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

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/704

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

704 Cards in this Set

  • Front
  • Back
What organs make up the physiologic anatomy of the female sexual organs?
ovaries, fallopian tubes, uterus, vagina
Describe the changes of composition of the follicular pool with age.
the number of viable follicles DEC with INCing age, during prepuberty ~17% are atretic (dead) follicles while the rest are viable, as a fertile adult, the number of atretic follicles INC, the follicular pool DEC and there are also growing and ovulated follicles present, at postmenopausal period, most are atretic and only a small portion are ovulated
What is menopause?
it is triggered by the faltering and shutting down of the ovaries, which are a part of the body’s endocrine system, shuts down hormones which make reproduction possible and influence sexual behavior
What is GnRH?
gonadotropin-releasing hormone 1, AKA LHRH, responsible for the release of FSH and LH from the anterior pituitary which is controlled by the size and freq. of GNRH1 pulses as well as feedback from androgens and estrogens, low freq. GNRH1 pusles lead to FSH release whereas high freq. GNRH1 pusles stimulate LH release, is synthesized and released by the hypothalamus
What is FSH?
follicle stimulating hormone, synthesized and secreted in the anterior pituitary
1. stimulates the growth and development of secondary follicles, as follicle grows it releases inhibin which shuts off FSH production
2. stimulates granulosa cells to convert androgens to estrogen
3. stimulates the synthesis of LH receptors on granulosa cells
What is LH?
lutenizing hormone, synthesized and secreted in the anterior pituitary
1. triggers primary oocyte to complete meiosis I and enter meiosis II
2. initiates ovulation with LH surge
3. affects the transformation of remaining granulosa and thece internal cells to luteal cells
4. Lh is necessary to maintain luteal function for the first two weeks
What is estrogen?
steroid
1. maintains the female reproductive tract
2. responsible for the secondary sexual characteristics of the female
3. responsible for the rebuilding of the uterus (proliferative phase) after menses
4. INCes inhibits FSH (granulosa cells secrete inhibin, follistatin and activin which also regulate FSH secretion)
5. induces a surge in LH levels
What is progesterone?
1. initiates the conversion from a proliferative uterus to a secretory uterus for implantation
2. maintains the uterus in a secretory phase in pregnancy
3. inhibits LH production
4. hormone of pregnancy
5. if pregnancy does not occur, progesterone levels will DEC, leading to menstruation
Describe the approx. plasma concentrations of estradiol.
from days 0-14 (with 14 days being ovulation), estradiol INC then reaches a INCing and DECing period, at ovulation DEC rapidly then INC and DEC from day 16-28
Describe the approx. plasma concentrations of progesterone.
from days 0-14, very low, INC rapidly a day or two after ovulation when estradiol reachces its lowest point after ovulation, then DEC at about day 22
Describe the approx. plasma concentrations of FSH and LH.
from days 0-12 FSH higher than LH, then at day 12 FSH INC but not as rapidly as LH which overtakes it, corresponds with DEC in estradiol, at ovulation, LH and FSH both DEC and LH becomes lower than FSH again, large surge of LH is what triggers ovulation, progesterone is stimulated by LH, LH receptor located on the theca interna cells
Give a summary of the levels of hormones during menstrual cycle.
1. follicular development depends on FSH levels, early in the menstrual cycle, FSH levels INC, LH stimulates the production of androstenedione by theca interna cells which is transferred to follicular cells for its aromatization into estrogen (this event is known as theca interna-follicular cell synergism)
2. estrogen and FSh stimulate the synthesis of LH receptor by follicular cells late in the follicular phase, LH stimulates the production of progesterone by follicular cells
3. progesterone and estrogen secretion from the rapidly luteinizing follicle INC in response to LH stimulation
4. high levels of progesterone and estrogen inhibit LH and FSH secretion, the corpus luteus lasts only a few days in the absence of LH, LH is luteotrophic
5. if pregnancy does not occur, luteolysis begins 7 days after ovulation, the production of progesterone, estrogen and inhibin DEC and FSH levels INC gradually, menstruation begins
What are some characteristics of the monthly ovarian cycle?
duration is on average 28 days, anywhere from 20-45 days, in every cycle one ovum is montly released, uterine endometrium is prepared in advance for implantation
What effects do the gonadotropic hormones have on the ovaries?
LH and FSH are the main gonadotropic hormones, important in the menstrual cycle, pituitary secretion of LH and FSH progressively INCes starting form 9-12 years of age, normal sexual cycle begins at puberty (ages of 11-15 years), menarche is the first menstrual cycle
What are the stages of follicular growth in the ovary?
primordial follicle -> primary follicle -> vesicular follicle -> mature follicle -> early, developing and final corpus luteum, ALSO primary follicles -> secondary (preantral) follicles -> antral follicles -> preovulatory (Graafian follicle) -> ovulation
Describe the follicular phase of the ovarian cycle.
during childhood, primordial follicles (ovum surrounded by a single layer of granulosa cells) are present, at puberty, FSH and LH INC greatly leads to follicle growth, 6-12 primordial follicles are recruited per month
Describe the functional synergism between follicular cells and theca interna cells during early folliculogenesis.
in the primary and secondary follicle, follicular cells have FSH receptors, in the graafian follicle, LH receptors appear and coexist with FSH receptors, the acquisition of LH receptors is essential for the luteinization of the ruptured follicle following ovulation
Describe the production of estradiol by follicular cells.
estradiol is the major steroid produced by follicular cells under stimulation by FSH, however, follicular cells depend on the supply of androstenedione by theca interna cells-regulated by LH-to produce estradiol (by aromatization of the androgen) since follicular cells lack the required enzymes for producing the precursor of estradiol
Describe the hormonal changes during the cycle.
1. FSH causes change from primary to antral follicle
2. estrogen (E2) causes FSH receptors on granulosa cells to INC leading to more stimulation from FSH
3. FSH + H2 leads to an INC in LH receptors on granulosa cells leading to stimulation from LH
4. FSH + LH + E2 leads to rapid growth of the follicles
5. only one follicle matures each month and the remainder undergo atresia
Describe ovulation.
1. surge of LH is essential for ovulation: 6-10 fold INC with a peak at ~16 hours before ovulation
2. FSH levels also INC (2-3 fold) but not sufficient to induce ovulation
3. LH surge also converts granulosa and thecal cells to progesterone-secreting cells -> E2 levels DEC 1 day before ovulation
4. environment when ovulation occurs (rapid growth of follicles, diminishing E2 levels after a prolonged phase of excessive secretion, onset of progesterone secretion)
Describe the physiological steps involved in the release of the egg during ovulation.
1. blood vessels of the theca intima proliferate (angiogenesis)
2. at the stigma, proteases from fibroblasts-induced LH surge-degrade collagen fibers of the tunica albuginea and theca externa
3. the follicular cell layer will begin to fold soon after ovulation
4. the basement membrane breaks down, blood vessels from the theca interna invade the follicular cell layer and the antral cavity is filled with blood (corpus hemorrhagicum)
Describe the initiation of ovulation.
LH INC in significant amounts which leads to an INC in progesterone, this leads to (1) a release of proteolytic enzymes from lysosomes of the theca externa which leads to swelling and degeneration of the stigma and (2) rapid growth of new blood vessels into the follicle wall, this leads to plasma transudation into the follicle which leads to follicle swelling, follicle ruptures and discharges the ovum
Describe the formation of the corpus luteum (luteinization).
following ovulation the follicular cell layer of the preovulatory follicle becomes folded and is transformed into part of the corpus luteum, a surge in LH is correlated with luteinization
Describe the transformations that occur during luteinization.
1. the lumen previously occupied by the follicular antrum is filled with fibrin which is then replaced by CT and new blood vessels piercing the basement membrane
2. follicular or granulosa cells enlarge and lipid droplets accumulate, they become follicular or granulosa lutein cells
3. the spaces between the folds of the follicular cell layer are penetrated by theca interna cells, blood vessels and CT, theca interna cells also enlarge and store lipids, they are now theca lutein cells
what is the function of the corpus luteum and what role do FSH and LH play in that function?
1. FSH stimulates the production of progesterone and estradiol by follicular lutein cells
2. LH stimulates the production of progesterone and androstenedione by theca lutein cells, androstenedione is translocated into follicular lutein cells for aromatization into estradiol
3. during pregnancy, prolactin and placental lactogens upregulate the effects of estradiol produced by follicular lutein cells by enhancing the production of estrogen receptors
4. estradiol stimulates follicular lutein cells to take up cholesterol from blood, which is then stored in lipid droplets and transported to mito for progesterone synthesis
What happens to the corpus luteum if fertilization does not occur?
luteolysis, involves a programmed cell death sequence, includes:
1. a reduction in the blood flow within the corpus luteum causes a decline in oxygen (hypoxia)
2. T cells reach the corpus luteum and produce interferon-gamma which in turn acts on the endothelium to enable the arrival of macrophages
3. macrophages produce tumor necrosis factor and the apoptotic cascade starts
What are the things that happen during the luteal phase of the ovarian cycle when fertilization does not occur?
1. involuation of the corpus luteum-E2 + progesterone -> FSH and LH DEC and inhibin from lutein cells leads to FSH DEC
2. onset of the next ovarian cycle-E2 + progesterone DEC -> FSH and LH INC
What is the corpus albicans?
regressed form of the corpus luteum, formed by macrophages laying down collagen, occurs 12 days after ovulation if not pregnant
What happens to the corpus luteum if fertilization does occur?
if pregnancy occurs, the corpus luteum survives for 2-4 months under the stimulation of HCG
Describe the chemistry of estrogen.
in the non-pregnant women it comes mainly from the ovary by ovarian follicular and thecal internal cells, small amount from the adrenal cortices, in the pregnant woman comes from the placenta
What are the three types of estrogens?
