Download Medical Physiology

Document related concepts

Myokine wikipedia , lookup

Ketosis wikipedia , lookup

Endocannabinoid system wikipedia , lookup

Lipid signaling wikipedia , lookup

Fatty acid metabolism wikipedia , lookup

Signal transduction wikipedia , lookup

Hemorheology wikipedia , lookup

Transcript
Hoang 1
MEDICAL PHYSIOLOGY
2005
ENDOCRINE PHYSIOLOGY
I. Hormones
Highest bound/free radioactive hormone = zero unlabeled hormone present
Lowest bound/free radioactive hormone = highest hormone concentration present
Rough endoplasmic reticulum = preprohormone & prohormone
Golgi apparatus = prohormone & hormone
G-Proteins
GTP-binding proteins = intrinsic GTPase activity
Adenylate cyclase, IP3, and Ca2+-calmodulin
A. Adenylate cyclase
Hormone bind receptor on cell membrane
Gs = stimulatory
Gi = inhibitory
GDP released from G-protein at -subunit
GTP binds to G-protein at -subunit
Activate adenylate cyclase
Adenylate cyclase converts ATP to cAMP
Intrinsic GTPase converts GTP to GDP
Inactive G-protein
cAMP activates protein kinase A (PKA)
Physiologic actions
Phosphodiesterase degrade cAMP to 5’-AMP
Inhibited by caffeine and theophylline
B. IP3
Hormone bind receptor on cell membrane
G-protein activates phospholipase C
Phospholipase C frees diacylglycerol (DAG) and IP 3 from membrane lipids
IP3 recruit Ca2+ from ER
IP3 and DAG activate protein kinase C (PKC)
PKC phosphorylate proteins
C. Ca2+-calmodulin
Hormone bind receptor on cell membrane
G-protein increase intracellular Ca2+
Open cell membrane calcium channels
Release calcium from ER
Calcium bind calmodulin
Physiologic actions
D. Steroid and thyroid hormone
Steroid and thyroid hormones diffuse cell membrane
Bind cytosolic or nuclear receptor
Receptor has conformational change
Expose DNA-binding domain
Interact with hormone-regulatory elements
Transcription initiated
mRNA translated in cytoplasm
Production of proteins
Hoang 2
Hormone
Type
Derivation
Storage
Transport
Receptor
Administration
Character-is
tic
Catechol-a
mines
Tyrosine
Membranebound
vesicles
Free or
loosely
associated
w/ proteins
Cell surface
receptors
Ineffectively
orally in native
form; effective
when injected
AA derived
Thyroid
Tyrosine
Follicles
Bound to
proteins
Intracellular;
cross cell
membrane
Orally
AA derived
Protein/
Peptide
Synthesize as
pre-prohomo
ne in RER
Membranebound
granules
Unbound,
exocytosis
upon
stimulus
Cell
membrane
NOT orally
Polar
Hydrophilic
Steroid
Cholesterol,
Esters,
synthesized
in SER and
mitochondria
inner
membrane
No storage,
released by
diffusion
after
synthesis
Bound to
proteins
Intracellular
and nuclear
membrane
receptors
Orally
Hydrophobic
Lipophilic
G protein-coupled receptors (GPCR)
Consist of 7 transmembrane segments
GTP-binding domain b/w 5,6 cytoplasmic loop
Subunits / modulate signaling
Increase K+ channel activity and phospholipase A2 activity
C-terminus binds intracellular proteins
Diversity of receptors
Adrenegic = Epi and NorEpi
Muscarinic = Acetylcholine
Peptide/Glycoprotein
Follicle Stimulating Hormone
Angiotensin II
Somatostatin
Thyroid Stimulating Hormone
Vasopressin
Parathyroid Hormone
Calcium
Ionotropic receptors (Ligand-gated ion channel)
Consist of membrane spanning subunits that form ion channel
Allows cations entry
Hoang 3
Calcium = EPI secretion in adrenal
Sodium = muscle contraction
ACH nicotinic and IP3 receptor
Catalytic receptors
Extracellular domain = bind hormone
May result in receptor dimerization
Ligand binds several extracellular domains
Activation of intracellular catalytic domain
Tyrosine kinase-associated receptors
Growth hormone receptor
No intrinsic kinase activity
Intracellular domain associate with JAK and Src
Dimerization to activate tyrosine kinase
Receptor guanylyl cyclase
Atrial natriuretic peptide (ANP) GC-A receptor
Ligand bind to dimerize intracellular guanylyl cyclase domains
Becomes activated
Convert GTP to cGMP
cGMP activate PKG, phosphodiesterase, or ion channels
Receptor serine/threonine kinases
Receptor tyrosine kinases
Insulin receptor, IGF-1 receptor
Extracellular domain = -chain
Ligand bind to -chain
Activated -chain kinase direct tyrosine phosphorylation
Insulin receptor substrate-1
Receptor tyrosine phosphatases
Intracellular Receptors
Steroid hormones
The only homodimer when binding to DNA
Thyroid hormones
Heterodimer
Vitamin D3
Heterodimer
Bind intracellular receptors that are transcription factors
II. Hypothalamus
Anterior hypothalamic nucleus
Temperature = dissipation of heat
Sexual behavior
Posterior hypothalamic nucleus
Temperature = conservation of heat
Paraventricular nucleus (PVN) & Supraoptic nucleus (SON) = magnocellular neurons
Arginine vasopressin
Oxytocin
Arcuate, AH, MPOA, PVN
CRH = Corticotrophin releasing hormone
TRH = Thyrotrophin releasing hormone
LHRH = Lactotrophin releasing hormone
GHRH = gonadotrophin releasing hormone
SS = somatostatin
DA = dopamine
Hoang 4
Paraventricular nucleus
Hypoglycemia & morning time
Stimulate secretion of CRH and AVP
Nighttime inhibit hypothalamus
CRH and AVP stimulate anterior pituitary to secrete ACTH
ACTH stimulate adrenal to secrete cortisol
ACTH inhibit pituitary and hypothalamus
Cortisol stimulates liver to secrete glucocorticoids = increase blood glucose
Highest levels during night
Prolactin and Growth hormone
Highest levels during day
Cortisol
Target Hormone Level
(ie. liver cortisol)
Low
High
Pituitary
Hormone
Level
Normal
Primary failure of
target endocrine
organ
Autonomous
secretion of
pituitary hormone
or resistance to
target hormone
action (pituitary
receptor)
(ie. ACTH)
Normal
Low
High
Normal range
Pituitary failure
Autonomous
secretion by target
endocrine organ
III. Pituitary Gland
Anterior lobe
Linked to hypothalamus via hypothalamic-hypophysial portal system
Thyroid-stimulating hormone (TSH)
Stimulate synthesis and secretion of thyroid hormones
Luteinizing hormone (LH)
Stimulates ovulation, formation of corpus luteum, and estrogen/progesterone synthesis
Follicle-stimulating hormone (FSH)
Stimulates growth of follicles and estrogen secretion
Adrenocorticotropic hormone (ACTH)
Stimulate synthesis and secretion of adrenal cortical hormones
Melanocyte-stimulating hormone (MSH)
Stimulates melanin synthesis
Hoang 5
-endorphin
Growth hormone
Stimulate protein synthesis and overall growth
Homologous to human placental lactogen and prolactin
Pulsatile release
Increased by sleep, stress, starvation, and hypoglycemia
Decreased by somatostatin, somatomedins, obesity, hyperglycemia, and pregnancy
Prolactin
Homologous to GH
Lactogenesis and breast development
Decrease synthesis and release of GnRH and inhibits ovulation / spermatogenesis
Secretion inhibited by dopamine from hypothalamus
Prolactin stimulate release of dopamine
Stimulated by TRH
Posterior lobe
Derived from neural tissue
Nerve cell bodies located in hypothalamic nuclei
Supraoptic nuclei = ADH
Stimulates water reabsorption by renal collecting ducts
Paraventricular nuclei = oxytocin
Milk ejection and uterine contraction
Used to reduce postpartum bleeding
Increased secretion by dilation of cervix and orgasm
IV. Growth Hormone
Highest concentration during sleep
Average = 1.5-1.6 microgram/L in adults
Peak = 15 microgram/L
Found on chromosome 17
3 variants
20 kDa
22.5 kDa = major variant
45 kDa = combination of two 22.5 kDa segments
Derivations of GH
Placental Variant GH (PVGH)
Same functions as GH
Synthesized in placenta
Function during fetal period
Placental lactogen
Similar to GH
Weaker affinity to GH receptor
100-1000 fold weaker
Prolactin
Pituitary produced
19-20% homologous to GH
No growth promoting function
GH binding protein
40% GH bound (inactive)
60% GH free (active)
Bind two GH receptors (dimerization) in liver
Increase IGF and IG Binding protein synthesis in liver
IGF bound to IGBP with ALS
IGF = directly affect growth in bone
IGF-1 receptor similar to insulin receptor
Hoang 6
Heterotetramer
IGF-1 = insulin-like effects
Found in growing tissue especially muscle
Function in childhood growth and adult maintenance
IGF-II receptor = mannose-6-phosphate
Clearance for mannose-linked proteins
Function in fetal growth
Childhood
Excessive GH
Gigantism
Deficient GH
Dwarfism
Adulthood
Acromegaly
Growth hormone direct effects
Increased muscle mass and decreased adiposity (lean body mass)
Decreased glucose uptake
IGF-1 direct effects
Increased linear growth
Increased tissue growth and organ size
V. Male Reproduction
GnRH
10 amino acids = peptide hormone
Pulsated release
Chronic infusion = desensitizing effect of GnRH receptors
Bind to G-protein linked receptor
Activate PLC
IP3 increase intracellular calcium concentration
DAG activate PKC
PKC activate synthesis of  and -subunits of LH and FSH
-subunit determine functionality since -subunits are identical
FSH, LH, hCG, and TRH
Dimerization of subunits
Release due to calcium increase
LH
Target Leydig cells
Bind G-protein linked receptor
Activate adenylate cyclase
Adenylate cyclase converts ATP to cAMP
Activate PKA
PKA stimulate StAR in converting cholesterol to pregnenolone
Cytosol to mitochondria to sER
Testosterone
Bound to SHBG (45%) and albumin (50%)
Free testosterone (5%) degraded by liver or attaches to target cell
Synthesized and secreted by Leydig cells
Does not contain 21-hydroxylase or 11-hydroxylase
Stimulated by LH
LH stimulate synthesis of cholesterol desmolase
Hoang 7
Testosterone inhibit GnRH release and LH release
Accessory sex organs contain 5-reductase
Convert testosterone (inactive) to dihydrotestosterone (active)
Finasteride = 5-reductase inhibitors
Differentiation of wolffian ducts and external genitalia
Development of male secondary sex characteristics
Pubertal growth spurt and libido
FSH stimulate Sertoli cells
Maintain spermatogenesis
Sertoli cells secrete inhibin = inhibit FSH secretion from anterior pituitary
Sertoli cells = Mullerian inhibiting hormone (MIH)
Mullerian ducts regression
Blood-testes barrier
Nourishment of developing germ cells
Produce seminiferous tubular fluid
Remove damaged germ cells
Synthesis of androgen-binding protein and inhibin
Maintain high levels of testosterone in testes
Inhibit FSH secretion from anterior pituitary
Leydig cells = Testosterone
Wolffian ducts transformation
Epididymis, ductus deferens, and seminal vesicles
Paracrine effect on development (unilateral)
Convert to DHT by 5-reductase
Development of penis and scrotum
Absence of MIH
Mullerian ducts transform
Uterus and uterine tubes
Absence of testosterone
Wolffian ducts regression
Development of vagina and female external genitalia
Klinefelter syndrome
XXY
Enlarged breasts, overweight, and infertile
Turner’s syndrome
XO
Nonfunctional ovaries and short stature
Testicular feminization
XY but androgen insensitivity
Male phenotype and ambiguous genitals
True hermaphrodite
XX with SRY gene due to crossing over
Reproductive tract
Seminiferous tubules – rete testis – epididymis – vas deferens
Seminiferous tubules = immature sperm and incapable of fertilization
Epididymis = stored for 2-3 weeks for maturation to become fertile
Spermatogonium – Spermatocyte – Spermatid
Steroidal effects
LH and FSH
Stimulate penile growth, pubic hair, and growth spur
Testosterone
Anabolic effects on muscle
Hoang 8
Inhibit breast development
Stimulate libido
DHT
Embryonic development of prostate
Descent of testes
Phallic growth
Balding and pubic hair
17-estradiol
Epiphyseal closure
Prevent osteoporosis
Feedback regulation of GnRH secretion from CNS
VI. Female Reproduction
Estrogen secretion
Low levels before puberty
No FSH or LH
Increase at puberty (age 12)
FSH and LH spikes at night
Established at age 13
FSH and LH become monthly surges
Decrease to low levels at menopause
Constant high levels of FSH and LH due to loss of inhibition from estrogen
Menarche
First onset of menstrual cycle
Thelarche
Beginning of breast development
Adrenarche (pubarche)
Maturation of adrenal gland cortex
Development of pubic hair
Menstruation
Primordial follicle
Grow to primary follicle
Follicule occurs several cycles before = gonadotropin-independent
Primary follicle divides to secondary follicle
Form zona pellucida and theca cells
Graffian follicle
Theca interna produce hormone
Cumulus oophorus surrounds oocyte
Ovulate into corpus luteum
Secrete progesterone
Degenerate to corpus albicans
Atresia
Theca cells = LH
LH stimulate StAR via adenylyl cyclase
Cholesterol to progesterone
Progesterone converts to Androstenedione and testosterone
No aromatase in theca cells
Granulosa cell = FSH
FSH promote aromatase synthesis
Androstenedione endocytose from theca cells
Converted to testosterone and estradiol via aromatase
Estrogen receptors = intracellular (nuclear), membrane, and cytoplasmic receptors
Hoang 9
Luteal phase
LH receptors synthesized
Cholesterol converts to progesterone via LH stimulation
Progesterone exported to theca cells
Absent in 17-hyroxylase and 17,20-desmolase
LH
Ovulation
Androgen production in theca cells
Luteinization and corpus luteum maintenance
Progesterone secretion from corpus luteum
Granulosa cells
FSH
Growth and development of ovarian follicles
Granulosa cells
Estradiol production
Increase in LH receptors
Proliferative phase
Estrogen induce endometrial cell proliferation
Increase in Estrogen and progesterone receptors on endometrium
Early follicular phase
Granulosa cells few
Have only FSH receptors
Secrete inhibin = inhibit FSH
Stimulated by FSH and estradiol
Midfollicular phase
Granulosa cells have FSH and some have LH receptors
Late follicular phase
Large number of granulose cells
Increase in aromatase
Responsive to both LH and FSH
Estradiol induce LH surge (ovulatory surge)
Progesterone induce collagenase, prostaglandins, and follicular hyperemia
Metalloproteinases and plasminogen activator weaken follicle wall
Follicle rupture with cumulus cells
Evagination of ovum
Secretory phase
Progesterone induce differentiation of epithelial cells to secretory cells
Luteal phase
Estrogen and inhibin increases
Decreases before menstruation
LH and FSH decrease from ovulatory surge
Menstrual phase
Decline of estrogen and progesterone
Activates phospholipase = arachidonic acid
Prostaglandins induce vasoconstriction and ischemia
Endometrial desquamation and bleeding
Shedding of endometrium
Due to proteolysis and ischemia
Lysis of glandular and stromal cells
Degeneration of vascular endothemlium from decreased hormonal support
Hoang 10
Estrogen Functions
Fetal
Sexual
differentiation of
brain
Puberty
Adult-Menstruation
Growth and
development of
oviducts, uterus,
vagina, and external
genitalia
Pregnancy
Stimulate
gonadotropin in
follicular phase
Increase
progesterone
receptors
Inhibit gonadotropin
in luteal phase
Increase oxytocin
receptors
Menopause
[Lack of estrogen]
Vaginal mucosa
dries
Osteoporosis
Breast development
Clear cervical mucus
Breasts atrophy
Growth spur and fat
Closure of epiphyses
Endometrial
proliferation
Increase
progesterone
receptors
Synthesis of SHBG
and TBG
Increase HDL
Decrease LDL and
cholesterol
Progesterone Functions
CNS
Increase body temp
Decrease GnRH release
Mood and behavior
Reproductive tract
Increased growth and
development of oviduct
and endometrium
Decreased estrogen
receptors and uterine
contractility of
myometrium
Decreased oxytocin
sensitivity of myometrium
Limit prepartum
lactogenesis
Uterus
Lack of progesterone
Quiescent
Menstruation
Increased differentiation
and secretion
Parturition (delivery)
Lactation
Dense viscous secretions
Postpartum depression
Increased differentiation of
vagina
Decreased proliferation of
vagina
Hoang 11
Implantation
Acromosal reaction via increased intracellular calcium
Cortical reaction to harden zona pellucida via increased intracellular calcium
Second meiotic division after sperm penetration
Hatching
Zona pellucida degenerates
Apposition
Blastocyst implant in endometrium
Adhesion
Invasion
Syncytiotrophoblast divides
Multinucleate
Forms lacuna
Bathed by maternal blood
Mature chorionic villus
Covered by syncytiotrophoblast
Syncytiotrophoblasts
Secrete hCG on day 8
Rescues corpus luteum
Stimulates production of fetal DHEAS and testosterone
Stimulate relaxin from endometrium = quiescent uterus
Inhibits hypothalamus and pituitary
Maintain low LH and FSH
Pregnancy test = detect hCG on day 9
Corpus luteum
Secrete progesterone from 2-5 wks of pregnancy
Corpus luteum and placenta
Secrete progesterone from 5-12 wks of pregnancy
Placenta only
Secrete estrogen and progesterone
Secrete human chorionic somatomammotropin (human placental lactogen)
Structurally and anabolically similar to GH
Antagonize insulin effects = spare glucose for fetus
Cause maternal hyperglycemia
Growth of mammary gland
Parturition = triggered by fetus
Increased prostaglandins
Increased oxytocin receptors
Increased E/P ratio
Decreased progesterone (functions to inhibit contractions)
Prolactin function
Breast differentiation
Duct proliferation and branching
Glandular development
Milk protein synthesis
Lactogenic enzyme synthesis
Lactation
Suckling stimulus travel via spinal cord to hypothalamus
Inhibits dopamine release from arcuate nucleus
Leading to prolactin release from lactotrophs (ant. Pituitary) = milk production
Inhibits arcuate and preoptic nucleus of hypothalamus
Drop in GnRH production = inhibits ovarian cycle
Stimulate release of oxytocin from PVN and SON = post. Pituitary
Milk release
Hoang 12
VII. Adrenal Gland
Adrenal Cortex
Zona glomerulosa = mineralocorticoids (aldosterone)
Hypovolemia stimulate renin secretion
Renin convert angiotensinogen to angiotensin I
ACE convert angiotensin I to angiotensin II
Angiotensin II stimulate zona glomerulosa
Increase conversion of corticosterone to aldosterone
Aldosterone increase renal sodium reabsorption
Hyperkalemia increase aldosterone secretion
Zona fasciculate = glucocorticoids (cortisol)
Oscillates with circadian rhythm
Corticotropin-releasing factor (CRF) stimulate anterior pituitary via cAMP
Corticotropes synthesize POMC in production of ACTH
ACTH stimulate steroidogenesis in zonae fasciculate and reticularis
Increase conversion of cholesterol to pregnenolone
Zona reticularis = gonadocorticoids (androgens)
Glucocorticoids
Increase gluconeogenesis
