Metabolism]

5. What class of receptor/signal transduction pathway does Cortisol exert its effect.
* bind intracellular receptors or binding proteins

– move to the nucleus & interact with chromatin
– change the rate of gene transcription on the target cell
–stress hormone
[DS – Metabolism]

5. What class of receptor/signal transduction pathway does NE/Epi exert its effect.
* act as neurotransmitter or hormone

– bind to adrenergic receptors
– work through the cAMP system (GPCRs)
[DS – Metabolism]

6. Which hormone would be secreted during:

An Overnight Fast
Decreased insulin

increased glucagon
[DS – Metabolism]

6. Which hormone would be secreted during:

Prolonged Starvation
Decreased insulin

increased glucagon
[DS – Metabolism]

6. Which hormone would be secreted during:

Exercise
increased glucagon
[DS – Metabolism]

6. Which hormone would be secreted during:

Surgery, trauma, or severe infection
increased Cortisol
[DS – Metabolism]

6. Which hormone would be secreted during:

A carbohydrate rich meal
increased insulin
[DS – Metabolism]

6. Which hormone would be secreted during:

A protein rich meal
increased insulin
[DS – Metabolism]

6. Which hormone would be secreted during:

a "fight or flight" situation
epinephrine
[Intro to Metabolism]

What is the main anabolic hormone?
Insulin
[Intro to Metabolism]

Which is considered the "fasted state" hormone?
Glucagon
[Intro to Metabolism]

How many days (about) before the brain begins to utilize Ketones & whatever glucose is available?
~4 days
[Intro to Metabolism]

When will the liver produce Ketones?
only when fasted & only when there is excess acetyl CoA
[Intro to Metabolism]

This is an important source of Amino Acids during the fasted state
Skeletal Muscle
[Intro to Metabolism]

What systems can export glucose into the blood stream?
Liver [& kidney]
[Intro to Metabolism]

The pathway that reduces NADP back to NADPH
Pentose Phosphate Pathway
[Intro to Metabolism]

Are important for energy storage & the transport of dietary fat
Triglycerides
[Intro to Metabolism]

What tissues can make glycogen?
liver
muscles
* all tissues in very small quantities
[Intro to Metabolism]

What tissues can make fatty acids
Liver
* a little in the adipose tissue
[Intro to Metabolism]

What tissues are only capable of using glucose
Red Blood Cells

Brain (until starvation, then Ketones + glucose)
[Intro to Metabolism]

Oxidative phosphorylation requires these:
Mitochondria & Oxygen
[Intro to Metabolism]

The end result of metabolism without oxygen
Lactate
[Intro to Metabolism]

Glucose can be metabolized into:
glucose–6–p
[Intro to Metabolism]

Glucose–6–p can be metabolized into:
Glucose
Glycogen (glycogenesis)
Pyruvate (glycolysis)
Reduces NADPH via PPP
[Intro to Metabolism]

What is the function of the pentose phosphate pathway
recycle NADPH
[Intro to Metabolism]

Glycogen can be metabolized into:
glucose–6–p (glycogenolysis)
[Intro to Metabolism]

Pyruvate can be metabolized into:
Glucose–6–p (gluconeogenesis)
Lactate
Acetyl CoA
[Intro to Metabolism]

Lactate can be metabolized into:
Pyruvate
[Intro to Metabolism]

Amino Acids can be metabolized into:
Pyruvate

Acetyl CoA
[Intro to Metabolism]

Ethanol can be metabolized into:
Acetyl CoA
[Intro to Metabolism]

Triacylglycerols can be metabolized into:
Fatty Acids [lipolysis] (+ glycerols)
[Intro to Metabolism]

Fatty Acids can be metabolized into:
Triacylglycerols (synthesis w glycerol)

Acetyl CoA (B–Oxidation)
[Intro to Metabolism]

Ketones can be metabolized into:
Acetyl CoA
[Intro to Metabolism]

Acetyl CoA can be metabolized into:
Fatty Acids (Fatty Acid Synthesis)

Ketones

utilized by TCA cycle to Oxidative phosphorylation
[Intro to Metabolism]

Describe RMR
– Resting Metabolic Rate

– measurement of the amount of energy needed to maintain normal function

– energy production requires Oxygen, thus Oxygen can be measured to calculate RMR/BMR
[Intro to Metabolism]