1. beta-estradiol: from ovaries
2. estrone-peripheral tissues (from adrenal and ovarian androgens)
3. estriol-weak estrogen, derivative from estradiol and estrone, in liver
Describe the different potencies between the three estrogens.
beta-estradiol > estrone > estriol
Describe the chemistry of progestins.
in the non-pregnant women, from corpus luteum, latter half of each ovarian cycle, in the pregnant women comes from the placenta
What are the two types of progestins?
1. progesterone: major
2. 17-alpha-hydroxyprogesterone-small amount (same effects)
Describe the synthesis of the estrogens and progestins.
1. in the ovaries-mainly cholesterol from blood
2. during the follicular phase-progesterone + adnrostenedione (thecal interna cells) -> conversion to E2 in follicular (granulosa) cells
3. during the luteal phase-follilcular lutein and thecal lutein cells produce progesterone
How are estrogens and progesterones transported in the blood?
estrogens and progesterone bound to plasma albumin and specific binding globulins in the blood
Describe the degradation of estrogen in the liver.
1. conjugation to form glucuronnides and sulfates -> mostly excreted in the bile, some from the urine
2. conversion from E2/estrone to estriol
3. diminished liver function INC estrogen activity -> hyperestrinism
Describe the degradation of progesterone in the liver.
1. quick turnover rate-a few minutes
2. major degradation product-pregnanediol
What functions do estrogens have on the uterus and external sex organs?
1. INC in size-internal (ovaries, fallopian tubes, uterus), external (vagina, fat deposition in the mons pubis and labia majora, enlargement of the labia minora)
2. vaginal epithelium-cuboidal -> stratified, more resistant to trauma and infection
3. uterus-size INC 2-3 fold, proliferation of the endometrial stroma, development of the endometrial glands
What functions do estrogens have on the fallopian tube?
1. proliferation of the epithelium
2. INC in the number of the ciliated epithelial cells
3. enhancement of cilia activity (beating)
What functions do estrogens have on breasts?
1. development of the stromal tissues
2. growth of an extensive ductile system
3. deposition of fat in the breasts
4. initiation of the growth of the breasts and of the milk-producing apparatus
5. further growth and function depend on progesterone and prolactin
What functions do estrogens have on the skeleton?
1. stimulates bone growth-INC in height at puberty
2. uniting of the epiphyses with the shafts of the long bones
3. effect on epiphyses-E2 >> T, girls stop growing earlier
4. females with earlier loss of E2 production tend to be taller
Describe osteoporosis in aging women.
after menopause, E2 levels DEC which leads to:
1. INC osteoclastic activity
2. DEC bone matrix
3. DEC deposition of Ca2+ and phosphate
What are some other functions of estrogen?
1. effect on protein deposition-slight INC
2. effect on body metabolism and fat deposition-metabolic rate slightly INC and fat deposition in subQ region, buttocks and thighs
3. effect on hair distribution-results from androgens from the female adrenal glands
4. effect on skin-soft and smooth, more vascular
5. effect on electrolyte balance-retention of Na+ and H2O in the kidney tubules (mainly during pregnancy)
What is the function of progesterone on the uterus?
1. promotes secretory changes in the uterine endometrium during 2nd half of the cycle-ready for implantation of the fertilized ovum
2. DEC the freq. and intensity of uterine contractions: prevents expulsion of implanted ovum
What is the effect of progesterone on the fallopian tubes?
promotes secretion of the Peg cells-nutrition for ovum
What is the effect of progesterone on the breasts?
1. development of the lobules and alveoli-proliferation and enlargement of alveolar cells -> secretory status
2. no milk production without prolactin
Describe the proliferative phase.
it occurs before ovulation under the influence of estrogens, stromal and epithelial cells proliferate rapidlye (re-epithelialization with 4-7 days after the beginning of menstruation), next 1.5 weeks, stromal cells INC, growth of epithelial glands and new blood vessels into endometrium -> thickness INC to 3-5 mm
Describe the secretory phase.
occurs after ovulation under the influence of progesterone and estrogens, E2 -> futher cellular proliferation in the endometrium, progesterone -> marked swelling and secretory development of the endometrium
What role does progesterone have secretory phase?
1. glands more tortuous
2. excess of secretory substance in glandular epithelial cells
3. stromal cell cytoplasm INC with deposition of glycogen and lipid
4. blood supply INC
Describe the thickness of endometrium.
peak at 1 week after ovulation, 5-6 mm
What is the physiologic meaning of the morphologic changes?
1. highly secretory endometrium provides large amount of stored nutrients for fertilized ovum from entry to the uterus (3-4 days after ovulation) to implantation (7-9 days after ovulation)
2. once implanted, trophoblast cells digest and absorb nutrients to provide to early implanting embryo
what affects the functional layer of the endometrium?
1. changes in the blood levels of estrogens and progesterone
2. the blood supply from spinal arteries
3. this layer is partially or totally lost after menstruation
What happens to basal layer with changes in the blood levels of estrogens and progesterone?
the basal layer is not affected by changes in the blood levels of estrogens and progesterone, the blood supply derives from basal arteries rather than spiral arteries, this layer is not lost after menstruation, the functional layer regenerates after menstruation from the basal-functional layer boundary
Describe menstruation.
occurs when no fertilization occurs, the corpus luteum degenerates -> E2 and P DEC -> (rapid involution of endometrium, vasospasm of blood vessels: release of prostanglandins), necrosis of the endometrium especially blood vessels, blood -> vascular layer -> hemorrhage areas expand for 24-36 h, necrotic outer layers separate from the uterus, ~48 h after the onset of menstruation all the superficial layers desquamated, uterine contraction-to expel the contents, ~40 mL blood and ~35 mL serous fluid are lost
What happens during the premenstrual or ischemic stage?
1. periodic contractions of the spiral artery-triggered by a reduction in progesterone-deprive the supply of O2 (hypoxia) to the functional layer
2. a breakdown of the spiral artery floods the lamina propria with blood
3. the functional layer-consisting of glands and decidual-like cells-detaches and sheds into the uterine cavity (menses)
4. the basal layer is not affected because basal straight artereies provide independent blood supply to this layer
Describe the regulation of the female monthly rhythm-interplay between the ovarian and hypothalamic-pituitary hormones (GnRH).
1. intermittent, pulsatile secretion of GnRH (5-25 min/1-2 hours) -> pulsatile release of FSH and LH (every 90 min)
2. pulsatile secretion of GnRH is essential for its function
3. in mediobasal hypothalamus, especially in the arcuate nuclei
4. regulated by signals from other centers
Describe the regulation of the female monthly rhythm-interplay between the ovarian and hypothalamic-pituitary hormones (negative feedback effects of E2 and P).
1. estrogen inhibits both FSH and LH
2. progesterone alone-little effects
3. P + E2-strong inhibitory effects on FSH and LH production
4. act on both pituitary and hypothalamus levels
5. inhibit-secreted by granulosa cells, inhibit FSH and LH levels
Describe the regulation of the female monthly rhythm-interplay between the ovarian and hypothalamic-pituitary hormones (positive feedback effect of E2 before ovulation).
1. LH surge 1-2 days before ovulation
2. FSH shows a smaller surge
3. during 1st half of the cycle, FSH and LH levels are first slightly suppressed and then rapid INC in LH (6-8 folds) and FSH (2 folds) right before ovulation
4. without the LH surge no ovulation
5. mechanism: unknown (positive feedback effect of estrogen, onset of progesterone production by granulosa cells)
What happens with an insufficient LH surge?
cycle without ovulation = anovulatory cycle, corpus luteum fails to develop: no P production, several days shorter, the first couple of cycles at puberty or cycles several months or a year before menopause, P control the rhythm but not the cycle itself
Describe GnRH secretion during puberty and menarche.
during childhood there is no GnRH secretion due to suppressive factors in the brain, during puberty there is a gradual INC of gonadotropins, there is an INC in total urinary gonadotropin release from puberty to menopause, when reach menopause INC very rapidly, then starts to DEC, INC before menopause is oscillatory
Describe menopause.
1. cycle ceases and female hormone production diminishes
2. ovarian burning out
3. before 45, 400 follicles mature and being ovulated
4. after 45, only a few remain to be responsive to FSH and LH -> estrogen levels DEC -> FSH and LH INC
5. loss of estrogen which leads to menopausal syndrome
What are the symptoms of menopausal syndrome?
1. hot flashes
2. psychic sensation of dyspnea (shortness of breath)
3. irritability
4. fatigue
5. anxiety
6. DEC strength and calcification of bones
What is hypogonadonism?