Increased protein catabolism
Decreased glucose utilization and insulin sensitivity
In adipose tissue
Increased lipolysis
Increase synthesis of lipocortin
Inhibitor of phospholipase A (prostaglandins and leukotrienes)
Inhibit IL-2 synthesis and T lymphocyte proliferation
Inhibit histamine and serotonin release
Mineralocorticoids
Increase renal Na+ reabsorption, K+ and H+ secretion
21-carbon steroids
Progesterone is precursor
19-carbon steroids
Androgenic activity
Precursors for estrogens
Testes = androstenedione converted to testosterone
18-carbon steroids
Estrogenic activity with unsaturated A ring
Addison’s disease
Adrenocortical insufficiency
Hypoglycemia
Hyperpigmentation
Increased POMC and ACTH = low cortisol feedback
Hyperkalemia and metabolic acidosis
Cushing’s syndrome
Bilateral hyperplasia of adrenal glands
Overproduction of ACTH = hypertension
Excess cortisol and androgens = hyperglycemia and osteoporosis
Central obesity = increased protein catabolism and muscle wasting
Conn’s syndrome
Aldosterone-secreting tumor
Hypertension
Hypokalemia
Hoang 13
Metabolic alkalosis
Decreased renin secretion
Congenital 21-Hydroxylase deficiency
Decreased cortisol and aldosterone = increased ACTH
Virilization in females
Early linear growth and pubic hair
Increased adrenal androgens
Adrenal Medulla
Chromaffin Cells = catecholamines (Epi and NE)
Majority in blood = Epinephrine (90%)
Innervated by sympathetic splanchnic nerve via acetylcholine on nicotinic receptor
Influx of calcium
Increase synthesis and secretion of Epi and NE via exocytosis into blood
Stored in granules
Epi and NE produce negative feedback on tyrosine hydroxylase (TH)
Acute regulation = increased TH activity
Chronic regulation = increased TH synthesis
Epinephrine carried in free form (no binding protein)
Short half-life = 1-3 minutes
Stimulated by hypovolemia, hypothermia, hypoglycemia, trauma, anxiety, and pain
Chromaffin granules
0.7 M Epi and NE
ATP and ADP
Ca2+
Dopamine -hydroxylase
Chromogranin A
Catecholamine metabolism
Liver, kidney, and sympathetic nerve endings
COMT and MAO
Form endproducts VMA, Metanephrine, and normetanephrine
Urine excretion
VMA, metanephrine, and normetanephrine
Unmetabolized Epi and NE
Epi and NE sulfates
Catecholamine receptors
Adrenergic receptors
1, 2, 1, and 2
 = NE
1 = increase phosphoinositide hydrolysis
2 =decrease adenylyl cyclase and cAMP
 = Epi
1, 2, 3 = increase adenylyl cyclase and cAMP
Cross-reactive receptors
Continuous release of catecholamine = down-regulates response
Via internalization of receptors
Sympathectomy = enhance sensitivity and response
Acute release of catecholamine = down-regulate response
Via desensitization of receptors
T3 enhance response via increase in adrenergic receptor and secondary messenger production
Hoang 14
Catecholamine
receptors
Metabolic
effects
1
Increase gluconeogenesis
Cardiovascular
effects
Vasoconstriction at
splanchnic, renal,
cutaneous, and genital
vascular beds
Other
effects
Pupil dilation
GI and Bladder sphincter
contraction
Increase blood pressure
2
Decrease insulin
secretion and stimulate
glucose uptake in muscle
and fat
1
2
Increase liver
glycogenolysis
Increase platelet
aggregation
Increase heart rate,
cardiac contractility, and
conduction velocity
Increase renin secretion
Vasodilation in liver and
skeletal muscle vascular
beds
Increase bladder,
bronchial, and
gastrointestinal muscle
relaxation
Increase gluconeogenesis
Increase glucagons
secretion
Decrease glycogen
synthase
Pheochromocytoma
Adrenal chromaffin cell tumor
Increase secretion of NE and Epi
Severe hypertension and headache
Sweating
Anxiety
Feeling hot
Hands and feet are cold
Large increase in VMA levels in urine
VIII. Thyroid Gland
Bilobed and anterior to trachcea
Level 5, 6, and 7 vertebrae
Thyroid arteries
Thyroxine and 3, 5, 3’-Triiodothyronine = T4 (inactive) and T3 (active)
Tyrosine combine with iodine (I2) to form MIT and DIT
DIT and DIT transform to T 4 (non-calorigenic)
DIT and MIT transform to T 3 (calorigenic)
Hoang 15
Follicular cells
Stimulated = elongated
Calorigenesis and growth effects
Utilization of energy substrates
O2 Consumption
Increase heat production and basal metabolic rate
Calcitonin
Parafollicular cells
Calcium regulation
Need 1 mg iodide per week
Seafood, milk, bread, iodized salt
30 % goes to thyroid
50-70% excreted out via kidneys
Thyroid gland use iodide trapping
Against concentration and electrochemical gradients
Basal level of thyroid hormone synthesis
Maintained by sympathetic nervous system
Thyroid peroxidase
Convert iodide ions to I2
Coupling to form T3, T4, and rT3
Iodinase
Combine tyrosine with I2
Thyroid stimulating hormone (TSH)
Increases iodide uptake
Activates thyroid peroxidase
Increases thyroglobulin synthesis
Activates iodinase
Increases secretion (not storage)
Most sensitive monitor of thyroid function
Free T4 decrease 50% = TSH increases 100%
Negative feedback controlled by free T4
Goiter = enlarged thyroid via stimulation of TSH-receptors
Normal euthyroid
90% T4
10% T3
< 0.1% reverse T3
T4 convert to T3 majority
In starvation = T4 converts to r T3 (non-calorigenic)
Conserve food energy stores
Iodide deficiency
Total T4and T3 production/secretion decrease
More T3 synthesized and secretion relative to T 4
50% T3 and 50% T4
Lack of iodine
No T3 or T4 production/secretion
T3 and T4
70% bound to thyroxine binding globulin (TBG)
20% bound to transthyretin (TBP)
10% bound to albumin
0.03% total plasma T4 in free state
Hoang 16
0.3% total plasma T3 in free state
Binding proteins maintain large storage pool of T 4 and T3
Prevent metabolism in liver or excreted in urine
Target cell
Liver – Kidney – Muscles
Entry via diffusion or carrier-mediated transport
T3 = nuclear receptor, biologically active, multiple cellular actions
Calorigenesis
Stimulate appetite
Growth effects
Bone growth and tooth eruption
Stimulate GH and IGF-1 production
Nervous system
Normal development
Neural branching and differentiation
Nerve myelination
Enhances effects of epinephrine and glucagons = substrates for oxidation
Inhibit pituitary (major) and hypothalamus (minor)
Hyperthyroidism = increased metabolism
Increased appetite
Weight loss
Heat intolerance
Increased sweating
Grave’s Disease
Autoantibody to TSH-receptor
Mimics TSH = agonist
Thyroid-stimulating immunoglobulins
Increased T3/T4 in plasma
Decreased TSH in plasma
Goiter
Exophthalmos = protrusion of eyes
Stimulate extraocular muscles = increased mucopolysaccharides
Double vision
Hypothyroidism = decreased metabolism
Reduced appetite
Weight gain
Cold intolerance
Deceased sweating
Autoantibody to iodinase and thyroid peroxidase
Decreased T3/T4 in plasma
Increased TSH in plasma
Goiter
Increased production of thyroglobulin
Autoantibody to thyroid = Hashimoto thyroiditis
Destruction of thyroid follicular cells
No production of T3/T4 = no negative feedback to anterior pituitary
Increased TSH in plasma
No goiter long term
Subclinical hypothyroidism
Increased TSH
Normal T4/T3
Hoang 17
Subclinical hyperthyroidism
Decreased TSH
Normal T4/T3
Iodide deficiency
Decreased T3/T4 secretion in plasma
Decreased negative feedback to anterior pituitary = thyrotrophes
Goiter
Increased thyroglobulin production
IX. Calcium and Phosphate Regulation
Blood plasma
Ca2+ free = 50% biologically active
Ca2+-Albumin = 40%
Ca2+-Citrate and Ca2+-Phosphate = 10%
Normal range = 8.8 – 10.6 mg/100ml
Parathyroid Hormone
84 amino acid peptide
MW 9,600
Synthesized by chief cells
rER
Pre-Pro-hormone = 115 aa
Pro-hormone = 90 aa
Golgi
PTH = 84 aa
Stored in secretory granules or degraded to amino acids
Function to increased plasma Ca2+
Kidney
Stimulate calcium reabsorption
Distal tubule and loop of Henle
Result in decreased excretion of calcium
Decrease phosphate reabsorption
At proximal and distal tubules
Result in increased excretion of phosphate
Increase level of free Ca2+
Low phosphate levels = activate renal-1 hydroxylase
Convert vitamin D to calcitrol
Bone
Increase osteocytic osteolysis (osteocytes) = rapid
Increase osteoclast activity = long-term
Remove Ca2+ and phosphate
Amplified via calcitrol
Intestine
Via calcitrol = increase Ca2+ and phosphate absorption from diet
Stimulated by epinephrine via adenylyl cyclase and cAMP
Act on bone, kidney, and intestine (indirectly via calcitrol)
Induce osteocytic osteolysis
Osteocytes remove calcium from bone via canaliculi
Inhibited by high plasma calcium levels
Ca2+ bind Ca2+-sensing receptors
Activate PLC into IP3 and PKC
PKC inhibits PTH transcription and release
Stimulate degradation of PTH and Pro-PTH
Hoang 18
Low plasma Ca2+ levels
Stimulate PTH synthesis and secretion
7-dehydrocholesterol converted to Vitamin D3 via UV light
Vitamin D3 binds to 2-globulin binding protein
Transported to liver
Vitamin D3 converted to 25-(OH)-D3 via 25-hydroxylase
Transported by 2-globulin to kidney
25-(OH)-D3 converted to calcitrol
Transported to bone and intestine by transcalciferin 2-globulin binding protein
Renal-1-hydroxylase = convert 25-(OH)-D3 to 1,25-(OH)2D3
Stimulated by vitamin D deficiency, PTH, low renal phosphate levels
Renal-24-hydroxyalse = convert 25-(OH)-D3 to 24,25-(OH)2D3
Stimulated by vitamin D sufficiency, low PTH, high renal phosphate levels
Vitamin D metabolite [1,25 (OH)2D3]
Found in skin and diet
Liver, fish, and milk
Vitamin D2
Plants and diet
Long-term negative feedback on PTH synthesis
Inhibit transcription in chief cells
Epinephrine
Bind b-adrenergic receptor
G-protein activate adenylyl cyclase
Increase cAMP levels stimulate PTH secretion
Pheochromocytoma = hypercalcemia
Calcitonin
32 amino acids (MW = 3200)
Synthesized by parafollicular cells of thyroid
Function to decrease plasma Ca2+
Act primary at bone
Inhibit bone resorption
Inhibit osteocytic-osteolysis and osteoclast activity
Stimulate osteoblasts
Weak acting at kidneys
Secretion occurs at 10mg/ 100mls plasma Ca2+
Rare in adults due to slow bone remodeling
Greater role in children
Osteoblast secrete collagen
Collagen polymerizes to osteoid matrix with calcium, phosphate, hydroxyl, and bicarbonate ions
Hydroxyapatite becomes new bone
Primary hyperparathyroidism
Parathyroid adenoma = secrete excess PTH
Increased
Serum Ca2+ = hypercalcemia
Serum urinary phosphate excretion
Urinary cAMP
Decreased
Serum P = hypophosphatemia
Urinary Ca2+ excretion = increased Ca2+ reabsorption (hypercacinuria long-term)
Kidney stones
Hoang 19
Secondary hyperparathyroidism
Renal failure
Increased
PTH secretion
Bone resorption
Decreased
Lack of 1,25-(OH)2D3 production
Lack of negative feedback to PTH
Serum Ca2+
Malignancy-associated hypercalcemia
Carcinomas = secrete PTH-related peptide (PTH-rp)
Mimic PTH
Increased
Serum Ca2+ = hypercalcemia
Hypoparathyroidism
Thyroid surgery or congenital
Increased
Serum P
Decreased
Serum Ca2+ and tetany
Urinary phosphate excretion
Pseudohypoparathyroidism type Ia
Defective G protein in kidney and bone
End-organ resistance to PTH
Hypocalcemia and hyperphosphatemia
Elevated PTH levels
Rickets (children) /Osteomalacia (adults)
Lack of Vitamin D
Inability to form 1,25-(OH)2D3
Vitamin D resistance
Increased
PTH secretion
Increased phosphate excretion
Increased Ca2+ reabsorption
Decreased
Absorption of Ca2+ and phosphate
Plasma phosphate levels
Osteoporosis
Decrease in bone mass
Estrogen
Inhibits PTH stimulated bone resorption
Inhibits osteoclasts
Promote PTH-stimulated 1,25-(OH)2D3 synthesis
Testosterone
Cushing Syndrome
Excessive glucocorticoids
Decreased Ca2+ reabsorption in kidney
Decreased Ca2+ absorption in intestine
Hyperparathyroidism = increased PTH secretion
Hoang 20
Calcium Regulatory Hormones and Action
PTH
(peptide)
Stimulus
Vitamin D
(peptide)
Calcitonin
[1,25 (OH)2D3]
Decreased serum Ca2+
Decreased serum Ca2+
Decreased serum P
Increased PTH
Increased serum Ca2+
Increased resorption
Increased resorption
Decreased resorption
Decreased P reabsorption
Increased Ca2+ reabsorption
Increased P reabsorption
Increased Ca2+ reabsorption
Increased Ca2+ absorption via
vitamin D
Increased Ca2+ absorption
Increased P absorption
Increase Ca2+
Increase Ca2+
Decrease P
Increase P
Action
Bone
Kidney
Intestine
Overall effect on
Serum Ca2+
Serum Phosphate
X. Plasma Glucose Regulation
Islets of Langerhans = interconnected by gap junctions with portal blood supply
Alpha cells
Secrete glucagon
Beta cells
Secrete insulin
Delta cells
Secrete somatostatin and gastrin
Pancreatic polypeptide cells
Secrete pancreatic polypeptide
Blood flows from center to periphery of islet of Langerhans
Beta cells influence Alpha and Delta cells
Glucagon (Alpha)
Peptide hormone
Prohormone cleaved to glucagons and GLP (glucagons-like product)
GLP-1 and GLP-2 are incretins = stimulate insulin release
Acts only on liver
Via cAMP
Increase blood glucose
Increase glycogenolysis
Prevent recycling of glucose into glycogen
Increase gluconeogenesis
Decrease production of fructose 2,6-bisphosphate
Decrease phosphofructoskinase
Inhibit glucose reuptake in liver
Increase blood fatty acid and ketone levels
Decrease Ca2+
Hoang 21
Increase lipolysis
Ketone production from acetyl-CoA
Increase urea production
Amino groups incorporated into urea
Stimulus
Hypoglycemia
Decrease glucose in plasma
Exercise
Sympathetic = NE and Epi
Amino acids
Inhibition
Increase blood glucose
Somatostatin
Ketones and fatty acids
Parasympathetic = acetylcholine
Insulin (Beta)
A and B chain = joined by two disulfide bridges
Proinsulin synthesized as single chain
Connecting peptide is cut in storage granules
C-peptide cleaved and combines with zinc
Measure C-peptide as indication of Beta cells efficiency
Secretion from mature granules only
Decrease blood glucose
Glucose metabolized by beta cells = stimulate insulin secretion
GLUT-2 = insulin-independent glucose receptor
Increase in intracellular ATP
Ca2+ channels out with influx of Ca2+
Promote glycogen synthesis in muscle and liver
Inhibit glycogenolysis
Decrease gluconeogenesis
Increase production of fructose 2,6-bisphosphate
Increase phosphofructokinase activity
Decrease blood fatty acid and ketone levels
Inhibit lipolysis
Modest increase in plasma glucose = drastic increase in insulin
IV Glucose = biphasic
Initial burst of insulin from stored granules
Slow release aftermath result from insulin synthesis
Oral Glucose = monophasic
Glucose stimulate incretins (CCK, GLP-1, GTP)
Diabetic with oral glucose
Slow increase in glucose
Little insulin response
Stimulus
Increase glucose in plasma
Amino acids
Cortisol and GH
Parasympathetic = acetylcholine
GI hormones = Gastrin, secretin, CCK
Inhibition
Decrease glucose in plasma
Exercise
Somatostatin from liver
Sympathetic = NE and Epi
Hoang 22
Glucagon
Insulin
Decrease glucose uptake
Increase glucose uptake
Increase glycogenolysis
Increase glycogen synthesis
Increase gluconeogenesis
Decrease gluconeogenesis
Increase fatty acid uptake and ketones secretion
Decrease fatty acid and ketones
Increase urea production
Increase fat deposition
Increase lipolysis via hormone-sensitive lipase
Decrease lipolysis
Decrease blood amino acid levels
Decrease blood K+ levels
Insulin receptor
Tetramer of two  and two  subunits
 subunit = tyrosine kinase activity
Autophosphorylation
Insulin-receptor complex become internalized
Down-regulates receptors
Increase in receptors during starvation
Decrease in receptors during obesity
Receptor regulation mechanisms
Internalization of receptor when insulin binds
Recycling of internalized receptor (or degraded)
Synthesis of new insulin receptors
Insulin translocates inactive glucose transporters to cell membrane to become active
GLUT-4 = only insulin-sensitive glucose transporter
Normal controls = 5 % of receptors are activated for maximum response
Type 2 diabetic = higher insulin concentration for the same maximum response
Down regulation of receptors
Impairment of postreceptor signaling
Hepatocytes
GLUT-2 = insulin independent
Insulin binds to insulin receptor
Stimulates
Glucose to G-6-P
Glycogen synthesis
Glycolysis
G-6-P to pyruvate to acetyl CoA
Lipogenesis
Acetyl CoA to malonyl CoA to free fatty acids to triglycerides
Protein synthesis
Inhibits
G-6-P to glucose
Glycogenolysis
Glucogenogenesis
Lipolysis
Fatty acids to ketones
Protein metabolism
Hoang 23
Secrete LDL into blood for transport to adipose storage
Muscle
GLUT-4 = insulin dependent
Insulin binds to insulin receptor recruit GLUT-4 transporters
Stimulates
Glucose uptake
Glycolysis
Glucose to G-6-P
Glycogen synthesis
Protein synthesis
Inhibits
Lipolysis
Fatty acids to acetyl CoA
Protein metabolism
Adipocyte
GLUT-4 = insulin dependent
Insulin binds to insulin receptor recruit GLUT-4 transporters
Stimulates
Glucose uptake
Glycolysis
Glucose to G-6-P
Pyruvate to acetyl CoA
Lipogenesis
Malonyl CoA to fatty acids to triglycerides
Free fatty acids uptake
Lipoprotein lipase synthesis
Move to endothelial cells
Convert LDL to fatty acids for uptake
Inhibits
Lipolysis
Hormone-sensitive lipase
Triglycerides to fatty acids and glycerol
Insulin Deficiency
Glucose-sparing
Liver secrete glucose into blood
Glucose is inhibited for uptake in liver, muscle, and adipose
Liver = secrete glucose and ketones
Uptake amino acids for gluconeogenesis
Increased nitrogen into urea synthesis
Uptake fatty acids for ketogenesis
Blood ketone level increase
Adipose = secrete fatty acids
Muscle = secrete amino acids
Uptake ketones for energy
Hypoglycemia
Activate VM hypothalamus
Increased sympathetic activity
Activate alpha (glucagons release)
Inhibit beta (decrease insulin release)
Increased epinephrine release
Muscle
Increase glycogenolysis
Decrease glucose uptake
Adipose
Hoang 24
Increase lipolysis
Liver
Increase glycogenolysis
Increase gluconeogenesis
Hyperglycemia
Insulin deficiency
Decreased glucose uptake in cells
Hypotension
ECF volume contraction
Metabolic acidosis
Overproduction of ketoacids
Hyperkalemia
Lack of insulin
Decreased potassium uptake in cells
Stomatostatin (Delta)
Pancreas
Inhibit secretion of insulin, glucagons, and gastrin
Inhibits absorption of glucose
XI. Steroidogenesis
Rate limiting step
Cholesterol to pregnenolone via P450scc
Zona glomerulosa
Mineralocorticoid = aldosterone
Zona fasciculata
Glucocorticoids = cortisol