Define Digestion
breaking food down into absorbable components
[Intro to Metabolism]

Define Absorption
transfer of digested components from the gut to the blood
[Intro to Metabolism]

Define Metabolism
What happens after the molecule has been absorbed
[DS – Byproducts of Metabolism]

Describe the benefits of lactate and ketone production
lactate can be used for energy or in gluconeogenesis. Ketones can be used as a source of fuel and is a normal production by the liver.
[DS – Byproducts of Metabolism]

Recall that amino acid catabolism generates ammonia and name the process through which the ammonia is detoxified and eliminated. Name the organ in which this occurs
the ammonia is turned into urea in a process called the urea cycle which occurs primarily in the liver
[DS – Byproducts of Metabolism]

Interpret BUN lab results
urea in the blood is measured by blood urea nitrogen (BUN)
[DS – Byproducts of Metabolism]

Describe the importance of the glutathione peroxidase system in neutralizing reactive oxygen species including the role of NADPH and the pentose phosphate pathway
it neutralizes ROS by acting as a reducing agent in a process catalyzed by glutathione peroxidase. NADPH is an electron donor used to regenerate reduced glutathione which can then neutralize another molecule of peroxide. Pentose phosphate pathway is the ONLY pathway that can produce NADPH in red blood cells.
[DS – Byproducts of Metabolism]

Define the term methemoglobin and explain the roles of the glutathione peroxidase system and methemoglobin reductase in preventing it from forming and regenerating ferrous hemoglobin
Methemoglobin is when the iron in heme is oxidized from its ferrous (Fe2+) to its ferric state (Fe3+). Methemoglobin reductase uses NADH to re–reduce the heme iron back into the ferrous state.
[DS – Byproducts of Metabolism]

Explain how bilirubin is eliminated and interpret bilirubin lab results
Bilirubin will travel to the liver on albumin and once it's there, the liver will convert it to bilirubin diglucuronide via an enzyme called glucuronyl transferase. It will then be excreted in the bile. Bilirubin diglucuronide is called conjugated bilirubin because it has been conjugated to something else. Indirect bilirubin is not conjugated and if there is a problem with the bile duct then the excess bilirubin will go into the blood.
[DS – Byproducts of Metabolism]

Alanine and aspartate aminotransferase (ALT and AST)
Enzymes that are important for amino acid and nitrogen metabolism. Present at high concentrations in the liver.
[DS – Byproducts of Metabolism]

Alkaline phosphatase
disorders affecting the bile duct can result in elevated serum levels of this enzyme
[DS – Byproducts of Metabolism]

Bilirubin (direct and indirect)
if a bile duct is blocked then you will find bilirubin in the blood and indirect bilirubin would be elevated if you have a genetic defect in glucuronyl transferase or if the liver is damaged
[DS – Byproducts of Metabolism]

Albumin
albumin is produced by the liver, it will often be present at reduced amounts in patients with suboptimal liver function
[Intro to Bacteria I & II]

Nucleoid
Bacterial chromosome of coiled DNA (~2000–4000 genes)
[Intro to Bacteria I & II]

Plasmid
Smaller circle of DNA
Carries genes for antibiotic resistance, toxin production, etc.
~5–100 genes
[Intro to Bacteria I & II]

Cytoplasm
PACKED with ribosomes (most: polysomes)
Ribosomes (50S and 30S) are sites for some antibiotics
Also has Inclusion Bodies (storage granules and gas vacuoles)
[Intro to Bacteria I & II]

Cytoplasmic Membrane
Osmotic barrier
Specific transport (nutrients)
generates ATP through respiration
Senses environmental changes
[Intro to Bacteria I & II]

Cell Wall
Peptidoglycan (aka murien)
Site for lysozyme (tears)
Synthesis is site for many antibiotics
Strength and shape to the cell
[Intro to Bacteria I & II]

Outer Membrane
Gram Negative ONLY
Contain Porins (entry of nutrient molecules)
Lipoprotein (anchors outer membrane to cell wall)
Composed of Lipopolysaccharides (specific O–side chain, core polysaccharide)
[Intro to Bacteria I & II]