1. causes irregularity of menses and amenorrhea-estrogen levels must reach certain points to maintain normal cycle
What happens if the ovaries are absent from birth or they become non-functional before puberty?
female eunuchism, lack of 2nd sexual characteristics, sex organs remain infantile, prolonged growth of long bones: delayed epiphyses fusion with the shafts -> taller
What happened if the ovaries of a fully developed woman are removed?
sex organs regress, similar to women after menopause
Describe hypersecretion by the ovaries.
rare, often associated with granulosa cell tumor (inhibin deficiency)
Describe the stimulation of female sexual act.
psychic stimulation, local stimulation
Describe female erection and lubrication.
under parasympathetic signals, erection of clitoris (same mechanism as the penile erection), lubrication (mainly by mucus secreted by the Bartholin’s glands (located beneath the labia minora) and vaginal epithelium)
Describe the female orgasm.
analogous to male emission and ejaculation, important for fertilization, includes:
1. perineal muscle contract rhythmically caused by spinal cord reflexes -> uterus and fallopian tube motility INC to help transport sperm upward to the ovum
2. dilation of the cervical canal for up to 30 min: easy entry of sperm
3. oxytocin secretion by posterior pituitary -> contraction of the uterus -> facilitate sperm transport
Describe the fertile period of each sexual cycle.
1. within 24 hour after ovulation
2. sperm in the female tract can survive up to 5 days
3. sperm have to be there from 4-5 days before ovulation and a few hours after ovulation
Describe the rhythm method of contraception.
1. interval from ovulation to menstruation-13-15 days
2. if a 28 day cycle, ovulation usually occurs within 1 day of the 14th day of the cycle
3. if a 40 day cycle, ovulation should be within 1 day of the 26th day of the cycle
4. avoid intercourse for 4-5 days before the calculated day of ovulation and 3 days afterwards
5. prerequisite-regular periodicity of the menstrual cycle
Describe hormonal suppression of fertility-contraceptive pills.
1. theory-appropriate quantitiy of estrogen and progesterone in the first half of the cycle inhibits LH surge leading to no ovulation
2. too much of either can cause abnormal menstrual bleeding pattern
3. synthetic estrogens + synthetic progestrins (because avoids liver degradation)
4. taken in the early stage of the cycle and continue beyond the time point of normal ovulation and then stopped to allow menstruation
Describe female infertility.
incidence of 15% of married couples, 50% are female factor, causes include endocrine, structural and genetic
Describe an endocrine abnormality.
anovulation due to hyposecretion of gonadotropins which is insufficient to induce ovulation, test urine pregnanediol levels (no surge, no ovulation), body temperature (secretion of progesterone INC body temperature ~0.5F), treatments include HCG, ovulation from multiple follicles, multiple births
Describe a structural abnormality.
1. ovarian anatomic abnormalities
2. endometriosis-endometrial tissue surrounding uterus, fallopian tubes and ovaries grows in pelvic cavity and even menstruates leads to fibrosis throughout the pelvis which leads to obstruction of ovulation, capture by the fimbriae of the fallopian tubes
3. abnormal secretion of mucus by the uterine cervix
What is salpingitis?
inflammation of the fallopian tubes
Describe a genetic abnormality.
mutations in genes essential for follicular genesis
Describe the hormonal control of ovarian function.
GnRH from the hypothalamus, FSH and LH from the anterior pituitary under the stimulation of GnRH, estrogen from the granulosa and thecal cells
what does estrogen do for controlling ovarian function?
1. proliferation of endometrium
2. thinning of the cervical mucus
3. stimulate pituitary to secrete LH
4. LH surge at mid-cycle which leads to ovulation
5. stimulate progesterone production
Describe ovulation.
LH surge causes completion of meiosis I leading to a preovulatory follicle, is then arrested in metaphase of meiosis II 3 hours before ovulation
On the ovarian surface, what does an INC in LH do?
1. LH INC leads to INC in collagenase, which leads to digestion of fibers around the follicle
2. LH INC leads to INC in prostaglandin which leads to muscular contraction
Describe the corpus luteum.
composed of lutean cells (granulosa cells and thecal cells), secretes progesterone, progesterone + estrogen leds to uterine mucosa into progestational/secretory stage in preparation for implantation
Describe oocyte transport.
ovulated oocyte is captured by sweeping movement of the fimbriae of the uterine tube (oviduct) and by the motion of cilia on the epithelial lining, from ampulla to uterine lumen (3-4 days), fertilization occurs at the ampulla of uterine tube
What are the different fates of the corpus luteum?
1. no fertilization-lutena cells apoptosis leads to corpus albicans which leads to DEC in progesterone which leads menstrual bleeding
2. fertilation-hCG secreted by the syncytiotrophoblast which leads to further growth
3. progesterone production until the end of 4th month leads to trophoblastic secretion of progesterone becomes adequate
Describe the transport of spermatozoa in the male.
1. passive transfer to epididymis via testicular fluid
2. maturation in epididymis (up to 2 weeks)
3. rapid transit through ductus deferens
4. addition of fluid from seminal vesicle
5. addition of prostatic fluid
Describe the transport of spermatozoa in the female.
1. sperm depositied in upper vagina (rapid elevation of pH) (10^7)
2. passage through cervix (fast and slow phases) 10^6
3. passage through uterus 10^5
4. entry into uterine tubes 10^4
5. passage up uterine tube by swimming and contractions of tube
6. only a small number of sperm near egg at a given time (10^2-10^3)
Describe the transport of the egg.
1. delay at amupllary-isthmic junction where fertilization occurs (days 1-2)
2. delay at utero-tubal junction (days 2-3)
3. egg enters uterine cavity as a morula (days 3-4)
4. blastocyst implants (day 7)
Describe the steps in fertilization.
1. capacitation-maturation of sperm in female reproductive tract leads to removal of the glycoprotein coat and seminal plasma protein form the sperm plasma membrane (important for IVF and IUI)
2. acrosome reaction-release of enzyme by sperm to dissolve zona pellucida (ZP)
3. fertilization occurs at the ampulla of the uterine tube
Describe the phases of fertilization.
1. phase I-penetration of the corona radiate
2. phase II-penetration of zona pellucida
3. phase III-fusion of the oocyte ad sperm cell membranes
Describe the acrosome reaction.
sperm reaches the corona radiate, releases its acrosome contents and passes through the corona radiata, then passes through the zona pellucida and plasma membrane of the egg
Describe the time course of fertilization.
1. zona pellucida (15-25 min)
2. perivitelline space (<1 sec)
3. perivitelline membrane (<1 min) (release of cortical granules and completeion of second meiotic division and formation of polar body)
4. male and female pronuclei visible (2-3 hours)
5. mitotic spindle and first cleavage (24 hours)
What does entry of a sperm into the oocyte trigger?
1. cortical and zona reactions to prevent polyspermy
2. oocyte completes meiosis II
3. egg is activated
Describe the events that occur to the membrane during fertilization.
after sperm enters the egg and rapid depolarization of plasma membrane of egg, zona pellucida hardens and sperm receptors inactivate
What are the results of fertilization?
1. restoration of diploid number of chromosomes
2. sex determination
3. initiation of cleavage
Describe what happens during cleavage and formation of the blastocyst.
1. blastomeres are formed-2, 4, and 8 cell stages, the cells get smaller and smaller
2. compaction occurs-goes from a loose form then compacted form which leads to segregation of inner and outer cells
3. morula forms at day 3 (a 16 cell form), the inner cell mass leads to the embryo proper which eventually forms tissues while the outer cell mass is called the trophoblast which leads to the placenta
4. blastocyst forms-zona pellucida degenerates and the uterine fluid penetrates, this forms the blastocele (cavity), the inner cell mass is now called embryoblast and outer cell mass is called the trophoblast + epithelial wall
Describe implantation.
occurs during the secretory phase, there are three layer in the endometrium (compact layer, spongy layer, and basal layer), implants along the anterior or posterior wall
What effects does progesterone have on the uterine endometrium?
make stromal cells highly secretory, secrete glycogen, proteins, lipids and minerals to decidual cells
What do trophoblast cells invade?
trophoblast cells invade the decidua, this causes to the release of nutrients which leads to embryo growth and development, it is the main source of nutrition for 1-8 weeks, it is gradually replaced by placenta nutrition
Describe the different types of villi that occur during the development of the trophoblast.
1. primary villus = cytotrophoblastic core + syncytial layer
2. 2nd villus = mesodermal cells penetrate the core of 1st villi
3. 3rd (definitive placental) villus = mesoderm cells -> blood cells + blood vessels
Following formation of the villi, describe the further development of the trophoblast.
at the end of the 3rd week, maternal vessels penetrate the cytotrophoblastic shell to enter intervillous spaces, which surround the villi, capillaries in the villi are in contact with vessels in the chorionic plate and the connecting stalk, which in turn are connected to intraembryonic vessels
What else happens during the further development of the trophoblast?
at the end of the 3rd week, 3rd and 2nd villi give the trophoblast a radial shape, intervillous spaces are lined with syncytium, cytotrophoblastic cells surround the entire trophoblast and in contact with endometrium, the embryo is suspended in the chorionic cavity by the connecting stalk
What are the functions of the placenta?
1. diffusion-from the mother to the fetus (foodstuffs and O2), from the fetus to the mother (excretory products)
2. O2 diffusion-fetal hemoglobin displays high O2-carrying capability
3. 50% higher concentration of fetal hemoglobin
4. Bohr effect-hemoglobin can bind more O2 at a low PCO2 than a high PCO2
5. CO2 diffusion-foodstuffs, wastes (nonprotein nitrogens including urea, uric acid, creatinine
Describe human chorionic gonadotropin (HCG).
1. origin-syncytial trophoblast cells
2. onset-8-9 days after ovulation
3. peak-10-12 weeks
4. function-prevention of involution of corpus luteum
Describe the rescue of the corpus luteum by HCG.
note rise of progesterone ~7-10 days after ovulation (arrow in right panel), this marks the first recognition of pregnancy by the mother, the second peak represents placental progesterone secretion, the corpus luteum is no longer necessary for maintaining pregnancy
Describe the composition of hCG.
common alpha + hCG beta chain + CHO chains
hCG beta-very similar to LH-beta, but has more CHO -> much longer half-life than LH
Describe the target of hCG.
corpus luteum, fetal Leydig cells -> T -> stimulation of Wolffian ducts
Describe the signaling sequence of hCG.