Zona reticularis
Androgens = DHEA and androstenedione
Acetate converted to mevalonic acid via HMG-CoA reductase
Mevalonic acid form cholesterol LDL
Excess LDL negative feedback on HMG-CoA reductase
Cholesterol esters convert to cholesterol in lipid droplet
Cholesterol with SCP2 transport to mitochondrial via StAR
Cholesterol converts to Pregnenolone via P450scc, Adx, and AdRex
Chronic steroidogenic response
Slow induction by cAMP
Synthesis of steroidogenic enzymes
Occurs hours-days
In all steroidogenic tissue
Acute steroidogenic response
Rapid induction by cAMP
Substrate availability
Occurs in 15 mins
In adrenal and gonads
Inhibited by CHX
Regulation of Steroidogenesis
Hypothalamus
CRF and AVP
Pituitary
ACTH and proopiomelanocorticortin
Adrenal
Glucocorticoid feedback
Hoang 25
Fast feedback
Seconds to minutes
Proportional to dose and duration
Delayed feedback
Hours to days
Proportional to dose and duration
Administration
Mechanism by glucocorticoid receptor = inhibit transcription of genes
Mineralocorticoid = renin-angiotensin
Androgens and adrenarche
Lipoid congenital adrenal hyperplasia
Inability to convert cholesterol to pregnenolone
Enlarged adrenals = cholesterol esters
Low steroid hormones
Salt-wasting syndrome = lack of aldosterone
Males and females both have normal female external genitalia
XI. Contraception
Reproductive age death
Highest relative risk = pregnancy
Oral contraceptive
Induced abortion
No contraception
Small absolute risk
Contraceptive use
Highest = birth control pills
Lowest = female condoms
Oral contraceptive
Venous thrombic embolism
Oral contraceptives increase 3 fold
Pregnancy increase 6 fold
Increase risk of cervical cancer over time of use
Contraindications
Vascular disease
Diabetes
Older than 35
Smoker
Hypertension
Pregnancy
Estrogen dependent neoplasia
Vasectomy
Highly effective
No increase in prostate cancer
Anti-sperm antibodies
Human papilloma virus (HPV)
Type 16 = most associated with cervical cancer
Intrauterine device (IUD)
Highly effective
Spermicidal
Pregnancy = risk of septic abortion
Paragard = 10 years
Hoang 26
RESPIRATORY PHYSIOLOGY
I. Functional Anatomy
Anatomic dead space = 0.15 L
Alveolar ventilation = (tidal volume – dead space) x breaths/min
Muscles of inspiration
Diaphragm
Push abdominal contents downward
External intercostals
Pull ribs up and out
Used during exercise
Muscles of expiration
Passive movement
Abdominal muscles
Push diaphragm up
Internal intercostals muscles
Pull ribs down and in
Blood flow of pulmonary capillary = 0.75 sec transit time past alveoli
Normal = blood equilibrate with alveolar gas in 0.25 seconds
Z= 10
End of cartilage bronchi
Z = 16
End of terminal bronchiole
End of conducting airway
Z= 17
Beginning of alveolar air spaces = 3.0 L
Beginning of respiratory bronchioles
Surfactant = lipid dipalmitoyl phosphatidyl choline
Type II alveolar cells
Increase compliance
Diffusion layers of the alveoli = total distance of ½ m
Alveolar epithelium
Basement membrane
Interstitial space
Capillary basement membrane
Capillary epithelium
II. Ventilation, Compliance, and Resistance
Minute ventilation = tidal volume x breaths/minute
Tidal volume = 0.5 L
Volume of exhaled air with each breath
Vital capacity/ Forced vital capacity = 4.8 L
Volume of air that can be exhaled after maximal inspiration
Tidal volume + Inspiratory reserve volume + Expiratory reserve volume
Residual volume = 1.2 L
Volume of air remaining in lungs after maximal expiration
Expiratory reserve volume = 1.2 L
Maximal volume of air that can be exhaled at end of tidal volume
Inspiratory reserve volume = 3.1 L
Hoang 27
Volume of air that inhaled at end of a normal inspiration
Inspiratory capacity = 3.6 L
Maximal volume of air inhaled after normal expiration
Tidal volume + Inspiratory reserve volume
Functional residual capacity = 2.4 L
Volume of air remaining in the lungs at end of normal expiration
Expiratory reserve volume + Residual volume
Total lung capacity = 6.0 L
Total volume of air in the lungs after maximal inspiration
Forced expiratory volume
Volume of air expired in one second following maximal inspiration = 80 % of FVC
FEV1 / FVC = 0.8
Compliance
Change in lung volume per change in transpulmonary pressure
Greatest compliance in middle pressure range
Lowest compliance at high expanding pressures
C = Distensibility / Elastic recoil
Rest
Alveolar pressure = 0 cm H2O
No air movement
Pleural pressure = -5 cm H2O
Lung volume = FRC
Mid-inspiration point
Alveolar pressure = -1 cm H2O
Max air movement
Pleural pressure = -7 cm H2O
Lung volume > FRC
Maximal inspiration
Alveolar pressure = 0 cm H2O
No air movement in
Pleural pressure = -8 cm H2O
Lung volume = FRC + tidal volume
Mid expiration point
Hysteresis
Velocity greater at start of expiration than end
Alveolar pressure = 1 cm H2O
Max air movement out
Pleural pressure = -6 cm H2O
Lung volume > FRC
Rest
Airway resistance
Bronchial smooth muscle = radius of airway
Parasympathetic = constrict and decrease radius
Sympathetic = dilate and increase radius via  2 receptors
High lung volumes
Decrease airway resistance
Low lung volumes
Increase airway resistance
Density and viscosity of gas
Low-density helium reduce resistance
Highest resistance in medium-sized bronchi
III. Gas Exchange
Hoang 28
Diffusion rates
Dependent on partial pressure differences & diffusion surface area
Perfusion-limited exchange
Gas equilibrates along length of pulmonary capillary
Diffusion of gas increases when blood flow increases
Diffusion-limited exchange
Gas do not equilibrate along length of pulmonary capillary
Seen in fibrosis and emphysema
Diffusion of gas increases when capillary length increases
Increased lung volumes = expand alveoli = collapse pulmonary vessels
P50 = P02 at 50 % saturation
26 mm Hg
IV. Oxygen and CO2 Transport
Oxygen transport
Carried via dissolved in solution and bound to hemoglobin
Hemoglobin = 22
Fe2+ binds oxygen reversibly (Fe3+ binds irreversibly)
H+ from histidine is released = function in acid/base buffering
Increase oxygen-carrying capacity of blood 70x
1 gram Hb = binds 1.39 mL O2
15 grams Hb per deciliters (100 ml) of blood
20.85 mL O2 per dl of blood at 97.5 % Hb saturation
0.3 mL O2 dissolved in blood
In tissues (venous blood)
14.4 mL O2 per dl of blood at 75 % Hb saturation
Chronic hypoxemia
Occurs at high altitudes
Increase synthesis of 2,3-DPG
DPG bind hemoglobin to unload oxygen in tissues
CapacityO2 = (1.39 ml O2/g Hb/dl x [Hb] x % saturation) + (0.003 ml O 2/dl/mm Hg PO2)
% Saturation = (HbO2 bound / HbO2 capacity) x 100 %
Solubility of O2 at 37oC and 100 mm Hg
0.3 mL O2/dl of blood
Hemoglobin-O2 Dissociation Curve
Shifts Left
Shifts Right
Increased pH
Decreased pH
Decreased PCO2
Decreased temperature
Decreased 2,3-DPG
Increased PCO2
Increased temperature
Increased 2,3-DPG
Hemoglobin F
Carbon monoxide
CO2 transport
Hoang 29
Dissolved CO2
Carbaminohemoglobin
HCO3- (major form)
CO2 in tissues = diffuse to plasma
CO2 combines with H2O forming H2CO3
Via carbonic anhydrase
Dissociates into H+ and HCO3Chloride shift
HCO3- leaves RBC to plasma in exchange with ClHaldane effect
Carbamino compound formation weakens affinity to O 2
Unload O2 from hemoglobin in high levels of CO2
Body regulates [H+] NOT pH
Buffer systems
Phosphate (weak)
pK = 6.8
Stronger system in kidneys
Protein (strong)
Hemoglobin = highest levels in blood
Imidazole group = buffer
Myoglobin
Bicarbonate
Open system
Lungs = CO2
Kidneys = H+ and HCO3PaCO2 = k x (VCO2/VA)
VA (alveolar ventilation) regulate [H+] via its control of PaCO2
V. Pulmonary Circulation
Pulmonary pressure much lower than systemic circulation
Balanced by a lower resistance than systemic
Equal cardiac output for systemic circulation
V/Q = 1 at 3rd rib level
Q changes more rapidly than V
Zone 1
Blood flow lowest at apex of lung
Highest V/Q = due to gravity effects
Highest PO2 = 132 mm Hg
Lowest PCO2 = 28 mm Hg
Alveolar pressure > arterial pressure > venous pressure
Zone 2
Arterial pressure > alveolar pressure > venous pressure
Zone 3
Blood flow highest at base of lung
Lowest V/Q = due to gravity effects
Lowest PO2 = 89 mm Hg
Highest PCO2 = 42 mm Hg
Arterial pressure > venous pressure > alveolar pressure
Hypoxia
Local vasoconstriction in pulmonary blood flow
Vasodilation in systemic circulation
Fetal pulmonary vascular resistance
High = low ventilation in fetal lungs
Hoang 30
Shunts
Right-to-Left
Decrease in arterial PO2
Left-to-Right
Increase in PO2 on right side of heart
VI. Ventilation/Perfusion Defects
Normal V/Q = 0.8
Arterial PO2 = 100 mm Hg
Arterial PCO2 = 40 mm Hg
Airway obstruction
Low V & V/Q
No gas exchange
Blood flow obstruction
Low Q & high V/Q
No gas exchange
R = respiratory quotient (rate of CO2 production per O2 consumption)
R = 0.7 for fat
R = 0.8 for proteins
R = 1.0 for carbohydrates
R = 0.85 for mixed protein, fat, and carbohydrate diet
VII. Breathing Regulation
Central control of breathing
Medullary respiratory center = reticular formation
Dorsal respiratory group
Nucleus tractus solitarius
Rhythmic drive to contralateral phrenic motor neurons
Project to VRG
Inspiration & generates basic breathing rhythm
Afferents = vagus (X) & glossopharyngeal (IX) nerve
Efferents = phrenic to diaphragm
Ventral respiratory group
Nucleus ambiguous & nucleus retroambiguous
2/3 of VRG neurons = expiratory (active)
1/3 of VRG neurons = inspiratory
Nucleus ambiguous
Vagus nerve
Auxiliary respiratory muscles
Larynx via recurrent laryngeal nerve
Nucleus retroambiguous
Stimulate external, internal intercostals, and abdominal expiratory muscles
Project to pons and DRG
Apneustic center = lower pons of cerebral peduncles
Stimulate inspiration
Apneusis = deep prolonged inspiratory grasp
Tonic diaphragmatic contraction
Pneumotaxic center = upper pons of nucleus parabrachialis
Inhibit inspiration and stimulate expiration
Regulate inspiratory volume and respiratory rate
Cortex
Voluntary control
Slowly adapting pulmonary stretch receptors (PSR)
Hoang 31
Smooth muscle of airways = mechanoreceptors
Myelinated neurons
Stimulated by distention or lung inflation
Over-inflation of lungs negative feedback
Decreased activity with deflation
Decreased activity with increased airway CO2
Continuous activity with sustained inflations
Hering-Breuer reflex = decrease in breathing frequency by distention
Inspiratory inhibitory
Insensitive to chemicals
Rapidly adapting pulmonary stretch receptors (RAR)
Between airway epithelial cells = mechanoreceptors
Stimulated by rate of change of lung volume (I to E, E to I)
Some discharge with normal tidal ventilation
Myelinated neurons
Irritant receptors
Stimulated by noxious substances
Coughing
Inspiratory excitatory
Augment inspiration
Shorten expiratory duration
C-Fibers
Unmyelinated slow fibers
Between alveolar membrane and capillary
Chemosensitive
Bronchial C-fibers = bronchial circulation
Respond to histamine and prostaglandins
Cardiac slowing
Increased blood pressure
Hyperpnea & coughing
Pulmonary C-fibers = pulmonary circulation
J receptors
Alveolar walls close (juxtaposition) to capillaries
Stimulated by engorgement of pulmonary capillaries
Result in rapid, shallow breathing
Seen in pulmonary edema
Respond to histamine, prostaglandins and pulmonary congestion
Cardiac slowing
Decreased blood pressure
Apnea
Joint and muscle receptors
Activated during limb movements
Exercise
Chemoreceptors = CO2, H+, and O2
Central chemoreceptors = medulla
pH of CSF
H+ do not cross blood-brain barrier
CO2 diffuse readily through blood-brain barrier
CO2 combines with H2O to produce H+ and HCO3Resulting H+ act on central chemoreceptors
Chronic high levels of CO2 = desensitization
Kidneys increase blood bicarbonate = lower H+ levels in CSF
Peripheral chemoreceptors = carotid and aortic bodies
Decreased arterial PO2 = stimulate breathing
PO2 below 60 mm Hg
Denervation possible with inhalation of 100% O2
Hoang 32
Increased arterial PCO2 = stimulate breathing
Increased arterial [H+] = stimulate breathing and carotid bodies
Carotid bodies = high metabolic rate
Greatest blood flow per gram = small arterial-venous difference
Exercise
Increase in ventilation and perfusion
Pulmonary blood flow increases
Cardiac output increases
Decrease in physiologic dead space
No change in arterial PO2 and PCO2 during exercise
Strenuous exercise cause lactic acidosis = drop in arterial pH
Venous PCO2 increases
High altitudes
Decreased alveolar PO2 and arterial PO2 (hypoxemia)
Stimulate peripheral chemoreceptors
Increase ventilation = hyperventilation
Respiratory alkalosis = treated by acetazolamide
Stimulate erythropoietin = increase RBC production
Increased O2-carrying capacity
2,3-DPG increases
Pulmonary vasoconstriction = increase pulmonary artery pressure
Hypertrophy of right heart
VIII. Pathology
Fibrosis
Restrictive lung disease
FEV and FVC reduced
Could have higher-than-normal expiratory flow rate
Low compliance
“Cement lung”
Need greater pressure for same volume change
Asthma
Obstructive lung disease
FEV reduced more than FVC
FEV/FVC decrease
Decreased ventilation
Decreased V/Q
Easier to breathe at high lung volumes = decrease airway resistance
High compliance
No elastic tissue = no elastic recoil
Inspiration easy but exhalation difficult
Albuterol
Sympathetic agonist
Dilate bronchi via relaxing smooth muscle
Chronic obstructive pulmonary disease (COPD)
High airway resistance
Decreased V/Q
Emphysema
High compliance
Higher FRC = barrel-shaped chest
Chronic infection – Airway obstruction – Destruction of alveolar walls
Increased airway resistance – Abnormal V/Q – Increased pulmonary vascular resistance
Pulmonary hypertension
Pulmonary hypertension
Smooth muscle hyperplasia and hypertrophy of pulmonary capillaries
Hoang 33
Increased pulmonary capillary resistance
Right heart backup
Low Q
Left heart failure
Blood back up in pulmonary circulation
Increased Q
Edema into alveoli
Pneumonia
Inflammation of lungs due to bacterial/viral infection
Consolidation = alveoli become filled with fluid and blood cells
Post-praudial dyspnea (“after eating” SOB)
Shortness of breath
GI tract filled = block diaphragm from contracting
Decreased V
Blood pool in lungs
Edema of alveoli
Fen-Phen
Weight losing drug via serotonin production and increased a-adrenergic activity
Reduce appetite
Act on endothelial cells of lungs
Serotonin cause vasoconstriction
Increased vascular resistance
Decreased Q
Cheyne-Strokes breathing
Respiratory disorder of periodic breathing
Cycle = breathes deeply then decrease to almost no breathing
Due to slow blood flow (cardiac failure) or increased negative feedback (brain damage)
Atelectasis = collapse of alveoli
Bronchial obstruction = air in alveoli is absorbed (negative pressure)
Lack of surfactant = seen in hyaline membrane disease
Hypoxemia
Decreased PaO2 or decreased PIO2 = low PaCO2
High altitudes
Hypoventilation = high PaCO2
Airway obstruction = asthma
Weakness of respiratory muscles
Decreased respiratory drive from CNS
Diffusion limitation
Pulmonary edema
Shunts
Right-to-left = atrial septal defect
Bronchial circulation via thebesian veins
Patient inhales 100% O2
1% shunt for every 20 mm Hg difference between P AO2 and PaO2
Metabolic = change in body metabolism resulting in abnormal pH
Respiratory = change in breathing resulting in abnormal pH
Respiratory academia
Pulmonary perfusion inadequacy
Insufficient ventilation
Severe pulmonary edema
Pneumonia
Hypoventilation
CNS defect
Depressants and anesthesia
Hoang 34
Cervical spinal cord injury
Metabolic academia
Retention of acid
Ketoacidosis
Chronic renal failure
Addison’s disease
Loss of base
GI disorders
Acid ingesting
Renal tubular acidosis
Respiratory alkalemia
Hyperventilation = CNS
Congestive heart failure
Metabolic alkalemia
Loss of acid
GI disorders
Intake of base
Excessive minerocorticoid activity
Potassium depletion
Blood transfusion
Normal
pH
PCO2
[HCO3-]
7.4
40 mm Hg
24 mEq/L
[H+] = 40 nEq/L
Respiratory acidosis
Decrease
Increase (primary)
Increase (secondary)
Metabolic acidosis
Decrease
Decrease (secondary)
Decrease (primary)
Respiratory alkalosis
Increase
Decrease (primary)
Decrease (secondary)
Metabolic alkalosis
Increase
Increase (secondary)
Increase (primary)
IX. Laws
Boyle’s Law
PiVi = PfVf while temperature is constant
Poiseuille’s Law
Air flow V =  Pr4 / 8l
Fick’s Law
Diffusion = SA / thickness of membrane
Laplace’s Law
P = 2 x Surface Tension / radius of alveolus
Dalton’s Law
Partial pressure = total pressure x fractional concentration
Hoang 35
COPD with fatigue, dyspnea at rest, & peripheral edema
Low PaCO2 and PaO2
pH = 7.6
Decreased diffusion capacity
Hypoxemia
V/Q mismatch
Increased alveolar ventilation
Increased RBC production
Increased right atrial pressure
Respiratory alkalosis
Increased Hematocrit
Increased bicarbonate
Right heart failure & respiratory acidosis
Normal A-a gradient
Right heart failure
Pulmonary edema
Hypoventilation
Anaerobic metabolism
Metabolic acidosis
Increased PaCO2
Increased bicarbonate
Respiratory acidosis
Normal anion gap
Decreased chloride
High A-a gradients are associated with oxygen transfer / gas exchange problems. These are usually associated with
alveolar membrane or interstitial disease.
High gradients result from impaired diffusion or, more commonly, by ventilation-perfusion inequality of the
"shunting" variety.
Hypoxemia in the face of a normal A-a gradient implies hypoventilation with displacement of alveolar O2 by CO2 or
other substance.
Hoang 36
Respiratory muscle weakness
Decreased tidal volume & vital capacity
Increased PaCO2
Hypoventilation
Decreased hemoglobin saturation
Decreased arterial pH
Severe acute asthma
Non-anion gap metabolic acidosis
Asthma attack
Decreased bicarbonate
Non-anion gap
Hyperventilation
Decreased PaCO2
[3 days later]
Metabolic acidosis
Hypocapnia
Respiratory alkalosis
Hoang 37
CARDIOVASCULAR PHYSIOLOGY
I. Functional Anatomy
Endocardium
Myocardium (Cardiac muscle)
Striated with intercalated discs at Z line
Gap junctions
Low electrical resistance
Maintain cell-to-cell cohesion
Sarcomere
From Z line to Z line
Thick filaments = myosin
Thin filaments = actin, troponin, tropomyosin
Atrioventricular bundle = conducts impulses from atria to ventricles