Periplasmic Space
Space between outer and inner membrane
Gel contains loose network of murien ("cell wall")
Hydrolytic and degradative enzymes
Other enzymes involved in peptidoglycan synthesis, nullification of toxins, etc.
[Intro to Bacteria I & II]

Cell Envelope
Entire outer complex (cell wall, inner and outer membranes)
[Intro to Bacteria I & II]

Bacterial Capsule
aka Glycocalyx
Composed of polysaccharides
Viscous, fibrous matrix
Antiphagocytic
*Encapsulated bacteria = smooth, nonencapsulated = rough
[Intro to Bacteria I & II]

Pili (fimbriae)
Responsible for attachment to specific cell types (e.g. Streptococcus to throat cells)
Used in sexual genetic transfer
[Intro to Bacteria I & II]

Flagella
Motility via proton motive force
Long helical filament, flexible hook, basal body
Spins counterclockwise (straight); clockwise (tumbles)
[Intro to Bacteria I & II]

Spores
Metabolically inactive bacteria
Survive literally almost everything
[Intro to Bacteria I & II]

Gram +
Thick peptidoglycan layer over cell membrane
[Intro to Bacteria I & II]

Gram –
Outer membrane, thin peptodiglycan layer, inner membrane
[Intro to Bacteria I & II]

Difference between prokaryotes and eukaryotes?
Eukaryotes have membrane–bound organelles; prokaryotes do not
[Intro to Bacteria I & II]

Main targets for antimicrobial activity
Cytoplasm
Peptidoglycan cell wall
[Intro to Bacteria I & II]

Symbiosis
An association between two organisms that live together
[Intro to Bacteria I & II]

Mutualism
Mutually beneficial association
[Intro to Bacteria I & II]

Commensalism
One organism benefits, but neither is harmed
[Intro to Bacteria I & II]

Parasitism
One organism benefits at the expense of the other
[Intro to Bacteria I & II]

Actions of normal human flora
–Occupy tissue receptors
–Antagonize other bacteria (bacteriocins)
–Synthesize vitamins and growth factors (B complex, K, E)
–Help assimilate ruffage from glycosidase production
–metabolize cholesterol and bile acids
–Stimulate proliferation of Gut–Associated Lymphoid Tissue (GALT)
[Intro to Bacteria I & II]

athogenicity
Ability to inflict damage
[Intro to Bacteria I & II]

Invasiveness
–Colonization
–Ability to bypass or overcome host defenses
–Production of extracellular substances that facilitate invasion
[Intro to Bacteria I & II]

Toxigenesis
–Soluble
–Transported by blood and lymph
–Cause cytotoxic effects at tissue sites
[Intro to Bacteria I & II]

Gram + Cocci
–Staphylococcus
–Streptococcus
[Intro to Bacteria I & II]

Gram + Rods
–Corynebacteria
–Bacillus
–Listeria
[Intro to Bacteria I & II]

Gram + Spiral
None! HAHAHAHAHA
[Intro to Bacteria I & II]

Gram – Spiral
–Treponoma
–Borrelia
[Intro to Bacteria I & II]

Gram – Rods
–Pseudomonas
–E. coli
–Haemophilus
–Bacteroides
[Intro to Bacteria I & II]

Gram – Cocci
Neisseria
[Intro to Bacteria I & II]

Acid Fast Organisms
Mycobacteria
[Intro to Bacteria I & II]

Intracellular (obligate parasites)
–Chlamydia
–Rickettsia
[Intro to Bacteria I & II]

Wall–less
–Mycoplasma
[Intro to Bacteria I & II]

Bacterial Metabolism
–HIGHLY efficient
–Building blocks synthesized in amounts proportional to the needs of making macromolecules
–Toxic intermediates don't accumulate
–Optimal levels of enzymes and cellular organelles are available
–Unnecessary enzymes are not made
–Cell senses and responds to environment
–Cell grows at maximum growth rate allowed by environmental conditions
[Intro to Bacteria I & II]

How is Proton Motive Force (pmf) generated?
Reducing power (ability to donate electrons) forces electrons to pass through Electron Transport Chain located in cytoplasmic membrane; protons are ejected, forming a concentration gradient of protons known as the proton motive force (pmf) (respiration)