LHR -> cAMP -> PKA
Describe the secretory pattern of hCG.
exponential rise is critical (LH administered in the same manner can also resuce the CL), hCG remains elevated after Cl involutes at the end of the 1st trimester
Describe the clinical utility of hCG.
diagnosis of pregnancy 7-10 days after fertilization
Describe the cell origin, synthesis and significance of estrogens.
1. cell origin-syncytial trophoblast cells
2. synthesis-from androgens produced by the fetal and maternal adrenal glands-the placenta cannot convert progesterone to androgens
3. significance of large scale production of estrogens-enlargement of the mother’s uterus, enlargement of the breasts and breast ductile structure, enlargement of external genitalia, relaxation of the pelvic ligament
Describe the importance of progesterone.
essential for implantation and the maintenance of pregnancy, relaxation of the uterus avoiding spontaneous abortion, decidualization of the endometrial stroma (nutrition), INC secretion from the fallopian tubes (nutrition to the early embryo)
What do progesterone and estrogens do to GnRH?
inhibits the GnRH pulse generator which reduces LH, FSH, corpus luteum is still alive which leads to blockade of new wave of folliculogenesis
Describe the origin of human chorionic somatomammotropin/Human placental lactogen.
syncytial trophoblast
Describe the composition of human chorionic somatomammotropin/Human placental lactogen.
polypeptide, short half-life
Describe the actions of human chorionic somatomammotropin/Human placental lactogen.
growth of the mammary gland (PRL-like), fetal growth (GH-like, via IGF-II), DEC glucose tolerance (GH-like), not indispensable
Describe the secretory pattern of human chorionic somatomammotropin/Human placental lactogen.
INC until the 36th week, large amounts
Describe the clinical utility of human chorionic somatomammotropin/Human placental lactogen.
diagnosis of DEC placental mass (e.g. due to placental infarction)
Describe other hormonal factors in pregnancy.
1. corticoid secretion
2. relaxin secretion-corpus luteum and placenta
3. pituitary-INC corticotrophin, thyrotropin and prolactin, DEC in FSH and LH
What is the response of the mother’s body to pregnancy?
1. weight gain: ~24 lbs.
2. metabolism during pregnancy-INC 15%
3. CO INC 30-40% by 27th week followed by a fall in the last 8 weeks for unknown reasons
4. blood volume is 30% above normal due to fluid retentition
5. respiration is 20% above normal
6. amniotic fluid is 500-1000 mL
7. preeclampsia (toxemia of pregnancy)-5% HTN + leakage of protein into urine
8. eclampsia-extreme degree of preeclampsia
Describe what happens during parturition (birth of the baby).
1. INC uterine excitability which leads to strong rhythmical contractions
2. hormonal factors that INC the uterine contractility
3. mechanical factors that INC the uterine contractility
What are the hormonal factors that INC the uterine contractility?
1. INC ratio of estrogens to progesterone-after 7th month, estrogens continue to INC while progesterone levels remain
2. oxytocin secreted by the neurohypophysis causes uterine contraction (oxytocin receptors on uterine muscles cells INC, secretion INC and stretching and irritation of uterine cervix which leads to INC in oxytocin
3. fetal hormones-oxytocin from the fetus’s pituitary, cortisol from adrenal glands, prostaglandins
What are the mechanical factors that INC the uterine contractility?
1. stretch of the uterine musculature
2. stretch of the irritation of the cervix
Describe the positive feedback mechanism for the initiation of labor.
1. baby’s head stretches cervix
2. cervical stretch excites fundic contraction
3. fundic contraction pushes baby down and stretches cervix some more
4. cycle repeats over and over again
Describe the contractions at the onset of labor.
Braxoton Hicks contractions occur which are weak and slow labor contractions
What is the role of oxytocin in parturition?
oxytocin administration can induce labor but oxytocin only starts to INC after the beginning of labor (stretch of the cervix), it hastens delivery, promotes the delivery of the placenta and reduces bleeding
Describe the abdominal muscle contractions during labor.
1. uterine contraction leads to pain, this pain elicits a neurogenic reflex from the spinal cord to the abdominal muscle that initiates more contraction
2. contraction in uterine fundus >> body >> lower segment near the cervix
3. every 30 mins -> every 1-3 minutes
4. 25 pounds of force each contraction
5. 95% head first
Describe the stages of contraction during labor
1. 1st stage-cervix dilation to the size of the head of the fetus, ~8-24 hours for the 1st pregnancy, can be a few minutes after
2. 2nd stage-membrane rapture, loss of amniotic fluid, head out, 1-30 mins
Describe separation and delivery of the placenta.
10-45 min after birth, contraction of the uterus leads to a shearing effect that separates the placenta from the uterine wall, bleeding occurs and is < 350 mL, contraction and prostaglandins
Describe labor pains.
1st stage-visceral sensory hypogastric nerves, 2nd stage-somatic nerves to spinal cord and brain
Describe the involution of the uterus after parturition.
4-5 weeks
What hormones are involved in the development of the breast and what is their target/action?
1. estrogens-target the lactiferous ductrs, stroma and fat
2. progesterone (w/ E2)-target the lobulo-alveolar system
3. cortisol, GH, IGF-I, PRL (nonlactating-levels)-permisive for other hormones
4. hCS/hPL-targets alveoli for milk secretion
5. PRL-targets alveoli for milk secretion
What do estrogens and progesterone do to milk secretion?
inhibit it
What does prolactin do for milk secretion?
activates milk secretion
Describe the levels of estrogen and progesterone during pregnancy.
10-20 time INC during 5th week to brith leading to no milk
What supports prolactin?
hCS
what happens to estrogen and progesterone levels after birth?
loss of estrogens and progesterone after birth -> PRL starts to work, leads to milk production (also requires other hormones including GH, cortisol, insulin and parathyroid hormone
What happens to the levels of PRL during breast feeding?
each time breast feeding, 10-20 fold surge of PRL lasts for ~1 hour
What happens to the ovarian cycle after delivery?
suppression of the femal ovarian cycles in nursing mothers for many months after delivery, PRL secretion inhibits gonadotropin secretion which leads to suppression of the ovarian cycle
What is lactational anovulation?
patterns of pulsatile secretion of LH and FSH over a 24 hour period in a fully breastfeeding woman at 4 weeks and 8 weeks postpartum, note suppressed GnRH pulse generator activity
Describe the ejection process in milk secretion.
is an ejection, not a leak, suckling leds to sensory impulses from nipples to spinal cord and then to hypothalamus, this leads to secretion of oxytocine from posterior pituitary, milk ejection occurs in about 30 sec-1 min, ejection reflex does not diminish with time
What does oxytocin do during milk secretion?
leads to contraction of myoepithelial cells which releases milk from alveoli to ducts
What is required for maintenance of lactation?
milk removal
When does spermatogenesis start?
starts at puberty (~12-14)
What are the three phases of spermatogenesis?
1. mitiotic-spermatogonial proliferation and differentiation
2. meitotic division of spermatocytes into spermatids
3. haploid-differentiation and morphogenesis of spermatids to spermatozoa
Describe the different cell types involved in spermatogenesis.
primordial germ cell -> spermatogonia -> proliferation of spermatogonia by mitotic cell division inside testis -> primary spermatocyte -> secondary spermatocyte -> spermatids -> mature sperm
What role does testosterone have on stimulating spermatogenesis?
1. secreted by the interstitial Leydig cells
2. essential for spermatogenesis, especially for meitotic and haploid phases
What role does LH have on stimulating spermatogenesis?
1. secreted by the anterior pituitary gland
2. stimulate the Leydig cells to secrete testosterone
What role does FSH have on stimulating spermatogenesis?
1. secreted by the anterior pituitary gland
2. stimulates the Sertoli cells to secrete factors to maintain quantitative spermatogenesis
What role does estrogen have on stimulating spermatogenesis?
1. aromatized from testosterone in Sertoli cells
2. re-absorption of luminal fluid in the head of the epididymis, allowing sperm to enter the epididymis concentrated rather than dilute
3. ER-beta on spermatogonia, spermatocytes and Sertoli cells may mediate the actions of xenoestrogens (estrogenic endocrine disruptors)
What role do growth factors have on stimulating spermatogenesis?
1. secreted from the Sertoli cells or germ cells
2. act on germ cells or somatic cells via their receptors
3. effectors of major hormonal signals
4. paracrine and autocrine regulation under the control of endocrine signals
Describe the maturation of sperm in the epididymis.
there are 120 million/day, no motility in the head of (caput) epididymis, gain during the transition from the caput to corpus (the middle) epididymis, moderate motility in the cauda (tail of) epididymis
What role does the vas deferens have on maturation of sperm?
most stored in the vas deferens where motility is remained >1 month, motility is suppressed in the vas deferens
Describe the maturation of motility of upon ejaculation.
motile upon ejaculation-initial motility, more motile in the female reproductive tracts-hyperactivated motility, survives ~1-2 days in the female tract, can survive in a neutral or slightly alkaline medium but an acidic medium leads to loss of motility
What is the function of the seminal vesicles?