Resting membrane potential = -85 to –95 millivolts
Action potential = 105 millivolts
Fast sodium channels
Open at initiation of action potential
Slow calcium-sodium channels
Maintain plateau
Calcium stimulate muscle contraction of myofibrils
Transverse tubules cause sarcoplasmic tubules to release calcium into reticulum
Well developed in ventricles rather than artia
Epicardium
Cardiac cycle
P wave
Depolarization of atria
Initiates atrial contraction (systole)
Contribute to ventricular filling
Atrial pressure increases after P wave
QRS wave
Ventricular depolarization = 0.16 sec after P wave
Initiates ventricular contraction (isovolumetric)
T wave
Repolarization of ventricle
Aortic & pulmonic valve close
Causes dicrotic notch
Atria function as primer pump for ventricles
75 % ventricular filling occurs during diastole before atrial contraction
25 % ventricular filling occurs via atrial contraction
Ventricles fill during diastole
During systole
A-V valves closed
Atria fill
Beginning diastole
Ventricular pressure decreases below atria
A-V valves open
First 1/3 diastole
Rapid filling of ventricles
Last 1/3 diastole
Hoang 38
Atrial contraction = 25 % of ventricular filling
Diastasis = slow ventricular filling
Beginning systole
Ventricular contraction
A-V valves close & pressure builds in ventricles
Period of isovolumic contraction = 0.2 to 0.3 seconds of ventricular contraction
Middle systole
Left ventricular pressure > aortic pressure (80 mm Hg)
Right ventricular pressure > pulmonary artery pressure (8 mm Hg)
Aortic & pulmonary valves open
Period of rapid ejection to period of slow ejection
Aortic pressure > ventricular pressure
Momentum push into aorta
Last systole
Period of isovolumic relaxation
Ventricular pressure < aortic & pulmonary artery pressure
Aortic & pulmonary valves close
Ejection fraction = fraction of end-diastolic volume ejected (60 %)
End-diastolic volume = 110 to 120 mL
Stroke volume = 70 mL
End-systolic volume = 40 to 50 mL
Systolic pressure
Ventricular ejection increases aorta pressure
Exceeds diastolic pressure in aorta
Opens aortic valve
Aorta pressure increases to 120 mm Hg
Afterload = pressure in artery exiting ventricle (aorta or pulmonary)
Diastole pressure
Blood flow to peripheral circulation
Arterial pressure decreases to 80 mm Hg
Preload = end-diastolic pressure
Atrial pressure waves
a wave = atrial contraction
c wave = ventricular contraction
v wave = in-filling of atria from venous return
Frank-Starling mechanism
Increased venous return = stretches heart muscle
Heart pumps with greater force of contraction
Heart pumps all blood that comes to it
Without allowing excess accumulation of blood in veins
Sympathetic
Heart rate increase from 72 to 180-200 beats per minute
Parasympathetic
Mainly affects atria
Decrease heart rate and force of contraction of ventricles
Inhibit sympathetic stimulation
Ion responses
Excess potassium
Flaccid heart
Reduced heart rate
Decrease in contractility
K+ decreases resting membrane potential = decreased action potential intensity
Hoang 39
Excess calcium
Spastic contraction
Low calcium
Flaccid heart
Acute increased temperature = increased heart rate
Increased permeability of muscle membrane to ions
Decreased temperature = decreased heart rate
II. Heart Rhythm and Conduction System
Sinoatrial node
Initiates cardiac impulse
Controls rate of heartbeat = pacemaker
Discharge faster than other cardiac tissues
Membrane potential = -55 to -60 mV
Due to fast sodium channels = slow leakage of sodium into cell fiber
Until potential reaches -40 mV
Calcium-sodium channels activated
Rapid entry of sodium and calcium = action potential
Potassium channels open = potassium escape cell
Membrane potential return to resting potential
Internodal pathway = conduct impulses from sinus node to A-V node
Atrioventricular (A-V) node
Delays impulses from atria to ventricle = 0.09 seconds
Allows atria to empty into ventricles
Delay resulting from:
1. Resting membrane potential is less negative in A-V node & bundle than normal cardiac
muscle
2. Fewer gap junctions between cells in A-V node & bundle = greater resistance
A-V bundle = conducts impulse from A-V node to ventricles
Purkinje fibers = left and right bundles conduct impulses to all parts of ventricles
Lie under endocardium
Rapid transmission of impulse
High permeability of gap junctions at intercalated discs
Atrial and ventricular syncytia
Separated and insulated from one another
Fibrous barrier that acts as insulator
Force atrial impulses to enter ventricles through A-V bundle
Parasympathetic = Vagal (acetylcholine)
Decrease rate of sinus node discharge
Decrease excitability of fibers between atrial muscle and A-V node
Increase permeability of potassium in sinus node and A-V fibers = hyperpolarization
Decreased membrane potential of sinus nodal fibers to -65 to -75 mV
Sympathetic (norepinephrine)
Increase rate of sinus node discharge
Increase cardiac impulse conduction rate
Increase force of contraction of atria and ventricles
Increase permeability to sodium and calcium
Increase resting membrane potential = more excitable
Ventricular escape
Lack of impulses from atria
Purkinje fibers develop own rhythm at 15-40 beats per minute
Hoang 40
III. Electrocardiogram
Depolarization
Electrical current flows from base toward apex of heart
Normal P-Q = 0.16 seconds
Normal Q-T = 0.35 seconds
Einthoven’s law
Electrical potential of any limb equals the sum of potentials of the other two limb leads
Abnormal waveforms
QRS
Increased voltage
Hypertrophy of heart
Decreased voltage
Old myocardial infarctions (decreased cardiac muscle mass)
Fluid in pericardium = pleural effusion
Prevents voltage from reaching body surface
Pulmonary emphysema
Prolonged conduction
Hypertrophied and dilated hearts (one or both ventricles)
Longer distance for impulse to travel
Blockade of impulses in Purkinje system
T
Inverted
Ischemia
IV. Heart Sounds
First heart sound
Closure of A-V valves
Beginning of ventricular systole
Ventricular contraction
Louder with higher pitch
Second heart sound
Closure of aortic and pulmonary valves
Semilunar valves
End of ventricular systole
Third heart sound
Maximal ventricular dilation
End of rapid ventricular filling
In-rushing of blood into ventricles
Beginning of middle third of diastole
Fourth heart sound
Atrial contraction
Abnormal = due to stiff heart
Failure of ventricle to fill during rapid ventricular filling
Large blood volume left in atrium
Acceleration of blood into ventricles cause sound
Last third of diastole
Pathologic valvular disease
Rheumatic fever
Stenotic valves
Insufficient valves
Heart murmurs
Hoang 41
Aortic stenosis
Left ventricular hypertrophy
Chronic increase in blood volume
Chronic increase in left atrial pressure
Due to hypervolemia
Angina pectoris pain
Aortic regurgitation
Increased stroke volume
Left ventricular hypertrophy
Decreased diastolic pressure
Increased blood volume
V. Cardiac Arrhythmias
Pathological ECGs
Etiology
ECG Signs
First-degree block
Longer PR interval (normal rate and rhythm)
Second-degree block: Mobitz Type I
(Wenckebach block)
PR – Long PR – Longer PR – No PR – cycle starts over
Second-degree block: Mobitz Type II
Normal PR – P wave but no QRS – cycle starts over
Third-degree block
P at regulate rate & rhythm
QRS-QRS at its own rate & rhythm
Wolff-Parkinson-White Syndrome
Shortened PR due to delta wave
Bundle Branch block
Right bundle branch = Wide QRS with dip in middle
Left bundle branch = Wide QRS with little or no dip
Atrial Fibrillation
Irregular R-R intervals
No detectable P waves (cancel each other out)
Atrial Flutter
Several P waves with normal QRS-T complex (2-3:1)
Tachycardia
Increased in heart rate greater than 100 beats per minute
Increased body temperature
Sympathetic stimulation
Due to blood loss
Digitalis intoxication = biphasic T waves (normal T wave followed by inverted T wave)
Bradycardia
Decreased heart rate less than 60 beats per minute
Carotid sinus syndrome
Artherosclerotic plaque stimulate vagus
A-V blocks
Inhibits impulses from SA node
Inflammation or ischemia of A-V node or bundle
Compression of A-V bundle
Strong vagal stimulation
Hoang 42
Atrial paroxysmal tachycardia
Inverted P wave
Tachycardia originate in atrium
Supraventricular tachycardia
Ventricular paroxysmal tachycardia
Originate in ventricles
Ventricular fibrillation
Impulse stimulates ventricular muscles
Then stimulate itself
Portions of ventricles contract at same time and other relax
Atrial fibrillation
Enlarged atrium caused by heart valve lesions
Atrial flutter
Single large wave front travels around atria
Atria contract at 250 to 300 times per minute
Small amount of blood can be pumped
VI. Circulation: Pressure, Resistance, and Distensibility
Principles
Vasoconstriction must be accompanied by vasodilation
Blood flow to tissue is proportional to tissue needs
Velocity of flow is inversely proportional to vascular cross-sectional area
Lowest velocity in capillaries
Highest in aorta
Blood flow Q = P / R
Poiseuille’s Law
Vascular Resistance = [Viscosity x Length] / Radius 4
Distensibility = Volume / [Pressure x Original Volume]
Vascular compliance = Volume / Pressure = Distensibility x Volume
Veins = 24x as compliant as arteries
Delayed compliance
First increase in pressure when there is an increased in volume
Delayed stretch of vessel
Then decreased in compliance
Allows continuous pulsatile flow of blood
Hematocrit = 40
Percentage of RBC in blood
Total blood volume
84 % in systemic circulation
64 % in veins
13 % in arteries
7 % in systemic arterioles and capillaries
7 % in heart
9 % in pulmonary vessels
Pulse pressure = systolic pressure – diastolic pressure
Increase pulse pressure can be caused by
Increased stroke volume
Decreased arterial compliance (arteriosclerosis)
Aortic valve stenosis
Patent ductus arteriosus
Aortic regurgitation
Pulmonary circulation has lower pressure than systemic circulation
Pulmonary = 25 mm Hg & 8 mm Hg
Systemic = 120 mm Hg & 80 mm Hg
Hoang 43
Pulmonary vascular resistance < systemic vascular resistance
Increased right atrial pressure = increased peripheral venous pressure
Increased venous resistance = increased peripheral venous pressure
Blood reservoirs
Spleen
Liver
Large abdominal veins
Venous plexus beneath skin
VII. Microcirculation
Precapillary sphincters
Smooth muscle fibers encircle vessel
Capillary wall
Thin, single layer of endothelial cells
Porous
Vasomotion = blood flows intermittently through capillaries
Blood flow through capillaries is discontinuous
Diffusion = transfer of substances between plasma and interstitial fluid
Influenced by pore size, molecular size, & concentration difference
Interstitium = spaces between cells
Collagen fiber bundles
Proteoglycan filaments
Fluid filtration through capillary membrane
Capillary hydrostatic pressure = force fluid outward through capillaries
Average of 17 mm Hg
Interstitial fluid hydrostatic pressure = force fluid inward
Interstitial fluid colloid osmotic pressure = osmosis of fluid outward
Averages of 8 mm Hg
Plasma colloid osmotic pressure == osmosis of fluid inward
Averages of 28 mm Hg
Edema = outward pressure exceeds inward pressure
VIII. Local Control of Blood Flow
Tissues autoregulate blood flow
Supply nutrients
Remove waste
Determine heat loss = control body temperature
Greater rate of metabolism = greater blood flow
High in thyroid and adrenal
Low in resting skeletal muscles
Decreased oxygen availability = increases local blood flow
Occurs in cyanide poisoning, carbon monoxide, pneumonia
Acute control
Seconds to minutes
Vasoconstriction or vasodilation
Increase local blood flow
Accumulation of vasodilator metabolites
Lack of nutrients = glucose, amino acids, or fatty acids
Beriberi
Hoang 44
Lack of oxygen
Increased tissue metabolic rate
Long-term control
Days, weeks, or months
Due to change in physical size and metabolic need
Angiogenesis = increase tissue vascularity
Stimulated by low oxygen
Ischemic tissues
Tissues growing rapidly
Tissues with excessively high metabolic rates
Angiogenic factors
Vascular endothelial growth factor (VEGF)
Fibroblast growth factor (FGF)
Angiogenin
Rarefraction = decrease tissue vascularity
Humoral control
Norepinephrine/epinephrine
Vasoconstrictors via -adrenergic receptors
Epinephrine cause be vasodilator via -adrenergic receptors
Angiotensin II
Vasoconstrictor
Form in response to volume depletion or decreased blood pressure
Vasopressin (ADH)
Vasoconstrictor
Form in response to decreased blood volume or increased plasma osmolarity
Endothelin
Vasoconstrictor from endothelial cells after damage
Eta & Etb receptors on capillaries = constriction
Etb receptors on endothelial cells = relaxation
Prostaglandins
PGE2
Vasodilator and vasoconstriction
Fever and pain
PGF2a
Vasoconstrictor
Uterine contraction
PGI2
Vasodilation
TxA2
Vasoconstrictor
Platelet aggregation
Bradykinin
Vasodilator
Can cause edema
Histamine
Vasodilator from mast cells and basophils
Increase capillary permeability
Nitric oxide (via nitric oxide synthase)
Calcium dependent
nNOS
Neurons and macula densa
eNOS
Endothelial cells in caveoli
Calcium independent
iNOS
Macrophages and microglia
Hoang 45
Vasoconstrictors
Calcium
Vasodilators
Potassium
Magnesium
Sodium
Increased osmolarity of blood
Increased [H+]
Increased [CO2]
Constrictors
Hormone
Receptor
Norepinephrine & epinephrine
-adrenergic
Angiotensin II
AT1
Vasopressin
V1a
Endothelin
Et a & Et b2
Prostaglandin E2
EP1 & EP3
Thromboxane A2
TP
Dilators
Hormone
Receptor
Epinephrine
-adrenergic
Endothelin
Et b1
Prostaglandin I2
IP
Prostaglandin E2
EP2 & EP4
Nitric oxide
Soluble guanylate cyclase
Bradykinin
BK
Histamine
H1
Reactive hyperemia
Blood supply blocked to tissue
Then unblocked
Blood flow increases 4-7 times normal
Due to accumulation of vasodilator metabolites & oxygen deficiency
Active hyperemia
Tissue metabolic rate increases
Hoang 46
Accumulation of vasodilator substances & oxygen deficiency
Local vasodilation = decreased total peripheral resistance
Increased venous return = blood flow increases to compensate
Arterial pressure
Increase in arterial pressure = increase in blood flow
Metabolic theory of autoregulation
Arterial pressure increases too great
Excess oxygen and nutrients to tissue
Constriction of vessels
Blood flow decrease to normal levels
Myogenic theory of autoregulation
Increased arterial pressure = stretch blood vessels
Cause smooth muscle of vessels to contract
Stretched blood vessels release endothelium-derived relaxing factor (NO)
Dilate upstream larger arteries
Decreased arterial pressure = relaxes smooth muscle
Decrease vascular resistance
Maintain normal blood flow even at lower blood pressure
Vasoactive hormones
Histamine
Arteriolar vasodilation & venous vasoconstriction
Increase Pcapillary = increased filtration out of capillaries = local edema
Bradykinin
Arteriolar vasodilation & venous vasoconstriction
Increase Pcapillary = increased filtration out of capillaries = local edema
Serotonin
Arteriolar vasodilation
Released in vessel damage
Migraine headaches
Prostaglandins
Vasodilator & vasoconstrictor
IX. Regulation of Cardiac Output and Venous Return
Cardiac output
Amount of blood pumped into aorta per minute
Peripheral circulation blood
Normal adult = 5 L/min
Venous return
Amount of blood flows back to right atrium per minute
Venous return resistance
Average resistance between peripheral vessels and heart
Increased by sympathetic stimulation
Frank-Starling Law of the Heart
Increased in venous return = increased in diastolic filling pressure = greater contractile force of ventricles
Due to stretching of cardiac muscle
Pump out all excess venous return
Cardiac output = venous return
Factors controlling venous return also control cardiac output
Increased venous return
1. Stretch sinus node
Increased rhythmicity of node
Hoang 47
Increased heart rate to pump extra blood
2. Bainbridge reflex
Increase in atrial pressure
Impulses go to vasomotor center then back to heart via sympathetic and vagus nerves
Increased heart rate & contractility
Prevent damming of blood in veins, atria, and pulmonary circulation
Ohm’s Law
Decreased total peripheral resistance = increased cardiac output
Increased total peripheral resistance = decreased cardiac output
Cardiac output curve
Increases plateau
Increased sympathetic stimulation
Decreased parasympathetic stimulation
Cardiac hypertrophy
Decreases plateau
Myocardial infarction
Cardiac valvular disease with stenosis
Abnormal cardiac rhythm
Measured by Fick’s principle
CO (L/min) = [O2 absorbed in lungs (mL/min)] / [Arteriovenous O 2 Diff (mL/liter of blood)]
Cardiac oxygen consumption increases via:
Increased afterload (aortic pressure)
Increased heart size
Increased contractility
Increased heart rate
Stroke work = primary source of energy is fatty acids
Pathologic cardiac output
Beriberi
Lack of thiamine
Increased vasodilation = decreased total peripheral resistance = increased cardiac output
Arteriovenous fistula
Shun between artery and vein
Decreased total peripheral resistance = increased cardiac output
Hyperthyroidism
Increased oxygen use = release vasodilatory products
Total peripheral resistance decreases = increased cardiac output
Anemia
Decreased total peripheral resistance = increased cardiac output
Lack of oxygen delivery = increased vasodilation
Decreased viscosity of blood from low RBC count
Contractility
Intrinsic ability of cardiac muscle to develop force at given muscle length
Estimated via ejection fraction = 0.55
AKA inotropism
Positive inotropic effects = increase contractility
Increased heart rate
Post-extrasystolic potentiation
Sympathetic stimulation
Increases Ca2+ entry during plateau of action potential
Increase Ca2+ pump activity of SR
Cardiac glycosides = digitalis
Inhibit Na+-K+ ATPase = increase contraction strength
Intracellular [Na+] increases = inhibit Ca2+-Na+ exchange
Increases Ca2+
Hoang 48
Negative inotropic effects = decrease contractility
Parasympathetic stimulation
Decrease Ca2+ entry = decrease contraction strength
Effects of Stimulation on CV Physiology
Cardiac Output
Venous Return
Contractility
Sympathetic