Pmf is used for cellular work
[Intro to Bacteria I & II]

How is ATP generated?
Substrate level phosphorylation
A phosphorylated intermediate is converted to a high energy phosphate bond, which reacts with ADP to produce ATP
[Intro to Bacteria I & II]

How is ATP generated?
Chemiosmosis
Pmf drives protons back into cell through ATPase generating ATP from ADP
[Intro to Bacteria I & II]

What happens when reducing power is in short supply?
ATP is hydrolyzed to ADP, expelling protons from the cell and creates pmf, driving electrons in opposite direction along ETC.
[Intro to Bacteria I & II]

Nutrient energy mechanisms
–Active transport
–Group translocation (phosphorylation)–most common
–Facilitated diffusion (metabolism of nutrients)– less common
[Intro to Bacteria I & II]

Binary Fission
–Cell grows to twice their size
–Divide by binary fusion
–Begins at origin and proceeds in both directions
–Takes ~40 mins at 37 degrees C
–In culture doubling every 20 mins, DNA replication must initiate every 20 mins
–Multifork replication used to accomplish this (already beginning second replication as first is completed)
[Intro to Bacteria I & II]

Phases of growth in bacteria
–Lag: Senses the environment and makes enzymes necessary to start growth
–Exponential: (log) rapid growth occurs
–Stationary: Growth halts as resources are used up from the environment
–Death: (log)
[Intro to Bacteria I & II]

Nutrition Requirements
–ALL cells are heterotrophic: need performed hydrocarbons like sugars in order to grow (gets from host)
–Some (ex E.coli) need only inorganic salts and a nitrogen source
–Others (streptococcus) require more complex media (vitamins, amino acids, purines, etc.)
–Others require VERY enriched medium––Fastidious
[Intro to Bacteria I & II]

Oxygen requirements
Strict Anaerobes
–Neisseria or Pseudomonas
–Respiration occurs via electron transport chain, with Oxygen as final e acceptor
–Cytochrome oxidase enzyme
[Intro to Bacteria I & II]

Oxygen requirements
Obligate anaerobes
–Clostridium
–Only grow in absence of CO2
–Fermentation: terminal e acceptor is an organic metabolic intermediate –> organic acids (lactic acids)
–Usually lack superoxide dismutase, catalase, Catalast, and peroxidase
[Intro to Bacteria I & II]

Oxygen requirements
Facultative
–E. coli (most pathogenic bacteria)
–Bacteria grow aerobically in presence of oxygen, anaerobically in absence
[Intro to Bacteria I & II]

Oxygen requirements
Aerotolerant
–tolerate oxygen, but grow fermentatively
[Intro to Bacteria I & II]

Iron Uptake
–Necessary for bacterial growth
–Free iron scarce in blood and tissue––bound
–Bacteria create siderophores: iron chelating compounds
[Intro to Bacteria I & II]

Difference between fermentation and aerobic respiration
Ferm: w/o O
Aerobic: w/ O
[Intro to Bacteria I & II]

Steps in Peptidoglycan synthesis
1. Uridine diphosphate (UDP) carrier activates N–acetylmuranic acid (NAM) and N–acetylglucosame (NAG)
2. Pentapeptide added to UDP–NAM
3. UDP–NAM–PEP transfers to bactroprenol phosphate
4. UDP–NAG added to UDP–NAM–PEP (peptidoglycan monomer)
5. bactroprenol phosphate transports monomer through cell membrane
6. Autolysins break glycosidic bond b/w peptidoglycan and peptide cross–linkages of existing cell wall
7. Transglycosidase (TG) enzyme inserts and links monomers into new ppg
8. Transpeptidases (TP) reform peptide cross–links
[Intro to Bacteria I & II]

Fosfomycin
Inhibits phosphoenopyruvate transferase; prevents formation of NAM
[Intro to Bacteria I & II]

Cycloserine
Analogue of D–ala; blocks addition of dipeptide to UDP–NAM
[Intro to Bacteria I & II]

Bacitracin
Blocks dephosphorylation of bactroprenol phosphate (prevents transport of ppg monomer across cell membrane
[Intro to Bacteria I & II]

Vancomycin:
binds to peptides of ppg monomers and blocks (TG)
[Intro to Bacteria I & II]