1. secrete a mucoid material enriched in fructose, citric acid, prostaglandins, fibrinogen and other nutrient substances
2. empty its contents in to the ejaculatory duct
3. provide nutrition to the sperm in the semen
4. prostaglandins-aid fertilization
What are the functions of the prostate gland?
1. secrete fluid containing Ca2+, citrate ion, a clotting enzyme, and a profibrinolysin
2. during emission, contract together with the vas deferens to add fluid to the semen
3. alkaline to neutralize acidic vas deferens fluid and vaginal secretion (pH 3.5-4.0)
Describe the composition of semen.
sperm and fluid from the vas deferens (~10%), seminal vesicle (~60%), prostate gland (~30%) and bulbourethral glands (small amount)
Describe some physical properties of semen.
pH = 7.5, milky appearance, coagulation (clotting enzyme) which dissolves in 15-30 min due to fibrinolysin derived from profibrinolysin, 1 month in vas deferens and epididymis but only 1-2 days in female tract, cryopreservation can keep sperm alive for years (sperm bank)
Describe capacitation.
1. making sperm capable of fertilizing the egg
2. occurs naturally in the female genital tract
3. takes 1-10 hours
What changes occur during capacitation?
1. washing off inhibitory factors that suppress sperm activity
2. removing cholesterol vesicles and exposing the acrosome so that acrosomal enzymes can be released
3. sperm membrane becomes more permeable and Ca2+ enter the sperm, triggering hyperactivated motility, which is essential for penetrating zona pellucida
What role does human tubular fluid (HTF) have on capacitation of sperm?
washing the semen with HTF or capacitating medium can capacitate sperm (first step in IUI)
What is the content of acrosome?
1. hyaluronidase-dissolves connective tissues among multiple layers of granulose cells
2. proteolytic enzymes-dissolve zona pellucida
Where is the acrosome derived from and what is its purpose?
derived from golgi apparatus during spermiogenesis, essential for sperm to fertilize eggs
What may cause abnormal spermatogenesis and male infertility?
temperature problem, requires a temperature ~2C or more below body temperature, scrotum has sweat glands, countercurrent heat exchange, reflex regulation of scrotal surface area, reflex regulation of distance from abdomen
What is cryptorchidism?
1. 1-3 months before birth
2. descent through inguinal canal into scrotum
3. testosterone from fetal Leydig cells triggers
4. if not usually descent within months after birth
5. testosterone treatment can induce descent
6. after one year need surgical help
What happens with patients who have abdominal testes?
spermatogenic block -> depletion of germ cells -> sterility, can lead to tumorigenesis
Describe low sperm count.
in 3.5 mL, average is 120 million/mL (25-200 million/mL), <20 million/mL, infertile, spermatogenic defects (genetic factors)
What are the different types of abnormal sperm motility?
structural (tail formation, deformation) and metabolic (energy supply, ion signaling)
Describe the neuronal stimulus for performance of the male sexual act.
1. sexual stimulations form the sex organs: glans penis or areas adjacent to penis
2. impulses -> pudendal nerve -> sacral plexus -> sacral portion of spinal cord
3. psychic stimulation from the brain-thinking, dreaming (nocturnal emissions)
4. impulses from brain and/or sex organs are integrated in the sacral and lumbar spinal cord
What are the stages of the male sexual act?
1. penile erection-role of parasympathetic nerves
2. lubrication-a parasympathetic function
3. emission and ejaculation-a sympathetic function
4. resolution-1-2 min after ejactulation
Describe penile erection.
1. parasympathetic impulses -> sacral portion of spinal cord -> pelvic nerves -> penis
2. parasympathetic nerve fibers release NO and/or vasoactive intestinal peptide as well as ACh
what does NO do?
relaxes penile arteries and trabecular meshwork of smooth muscle fibers in the erectile tissue of the corpora cavernosa and corpus spongiosum in the shaft of the penis (how Viagra works)
Describe the lubrication stage of the male sexual act.
parasympathetic impulses stimulate secretion of mucus from the urethral glands and the bulbourethral glands
describe the emission and ejaculation stages of the male sexual act.
1. culmination of the male sexual act
2. reflex center of spinal cord emits sympathetic impulses at T12-L2 -> hypogastric and pelvic sympathetic nerve plexuses -> genital organs -> emission
3. contraction
4. filling of internal urethra -> sensory signals
Describe the contraction that occurs during the emission and ejaculation stage of the male sexual act.
vas deferens -> expulsion of sperm into the internal urethra -> prostate gland -> seminal vesicle -> prostatic and seminal fluids into the urethra + sperm + mucus from the urethral glands and the bulbourethral glands
Describe the filling of the internal urthra during the emission and ejaculation stages.
1. rhythmical contraction of internal genital organs and pelvic muscle
2. thrusting movement of the pelvis and penis
3. propel semen
What is the nature and origin of testosterone?
steroids synthesized from cholesterol, comes from the Leydig cells = interstitial cells, < 5% of testosterone from the adrenals
What are the circulating binding proteins for testosterone?
SHBG/ABP (produced by Sertoli cells), albumin
Describe the conversions of testosterone.
conversion to dihydro-testosterone (DHT) by 5-alpha-reductase in liver (type I) and target tissues (type II), low abundance (5% of T in blood), high affinity for androgen receptors, T can also be aromatized -> E2
Describe the metabolism and excretion of testosterone.
reduction, then conjugation in the liver, excretion by the kidneys or by the gut
What is the function of testosterone during fetal development?
1. T production starts ~7th week
2. formation of male external and internal sex organs
3. descent of the testes-cyrptorchidism
What is the function of testosterone during the development of adult primary and secondary sexual characteristics?
1. further development of external and internal genitalia
2. development of secondary sexual characteristics:
a. distribution of body hair
b. baldness-genetic background + large amount of androgen
c. voice
d. INC skin thickness (acne)
e. INC muscle development (abuse by athletes)
f. INC bone matrix and Ca2+ retention
g. INC basal metabolism
What is the function of testosterone during the adult stages?
gonadal steroid actions are no longer concerned with establishing the individual as a male or female, rather they ensure that the reproductive system functions effectively
Describe the basic intracellular mechanism of action of testosterone.
ligand binds to androgen receptor which binds to a coregulator, this complex binds to a transcription initiation complex that leads to transcription of specific genes
Describe estrogen in men.
1. at early follicular phase levels
2. from Leydig cells, Sertoli cells and developing germ cells
3. 2/3 from peripheral conversion (skin, brain, fat and liver)
What are the roles of estrogen in men?
1. estrogen regulates the reabsorption of luminal fluid in the head of the epididymis, allowing sperm to enter the epididymis concentrated rather than diluted
2. full expression of gonadal negative feedback on pituitary gonadotropin secretion
3. pubertal growth spurt, epiphyseal closure, maintenance of bone mineralization and integrity
4. maintenance of insulin sensitivity
5. ER-beta on spermatogonia, spermatocytes and Sertoli cells may mediate the actions of xenoestrogens (estrogenic endocrine disruptors)
Describe GnRH.
1. 10 aa peptide
2. secreted by neurons whose cell bodies are located in the arcuate nuclei of the hypothalamus
3. GnRH is transported via blood to anterior pituitary gland
4. intermittently secreted a few minutes per 1-3 hours
5. freq. and amplitude
6. stimulates secretion of LH and FSH by the anterior pituitary
7. cyclic secretion of LH similar to that of GnRH
Describe gonadotropin hormones
LH and FSH, secreted by the same cells (gonadotropes) in the anterior pituitary, glycoproteins
What is the mechanism of action, action and target of LH?
1. mech of action-LHR -> cAMP -> PKA
2. actions-T production (direct), spermatogenesis (via T which is absolutely required (at a high concentration) -> Sertoli cells -> growth factors), LHR (down-regulated), Leydig cell growth and differentiation
3. target-Leydig cells
What is the mech of action, action and target of FSH?
1. mech of action-FSHR -> cAMP -> PKA
2. actions-spermatogenesis (needed for normal quantity and quality), growth, differentiation and maintenance of Sertoli cells, INC LHR on Leydig cells (indirect)
3. target-Sertoli cells
What is the role of the GnRH pulse generator and negative feedback control of LH secretion?
note the effects of testosterone deprivation on LH levels in blood and on the freq. of LH pulses, the site of T negative feedback is both at the level of the pituitary gland (DECing pulse amplitude) and at the hypothalamic GnRH pulse generator (DECing LH pulse freq.)
What is the role of inhibin in control of FSH secretion?
male monkeys were castrated and preoperative testosterone levels were maintained by exogenous T administration, note that while LH remained at preoperative levels, FSH levels gradually rose to high concentrations indicating that a testicular factor (inhibin) in addition to T normally controls FSH secretion, C is cryptorchidism
Describe inhibin.
1. Sertoli cells of the testes produce inhibin, whose role is to control the secretion of FSH
2. FSH secretion from cultured AP cells is inhibited when AP cells are co-cultured with Sertoli cells
3. this is further substantiated by the fact that prior exposure to inhibin suppresses the elevation of FSH release following GnRH administration
4. other evidence suggests that inhibin also suppresses FSH at the hypothalamus level
5. granulosa cells from the ovarian follicle secrete inhibin that acts directly on AP cell cultures derived from the male or female
6. administration of anti-inhibin antisera to rats of either sex causes an elevation in serum FSH, LH remains unchanged
What factors affect the hypothalamogonadal axis?
1. psychic factors
2. hCG secreted by the placenta during pregnancy stimulates T production by the fetal testes
3. onset of puberty-unknown! childhood no GnRH secretion due to stronger seuppressive effects of sex steroid hormones
Describe the male climacteric (andropause).