Mean Systemic
Pressure

Parasympathetic



-
Increased Total Peripheral Resistance


-
-
Decreased Total Peripheral Resistance


-
-
Hemorrhage


-

Excess Blood Volume


-

Parasympathetic effects
Negative chronotropic effect
Decreases heart rate via phase 4
Decreases If of Na+
Negative dromotropic effect
Decreases conduction velocity through AV node
Increases PR interval
Sympathetic effects
Positive chronotropic effect
Increases heart rate via phase 4
Increase If of Na+
Positive dromotropic effect
Increases conduction velocity through AV node
Decreases PR interval
Autonomic Effects on Heart
Sympathetic
Parasympathetic
Effect
Receptor
Effect
Receptor
Heart Rate

1

Muscarinic
Conduction velocity
(AV Node)

1

Muscarinic
Contractility

1

Muscarinic
Hoang 49
X. Nervous Regulation of Circulation
Vasomotor centers
Area C-1
Vasoconstrictor
Norepinephrine
Upper medulla
Area A-1
Vasodilator
Project to C-1 and inhibit vasoconstrictor activity
Lower medulla
Area A-2
Sensory
Medulla and lower pons
Tractus solitarius
Receive from vagus and glossopharyngeal nerves
Reflex control (i.e. Baroreceptor reflex)
Sympathetic regulation = rapid
Norepinephrine
-adrenergic receptors
Vasoconstriction
Arterioles constricted
Increased total peripheral resistance
Increased blood pressure
Veins constricted
Increase effective blood volume
Increased cardiac output
Heart enhance in cardiac pumping
Increased heart rate
Sympathetic stimulation
Inhibition of parasympathetics
Arterial baroreceptor reflex control system
Stretch receptors on carotid sinus and aortic arch
Carotid sinus – Herring’s nerve – glossopharyngeal nerve – tractus solitarius
Aortic arch – vagus nerve – tractus solitarius
Increased pressure – increased impulse firing of baroreceptor – inhibition of vasoconstrictor center
Excite vagal center
Vasodilation of veins and arterioles
Decreased heart rate and contraction strength
Decreased peripheral vascular resistance = decreased cardiac output
Decreases arterial pressure
Decreased baroreceptor activity = increased sympathetic activity
Baroreceptor system plays minor role in chronic control
System reset within 1-2 days to exposed blood pressure levels
Cardiopulmonary reflex
Stretch receptors on atrial and pulmonary arteries
Detect increase in heart and pulmonic circulatory pressure
Due to change in volume
Increased atrial stretch = decreased sympathetic activity of kidney & ADH secretion from hypothalamus
Renal vasodilation of afferent arterioles
Increases glomerular capillary pressure
Increase glomerular filtration rate = increased volume excretion
Decrease tubular reabsorption of sodium
Release of atrial natriuretic peptide
Hoang 50
Abdominal compression reflex
Stimulation of sympathetic vasoconstrictor system = increased tone of abdominal muscles
Compress venous reservoirs = increased effective blood volume
Central nervous system ischemic response
Cerebral ischemia
Decreased blood flow to vasomotor center of lower brain stem
Maximum increase in arterial pressure (250 mm Hg)
Due to lactic acid buildup and increased CO2
Potent sympathetic vasoconstriction of organs (i.e. kidneys stop urine production)
Cushing reaction
Increased pressure in cranial vault
Could compress blood vessels in brain
CSF pressure = arterial pressure
Initiate central nervous system ischemic response
Increase arterial pressure to 250 mm Hg
Maintain blood flow to vital centers
Orthostasis
Gravitational acceleration of blood to lower extremities
Compensation to decreased venous return
Vis a fronte = sucking action of right atrium on venous system
Vis a tergo = positive pressure transmitted from capillaries to venous bed
Intrapleural pressure
Abdominal pressure
Venous valves & skeletal muscle pump
Respiratory waves = small rhythmic changes in arterial pressure
Increase in arterial pressure during early expiration
Decrease in arterial pressure during the remainder of the cycle
Vasomotor waves = large rhythmic changes in arterial pressure
Oscillation of baroreceptor reflex
Oscillation of chemoreceptor reflex
Oscillation of CNS ischemic response
XI. Long-Term Regulation of Arterial Pressure via the Kidneys
Renal output of salt and water = intake of salt and water
Two determinants of long-term arterial pressure
1. Renal output of salt and water
2. Level of salt and water intake
Renal-body fluid feedback system
Arterial pressure rises excessively high
Pressure natriuresis
Kidney excrete sodium
Pressure diuresis
Kidney excrete water
Extracellular fluid volume and blood volume decreases
Blood pressure return to normal
Arterial pressure decreases excessively
Kidney reduce rate of sodium and water excretion
Increased total peripheral resistance
Do not elevate long-term arterial pressure
Unless fluid intake and renal function changes
Normal arterial pressure =120 / 80 mm Hg
Hoang 51
Hypertension
Diastolic greater than 90 mm Hg
Systolic greater than 140 mm Hg
Mean arterial pressure = 93 mm Hg
Volume loading effects and results
Hypertension
Increased total peripheral resistance
Complete return of extracellular fluid volume, blood volume, & cardiac output back to normal
Renin-angiotensin system
Decreased arterial pressure
Renal JG cells secrete renin
Catalyze angiotensinogen to angiotensin I
Angiotensin I convert to angiotensin II via ACE
ACE present throughout body
Predominately in lungs & kidneys
Angiotensin II constricts arterioles and veins & decreases salt and water secretion in kidneys
Increase total peripheral resistance
Decrease vascular capacity = increase venous return
Degraded by angiotensinase
Arterial pressure increase to normal
Angiotensin II effects on kidney
1. Constrict efferent arterioles
Diminishes blood flow and filtration
2. Stimulate renal tubules to increase reabsorption of sodium and water
3. Stimulate adrenal glands to secrete aldosterone
Aldosterone increases renal salt and water reabsorption
Especially in collecting tubule
Renal hypertension
Causes = excessive salt and water retention
Renal arterial stenosis
Constriction of afferent arterioles
Increased resistance to fluid filtration through glomerulus
Excessive angiotensin II formation
One-kidney Goldblatt hypertension
One kidney removed
Constrictor on renal artery of remaining kidney
Systemic arterial pressure increases
Renal arterial pressure increases back to normal levels
Due to renin-angiotensin system
Phase 1 = vasoconstriction type
Transient
Phase 2 = volume-loading type
Total peripheral resistance increased
Essential hypertension
Unknown cause
Kidney require high arterial pressure to maintain intake and output balance
Rapid blood pressure control mechanism (seconds)
Baroreceptor feedback mechanism
CNS ischemic mechanism
Chemoreceptor mechanism
Intermediate blood pressure control mechanism (minutes)
Renin-angiotensin vasoconstrictor mechanism
Hoang 52
Stress relaxation of vasculature mechanism
Capillary fluid shift mechanism
Long-term blood pressure control mechanism (hours to months)
Renal-body fluid feedback mechanism
XII. Congestive Heart Failure
Heart failure
Inability of the heart to maintain CO sufficient to support vital organs
Rapid compensation via sympathetic system
Increased MSFP, contractility, heart rate
Vasoconstriction of peripheral blood vessels
Chronic response via renal mechanisms (RAS)
Sodium retention and increased blood volume
Compensated failure
Recovery of cardiac output to normal levels after heart damage
Decompensated failure
Compensatory responses fail
Longitudinal tubules of SR fail to accumulate calcium
Increased blood volume overstretch sarcomeres
Decreases cardiac contractility
Edema of heart muscle
Systolic heart failure
Poor pumping leads to defect in expulsion of blood
Decreased ejection fraction
Diastolic heart failure
Impairment of ventricular filling
Leads to high venous pressure upstream from ventricle
Often concomitant with systolic heart failure
Hypertension – left ventricular hypertrophy
Diagnostic tests
ECG
Lower QRS amplitude = MI
Prolonged QRS = conduction block
Arrhythmias
Chest X-ray
Echocardiogram
Cardiac catheterization
Blood testing
Secondary effects of heart failure
Effects of therapy or contributing factors
Exercise testing
ACE inhibitors
First therapy in systolic dysfunction
Beta-blockers
Second therapy in systolic dysfunction
Diuretics
Decrease load tension on heart
Inotropic agents
Increase contractility and CO
Hoang 53
RENAL PHYSIOLOGY
I. Body Fluid Compartments
Total body water
60% of body weight
Muscle & skin have most of the water
Blood & kidneys have highest water % to body weight
Skeleton & adipose have lowest water % to body weight
42 L of water in a 70-kg adult
Water intake
Majority from food then drink
Minor production from oxidation
Water output
Majority in urine then insensible water lost
Insensible skin & insensible lungs
Minor from feces and macroscopic sweat
Intracellular fluid = 40% of body weight (28 L)
Extracellular fluid = 20% of body weight (14 L)
Interstitial fluid = 15% of body weight
Plasma = 4% of body weight
Lymphatic fluid = 1% of body weight
Decrease % fat = Increased water % of body weight
Obesity = low body water to weight ratio
Females has more subcutaneous fat than men
Older babies have more fat than newborns
Indicator-Dilution principle
Used to measure compartmental volumes
Substance A is injected into compartment 1
Substance A disperse equally to equilibrium
Volume 1 = [Volume A x Concentration A] / Concentration 1
Total body volume = antipyrine – D2O – HTO
Plasma volume = Evan’s blue, radioiodinate human serum albumin (RISA)
Extracellular volume = inulin, radiosulfate, radiosodium
Molarity (M) = moles of solute / liter of solvent
One mole = 6 x 1023 particles
Electrochemical equivalence (mEq/L) = M x valence
One equivalent = weight in grams that associate with 1 gram of H +
Osmolarity (osm/L) = Molarity x number of particles per molecule
Osmotic pressure of a solution through a semipermeable membrane
Osmolality (osm/kg) = Molality x number of particles per molecule
Water moves rapidly across cell membrane
Intracellular almost always equal extracellular fluid compartment
Addition of hypertonic solution to ECF
Increased ECF osmolarity
Osmosis to ECF
ECF volume > ICF volume
Addition of isotonic solution to ECF
No change in osmolarity
Hoang 54
Increased in ECF volume
Addition of hypotonic solution to ECF
Decreased ECF osmolarity
Osmosis to ICF
ECF volume < ICF volume
Capillaries
Filtration = Kf x [ Pc – Pif – c + if ]
Hydrostatic pressure & osmotic/oncotic pressure
Osmotic pressure () = [solutes] x R x Temperature
Edema
Intracellular edema
Depression of metabolic systems
Lack of adequate nutrition to cells
Sodium leak into interior of cells
Osmosis of water into ICF compartment
Extracellular edema
Abnormal leakage from plasma to interstitial space
Lymphatic insufficiency or blockage
Excessive blood flow or heart failure
Cirrhosis or loss of protein
Blockage of interstitial proteins reuptake into lymphatic system
Factors preventing edema = 17 mm Hg
Low compliance of tissues
Increased interstitial fluid = increases interstitial hydrostatic pressure
Lymph flow can increase 50 fold
Decreases interstitial protein concentration = decrease interstitial colloid pressure
Intracellular
Extracellular
Na+
145 mM
Cl-
108 mM
HCO3-
25 mM
Proteins
200 mg/dl
15 mg/dl
Ca2+
10-4 mM
1 mM
Mg2+
40 mM
2 mM
K+
140 mM
4.5 mM
Intracellular
Total = 400 mM but 300 mosm/L due to associative affects
98% of K+ is intracellular
Highest ion in cell
Extracellular
Total = 300 mM and 300 mosm/L
Major cation is Na+
NaCl dictates volume of ECF compartment
Hoang 55
Asymmetry in solute composition b/w ECF & ICF
Gibbs-Donnan Effect
Non-diffusible ion present only on one side of membrane
i.e. Non-diffusible protein anions of blood plasma
Resulting in slightly greater concentration of cations in plasma than ICF
Resulting in slightly greater concentration of anions in ICF than plasma
Electroneutrality of solutes conserved in each compartment
Number of diffusible cations greater in blood plasma (ECF) than ICF
Oncotic pressure
Osmotic pressure generated by large solutes
Proteins
Balanced by hydrostatic pressure
Primary active transport = Ionic pumps
Membrane proteins
Transport ions against concentration gradient
Na/K ATPase pump
2 K+ into cell = 3 Na+ out of cell
Favors passive diffusion of Na + into cell across luminal membrane
Favors passive diffusion of K+ out of cell into tubular lumen
Consume ATP to establish asymmetric distribution
Secondary active transporter
Transport solute against concentration gradient
Use Na+ gradient generated from Na/K-ATPase
Antiporters
Na+ inward = counter-ion outwards to ECF
Symporters
Na+ & accompanying ion into ICF
RBC
Permeable to urea
RBC burst when placed in same osmolar urea solution (hypotonic)
Impermeable to sucrose
RBC maintain normal volume (isotonic)
“To exert an osmotic pressure across a membrane, a solute must not permeate that membrane”
II. Renal Anatomy
Function of kidney
Monitor and regulate volume/concentration of body fluids
Excrete waste products & drugs
Make glucose – ammonia – bicarbonate
Maintain acid base balance
Synthesize renin – erythropoietin – 1,25-OH-VitD
Function of nephron
Modification of filtrate
Filtration, secretion, reabsorption, excretion
Anatomy
Retroperitoneal
Encased in fat
Cortex
Covered by fibrous capsule
Medulla
Pyramids
Inner medulla = papilla
Collecting duct – calyces – renal pelvis – ureter – bladder
Hoang 56
All glomeruli = located in cortex
Cortical glomeruli = 90% at outer 2/3 of cortex
Juxtamedullary glomeruli = 10% at inner 1/3 of cortex
Larger with higher GFR
Macula densa
Junction of thick ascending loop of Henle & distal convoluted tubule
Distal nephron = distal convoluted tubule + collecting duct
Blood supply = aorta – renal artery
Renal blood flow = 22% of cardiac output
Segmental – interlobar – arcuate – afferent – glomerular – efferent – peritubular (90%) or vasa recta (10%)
In series = 2 resistance beds & 2 capillary beds
Vasa recta = specialized peritubular capillaries & parallel to loop of Henle
Afferent arterioles = muscular
Contain granular cells = synthesize renin
Renal corpuscle
Glomerular capillary = no VSM
Contain mesangial cells
Structural support
Contractile
Regulation of surface area
Synthesis – phagocytic – immune functions
Epithelium
Podocytes
Foot processes create slit pores
Each pore covered by diaphragm
Bowman’s capsule
Surrounds glomerulus
Juxtaglomerular apparatus
Macular densa + extraglomerular mesangium + terminal afferent arteriole
Functional importance = tubule communicate with blood vessels
Tubule membrane transport
Asymmetric
Na/K ATPase on basolateral surface (peritubular)
Variable Na+ permeability on apical surface (lumen)
Proximal tubule
Thick microvilli
Leaky tight junction
Ascending loop of Henle
Tight tight junction
Distal nephron (distal tubule + collecting duct)
Tight-tight tight junction
Collecting duct
Principal cells & intercalated cells
Renal innervation
Sympathetic to arterioles & proximal and thick ascending loop of Henle
No significant parasympathetic innervation
Some non-adrenergic, non-cholinergic nerves (NANC)
NO neurotransmitter
Chemoreceptors
Composition of urine
Mechanoreceptors
Hoang 57
Degree of perfusion of renal tissue
III. Glomerular Function & Renal Clearance
Renal blood flow = [ Renal artery pressure – Renal vein pressure ] / Total renal vascular resistance
Resistance vessels
Interlobular arteries
Afferent arterioles
Efferent arterioles
Glomerular filtration barrier
Fenestrated endothelium (50-100 nm)
Glomerular basement membrane
Filtration slit diaphragms bridging between adjacent foot processes
Podocytes cytoplasmic processes
Glomerular capillary wall
Strong negative charge
Polyanionic molecules on surface of endothelial cells, podocytes, & within GBM
Negatively charged molecules filtered less
Mesangium
Mesangial cells and surrounding matrix
Structural support
Urinary excretion rate = Filtration rate – Reabsorption rate + Secretion rate
Glomerular filtration rate (GFR)
GFR = 80-200 ml/min or 115-290 L/day
20% of renal plasma flow
Increase body weight = Increase GFR
Glomerular filtrate = identical to plasma except it has no protein
Rate of filtration of X = plasma [X] x GFR
Assume X is freely filtered
Rate of excretion of X = urine [X] x Urine flow rate (V)
Renal clearance
Volume of plasma which all substance X is removed / unit time
CX = [ Ux/Px ] x V
Fractional clearance
Fractional clearance of X = [ CX/Cinulin ]
FC > 1 when secretion
FC = 1 when no reabsorption or secretion
FC < 1 when reabsorption
Glucose
All filtered & all reabsorbed via active transport
Seen in urine when Tmax is exceeded
Diabetes mellitus = plasma [glucose] abnormally high
Renal glycosuria = Tm abnormally low
Phosphate
All filtered but some reabsorbed via active transport at proximal tubule
Tm is low = PO4 usually seen in urine
CPO4 / Cinulin = 0.2
20% is excreted
H 2O
Filter 125 ml/min but excrete 1-2 ml/min
Passively reabsorbed
Water reabsorbed = urinary [inulin] increases
Hoang 58
Uinulin / Pinulin = 1 when no water reabsorbed
Uinulin / Pinulin = 2 when 50% filtered water reabsorbed
Uinulin / Pinulin = 100 when 99% filtered water reabsorbed
Free water (solute-free)
ClearanceH2O = Urine flow rate V – Cosm
Cosm = Urineosm x Urine flow rate V / Plasmaosm
Urineosm < Plasmaosm
Positive free-water clearance
Net excretion of free-water
Hypo-osmotic urine
Urineosm > Plasmaosm
Negative free-water clearance
Net absorption of free-water
Hyperosmotic urine
Urea
Waste product of protein metabolism
Produced in liver
Excreted by kidney
Freely filtered & passively reabsorbed via concentration gradient
Require transporter to cross membrane
Reabsorbed in proximal tubule and inner medullary collecting duct
Secreted in thin ascending limb of loop of Henle
Concentration gradient determined by water reabsorption
Water reabsorbed = urinary [urea] increases
Urine flow rate decreases = urea excretion decreases
Curea / Cin = 0.6
Protein
Filter 2 g/day but 5-6 kg flow through kidney per day
99% reabsorbed in proximal tubule
Para-aminohippuric acid (PAH)
Non-endogenous
Freely filtered
Secretion via peritubular capillaries
Proximal tubule
CPAH = renal plasma flow rate (RPF)
CPAH / Cin = 5
Creatinine
Freely filtered
Secreted but not reabsorbed
Ccr / Cin = 1.1
Measuring renal plasma flow
Fick principle
RPF = Urinary [X] x Urine flow rate V / ( Arterial [X] – Venous [X] )
Invasive
Urinary clearance method
PAH as ideal marker
Freely filtered & actively secreted
10-20% remains in renal vein
Due to vasa recta of juxtamedullary nephrons
No secretion of PAH in medulla
“Effective” RPF = CPAH = Urinary [PAH] x Urine flow rate V / Plasma venous [PAH]
Underestimates RPF by 10-20%
Measuring GFR
Inulin & iothalamate
Hoang 59
Non-endogenous ideal marker
Rate of filtration via plasma = rate of excretion via urine
PIn x GFR = UIn x V
Clearance = GFR = [UIn / PIn ] x V
Because inulin is freely filtered, no secreted or reabsorbed
Creatinine
Endogenous non-ideal marker
Freely filtered but is also secreted
Ccreatinine > Cinulin
Excretion is constant and equals production
Plasma creatinine is constant = 1 mg/dl
If GFR decreases ½ = Plasma [creatinine] increase 2 fold to compensate
Production is inconstant in pregnancy, aging, muscle wasting, & maturation
Cockcroft-Gault formula
Females is less than males
Incorporate age, weight, & gender
MDRD equation
Incorporates race in addition to previous formula
Females produce less; blacks produce more
GFR = Renal plasma flow x Filtration fraction
FF = GFR/RPF
0.2 = 120 ml/min / 600 ml/min
GFR = Kf x Net filtration pressure
1. PG = 60 mm Hg (promotes filtration)
2. G = 33 mm Hg (oppose filtration)
3. PB = 18 mm Hg (oppose filtration)
4. B = 0 mm Hg
Methods to increase G
Increase renal plasma flow = increase filtration fraction
Increase renal blood flow = decrease glomerular colloid osmotic pressure
Methods to increasing PG
Increase arterial pressure
Decrease afferent arteriolar resistance
Increase efferent arteriolar resistance
Excretion
Reabsorption
Glucose
Filtered
Proximal tubule
Phosphate
Filtered
Proximal tubule
Water
Filtered
Everywhere
Urea
Filtered
Secreted in thin ascending Henle
Proximal tubule
Inner medullary collecting duct
Protein
Filtered
Proximal tubule
PAH
Filtered
Secreted via peritubular capillaries
Not reabsorbed
Creatinine
Filtered
Secreted
Proximal tubule
Hoang 60
Neurohumoral systems of GFR
Sympathetic stimulation
Constrict renal arterioles = decrease renal blood flow & GFR
Norepinephrine & epinephrine
Constrict afferent & efferent arterioles = decrease renal blood flow & GFR
Endothelin
Constricts renal arterioles = decrease renal blood flow & GFR
Endothelium-derived nitric oxide (EDNO)
Decrease renal resistance = increase renal blood flow & GFR
Prostaglandins (PGE & PGI)
Decrease vasoconstrictor effects of sympathetic = increase renal blood flow & GFR
Angiotensin II