Beta–lactams
–Penicillins, Cephalosporins, Carbepenems
–bind to TP
[Intro to Bacteria I & II]

Linezolid
Blocks initiating complex (translation)
–30S inhibitors:
–Tetracycline, Aminoglycosides
–50S inhibitors:
–Macrolides, Chloramphenicol
[Intro to Bacteria I & II]

Fluoroquinolones
Inhibits DNA gyrase or topoisomerase
[Intro to Bacteria I & II]

Metronidazole
Disrupts DNA helical structure under anaerobic conditions
[Intro to Bacteria I & II]

Rifampin
Binds to RNA polymerase
[Intro to Bacteria I & II]

Sulfonamides
Blocks synthesis of dihydropteroic acid
[Intro to Bacteria I & II]

Trimethoprim
Blocks synthesis of tetrahydrofolate
[Intro to Bacteria I & II]

Polymyxins
Interact with with phospholipids
Name the 4 major dietary sources of fuel
Carbohydrates
Fats
Proteins
? unknown, clarify
Explain: Catabolism vs. Anabolism
Catabolism: breaking down for energy

Anabolism: constructing a molecule, requires
Monosaccharides
Glucose

Galactose

Fructose
Disaccharides
Lactose

Maltose

Sucrose
Sucrose is made up of what saccharides
glucose + fructose
Maltose is made up of what saccharides
glucose + glucose
Lactose is made up of what saccharides
glucose + galactose
Polysaccharides
Cellulose
Hemicelluloses
Pectin
beta–glucans
Fructans
The portion of the reducing sugar that is oxidized in a cupric compound
Aldehyde group
What color will the glucose or galactose oxidase tests turn in the presence of a reducing sugar?
Blue
What is the difference between an unsaturated & polyunsaturated fatty acid?
1 double bond vs. >1 double bond
Results from partial hydrogenation of vegetable oils
trans
Most abundant double bond arrangement in fatty acids
CIS
used to make vegetable oils more solid at room temperature
hydrogenation
structure of triglycerides
3 fatty acids
one glycerol
Importance of Triglycerides
energy storage

transport of dietary fats
Three main body fuel stores

proteins

carbohydrates

fats

[Fungal]

Describe fungal pathogenesis

{think definition, not process}

basic mechanism of fungal pathogenicity is the ability to adapt to tissue environment & temperature (body & fever range) & withstand the lytic activity of host cellular defenses

[Fates of a Cell]



Describe Oxygen-derived free Radicals,



specifically: Superoxide Ion



*boards only

production: incomplete reduction of O2 during oxidative phosphorylation; by phagocyte oxidase in leukocytes


inactivation: conversion of H2O2 & O2 by superoxide dismutase


path. effects: stimulates production of degradative enzymes in leukocytes & other cells; may directly damage lipids, proteins, DNA, acts close to site of production

[Fates of a Cell]



Describe Oxygen-derived free Radicals,



specifically: Hydrogen Peroxide



*boards only

production: generated by SOD from O2 and by oxidases in peroxisomes


inactivation: conversion to H2O & O2 by catalase (peroxisomes), glutathione peroxidase (cytosol, mitochondria)


path. effects:can be converted to *OH & OCL- ; which destroy microbes & cells; can act distant from site of production

[Fates of a Cell]



Describe Oxygen-derived free Radicals,



specifically: Hydroxyl Radical



*boards only

production: generated by H2O by hydrolysis (radiation) ; from H2O2 by Fenton Reaction; From superoxide ion


inactivation: conversion to H2O by glutathione Peroxidase


path. effects: most reactive oxygen-derived free radical; principal ROS responsible for damaging lipids, proteins & DNA

[Fates of a Cell]



Describe Oxygen-derived free Radicals,



specifically: Peroxynitrate



*boards only

production: produced by interaction of superoxide ion & NO generated by NO synthase in many cell types (endothelial cells, leukocytes, neurons, etc)


inactivation: conversion to HNO2 by peroxiredoxins (cytosol, mitochondria)


path. effects: damages lipids, proteins, DNA

[Fates of a Cell]



What is the most reactive oxygen derived free radical responsible for the most damage?

Hydroxyl Radical