1. starting from 40s, T levels decline
2. sometimes associated with symptoms of hot flashes, suffocation, and psychic disorders, similar to those of menopause syndrome
describe the growth of the prostate gland and changes that may lead to abnormalities.
1. at puberty, T stimulates growth of prostate until 20 years of age
2. after 50, the prostate starts to involute in some men
3. coincides with the DEC of T levels
4. not caused by T, but by its own tissue
5. T stimulates the growth of cancerous cells
6. benign prostatic fibroadenoma (urinary obstruction)
7. cancer-2-3% of all male deaths
Describe the prevalence and distribution of testicular tumors.
1. testicular cancer is most common among white males and rare among African Americans, worldwide incidence has doubled since the 1960s with the highest rates of prevalence in Scandinavia, Germany and New Zealand, T cancer is uncommon in Asia and Africa
2. incidence in blacks has doubled from 1988 to 2001 with a bias towards seminoma, the lack of any significant INC in the incidence of early-stage testicular cancer during this timeframe suggests that the overall INC was not due to heightened awareness of the disease
3. although testicular cancer is most common among men aged 15-40 years, it has three peaks (infancy, ages 25-40, and 60)
Describe the rates of occurrence of germ cell tumors of the testis.
1. seminoma-35%
2. embryonic carcinoma-20%
3. teratoma-5%
4. choriocarcinoma-<1%
5. mixed cell type-40%
What are some examples of non-germ cell tumors of the testis?
Leydig cell tumors, Sertoli cell tumors
What are the three basic types of treatment?
surgery, radiation therapy and chemotherapy
What are the effects of orchidectomy in the adult on accessory sex organs?
prostate, epidydimes, seminal vesicles involute, their epithelia shrink, secretory activity declines -> no seminal plasma
What are the effects of orchidectomy in the adult on muscular mass?
muscular mass DEC (reversible, role of T, abuse)
What are the effects of orchidectomy in the adult on brain/behavior?
DEC (not lost) libido (unlike after ovariectomy), chemical castration as treatment of antisocial patterns of sexual behavior, but T is not an aphrodisiac, importance of monitoring of nocturnal penile tumescence (NPT)
What are the effects of orchidectomy in the adult on other organs?
prevention of male baldness, DEC in kidney and liver weight and erythropoiesis, INC in thymus weight
What is the nocturnal penile tumescence (NPT) test.
The NPT test may also be called the stamp test or the rigidity test.
Most men have 3 to 5 full erections during deep (rapid eye movement or REM) sleep. Men who are unable to have an erection because of a psychological problem still have erections during deep sleep. Occasionally some sleep disorders or serious depression can prevent these nighttime (nocturnal) erections.
The NPT test determines whether a man is having normal erections during sleep. This test can be done at home or in a special sleep lab. One of two methods may be used:
Use a simple ringlike device called a snap gauge made up of plastic films that fit around the penis. The films break at predetermined pressures.
Use an electronic monitoring device. This method is more expensive than using the snap gauge, but it is more accurate and provides more detailed information about the number, duration, and stiffness of erections that occur.
Testing is usually repeated for at least two nights. If good erections occur during sleep, the major cause of the erection problems probably is not physical.
What is adiposogenital syndrome?
AKA Frohlich’s syndrome or hypothalamic eunuchism, the hypothalamus has a deficiency in GnRH production, abnormalities in the feeding center of the hypothalamus leading to overeating and obesity
Describe the pineal gland.
1. the pineal gland was the last endocrine gland to have its function discovered
2. its location deep in the brain seemed to indicate its importance
3. it is located in the geometric center of the brain, this correlates to the location of the great pyramid in the center of the physical planet
4. this combination led ot its being a mystery gland with myth, superstition and even metaphysical theories surrounding its perceived function
What are the functions of the pineal gland?
1. produces melatonin
2. it is large in children and begins to shrink at puberty
3. may play a significant role in sexual maturation, circadian rhythm, sleep induction, seasonal affective disorder and depression
4. in animals it is known to play a major role in sexual development, hibernation and seasonal breeding
Describe the growth and functional development of the fetus.
during the first 2-3 weeks the fetus remains almost microscopic, it then begins to grow in length, pretty steadily throughout the pregnancy, weight of the fetus remains minuscule during the first 12 weeks of gestation then begins to grow, during the last 2 months, weight gain in tremendous averaging 2 lbs. for both the 8th and 9th months of gestation
Give an overview of the development of the organ systems.
1. within a month of fertilization, all of the organ systems of the fetus have been blocked out
2. during the next 2-3 months, more details of the systems begin to form
3. after the 4th month, the organ systems are grossly the same as in the newborn
4. cellular development of these systems requires the remainder of gestation to complete
5. at birth, structures within the nervous system, kidneys and liver lack full development
In general, describe the development of the circulatory system.
the human heart starts beating at 26 days following fertilization at ~65 beats/minute, the heart rate INC steadily throughout gestation and reaches 140 beats/minute just before delivery
Describe the formation of blood cells.
1. nucleated blood cells formed in yolk sac and mesothelial layer of the placenta ~3rd week of fetal development
2. after 4-6 weeks, non-nucleated RBCs begin to form in fetal mesenchyme, fetal blood vessel endothelium and liver
3. from 3rd month to birth, bone marrow forms red and white blood cells, as gestation progresses, bone marrow INC its production of blood components, until birth, when other blood-forming tissues lose their ability to form blood cells
What are the time periods for fetal erythropoesis?
yolk sac (3-8 week) -> liver (6-30 weeks) -> spleen (9-28 weeks) -> bone marrow (28 week onward) (young liver synthesizes blood)
Describe the development of the respiratory system.
1. respiratory movements take place during 1st trimester
2. during last 3-4 months of gestation, these respiratory movements are inhibited, possible causes for this inhibition include (1) special chemical conditions in the body fluids of the fetus, (2) presence of fluid in the fetal lungs
3. this inhibition of respiration prevents the filling of the lungs with debris from the meconium secreted from the GI tract, additionally fluids secreted by the alveolar epithelium, keeping the pulmonary sacs with clean secretions
What is the meconium?
the first intestinal discharges of the newborn infant, greenish in color and consisting of epithelial cells, mucus and bile, composed of unabsorbed residue of the amniotic fluid and excretory products of the GI mucosa and glands
Describe the development of the nervous system.
skin reflexes develop in the fetus between 3-4 months, most of the higher function of the cerebral cortex are underdeveloped at birth, even myelinization of the major tracts of the nervous system takes ~1 year to complete
Describe the development of the GI tract.
after ~4.5 months gestation, the fetus ingests and absorbs large quantities of amniotic fluid, during last 2-3 months, GI function approx. that of a newborn infant, meconium is excreted by the fetus
Describe the development of the kidneys.
fetal kidneys are capable of excreting urine during the last half of gestation and urination occurs in utero, control systems regulating extracellular fluid electrolyte and acid-base balance don’t exist to any degree until ~4.5 months gestation, and don’t develop until months after birth
Describe fetal metabolism of Ca2+ and phosphate.
fetus accumulates ~23 g of Ca2+ and 14 g of phosphorus during gestation, about half of these total accumulated in the last 4 weeks of gestation, coincident with the period of rapid ossification of fetal bones and rapid weight gain, these Ca2+ totals represent ~1/50th of the maternal bone contents, so that nursing, not growth of the fetus represents the greater drain on the mother’s system
Describe the accumulation of iron in the fetus.
accumulates somewhat faster than Ca2+ and phosphorus, most of the iron is incorporated into hemoglobin which begins to form ~3 weeks after fertilization, about 1/3 of the iron in a term fetus is stored in the liver for future formation of hemoglobin
What is the importance of B vitamins to the fetus?
B12 and folic acid are necessary for RBC formation and overall growth
What is the importance of vitamin C to the fetus?
necessary for bone matrix and connective tissue fiber formation
What is the importance of vitamin D to the fetus?
necessary fro proper fetal bone growth and stored in liver, must be present in maternal GI tract for Ca2+ absorption
What is the importance of vitamin E to the fetus?
maintains normal development in early embryo, since deficiencies in experimental animals produce spontaneous abortions
What is the importance of vitamin K to the fetus?
used by fetal liver for formation of factor VII, prothrombin and other blood coagulation factors, since vitamin K is formed by bacterial action in the colon, only the mother can provide the vitamin, limited storage in the fetal liver is helpful in preventing hemorrhage, especially if the head is traumatized during birth
Describe the cause of breathing at birth.
if the fetus has not been depressed by anesthetics, a child will exhibit normal respiratory rhythm within seconds of birth, this promptness is probably produced by (1) low O2-a slightly asphyxiated state incident to the birth process. (2) low temperature-cooled skin, if no breathing, further hypoxia and hypercapnia will usually start respiration
What is hypoxia at birth and what can cause it?
it is delayed and abnormal breathing at birth, can be caused by umbilical cord compression, premature placental separation, excessive contraction of the uterus or excessive anesthesia
What is the danger of hypoxia?
1. intracranial hemorrhage or brain contusion causes a concussion syndrome
2. serious depression of the respiratory center
What degree of hypoxia can an infant tolerate?
newborn infants can survive from 10-15 minutes without respiration (adult ~4 min), permanent and very evident brain impairment develops after 8-10 minutes of breathing delay, specific lesions develop in the thalamus, inferior colliculi and other brain stem area thus affecting many of the motor function of the body
Describe the expansion of the lungs at birth.