Constricts efferent arterioles more than afferents
Increase PG & decrease renal blood flow
Low [Ang II]
Constricts both afferent & efferent arterioles = decreases RPF (& GFR)
Constrict efferent arterioles more = increases P GC (& GFR)
Net zero effect on GFR
Decrease sodium excretion
Stimulate sodium transport in proximal tubule
Via Na/H-antiporter
Stimulate aldosterone release
Increase sodium reabsorption in collecting duct
High [Ang II]
Intense renal vasoconstriction = decreases GFR
Atrial natriuretic peptide
Stimulated by volume overload (stretch fibers)
Increase RPF & PGC
Vasodilator on afferent arterioles
Vasoconstrictor on efferent arterioles
Tubuloglomerular feedback
Decreased bp = decreased NaCl sensed by macula densa cells
Decreased afferent arteriolar resistance
Increase PG = increased GFR
Increased renin release from JG cells of afferent & efferent arterioles
Increase Ang II = constricts efferent arterioles to increase P G & GFR
Myogenic autoregulation
Blood pressure increase = blood vessels auto-constricts
Increase vascular resistance = decrease renal blood flow & GFR
Blood pressure decrease = blood vessels dilate
Decrease vascular resistance = increase renal blood flow & GFR
High protein diet
Increase [amino acids] in blood
Reabsorbed in proximal tubule using co-transport of sodium
Decreased NaCl to macula densa = decreased afferent arteriolar resistance
Increase GFR
Hyperglycemia
Increase sodium reabsorption via co-transport at proximal tubule
Tubuloglomerular feedback = increase GFR
Glucocorticoids
Increase renal blood flow & GFR
Fever
Increase renal blood flow & GFR
Hoang 61
Aging
Reduction in functional nephrons
Decrease renal blood flow & GFR
IV. Renal Hemodynamics & Glomerular Function
Kidney
0.5 % of body weight
25 % of cardiac output
Regulates erythropoietin release
Stimulated by low pO2 via interstitial renal cells
Regulates RAS – water balance – countercurrent exchange
Renal blood flow
Flow = P / Resistance
Resistance vessels
Interlobular artery (cortical radial) & afferent artery
Efferent artery
Glomerular filtration
Size, charge, and shape
Exclude large, globular, and anionic molecules
Filter small, deformable, and cationic molecules
Proteins phagocytosed by mesangial cells
99 % filtered is reabsorbed in proximal tubule
Glomerular filtration rate determinants
1. Renal plasma flow (RPF)
2. Filtration rate of each glomeruli
3. Number of functioning glomeruli
Glomerular filtration fraction determinants
Transcapillary hydrostatic pressure gradient
Glomerular capillary ultrafiltration coefficient (Kf)
Oncotic pressure of blood at glomerulus
Due to nonfilterable plasma proteins
Renal plasma flow
Increase RPF = increased filtration
Via decreased afferent or efferent arteriolar resistance
Transcapillary hydrostatic pressure gradient
Increased capillary blood pressure = increased filtration
Increase filtration fraction = increased GFR
Afferent arteriole constriction
Decreases glomerular blood pressure
Decreased GFR
Efferent arteriole constriction
Increases glomerular blood pressure
Increased GFR
Constriction of afferent & efferent equally
No change in glomerular blood pressure
Decreased RPF
Glomerular capillary ultrafiltration coefficient Kf
Increase surface area
No effect on GFR if filtration pressure equilibrium was already present
Increase GFR if filtration pressure disequilibrium is present
Controlled by contractile intraglomerular mesangial cells
Contain receptors = respond to hormone & drugs
Synthesize vasoactive substances
Oncotic pressure of glomerular capillary A
Hoang 62
Decreased glomerular oncotic pressure = increase transcapillary pressure gradient
Increased GFR
Fluid Flux = P x water permeability x total area
P = hydrostatic & colloid osmotic pressure
Kf = water permeability x total area
Self-limiting during filtration
Single nephron GFR (SNGFR)
Filtration pressure
Area between P and  curves
V. Tubular Processing
Secretion = peritubular capillaries into tubules
Simple diffusion into renal interstitium
Active or passive transport across tubular epithelium into lumen
Reabsorption = from tubules into peritubular capillaries
Transported across renal tubular epithelial membrane into interstitial fluid
Then through peritubular capillary into blood
Transcellular route = through cell membranes
Active or passive transport
Paracellular route = through junctional spaces between cells
Ultrafiltration = bulk flow into peritubular capillaries
Mediated by hydrostatic & colloid osmotic pressure
Reabsorption rates are variable for different substances
Glucose & amino acids = completely reabsorbed
NaCl & bicarbonate = highly reabsorbed but changes depending on body needs
Urea & creatinine = poorly reabsorbed
Proximal tubules
High capacity (65%) for reabsorption
Water, Na+, Cl-, K+, & electrolytes
Loop of Henle
Descending thin segment
Highly permeable to water
20% of glomerular filtrate volume is reabsorbed
Resulting in hyperosmotic tubular fluid
Ascending thin segment & thick segment
Impermeable to water
Permeable to Na+, Cl-, & K+
Resulting in dilute tubular fluid
Early distal tubule (“diluting segment”)
Form justaglomerular complex
Feedback control on GFR & blood flow
Impermeable to water & urea
Reabsorb ions
Dilutes tubular fluid = 100 mOsm/L
Late distal tubule & cortical collecting tubule
Principal cells
2/3 of total cells in segment
Absorb Na+ & water into cell
Secrete K+ into lumen
Intercalated cells
1/3 of total cells in segment
Absorb K+ into cell
Secrete H+ into lumen
Controlled by aldosterone
Hoang 63
Permeability to water
Controlled by ADH
Impermeable to urea
Medullary collecting ducts
Permeability to water
Controlled by ADH
Highly permeable to urea
Secretes H+ against concentration gradient
VI. Sodium Transport & Balance
Na/K-ATPase is present in basolateral membrane
All segments of nephron
Proximal convoluted tubule
60-70% sodium reabsorption
Early PCT
Sodium reabsorption via Na/H antiporter
Na+ into cell
Down concentration gradient created by Na/K ATPase
3 Na+ out of cell
2 K+ into cell
+
H out into lumen
Sodium-linked bicarbonate, glucose, phosphate, amino acids & chloride reabsorption
Na symporter
More bicarbonate than chloride is reabsorbed
Increase [Cl-] downstream in late PCT
Reabsorption of CO2
H+ bind to HCO3- = form H2CO3
Split into CO2 and water
CO2 reabsorbed into cell
Reform carbonic acid via carbonic anhydrase
Late PCT
Na/H exchanger
Na+ from lumen into cell
H+ from cell into lumen
H+ binds with protein anion
Neutral H-anion diffuse back into cell
Cl/Anion exchanger
Cl- from lumen into cell
Anion from cell into lumen
H+ binds with protein anion
Neutral H-anion diffuse back into cell
Paracellular transport
[Cl-] higher in lumen than plasma
Net chloride diffusion into peritubular fluid
Via chemical gradient
Result in net positive charge in luminal fluid
Stimulate sodium, Ca2+, Mg2+, & K+ reabsorption
Via paracellular transport
Net NaCl reabsorption between cells (leaky tight junctions)
Avoid entering cells
Thin descending loop of Henle
No sodium reabsorption
Water reabsorption via aquaporins
Osmolarity gradient
Hoang 64
Thick ascending loop of Henle
25% sodium reabsorption
Na/Cl/K exchanger
Na+, 2Cl-, & K+ from lumen into cell
Via low intracellular [Na+] & [Cl-]
Against high intracellular [K+]
Chloride channel in basolateral membrane
Cl- from cell into peritubular capillaries
+
K channel
K+ from cell into lumen
Result in net positive charge in lumen
Stimulate Ca2+, Mg2+, & other cations reabsorption
No water reabsorption
No aquaproins in apical membrane
Early distal convoluted tubule
5-7% sodium reabsorption
Na/Cl co-transporter
Na & Cl from lumen into cell
Na+ exit cell to peritubular fluid via Na/K-ATPase
Cl- exit cell to peritubular fluid via chloride channel
Water cannot cross apical membrane
Only section that can generate hypotonic luminal fluid (dilute urine)
No aquaporins in apical membrane
Late convoluted tubule & collecting duct
1-4% sodium reabsorption
Via principal cell
2/3 of total cells in collecting duct
Function in Na/K/H2O transport
Sodium channel = epithelial Na+ Channel (eNaC)
Na+ diffuse from lumen into cell
Via negative intracellular charge
Created by Na/K-ATPase
Create negative charge in lumen
Stimulate K+ diffusion from cell into lumen
Via K+ channel
Hormonal regulation
Aldosterone
Early
Open exisiting ENaC
Late
Stimulate ATP production = activate intracellular proteins & transcription
Form new Na/K-ATPase on basolateral membrane
Determinants of aldosterone release
Primary mechanism = increase plasma [angiotensin II]
Renin release = rate limiting step
Increased PK
Increased ACTH
Decreased PNa
Decreased plasma ANP
ANP has tonic inhibition on aldosterone
Angiotensin II
Regulation of release
Renin release = rate limiting step
Renin
Synthesized in granular cells of afferent arteriole = part of JGA
Hoang 65
Determinants of renin release
Decreased renal perfusion pressure
Via baroceptors in afferent arteriole
-adrenergic sympathetic stimulation
Occur when blood pressure falls
Decrease in NaCl at macula densa
Occur when GFR falls
Regulation of release
Directly sense volume = indirectly relates to sodium content
Do not sense sodium concentration
Inhibited by Ang II
Atrial natriuretic peptide
Increase GFR
Increase UNaV
Rapid
Direct inhibition of sodium reabsorption
Proximal tubule & collecting duct
Decreased aldosterone release
Determinants of ANP release
Atrial stretch from volume expansion via sodium increased
Nitric oxide
Increase GFR
Direct inhibition of sodium reabsorption
Proximal tubulte & collecting duct
Systemic vasodilator
cGMP 2nd messenger
Determinants of NO release
High salt intake
Prostaglandins
Inhibit sodium reabsorption
Thick ascending loop of Henle & collecting duct
Stimulation of prostaglandin release
Increase in volume
Endothelin
Vasoconstrictor
Increase sodium excretion
Different from other natriuretic hormones (vasodilators)
Bartler’s Syndrome
Seen in newborns
Mutation of Na/Cl/K exchanger or K+ channel of thick ascending loop of Henle
Build-up of K+ intracellular compartment
Inhibit Na/K-ATPase
Decreased NaCl reabsorption = decreased urinary sodium excretion
Increased urine Na & K concentrations
Decreased serum K concentration & blood pressure
Hemorrhage
Less than 10% blood loss
BP maintained by SNS & Ang II
Between 10-25% blood loss
ADH + SNS + Ang II
ADH = vasoconstrictor
Greater than 25% blood loss
Maximal vasoconstriction
Shock & renal failure
Hoang 66
Septic Shock
Endotoxin = increase NO release
Massive vasodilation
Underfilled circulation
Volume depletion
Cause sodium retention = increase effective circulating volume
3rd spacing = edema
VII. Potassium Regulation
Potassium
Intracellular ion = 150 mEq/L
Plasma [K+] = 4 mEq/L
Low plasma [K+] = hyperpolarize cell
Less excitable = “flaccid paralysis”
High plasma [K+] = depolarization
Spastic paralysis
Excretion
Renal = 90%
GI = 10%
K+ uptake into cells
Insulin
Epinephrine = -adrenergic agonist
Aldosterone
ECF alkalosis
K+ into cell = H+ out of cell
K+ secretion out of cells
-adrenergic stimulation
Exercise
Increased plasma osmolality
Cell lysis = physical injury
ECF acidosis
K+ out of cell = H+ into cell
Proximal convoluted tubule
67% reabsorption of K+
K+ exit cell to peritubular space
Via K+ channel & K/Cl-cotransport
+
K channel for emergency secretion only
Early PCT
Na+ reabsorption cause H2O to diffuse via paracellular transport
Solvent drag = water diffusion drag K+ reabsorption via paracellular transport
Concentration gradient
Late PCT
Early bicarbonate reabsorption cause lumen to be positively charged
K+ diffuse via paracellular transport
Electric gradient
Thick ascending loop of Henle
20% reabsorption of K+
Na/2Cl/K-cotransport
K+ transport to peritubular fluid via K-channel
K-channel secrete K+ into lumen to recycle for pump (50% of total reabsorption)
K+ reabsorb by paracellular diffusion (50% of total reabsorption)
Driven by luminal positive voltage
Hoang 67
Distal nephron
Reabsorption with low K+ intake
Excretion with high K+ intake
Distal K+ secretion = primary determinant of K + excretion
Principal cells
Regulate UK & V (urine flow rate)
Not by GFR or rate of reabsorption
Cortical collecting tubule: Intercalated cell
K/H-ATPase
K+ from lumen into cell
K+ transported to peritubular fluid via K-channel
H+ from cell into lumen
H-channel
H+ out of cell when K+ reabsorption is not needed
Cl/HCO3-antiporter
Cl- into cell
HCO3- out of cell
Cortical collecting tubule: Principal cell
Aldosterone-regulated-channels
ENaC
Na+ from lumen reabsorbed into cell
+
K pump
K+ from cell secreted into lumen
K/Cl-cotransporter
K+ & Cl- from cell secreted into lumen
+
K pump on basolateral membrane
K+ from cell transported into peritubular fluid
K+ recruit from peritubular fluid into cell via Na/K-ATPase
CK/Uin < 1.0
Net reabsorption
Excreted K+ from secretion (not filtration) in distal nephron
Secretion across luminal membrane = passive
Increasing K+ secretion of collecting tubule
Increased intracellular [K+]
Increase plasma [K+] = increase aldosterone secretion
Increase ECF pH
Induction of Na/K-ATPase via aldosterone
Increased K+ from cell into lumen
Increase luminal membrane permeability via aldosterone
Increase negativity of lumen
Increase Na+ reabsorption
Increase luminal concentration of non-reabsorbable anions
i.e. Phosphate or bicarbonate
Increase tubular fluid flow rate
Diuretics result in K+ wasting
Potassium does not “escape” aldosterone unlike sodium
Conn’s syndrome result in hypokalemia
VIII. Calcium, Phosphate, & Magnesium Regulation
Excess calcium
Nausea – vomiting – polyuria – dehydration – hypotension – coma
Excess phosphate
Calcium deposits in skeletal tissues, cardiac system, & arteries
Hoang 68
Deficient phosphate
ATP deficiency – muscular weakness – respiratory failure
Plasma calcium
Protein (albumin) bound = 40%
Bicarbonate complex = 10%
Ionized in solution = 50%
Only kind that is filterable & active
Calcium
Proximal tubule = 70%
Paracellular diffusion into interstitial fluid
Via lumen positive electrical gradient
Transcellular transport (minor)
Thick ascending limb of loop of Henle = 20%
Paracellular diffusion
Via lumen positive electrical gradient
Resulting from Na/2Cl/K-cotransport & recycling of K+
Increased Na+ reabsorption = increased paracellular Ca2+ reabsorption
Loop diuretics = inhibit Na/2Cl/K-cotransporter
Increase urinary calcium excretion
Distal tubule = 9%
Paracellular diffusion = minor
Transcellular transport = major
Calcium channel = apical
Intracellular negative gradient & low intracellular ionized calcium concentration
Via electrochemical gradient
Ca-ATPase & Na/Ca-exchanger = basolateral
Collecting ducts = 1%
Thiazide diuretics = increase calcium reabsorption
Inhibit Na/Cl-symporter of distal tubule
Inhibit NaCl entry into cell = increase intracellular negativity gradient
Increases apical calcium entry & stimulate basolateral calcium exit
Increased sodium excretion = decreased intravascular volume
Stimulates Na/2Cl/K-symporter of loop of Henle
Increased sodium reabsorption = increase calcium reabsorption
Phosphate
Essential buffer in urine for proton excretion
Bone = 86% of body’s total [phosphate]
ICF = 14% of body’s total [phosphate]
ECF = 0.03% of body’s total [phosphate]
Protein bound = 10%
Free ionized = 90%
Proximal tubule = 90%
2Na/Pi-Symporter
Via sodium gradient = intracellular negative potential gradient
Distal tubule = 10%
Phosphate loading or hypocalcemia
Increased PTH
Cause parathyroid gland hyperplasia = increased PTH secretion
Decreased calcium sensitivity to inhibit PTH release
Increased phosphate excretion
Increased calcium reabsorption
Hoang 69
Phosphate depletion or hypercalcemia
Decreased PTH
Decreased phosphate excretion
Magnesium
Proximal tubule = 15% of filtered magnesium
Thick ascending limb of loop of Henle = 70-80% of filtered magnesium
Distal tubule = 10% of filtered magnesium
IX. Neurohormonal Regulation
Aldosterone
Secreted by adrenal cortex
Act on principal cells of cortical collecting tubule
Stimulate Na/K-ATPase = increases sodium reabsorption & potassium secretion
Excess aldosterone = hypokalemic alkalosis
Angiotensin II
Activated by renin & ACE in RAAS
Stimulate aldosterone secretion
Constrict efferent arterioles = decrease RPF
Reduce hydrostatic pressure peritubular capillaries = reduce renal blood flow to increase FF
Increase absorptive force of peritubular capillaries & sodium and water
Direct stimulation Na+ reabsorption at proximal tubules & TAH
Antidiuretic hormone
Secreted from posterior pituitary gland
Increase water permeability of distal tubules, collecting tubules, & collecting ducts
Atrial natriuretic peptide
Secreted by cardiac atria
Stimulated by distension of atrium from plasma volume expansion
Decrease sodium & water reabsorption
At proximal tubule & distal nephron
Parathyroid hormone
Increase calcium reabsorption
Predominantly in distal tubule & ascending loop of Henle
Decrease phosphate reabsorption
Stimulate renin release & Ang II formation
Nitric oxide
Increase sodium excretion
Increase GFR
Direct inhibition of sodium reabsorption on distal nephron & proximal tubule
Prostaglandins
Stimulated by increased ECF volume
Inhibit sodium reabsorption
At TAL & collecting duct
Sympathetic nervous system
Constricts afferent & efferent arterioles
Reduce GFR
Increase sodium reabsorption at proximal tubule & ascending loop of Henle
Stimulate renin release & Ang II formation
Hoang 70
Summary of Direct Neurohormonal Effects
Hormone
Source
Site of action
Filtration
Secretion
 K+
 H+
Reabsorption
 Na+
Aldosterone
Adrenal cortex
Distal nephron
TAH
Angiotensin II
ECF
Proximal tubule
TAH
 Na+
 H2O indirectly
ADH
Post. Pituitary
Distal nephron
 H2O
SNS
Peripheral System
(-Adrenergic)
Proximal tubule
TAH
Distal nephron
Parathyroid Hormone
Parathyroid gland
Distal tubule
TAH
ANP
Cardiac atria
Proximal tubule
Distal tubule
Collecting duct
 GFR
 Na+
 H2O
Nitric Oxide
Endothelial cells
Proximal tubule
Distal tubule
Collecting duct
 GFR
 Na+
 GFR
 Na+
 Ca2+
 PO43-
X. Urinary Concentration & Dilution
Serum osmolarity
Regulate between 285 mOsm/kg and 288 mOsm/kg
Serum osmolarity = (2 x Na) + (BUN / 2.8) + (glucose / 18)
Urine osmolarity
Regulated by the level of ADH
Concentration of urine
High ADH level
Increased water permeability of distal tubule & collecting duct
Dilution of urine
Low ADH level
Function
Proximal tubule
Tubule fluid does not change osmolarity
Thin descending limb of loop of Henle
Basal level of water reabsorption
Increased via ADH
Urea reabsorbed going toward medullary
Thick ascending limb of loop of Henle
Absorb Na+ & Cl- in absence of water
Active transport
TF is maximally concentrated at papillary tip
TF becomes diluted in the loop
TF becomes hypotonic to plasma at end of TAH
Hoang 71
Fluid entering distal tubule is hypoosmotic to plasma
Impermeable to water
Distal convoluted tubule = connecting segment
Na+, Cl-, & Ca2+ reabsorption
H2O reabsorption via ADH
Cortical collecting duct
Na+ & Cl- reabsorption
H+ & HCO3- balance
H2O reabsorption via ADH
Medullary collecting duct
H2O reabsorption via ADH
Sodium reabsorption via ENaC
Urea
Proximal tubule
Reabsorbed
Thin descending loop of Henle
Urea efflux to interstitum in inner medulla
Thin ascending loop of Henle
Urea influx into TF
Passive efflux of sodium into interstitium
Thick ascending loop of Henle
Impermeable to urea
Active efflux of sodium into interstitium via Na/2Cl/K-symporters
Dilute TF to 200 mOsm by end of TAL = TF is hypotonic
Distal convoluted tubule
Water reabsorption via ADH-stimulated aquaporins
Only occur in water restriction
Sodium reabsorption via Na/Cl-symporters
TF is further hypotonic (100 mOsm) by end of distal tubule
Cortical & outer medullary collecting tubule
Water reabsorption via ADH-stimulated aquaporins
Occur in water restriction
[Urea] = increases
Impermeable to urea
Inner medullary collecting tubule
Permeable to urea = high concentration gradient
Efflux into interstitium
Urea cycling
Urea reabsorbed in inner medullary collecting tubule
Urea diffuse from interstitium into thin descending & ascending limb of loop of Henle
Recycle back to inner medullary collecting tubule
Requirements for forming concentrated urine
High level of ADH
High osmolarity of renal medullary interstital fluid
Countercurrent multiplier
Create high osmolarity in renal medulla
Active transport of sodium with co-transport of K+, Cl-, et cetera
From ascending limb of loop of Henle into medullary interstitium
Active transport of ions from collecting ducts into medullary interstitium
Diffusion of urea from inner medullary collecting ducts into medullary interstitium
Diffusion of minute amounts of water from collecting tubules
Countercurrent exchange of vasa recta = preserves hyperosmolarity of renal medulla
Vasa recta blood flow = low
Blood more concentrated in medulla
Hoang 72
Highly permeable to water & solutes