1. tremendous force required to open the alveoli the first time, at least 25 mm Hg negative pressure
2. fortunately the first inspirations of the infant can produce as much as 60 mm Hg negative pressure in the intrapleural space
3. the second breath is much easier although breathin is rarely normal until ~40 minutes after birth
What is respiratory distress syndrome (RDS)?
AKA hyaline membrane disease, premature infants of those born to diabetic mothers develop respiratory distress produced by large quantities of proteinaceous fluid resembling plasma in the alveoli, the fluid also containes desquamated alveolar epithelial cells
What causes hyaline membrane disease?
1. caused by a lack of surfactant secretion
2. surfactant-a substance normally secreted by type II alveolar epithelial cells into the alveoli that DEC the surface tension of the alveolar fluid therefore allowing the alveoli to open easily
3. infants that lack surfactant have a tendency of collapsed alveoli and development of pulmonary edema
When do type II alveolar cells begin secretion?
do not begin secretion of surfactant until the last 1-3 months of gestation
What are the risk factors for hyaline membrane disease?
1. male sex
2. premature birth
3. second born twin
4. perinatal asphyxia
5. maternal diabetes
6. (Lecithin/sphingomyelin), L/S ratio < 2 9used to determine fetal pulmonary maturity
What are the clinical features of hyaline membrane disease?
1. tachypnea (rapid breathing)
2. nasal flaring
3. grunting
4. cyanosis
5. ground glass appearance on CXR
what are the complications of hyaline membrane disease?
intrinsic-pneumothorax, pulmonary emphysema
O2 therapy-bronchopulmonary dysplasia, retinopathy
Describe the functionality of the lung and liver in fetal circulation.
are non- or partially functional, not much blood needed, the placenta does have large quantities of blood
Describe the flow of O2 rich blood.
umbilical vein -> ductus venosus -> IVC -> right atrium -> foramen ovale -> left atrium -> left ventricle -> vessels of head and forelimbs
Describe the flow of O2 deprived blood.
SVC -> right atrium -> tricuspid valve -> right ventricle -> pulmonary artery -> ductus arteriosus -> descending aorta -> umbilical arteries -> placenta
Describe blood circulation within the placenta.
1. fetal circulation in the villous tree
2. maternal circulation in intervillous spae
3. maternal blood pathways
villous tree is anchored on the basal plate, pattern of maternal blood flow through the intervillous space (the cotyledons are separated from each other by placental septa
What are the primary changes in pulmonary and systemic vascular resistance at birth?
1. loss of tremendous blood flow to the placenta coupled with the approximate doubling of the system vascular resistance, these changes INC aortic pressure and pressures within the left ventricle and left atrium
2. pulmonary vascular resistance greatly DEC as a result of the expansion of the lungs, right ventricular and arterial pressure are reduced
Describe the closure of the foramen ovale.
the low right atrial and high left arterial pressures that occur cause blood flow to attempt to flow backward through the foramen ovale from the left atrium into the right atrium, a small valve that lies over the foramen ovale on the left side of the atrial septum closes over the opening right after birth
Describe the closure of the ductus arteriosus.
INC systemic resistance elevates aortic pressure while the DEC pulmonary resistance reduces pulmonary arterial pressure, the consequence is that blood begins to flow backward through the ductus arteriosus, the ductus arteriosus begin to constrict with 1-8 days, producing a functional closure, during the next 1-4 months, fibrous tissue grows to completely occlude the lumen
Describe the closure of the ductus venosus.
During fetal life, the portal blood from the fetus' abdomen join the blood from the umbilical vein and pass through the ductus venosus directly into the vena cave, bypassing the liver. Obviously, at birth, the umbilical vein blood flow ceases, but the portal blood flow continues into the ductus venosus. Within 1 3 hours, the walls of the ductus venosus begin to constrict, closing off this avenue of blood flow. Portal venous blood flow rises from 0 10 mm Hg, enough to force blood through the liver sinuses.
What are some fetal structures that are now fetal remnants?
1. epigastric arteries-lateral umbilical ligaments
2. umbilical arteries-medial umbilical ligaments
3. urachus-median umbilical ligament
4. umbilical vein-round ligament
5. ductus venosus-venous ligament
6. ductus arteriosus-ligamentum arteriosus
7. yolk stalk-Meckel’s diverticulum
Describe the nutrition of the newborn infant.
1. maternal glucose stored in the infant as glycogen only lasts a few hours after birth
2. the newborn liver is marginally functional, insignificant gluconeogenesis occurs, neonatal blood glucose can fall to 30-40 mg/dL, about one half of normal
3. the infant can however utilize the stored fats and proteins for the 2-3 days until mothers milk can be provided
4. since infant body fluid turnover is seven times that of an adult, most infants experience a 5-10% weight loss after birth until mothers milk supply becomes sufficient
Describe a special functional problem in the neonatal respiratory system.
Normal respiration ~ 40 breaths/minute, with tidal air volume ~16 ml. This yields a total minute respiratory volume of 640 ml/min, about 2x that of an adult. This is compensated for with a functional residual capacity of one half that on an adult in relation to body weight.
Describe a special functional problem in the neonatal blood volume.
Normal blood volume ~300 ml right after birth. If the infant is left attached to the placenta, this can increase to 375 ml. A combination of this with the fluid loss right after birth increases the hematocrit and reduces the blood volume to normal. This extra blood volume in some instance causes mild pulmonary edema with some degree of respiratory distress.
Describe a special functional problem in the neonatal CO.
Averages about 2x that of an adult, 550 ml/min.
Describe a special functional problem in the arterial pressure.
Averages 70/50 at birth and increases over the next several months after birth to 90/60. Blood pressure then slowly increases to adolescent values of 115/70.
Describe a special functional problem in the neonatal blood characteristics.
RBC count 4 x106/ul, which can increase to 4.75 x106/ul if the umbilical cord blood is stripped into the infant. This falls to 3.24 x106/ul as the hypoxic stimulus for RBC formation disappears. White blood cell counts 45,000/ul at birth, about 5x that of adult values.
Describe a special functional problem in the neonatal jaundice.
Bilirubin is excreted through the placenta by the fetus, which disappears at birth. Since the neonatal liver is marginally functional, plasma bilirubin rises from < 1 mg/dl at birth to 5 mg/dl after 3 days of life. This condition called physiologic hyperbilirubinemia and associated with mild jaundice.
Describe a special functional problem in the neonatal erythroblastosis.
Erythroblastosis fetalis produced by Rh incompatibility between the mother and fetus. The maternal system recognizes the infants' RBCs as non self, and destroy them with antibodies, releasing large quantities of bilirubin into the plasma.
Is physiologic jaundice unconjugated or conjugated and what are some characteristics about it?
unconjugated, 3 to 5 days postnatal, due to INC bilirubin production and relatively immature liver
Is breast milk jaundice unconjugated or conjugated and what are some characteristics about it?
unconjugated, 2 to 3 weeks after birth, due to INC bilirubin absorption, brief interruption of breast feeding (usually not necessary)
Is hemolysis unconjugated or conjugated and what are some characteristics about it?
UC, Rh, ABO incompatibility, spherocytosis, G6PDH deficiency
Is enzyme defects unconjugated or conjugated and what are some characteristics about it?
UC and C, Gilbert (mild, DEC hepatic bilirubin uptake), Crigler-Naijar (severe, deficient glucuronyl transferase) Dubin Johnson (impaired hepatocellular secretion)
Is cholestasis unconjugated or conjugated and what are some characteristics about it?
C, bile duct stenosis, biliary atresia, hepatitis, CF
Is kernicterus unconjugated or conjugated and what are some characteristics about it?
UC, unconjugated bilirubin > 20 mg/dl, staining of basal ganglia, lethargy, hypotonia, encephalopathy
Describe fluid and acid-base balance, and renal function.
1. Some of the most important problems in infancy: dehydration, acidosis, and overhydration.
2. Fluid intake and excretion in the infant is 7x that of an adult. Slight alterations in fluid balance cause large abnormalities in infants.
3. Metabolism 2x that of an adult in infants, which increases 2x the acid formation, making the infant slightly acidotic.
4. Functional development of the kidneys is not complete until ~1 month after birth. Infants can only concentrate urine 1.5x, whereas adults do so ~ 3 4 times.
What is some evidence of deficient liver function?
1. Conjugate bilirubin with glucuronic acid poorly, which reduces bilirubin excretion.
2. Reduced plasma protein formation, so that plasma proteins drop to 15 20% of older children. Can drop low enough for infant to develop hypoproteinemic edema.
3. Gluconeogenesis almost non existent, producing hypoglycemia in newborns.
4. Forms blood coagulation factors poorly
What are some exceptions to newborn’s ability to digest, absorb and metabolize nutrients?
through to be similar to older children except for:
1. Reduced pancreatic amylase, reducing starch utilization.
2. Reduced absorption of fats by gastrointestinal tract. High fat milks are not utilized well.
3. Imperfection liver function renders blood glucoses low and unstable
4. Neonates are especially good at utilizing amino acids. As much as 90% of ingested amino acids are utilized in the formation of body proteins
Describe the metabolic rate and body temperature of neonate.
1. Metabolic rate of the neonate in relation to body weight is 2X that of the adult (2x cardiac output; 2X minute respiratory volume)
2. Body surface area>body mass→ body temperature DEC
What are some specific problems concerning nutrition during the early weeks of life?