Water diffuse into vessel when ascending back to cortex
Osmoreceptor-ADH feedback system
ECF osmolarity increases above normal
Stimulate osmoreceptors of anterior hypothalamus
Signal release of ADH in posterior pituitary
ADH increase water permeability
Late distal tubules, cortical collecting tubules, & medullary collecting ducts
Increased water reabsorption = excrete concentrated urine
Antidiuretic hormone (ADH)
Synthesized in supraoptic & PVN of hypothalamus
Release from posterior pituitary
V1 receptor = vasoconstriction of blood vessels & endothelial cells
V2 receptor = intercalation of pre-formed aquaporin-2
G-protein stimulate AC = increased intracellular [cAMP]
Stimulate PKA = phosphorylation cascade
Fusion of aquaporin-2 with luminal membrane of principal cells
Water reabsorption
Stimulated by increased plasma osmolarity, decreased blood pressure or volume
Thirst
Stimulated by angiotensin II, increased osmolarity, decreased blood pressure or volume
Pathological urinary concentrating ability
Central diabetes insipidus
Inability to produce/release ADH from posterior pituitary
Nephrogenic diabetes insipidus
Inability of kidney to respond to ADH
XI. Regulation of Acid-Base Balance
ECF [H+] = 0.000 000 04 Eq/L = 40 nEq/L
Normal arterial blood pH = 7.4
Normal venous & interstitial fluids = 7.35
Nonvolatile acids
Sulfuric acid
Phosphoric acid
Organic acid
Volatile acid
CO2
Regulation
Buffer systems of body fluids
Bicarbonate buffer system
pH = 6.1 + log [HCO3-] / [0.03 x PCO2]
Proteins of cells
Phosphate buffer system
Respiratory system
Acidosis
Hyperventilation = decrease PCO2
Decrease carbonic acid in blood = decreased [H+]
Kidneys
Excretion of titratable acids H+ (phosphoric acid)
Hoang 73
Reabsorption of filtered HCO3About 100% filtered
PT = 80%
TAL = 15%
CCD = 5%
About zero % is excreted
Production of new HCO3Excretion of ammonium
Alkalosis
Excess bicarbonate ions
Bicarbonate reacts with available H+ to be reabsorbed
Excess HCO3- left in urine is excreted
Acidosis
Excess hydrogen ions
All filtered bicarbonate is reabsorbed
Excess hydrogen ions are excreted
Minimal urine pH = 4.5
Buffers of tubular fluid
Phosphate buffer
HPO42- & H2PO4For each H+ buffered = 1 bicarbonate is reabsorbed
Ammonia buffer
Ammonia (NH3) & ammonium (NH4+)
Synthesized from glutamine
Form 2 NH4+ into urine & 2 HCO3- into blood
“New” bicarbonate generation
Renal glutamine metabolism
Stimulated by acidosis & hypokalemia
Renal acid-base secretion & reabsorption
Proximal tubule & TAL of Henle
Apical membrane
Na/H-exchanger (2/3 of hydrogen secretion)
H-ATPase (1/3 of hydrogen secretion)
Basolateral membrane
Na/K-ATPase
High intracellular sodium concentration
Na/3HCO3-symport
Via electrical gradient = negative intracellular potential
HCO3/Cl-antiport
Proximal tubule & Thick ascending limb of loop of Henle
Hoang 74
Collecting duct: A-cell
Apical membrane = both pumps use ATP to make steep pH gradients
K/H-ATPase
Secrete H+ into lumen
H-ATPase
Basolateral membrane
HCO3/Cl-antiporter
Chloride depletion = inable to reabsorb bicarbonate
Cl-channel
Passive diffusion of chloride out of cell into interstitium
Via electrical gradient
Collecting duct: Acid-secreting intercalated cell
Collecting duct: B-cell
Apical membrane
HCO3/Cl-antiporter
Chloride into cell = bicarbonate into lumen
Driven by chloride concentration gradient
Can increase luminal pH to 8
Basolateral membrane
H-ATPase
Cl-channel
Collecting duct: Bicarbonate-secreting intercalated cell
Hoang 75
Renal generation of bicarbonate
Proximal tubule
Glutamine metabolize to 2 NH4+ & 2 HCO3Low potassium concentration stimulate ammonia production
Bicarbonate return to systemic circulation via renal veins
NH3 diffuses into lumen to reform ammonia
Medullary thick ascending limb of loop of Henle
NH4+ reabsorbed into interstitial space to collecting duct
Via Na/2Cl/K-symporter
NH4+ is mimic potassium
Collecting duct
NH3 diffuse to luminal fluid & excreted
Hypokalemia
Stimulate ammonia production and excretion
Increase bicarbonate generation
Signal collecting duct to stop secretion of potassium = principal cell
Via K/Cl-symporter & K-channel
Signal collecting duct to reabsorb potassium = -intercalated cell
Via K/H-antiporter
Long-term chronic acidemia
Significant increase in acid excretion
Short-term acute academia
Little or no increase in net acid excretion
XII. Nephropathy of Diabetes Mellitus
Diabetes mellitus
Chronic hyperglycemia
Polyuria – polydipsia – infection – weight loss
Type 1
Insulin deficiency
Type 2
Insulin resistance due to defective receptors
Macrovascular disease
Coronary arteries – carotid arteries – peripheral vasculature
Microvascular disease
Retinopathy & nephropathy
Hoang 76
Neuropathy
Mono- & polyneuropathy
Autonomic neuropathy
Renal complications of diabetes mellitus
Vascular disease
Increased infection
Hyperglycemia & inflammatory thrombosis
Atonic bladder = inable to empty bladder
Prone to infection
Renal ischemia & failure
Glomerular ischemic damage = hypertension & renal failure
Renal failure = 2nd leading cause of death in Type 1 diabetes mellitus
Chronic hyperglycemia
Glomerular basement membrane glycosylation
Decreased negative potential = decreased GBM selectivity
Leaky membrane = proteinuria
Mesangial proliferation
Increased pore size
Hypertension
Hyperfiltration
Intraglomerular hypertension
Glomerular damage
Thicken GBM & mesangial proliferation
Obliteration of capillary loops
Death of nephrons
Loss of surface area
Chronic renal failure
Stage I = Early hypertrophy-hyperfunction
100% progression to stage II
Treatment to control hyperglycemia & blood pressure
Hypertrophy = enlargement of kidneys
Expanded blood volume due to hyperglycemia
Hyperfunction = increased GFR
Expanded blood volume due to hyperglycemia
Stage II = Glomerular lesions without clinical disease
40% progession to stage III
Treatment same as stage I
“Silent phase”
Normal bp – no proteinuria – no microalbuminuria
Histological changes
Thickened GBM
Proliferated mesangial matrix
Stage III = Incipient diabetic nephropathy
Dipstick for protein = negative
Minimal albumin excretion = microalbuminuria
Special dipstick for MAU = cheapest test available
Elevated MAU requires second testing for confirmation
24 hour urine collection = gold standard
Mild hypertension
“Turning point”
Stage IV = Clinical diabetic nephropathy
100% progession to stage V
Dipstick-detectable albuminuria
Hoang 77
Decreased GFR
Measured via clearance of creatinine
Hypertension
Retinopathy always present
Stage V = End-stage renal disease
Azotemia
Uremia & olgiuria
Treatment = dialysis or transplantation
Histological findings
Nodular diabetic glomerulosclerosis
Kimmelstiel-Wilson nodules
XIII. Renal Disease & Failure
Acute renal failure
Abrupt loss of kidney function due to hemodynamics
Vasoconstriction of renal blood flow & plasma flow
Decreased RPF & GFR
Oliguria = low urine flow
Anuria = no urine flow
Acute tubular necrosis = tubular obstruction cause tubular damage
Can reduce urine
Can increase urine
Polyuria = tubular injury result in decreased sodium reabsorption
Occur even in low GFR
Category of acute renal failure
1. Prerenal: decreased renal perfusion
Hemorrhage – diarrhea – post partum ARF – hypotension
2. Renal: caused within kidney
Ischemic damage – surgery – radiocontrast – toxins
3. Obstructive: occlusion of renal system or vessels
Treatment
Increase volume & urine flow
Hemodialysis
Chronic renal disease
Progressive fall in renal function & GFR
Leads to end stage renal disease
Dialysis or renal transplant
Compromised sodium excretion & conservation
Edema & ECF volume expansion = congestive heart failure
Low sodium intake = becomes volume depleted
Compromised water concentration & dilution
Hyponatremia or hypernatremia
Compromised potassium regulation
Hyperkalemia = cardiac arrhythmias
Compromised mineral metabolism
Deficiency in 1,25-OH-Vitamin D
Abnormal levels of Ca2+ & PO43Hyperparathyroidism
Osteomalacia
Increased serum phosphate concentration due to decreased GFR
Increase binding of phosphate with calcium
Decrease serum ionized [calcium]
Stimulate PTH = demineralization of bones
Hoang 78
Uremia
Dysfunctional waste excretion = systemic poisoning
Anemia
Deficient erythropoetin
Acidosis
Deficit in reabsorption and generation of bicarbonate
Aging kidney
Decline in GFR
Male @ age 40 = females @ age 60
Falling RAAS
Decreased aldosterone
Sodium depletion
Diminished natriuretic response to sodium loading
Increased sodium sensitivity of blood pressure
Hypertension
Increased systolic
Diminished sensitivity to ADH
Case 1 = High salt intake
No significant  in GFR or blood pressure
Decreased Na+ reabsorption
Increase tubuloglomerular feedback
High salt intake
Thirst-receptors
Increase ECF osmolarity
Increase water intake
Increase plasma volume
Macula densa sense high salt levels
Decrease SNS stimulation
Decrease renin secretion
Dilate renal arterioles
Decrease angiotensin II
Increase secretion of ANP
Decrease Na+ reabsorption
Dilate efferent arterioles
Decrease aldosterone
Decrease H2O reabsorption
K+ & H+ retention
Hoang 79
GASTROINTESTINAL PHYSIOLOGY
I. Gastrointestinal Anatomy
Mouth – esophagus – stomach – small intestine – large intestine – colon – anus
Esophageal muscle
Striated = upper 1/3
Striated & smooth = middle 1/3
Smooth = lower 1/3
Intestinal wall
Serosa
Longitudinal muscle layer
Interstitial Cells of Cajal = non-neural cells
Circular muscle layer
Submucosa
Mucosa
Muscularis mucosae lies within deep mucosa
GI smooth muscle
Muscle fibers connected through gap junctions
Function as syncytium
Resting membrane potential = -56 mV
Depolarization
Stretching
Acetylcholine (parasympathetic nervous system)
Hormones
Hyperpolarization
Norepinephrine & epinephrine (sympathetic nervous system)
Enteric nervous system
Lies within gastrointestinal wall
Esophagus to internal anal sphincter
Composition
Myenteric plexus (Auerbach’s plexus)
Between circular & longitudinal muscle layers
Increase muscle tone
Increase contractile intensity
Increase rate of contraction
Increased conduction velocity
Neuron density = 50,000/cm2 gut surface
Submucosal plexus (Meissner’s plexus)
In submucosa
Integrate sensory signals from gastrointestinal epithelium
Regulation of secretion, absorption, and contraction (via myenteric plexus)
Plexuses are interconnected
Increased motility = increased secretion
Gastrointestinal movements
Myenteric reflex = Peristalsis by myenteric plexus
Distention cause contraction ring to move forward
Simultaneous receptive relaxation of gut
Law of the gut = myenteric reflex + unidirectional movement toward anus
Gastrointestinal blood flow
Splanchnic circulation = gut + spleen + pancreas + liver
Drain into liver via portal vein
GI blood flow = proportional to local activity
Hoang 80
Vasodilators
CCK – gastrin – secretin – kallidin – bradykinin
Tissue hypoxia = increase blood flow by 50%
Parasympathetic = increase blood flow
Sympathetic = decrease blood flow
Autoregulatory escape = local metabolic vasodilators elicited by ischemia redilate arterioles
II. Secretory Principles of Alimentary Tract
Contact of food with epithelium stimulate secretion
Tactile stimulation
Chemical irritation
Gut wall distention
Parasympathetic stimulation increase rate of glandular secretion
Salivary glands
Esophageal glands
Gastric glands
Pancreas
Brunner’s glands of duodenum
Secrete alkaline mucus with bicarbonate
Also stimulated by tactile and chemical irritants & GI hormones (secretin)
Sympathetic stimulation has dual effect on glandular secretion
Sympathetic alone increase secretion
Sympathetic superimposed on secretion decreases blood flow to gland = decrease secretion
Acetylcholine
Pepsinogen by chief cells
HCl by parietal cells
Mucus by mucous cells
Gastrin
HCl by parietal cells
Digestive enzymes by pancreas
Pepsinogen by chief cells
Histamine
HCl by parietal cells
GI Hormones
Gastric inhibitory peptide (GIP)
Secreted by duodenum & jejunum
Stimulated by fatty acids, amino acids, & oral glucose
Stimulate insulin release
Inhibits P-cell secretion of HCl
Vasoactive intestinal peptide (VIP)
Homologous to secretin
Released from mucosal nerves
Relaxation of GI smooth muscle
Stimulate pancreatic bicarbonate secretion
Inhibit gastric HCl secretion
Gastrin-releasing peptide (GRP)
Stimulated by vagus
Released from mucosal nerves
Stimulates gastrin release from G-cell
Enkephalins
Released from mucosal nerves
Stimulate GI smooth muscle contraction (sphincters)
Hoang 81
Inhibit intestinal secretion of fluid & electrolyttes
III. Salivary Secretion & Regulation
Daily = 1-1.5 L of saliva secreted
Active process
High concentrations of K+ & HCO3Low concentrations of Na+ & ClControlled mainly by parasympathetic signals
Salivatory nuclei via taste & tactile stimuli of tongue, mouth, & pharynx
Function of saliva
Facilitator
Chewing – swallowing – speech – taste
Protector
Dilute, neutralize and destroying acids & bacteria
Digestor
Salivary amylase
Lingual lipase
Salivon
Acini – intercalated ducts – striated duct – excretory duct – major excretory duct
Synthesize and secrete primary secretory fluid
Isotonic to blood plasma
Becomes hypotonic to major excretory duct = saliva
Ranges from 150 mOsm/L to 225 mOsm/L
Active reabsorption of NaCl
Active secretion of K+ & HCO3Hypotonicity = osmotic lysis of bacteria
Aldosterone = stimulate reabsorption of NaCl to create hypotonic saliva
High [carbonic anhydrase] at acini
Form H2CO3 = H+ reabsorbed into capillaries & HCO3- secreted in saliva
Bicarbonate from blood & acinar and intercalated duct cells
Increases salivary pH for optimum enzyme activity = pH 7.0
Neutralization of bacterial and acids from food
Increased rate of salivary secretion = increased pH due to high bicarbonate
Ion
Parotid Saliva
Blood Plasma
Na+
85 mM
145 mM
Cl-
40 nM
110 mM
K+
20 mM
10 mM
HCO3-
70 mM
24 mM
Composition of saliva
Serous
Primary electrolytes
Na+, K+, HCO3-, ClSecondary electrolytes
Hoang 82
Ca2+
Stimulate salivary amylase
SCNBacteriostatic agent
Organic
Mucin
Glycoprotein for lubrication during swallowing
Stimulated by K+ & attach to teeth
Amylase (Ptyalin)
Hydrolyze polysaccharides to maltose, glucose, & dextrins
Split 1,4-glucosidic linkage
Activation by chloride
Stimulation by calcium
Optimum pH = 7.0
Lingual lipase
Hydrolyze triglycerides to 2 free fatty acids & 1 monoglyceride
Optimum pH = 7.0
Lysozyme
Antibacterial = degrade bacterial cell walls
Lactoferrin
Adsorb Fe2+ & Fe3+
Compete with bacteria = antibacterial
IgA
Antibacterial
Secreted by salivon
Autonomic nervous system
Parasympathetic & sympathetic = stimulate secretion of saliva
Parotid – submaxillary – sublingual glands
Regulation by cerebrum, buccal cavity receptors, olfactory receptors
Parasympathetic nervous system = CN VII & IX
Secretion of serous saliva via cGMP & PIP 2
Vasodilation
Via neurotransmitter on vascular smooth muscle
Via bradykinin production from stimulation of secretory segment cells = long term
Myoepithelial constriction of basket cells around salivary gland
Acetylcholine
Sympathetic nervous system
Mucous (organic) secretion from mucous cell via cAMP
Vasoconstriction
Myoepithelial constriction of basket cells around salivary gland
Norepinephrine
Condition reflex secretion of saliva
Include cerebrum & memory
Unconditioned reflex secretion of saliva
Do not include cerebrum
IV. Gastric Secretion & Regulation
Stomach function
Denaturation
Expose peptide linkages
Tight-tight junctions between cells = no absorption occur
Secretion of intrinsic factor
Essential for life
Hoang 83
Digestion
Gastric secretory cells
Parietal cell
Only in body and fundus
Secrete HCl & Intrinsic Factor
Chief cell
Only body and fundus
Secrete pepsinogen
Mucous cells = protect stomach
Mucous neck cell
Secrete alkaline fluid with mucus
Mucous cells of cardiac and pyloric glands
Secrete alkaline fluid with mucus
Surface epithelial cells of stomach mucosa
Secrete alkaline fluid with insoluble mucus
Regulation of secretory cells
Acetylcholine
Secreted by postganglionic parasympathetic nervous system (PSNS)
Stimulate Auerbach’s plexus & Meissner’s plexus
Does not stimulate G-cells
Gastrin
Hormone secreted by G-cells of pyloric antrum
Released by gastrin-releasing peptide (GRP)
17 amino acid peptide = terminal tetrapeptide active fragment
Stimulate body and fundus
Cephalic phase of secretion
Caused by taste, smell, chewing, swallowing reflexes = psychic factors
Stimulate vagus efferents to stomach
Efferent vagi stimulate Auerbach’s & Meissner’s plexuses
Plexuses stimulate G-cells, chief cells, & parietal cells
G-cells secrete gastrin
Gastrin stimulate parietal cells to secrete HCl and IF
Gastric phase of secretion
Caused by distention of stomach = lasts as long as food is present in stomach
Vago-vagal reflex (long reflex)
Distention stretch nerves = influx of sodium into nerve cause stimulation
Vagal afferents transmit to efferents
Stimulate G-cells to secrete gastrin
Gastropancreatic reflex
Distension of stomach = signal vagal efferents
Stimulate exocrine pancreas secretion of digestive enzymes
Local reflex
Mediated by Meissner’s plexus
Local cholinergic reflex cause acid production
Caused by chemicals in food
Secretagogues in foods bind receptors in pyloric antrum
Meissner’s plexus stimulate G-cells via GRP
G-cell secrete gastrin to stimulate parietal cells
Secretagogues = amino acids, alcohol, caffeine, & calcium
Intestinal phase of secretion
Caused by food in intestine stimulating gastric pouch
Stimulation of duodenal mucosa cause secretion of Big Gastrin
Stimulate stomach secretion
Control of secretion
Hoang 84
Excess H+ from parietal cells inhibit G-cells
Excess H+ stimulate D-cells to release somatostatin
Somatostatin inhibit G-cell gastrin secretion = paracrine effect
Enterochromaffin-like cells (ECI) secrete enterogastrones
From intestine inhibiting stomach
Secretin, CCK, gastric inhibitory peptide, & vasoactive intestinal peptide
Direct inhibition of G-cells of pyloric antrum
Excess H+ stimulate S-cell to secrete secretin into blood
Inhibit G-cell from secreting gastrin
Enterogastric inhibitory reflex
Excess H+ stimulate nerves in mucosa & submucosa
Stimulate sympathetic celiac plexus to secrete norepinephrine
Inhibit Meissner’s plexus secretion of GRP = inhibit gastrin & HCl secretion
Signals from colon & small intestine inhibit stomach motility & secretion
Gastric juice
Secretions from parietal cells, chief cells, & mucous cells
Alkaline = neutralize parietal cell HCl acid
Histamine
Secreted by histaminocytes = gastric mucosal mast cells
Gastrin & acetylcholine receptors
Located adjacent to parietal cells
Potent stimulator of parietal cells via cAMP
Paracrine hormone
Multiplicative effect on acid secretion
Obligatory agent = acts directly on parietal cells
Acetylcholine & gastrin stimulate histaminocyte to secrete histamine
Ach & gastrin potentiate histamine effect on parietal cell
Formed from histadine via histidine decarboxylase
Parietal cell secretion
Maximum acidity = 150 mEq/L (pH 0.9)
pH increases during meal to pH 2 due to dilution & regurgitation
H/K-ATPase = antiporter on apical membrane
ATP from oxidative metabolism & anaerobic glycolysis
HCO3/Cl-antiporter on basolateral membrane
Isosmotic with blood plasma
Organic gastric secretion
Pepsin
Endopeptidase stored in chief cells as pepsinogen
Activation via HCl & pepsin
Ach, gastrin, histamine, & H+ stimulates chief cell secretion
Cleave basic-aromatic peptide bonds = form peptides
Optimal pH = 2-3
Mucopolysaccharide
Alkaline fluid with gel-like structure
Create 1.0-1.5 mm thick barrier
Protects stomach from mechanical & chemical injury
Denatures pepsin & neutralize H+ due to high pH
Secretion stimulated by mechanical and chemical irritations & vagus
R-protein
Synthesized by secretory segment cells of salivary glands
Binds B12 (extrinsic factor) after pepsin free it from protein complex in stomach
Gastric intrinsic factor (GIF)
Hoang 85
Synthesized by parietal cell & released when stimulated
Binds remaining B12 in stomach
Pancreatic proteases degrade R-protein in small intestine
GIF bind remaining B12
GIF-B12 complex bind high-affinity receptor in ileum brush border membrane to be absorbed
Required for heme biosynthesis
V. Exocrine Pancreas
Functions
Neutralize gastric juice = pH 3.3 in stomach increases to pH 6.1 in small intestine
Small volume secretion
Digestive carboxypeptidases secreted by pancreatic acinar cells
Endopeptidase = form peptones & oligopeptides
Trypsin – chymotrypsin – elastase
Trypsinogen activated by enterokinase on intestinal epithelium
Trypsin activate all other endopeptidases
Exopeptidase = form amino acids
Carboxypeptidase A – carboxypeptidase B
Trypsin inhibitor
Form 1:1 combination with trypsin = inactivation
Protect pancreas from autolysis
Lipase = form glycerol, fatty acids, & monoglycerides
Degrade fats
Amylase = form maltose, detrin, & glucose
Activated by chloride
Stimulated by calcium
Large volume secretion
Water & electrolytes secreted by intercalated ducts
Sodium & potassium concentrations equal that of plasma
Bicarbonate increases as secretion rate increases = always higher than plasma
Intercalated cells = high concentration of carbonic anhydrase
Neutralize gastric contents
Chloride decreases as secretion rate increases = always lower than plasma
[HCO3-]pancreas + [Cl-]pancreas = [HCO3-]plasma + [Cl-]plasma
Neural regulation of pancreatic secretion
Cephalic phase via efferent vagus
Stimulate secretion of enzymes (small volume secretion)
Gastropancreatic reflex
Secretion stimulated by distended stomach via long reflex
Afferent to efferent vagus fibers
Hormonal regulation of pancreatic secretion
Secretin
Secreted from duodenal mucosa S-cells
Stimulate pancreas to secrete large volume (bicarbonate)
Stimulate growth of exocrine pancreas
Inhibit G-cells & acid secretion
Inhibit gastric motility
Respond to duodenal pH 4.5 or less
Protect duodenum by stimulating secretion of alkaline juice (no enzymes)
Via intercalated cells of pancreas
Stimulated by H+, peptides, fats, triglycerides, & amino acids
Via cAMP
Hoang 86
Cholecystokinin
Secreted from duodenal mucosa I-cells
Stimulate pancreas to secrete small volume (enzymes)
Potentiates secretin stimulation of pancreatic bicarbonate
Stimulate growth of exocrine pancreas & gallbladder mucosa
Relaxation of sphincter of Oddi
Cause contractions of gallbladder
Stimulated by peptides, amino acids, fatty acids, & monoglycerides
Triglycerides do not cross mucosal cell membrane = no stimulation of CCK
Gastrin
Secreted from pyloric antrum G-cells
Stimulated by gastric distension
Gastrin induce pancreatic secretion of digestive enzymes with little aqueous juice
Gastrin induce P-cells secretion of acid
VI. Liver & Gallbladder
Bile
Synthesized by hepatocytes continuously
Secreted into bile canaliculus to biliary system
Provide osmotic force for production & flow of bile
Hepatic duct – common bile duct – sphincter of Oddi – ampulla of Vater
Recirculation in enterohepatic circulation = 4-12 times per day
Proximal & middle small intestine = reabsorbed passively
Distal ileum = reabsorbed actively
Most abundant organic anion secreted into canaliculi
Active transport of inorganic ions NaCl provide osmotic force
Function of bile
Solubilize biliary lipids
Lipid digestion & absorption
Lipid-soluble xenobiotics excretion
Toxins & waste products excretion
Cholesterol homeostasis
Gut immunologic function
Composition of bile
Lipids = lecithin & cholesterol
Major component
NaCl = via active transport
Bile acids = primary detergents & major organic solutes
HCO3- = minor
Proteins & bilirubin conjugated with glucuronic acid
Produced in reticuloendothelial system
Bile acids
Primary bile acids = cholic & chenodeoxycholic acid
Secondary bile acids = deoxycholic, lithocholic, & ursodeoxycholic via intestinal bacteria
Steriod nucleus & aliphatic side chain
C-terminus conjugated to glycine or taurine
Increase hydrophilicity – lowers pKa – resists degradation
High bile acids
Inhibits bile acid synthesis from cholesterol
Inhibits cholesterol synthesis
Actively secreted against concentration gradient
100 fold greater than in plasma
High canaliculi osmolarity = water flow passively into canaliculi
Bile acid dependent flow
Hoang 87
Electrolytes
Actively secreted into bile
Hepatocytes secrete bicarbonate into canaliculus
Water & NaCl follows passively = bile acid independent flow
Preventing kidney stones
Increased bile acid secretion = increases lipid secretion
Lecithin secretion is always higher than cholesterol
Mixed micelle = increase solubility of free cholesterol 2 million fold
Formation of kidney stones
Biliary cholesterol & lecithin forms unilamellar vesicles
Unilamellar vesicles coalesce into multilamellar vesicles
Becomes cholesterol crystals = eventually gallstones
Metastable zone = supersaturated cholesterol in bile but no crystals form
Low cholesterol (but higher than normal in micelle stage)
High lecithin
High bile salts
Gallbladder
Store bile during fasting
Neural control
Vagal stimulation cause contraction = cephalic phase
Hormonal control
CCK released from intestinal mucosa (fat or protein)
Gallbladder contraction & relaxation of sphincter of Oddi
Stimulate pancreatic secretion of enzymes (small volume)
Secretin released from intestinal mucosa (protons, fats, & proteins)
Stimulate biliary epithelium to secrete bicarbonate
Stimulate pancreatic secretion of biocarbonate (large volume)
Actively absorbs water & electrolytes from stored bile
Sodium actively reabsorbed with water following passively
Bicarbonate go with sodium = pH decreases in gallbladder
Identical osmotic pressure between gallbladder bile & hepatic bile and plasma
VII. Carbohydrate Digestion & Absorption
Small intestine
Villus function in absorption
Crypt function in secretion
Tight junctions between each enterocyte
Duodenum = minor in absorption
Function in neutralization and regulation of chyme movement
Jejunum & ileum = major in absorption
Carbohydrates
Salivary amylase & pancreatic amylase hydrolyze starch into dextrins & maltooligosaccharides
-Dextinase on brush border membrane degrade -limit dextrins into glucose
Glucoamylase on brush border degrade maltooligosaccharides into glucose
Disaccharides degrade into specific monosaccharides on brush border
Lactase make galactose & glucose
Sucrase make fructose & glucose
Hoang 88
Only monosaccharides are absorbed into enterocytes
Glucose via SGLT-1 = sodium-dependent active transporter
Water absorb with sodium and glucose/galactose
Galactose via SGLT-1
Fructose via GLUT-5 = sodium-independent transporter
Monosaccharides transport across basolateral membrane into portal blood
GLUT-2 = sodium-independent transporter
Glucose – galactose – fructose
Water exit via aquaporins on basolateral membrane
Disaccharides are not transported or absorbed in small intestine
Lactose is degraded by lactase on intestinal border
Lactase deficiency = lactose intolerance
Lactose intolerance = autosomal dominant
Deficiency in lactase
Increased concentrations of lactose in intestinal lumen
Increased osmotic effect = draws water into lumen
Cause osmotic diarrhea
Microflora degrade lactase = fermentation
Produce hydrogen gas = distention & bloating
Decreased pH of stool = acid stool
Detection
Disaccharide tolerance test
Lactose tolerant = increased plasma glucose from lactose breakdown
Peroral jejunal mucosa biopsy & enzyme assay
Breath hydrogen test
Adult onset
Worldwide & most common
Secondary lactose intolerance
Intestinal insult cause shut down of lactase
3 day enterocyte turnover period
Congenital
Rare
No breast-feeding
Osmotic diarrhea
Lactose intolerance
Sorbitol ingestion
Malabsorption
VIII. Protein Digestion & Absorption
Carboxypeptidase = pancreatic zymogens
Recognize C-terminus of proteins
Secreted by pancreas acinar cell
CCK from intestinal epithelial endocrine cell
Stimulated by proteins and fats
Vagus stimulation
Trypsinogen activated to trypsin via enterokinase
Trypsin activates other zymogens
Trypsinogen – chymotrypsinogen – proelastase = endopeptidases
Procarboxypeptidase A & B = exopeptidases
Digestion & absorption (via villus)
Gastric pepsin & HCl denature and hydrolyze protein
Hoang 89
Pancreatic C-terminal proteases breakdown protein
Degrade into free amino acids, peptides, & dipeptides
Peptides breakdown further via apical membrane amino-peptidases
Apical membrane H+ dependent active transporter of amino acids & dipeptides
Dipeptide transport is more efficient than free amino acid transport
Intracellular dipeptides degraded into amino acids
Via intracellular enterocyte aminopeptidase
Only amino acids are transported by basolateral membrane amino acid transporters
Enterocyte
Glutamine = major energy source of small intestine
Spare glucose
IX. Lipid Digestion & Absorption
Major classes of lipids
Triglycerides
Hydrolyzed by lingual & pancreatic lipase
Degrade to 2 free fatty acids & 2-mono-glyceride
Phospholipids
Hydrolyzed by pancreatic phospholipase A2
Lecithin degrade to 1 free fatty acid & 2-lysolecithin
Cholesterol esters
Hydrolyzed by pancreatic cholesterol esterase
Degrade to 1 free fatty acid & cholesterol
Step 1: Emulsification
Occur in stomach antrum & duodenum
Fats become fat droplets
Intact TG, lecithin, cholesterol esters
Some fatty acids, monoglycerides, lysolecithin, & cholesterol
Fats stimulate CCK release from I-cell
Contraction of gallbladder
Pancreatic enzyme secretion (small volume) & potentiates secretin on bicarbonate secretion
Growth of exocrine pancreas & gallbladder mucosa
Inhibit gastric motility = slows gastric emptying
Exocrine pancreas secrete trypsinogen, pro-colipase, & lipase
Trypsinogen converts to trypsin via duodenal enterokinase
Pro-colipase converts to colipase via trypsin
Colipase = positive charge
Displace bile from triglycerides & facilitate lipase binding
Lipase = negative charge
Gallbladder release bile salts
Unconjugated & deconjugated = not useful in metabolism
Conjugated = useful form for metabolism of fats
Negative charge at pH 7.5
Step 2: Hydrolysis
Hydrolysis on droplet surface
Lipase hydrolyze triglyercides to 2 fatty acids & 2-monoglycerides
Long-chain fatty acids = C17 or more
Short and medium chain fatty acids = C7 or less & C8-C16
Bile salts become deconjugated via microflora
Reabsorbed in distal ileum via enterohepatic circulation
Step 3: Packaging
Long-chain fatty acids packaged into mixed micelle
Contains conjugated bile & phospholipids with vitamins K, D, E, & A
Step 4: Absorption
Hoang 90
Short & medium-chain fatty acids with 2-monoglycerides absorb directly into portal blood system
Mixed micelle contact intestinal villus due to hydrophilic surface of micelle
Diffusion of 2-monoglycerides, lysolecithin, cholesterol, & Vitamins K, D, E, & A
Free fatty acid transporter required for FFAs
Bile salts stay in lumen until distal ileum via Na+-dependent active transport
Step 5: Chylomicron formation & transport
Long-chain FFA activated with CoA = FFA-CoA
sER = monoglyceride pathway
2 FFA-CoA join with 2-monoglyceride to form triacylglyceride
Packaged into chylomicron in cytoplasm with ApoA & ApoB
rER = Phosphatidic acid pathway
Form phospholipids
Packaged into chylomicron in cytoplasm
Lysolecithin + vitamins + cholesterol = packaged into chylomicron
Chylomicron transport to golgi
Chylomicrons absorbed into lymphatic system of villus
Cholesterol can also form VLDL
Short & medium-chain FFA reform triacylglycerides
Diffuse directly from enterocyte into portal vessel
X. Fluids & Electrolytes
Water
Output
Ingest 2000 mL/day
Secrete 1500 mL/day saliva
Secrete 2000 mL/day gastric juice
Secrete 500 mL/day bile
Secrete 1500 mL/day pancreatic juices
Secrete 1500 mL/day intestinal juices
Reuptake
Absorb 8500 mL/day in small intestine
Absorb 400 mL/day in colon
Excrete 100 ml/day of water
Jejunum enterocyte
SGLT-1
Active apical transport of sodium, water, & glucose/galactose
Glucose bind first – then sodium – then water (40%)
Sodium exit enterocyte via
GLUT-5
Apical transport of fructose alone
GLUT-2
Basolateral transport of glucose, fructose, & galactose
Water diffuse into enterocyte
Passively via SGLT-1 water channel = 25%
6 L = channel + coupled
Passive diffusion through membrane = 35% (3L)
Water exit enterocyte via aquaporin 4 & 3
Jejunum epithelial cell
Crypt of Lieberkuhn
Secrete chloride via CFTR = cAMP
Sodium follows via electrical gradient
Enters through basolateral membrane via Na/K/2Cl-transporter
Water diffuse into lumen
Hoang 91
Enters through basolateral membrane via AQP4
Ileum
Secrete bicarbonate for titration
Secretory diarrhea
Cholera = toxin-mediated
Neoplastic causes
Cholera toxin-induced secretory diarrhea
Excess secretion of chloride and water
Toxin
Inhibits GS-GTP degradation
Increased cAMP = increased chloride permeability via CFTR
Stimulate excess secretion of VIP from enteric nervous system
Activate adenylate cyclase
Treatment
Oral rehydration therapy
Increases villus absorption via SGLT-1 in crypt
High sodium, glucose, & chloride
Crypt secretions are independent of villus reabsorption
Simultaneous events
Iron absorption in duodenum & upper jejunum
Via heme transporter
Iron circulate in Fe3+ state
XI. Propulsion & GI Movement
Myogenic control
Mediated by basic electrical rhythm (BER)
BER = slow waves (basal electrical rhythm)
Cyclic depolarization of muscle membrane
Propogated aborally via nexuses to adjacent cells
Interstitial cells of Cajal
Form networks connecting GI musculature
Pacemakers for intestinal electrical slow waves
Slow wave of GI muscles
Phase 0 = K+ out
Resting membrane potential
Phase 1 = Ca2+ in
Upstroke depolarization
Phase 2 = K+ out
Transient repolarization
Phase 3 = K+ out & Ca2+ in
Plateau phase
Phase 4 = K+ in
Repolarization
BER frequency
Stomach = 3 waves / min
Duodenum = 11 waves / min
Ileum = 8 waves / min
Colon = 3-6 waves / min
Sympathetic nervous system
Hoang 92
Slows GI motility
Superior cervical ganglion & prevertebral ganglia
Parasympathetic nervous system
Increase GI motility
Vagal nerve & pelvic nerve
Enteric motor neurons = final pathway to GI musculature
Inhibited by VIP & NO
Excited by substance P & acetylcholine
Muscle contractions
Peristalsis = bolus propulsion
Ring of contraction moves abroad
Segmental = bolus mixing
Ring of contraction is stationary
Oropharyngeal phase
Voluntary initiation of swallow
Sealing of nasopharynx
Protection of airway
Clearing of bolus
Oropharyngeal dysphagia
Difficulty initiating swallow
Nasopharyngeal regurgitation
Pulmonary aspiration
Residual
Esophageal peristalsis
Primary peristalsis = initiated by swallowing
Lower esophageal sphincter relaxes as bolus arrives then contract at the end
Secondary peristalsis = esophageal distension
Lower esophageal sphincter relaxes longer and contracts later
LES = primary barrier against gastroesophageal reflux
Decrease LES pressure = increased reflux
Secretin – CCK – progesterone – VIP – GIP
Fat
Increase LES pressure = decreased reflux
Gastrin – Substance P – ADH – Ang II
Protein
Neural blockade = minimal effect on LES pressure
Primary myogenic in origin
Atropine (anti-cholinergic) decrease LES pressure slightly
Ca2+ decrease LES pressure effectively
LES relaxation
Mediated by non-cholinergic, non-adrenergic (NANC) motor neurons
VIP & NO
Stomach functional motor regions
Reservoir = body & fundus
Tonic contractions
Receptive relaxation to minimize change in pressure
Antral pump = antrum
Phasic contractions
Break foods into 1 mm before emptying into duodenum
Ileal brake = Lipids in distal small intestine delay gastric emptying
Pacemaker region = greater curvature of corpus
Determine max frequency – propagation velocity – propagation direction
Hoang 93
Gastric emptying factors
Increase
Isotonic solutions
Low caloric content
Small particle size
Erythromycin (Motilin agonist)
Solid foods
Decrease
Hypertonic solutions
High caloric cotent
Large particle size
Lipids
Stress
Liquid foods
Vagotomy
Liquids = increased emptying due to lost of receptive relaxation reflex
Discomfort threshold decreased
Solids = decreased emptying due to lost of antral pump
Gallbladder
CCK decrease sphincter of Oddi pressure & increase gallbladder contraction
Morphine increase pressure and frequency of contractions
Contraction is coordinated with onset of MMC in antro-duodenal region
Migrating motor complex = fasting state
Intestinal housekeeper
Seen during interdigestive state
Has phase of irregular contractile activity (phase II) before phase of regular contraction (phase III)
Migrates with decreasing velocity from duodenum to ileum
Return bile acids to enterohepatic circulation during interdigestive state
Ileus phase I
Contractile quiescence
Ileus phase II
Less than 100% of slow waves trigger peristaltic contractions
Ileus phase III
100% of slow waves trigger peristaltic contractions
Feeding state = no MMC
MMC switch to segmenting motility = keep food stationary
Colon movments
Mixing movements
Haustral migration
Mass movments
Colonic smooth muscle
Innervated by vagus
Cholinergic & non-cholinergic
Sympathetics inhibit colonic contraction & frequency
Anal sphincter
Allows defaction via dilation of internal sphincter
Provide continence via contraction of external sphincter
Aganglionosis
Internal anal sphincter inable to relax = cause pain
External anal sphincter is still able to contract
Hoang 94
XII. Gastrointestinal Pathology
Gastric ulcers
Mucus barrier impaired
H+ secretion lower than normal
But still injures gastric mucosa
Gastrin level is high in blood due to lower-than-normal acid concentration
Duodenal ulcers
Higher than normal H+ secretion
Increased number of gastric P-cells
Due to trophic effect of gastrin
Result in higher-than-normal gastrin levels in response to a meal
Zollinger-Ellison syndrome
Triad
Non-beta islet cell tumors = gastrinomas
Gastric acid hypersecretion
Severe ulcer disease
Hypergastrinemia = 1,000 pg/mL
Induce ECL (histamine) & P-cells (HCl) secretion
Trophic effects = prominent gastric mucosal folds
Gastrinomas
1/3 originate outside pancreas
Second portion of duodenum = most common
1/3 of patients develop diarrhea
50% are malignant
Peptic ulcer disease = developed in 95% of patients
First portion of duodenum = most common location of ulcers
Diagnose by fasting serum gastrin of 1000 pg/mL or higher
Treatment
H2-receptor antagonists
Omeprazole = H/K-ATPase inhibitor
Stomatistatin analog = inhibit gastrin release
Surgical removal of gastrinoma
Proximal vagotomy
Pernicious anemia
Atrophic gastric mucosa of body of stomach
Achlorhydria or hypochlorhydria
Most common cause of hypergastrinemia
Steatorrhea
Malabsorption of lipids
Pancreatic disease = cystic fibrosis or pancreatitis
Hypersecretion of gastrin = inactivates pancreatic lipase
Deficiency of bile acids = ileal resection
Decreased intestinal cells = tropical sprue
Inability to synthesize apoprotein B = inable to absorb chylomicrons
Scleroderma
LES hypotension due to decreased smooth muscle tone (lower 1/3 esophagus)
Achalasia
Loss of ganglion cells
Degeneration of dorsal motor nucleus & vagal fibers
Simultaneous contraction = spasms
Treated with nitrates & calcium channel blockers or pneumatic dilation
Diffuse esophageal spasm
LES inable to relax
Nutcaracker esophagus
Hoang 95
High contractions with long duration
Normal peristaltic rhythm
Hypertensive LES
Normal peristalsis
Exaggerated post-relaxation contraction of LES
Non-ulcer dyspepsia
Lost of receptive relaxation
More sensitive to gastric distension & pain
Familial visceral myopathy
Degenerative disease of smooth muscle
Can’t contract & push foodstuffs through intestines
Leads to pseudo-obstruction
Neuropathic pseudo-obstruction
Normal smooth muscle tissue
Abnormal innervation of muscle = irregular contractions
No phase I – II – III
Treated with prokinetic agents = erythromycin
Gallbladder diseases
Primary biliary cirrhosis
Destroys small bile ducts
Inable to absorb fat soluble vitamins and cholesterol homeostasis
Crohn’s disease
Disease of distal ileum
Interrupt enterohepatic circulation of bile acids
Inable to absorb fats & fat soluble vitamins
Gallstones = fat fertile forty female
Cholesterol stones
80% cholesterol
Composed of mucin glycoproteins & calcium
Nucleation promoters = high biliary proteins & mucin glycoproteins & stasis
Nucleation inhibitors = apolipoproteins
Black pigment stones
Bilirubin & mucin glycoproteins
Brown pigment stones
Calcium & bilirubin with cholesterol and proteins
Symptoms
Biliary colic pain
Cholecystitis
Gallbladder gangrene & perforation
Pancreatitis
Cholangitis
Treatment
Increase bile acid secretion = dissolution of gallstones
Chenodeoxycholic acid
Ursodeoxecholic acid
Decrease cholesterol secretion
Lithotripsy
Cholecystectomy
Gold-standard
Cholestasis leads to hepatic retention & intestinal deficiency
Retention of bile acids = hepatocyte injury
Retention of bilirubin = jaudice
Retention of cholesterol = xanthomata
Deficiency of bile acids = malabsorption of triacylglycerols & vitamins K, D, E, & A
Hoang 96
XIII. Helicobacter Pylori Infection
H. pylori cause chronic active gastritis
Leads to duodenal ulcer – gastric ulcer – gastric cancer – MALT lymphoma
Higher incidence in non-Caucasian groups
Whites = 19-30%
Blacks = 54%
Hispanics = 59%
Seropositivity decreasing = decreasing antibodies to H. pylori
Diagnosis
Serology test
Breath test = false negatives
Stool antigen test
Treat patients who are to be treated with antibiotics
Treat only patients who test positive for H. pylori
BMT = 14 days
LAC = 14 days
OAC = 10 days
Do not treat in non-ulcer dyspepsia
Treating GERD cause more heartburn
Secondary Messengers
cGMP
Parasympathetic
Acetylcholine
PLC/PIP2
Sympathetic
Epinephrine
Histamine
cAMP
Secretin (pancreatic large volume)
CCK (pancreatic small volume)
cGMP
IP3