1. Need for calcium and vitamin D: rapid ossification in the neonate→ Vitamin D DEC→ Calcium absorption DEC→ Severe rickets
2. Necessity for iron in the diet: if mother has adequate iron in her diet →enough for 4-6 months for blood cell formation. If not, anemia at ~ 3 month
3. Vitamin C deficiency in infants can result from low content in cow’s milks. Usually rectified with juice supplementation.
Describe immunity in the infant.
Immunity inherited from the mother protects the infant for ~6 months against major childhood infectious diseases including diphtheria, measles, and polio. Therefore, immunization against these diseases before 6 months is unnecessary.
Describe allergies in the infant.
While newborns are rarely subject to allergies, once the infant begins to form its own antibodies, allergies begin to appear.
What are some endocrine problems in the fetus?
1. Pregnant mothers treated with androgenic hormones or who develop androgenic tumors bear female children with masculinization of the sex organs, hermaphrodism.
2. Sex hormones from the placenta and mother often cause neonatal breast development and sometimes milk formation.
3. Infants born to diabetic mothers have considerable hypertrophy and hyperfunction of the islets of Langerhans, caused by the maternal hyperglycemia.
What happens in hyperthyroidism in the fetus/newborn?
1. Hypofunctional adrenal cortices.
2. Hyperthyroidism or thyroid hormone abuse produce a hyposecreting thyroid gland, temporarily. Also occurs in cases where mother has had thyroid gland removed and pituitary hypersecretes TSH.
3. A fetus lacking thyroid hormone secretion will exhibit poor bone growth and mental retardation. The condition is called CRETIN DWARFISM (congenital hypothyroidism) .
What are some special problems of prematurity?
1. Immature development of the premature infant
– Respiration: RDS
– GI: Low-fat and high calcium and vitamin D diet
– Other organs:
2. Instability of the control systems in the premature infant
– Body temperature: use of incubator
– Hypoproteinemic edema
3. Danger of blindness caused by oxygen therapy in the premature infant
– O2 INC→new blood vessels in retina DEC
– When stopped, rapid growth of blood vessels →great mass of vessels growing through the vitreous humor→ Blocking light from the pupil to the retina
What is APGAR scoring for newborns?
A score is given for each sign at one minute and five minutes after the birth. If there are problems with the baby an additional score is given at 10 minutes. A score of 7-10 is considered normal, while 4-7 might require some resuscitative measures, and a baby with apgars of 3 and below requires immediate resuscitation.
What are the five APGAR categories?
activity (muscle tone), pulse, grimace (reflex irritability), appearance (skin color), respiration
Describe the key roles that calcium plays in nerve and muscle exciation, muscle contraction, enzyme function and bone.
Calcium is a key actor in many physiologically important processes. It influences nerve excitability and is involved in neurotransmitter release from axon terminals and excitation-contraction coupling in muscle cells. It serves as a second or third messenger in several intracellular signal transduction pathways. Some enzymes use calcium as a cofactor, including some in the blood clotting cascade. Finally, calcium is a major constituent of bone.
What function of Ca2+ demands the most careful regulation of plasma calcium?
Of all these roles, the one that demands the most careful regulation of plasma calcium is the effect of calcium on nerve excitability. Calcium affects the sodium permeability of nerve membranes, which influences the ease with which action potentials are triggered. Low plasma calcium can lead to generation of spontaneous action potentials in nerve. When the motor neurons are affected, tetany of the muscles of the motor unit may occur (hypocalcemic tetany).
What does phosphate do?
Phosphate participates in pH buffering and is a major constituent of many macromolecules and of bone. Phosphorus (usually as phosphate) also participates in many important metabolic processes. Phosphate buffer and various metabolic intermediates, DNA, RNA and phosphoproteins all contain phosphate as an integral part of their structure. Phosphate is also a major component of bone.
Describe the distribution of calcium.
The table below shows the relative distributions of calcium and phosphate in a normal individual. Roughly 99% of the body calcium is in bones. Despite its critical role in E-C coupling, only about 0.3% of total body calcium is located in muscle. About 0.1% of total calcium is in extracellular fluid.
Describe the distribution of phosphorus.
Of the roughly 600 g of phosphorus in the body the majority (86%) is in bone. Compared to calcium, a much larger percentage of phosphorus is located in cells (14%). The amount of phosphorus in extracellular fluid is rather low (0.08% of body content).
Describe the ionic composition of bone.
Bone also contains a relatively high percentage of the total body content of several other inorganic substances (see Table). Roughly 80% of the total carbonate in the body is located in bone. This carbonate can be mobilized into the blood to combat acidosis, thus bone participates in pH buffering in the body. Long standing uncorrected acidosis can result in considerable loss of bone mineral. A significant percentage of the body’s magnesium and sodium and nearly 10% of its total water content is in bone.
In what forms and concentrations is Ca2+ found in the blood?
In humans, the normal plasma concentration is 9-10.5 mg/dl. Plasma calcium exists in three forms: ionized calcium (50% of the total), protein bound calcium (40%) and calcium bound to other anions (10%). The association of calcium with plasma proteins is pH-dependent. At an alkaline pH, more calcium is bound; the opposite is true in acidosis.
In what forms and concentrations is phosphorus found in the blood?
Plasma phosphorus concentration may fluctuate significantly during the course of a day, from 50% to 150% of what is “normal” for any particular individual. In adults, the normal range of plasma concentration is 3-4.5 mg/dl (expressed in terms of mg of phosphorus). Phosphorus circulates in the plasma primarily as orthophosphate (PO4). At normal blood pH of 7.4, 80% of the phosphate is in the HPO42- form and 20% is in the H2PO4- form. Nearly all of the plasma inorganic phosphate is ultrafilterable. In addition to free orthophosphate, phosphate is present in small amounts in the plasma in organic form, such as in hexose or lipid phosphates.
Describe the intake, absorption and excretion amounts of Ca2+.
The approximate tissue distribution and average daily flux of calcium between tissues in a normal adult are shown in the next figure. Dietary intakes may vary widely, but an average diet contains approximately 1000 mg/day of calcium. Intakes up to twice that amount are usually well tolerated, but excessive calcium intake can result in soft tissue calcification or kidney stones. Only about one third of ingested calcium is actually absorbed from the GI tract; the remainder is excreted in the feces. The efficiency of calcium uptake from the GI tract varies with the individuals physiological status. The percentage uptake of calcium may be increased in young growing children and pregnant or nursing women, while quite often it is reduced in the elderly.
How much Ca2+ actually enters the GI tract from the body?
Approximately 150 mg/day of calcium actually enters the GI tract from the body. This component of the calcium flux results in part from sloughing of mucosal cells that line the GI tract and also from calcium that accompanies various secretions into the GI tract. This component of calcium metabolism is relatively constant, so the primary determinant of net calcium uptake from the GI tract is calcium absorption. Intestinal absorption is an important site of regulation of calcium homeostasis.
How much Ca2+ is in bone?
Bone in an average individual contains approximately 1000 g of calcium. Bone mineral is constantly resorbed and deposited in a process called remodeling. As much as 500 mg/day of calcium may flow in and out of bone (see figure). Since bone serves as an important reservoir in determining plasma calcium concentration, both bone resorption and bone formation are important sites of regulation.
Describe the net uptake of Ca2+ in overall Ca2+ balance.
In overall calcium balance, the net uptake of calcium from the GI tract presents a daily load of calcium that will eventually require elimination. The primary route of elimination is via the urine, thus the kidneys play an important role in regulation of calcium homeostasis. The 150 mg/day of calcium excreted in the urine represents only about 1.5% of the calcium initially filtered by the kidneys; the remaining 98.5% is reabsorbed and returned to the blood. Therefore, small changes in the amount of calcium reabsorbed by the kidneys can have a dramatic impact on calcium homeostasis.
Describe phosphate handling by the GI tract, kidneys and bone.
The next figure shows the overall daily flux of phosphate in the body. A typical adult ingests approximately 1400 mg/day of phosphorous. In marked contrast to calcium, most (1300 mg/day) of this phosphorus is absorbed from the GI tract, typically as inorganic phosphate. There is an obligatory secretion of phosphate into the GI tract (approximately 200 mg/day), much like that for calcium, resulting in a net uptake of phosphorus of 1100mg/day and excretion of 300 mg/day via the feces. Note that less phosphorous exchanges with bone per day than calcium because bone mineral contains more calcium than phosphate. Because the majority of ingested phosphate is absorbed by the GI tract, phosphate homeostasis is greatly influenced by excretory mechanisms. Since the majority of circulating phosphate is readily filtered in the kidneys, phosphate reabsorption in the kidney is a primary site of regulation of phosphate homeostasis.
Describe Ca2+ absorption.
Calcium absorption in the small intestine occurs by both active transport and diffusion. The relative contribution of each process varies with the region and total calcium intake. Uptake of calcium by active transport predominates in the duodenum and jejunum, in the ileum, simple diffusion predominates. Transport across cells involves calcium channels (TRPV6) at the apical membrane, calcium binding proteins within the cell and two transporters (i.e., CaATPase and 3Na/Ca) at the basolateral membrane. The relative importance of active transport in the duodenum and jejunum versus passive diffusion in the ileum depends on a number of factors. At very high levels of calcium intake, active transport processes are saturated and most of the uptake occurs in the ileum, in part because of its greater length as compared to other intestinal segments. With moderate or low calcium intake, however, active transport predominates because the gradient for diffusion is low.
What is the regulated variable in control of Ca2+ uptake?
Active transport is the regulated variable in control of calcium uptake from the small intestine. Metabolites of vitamin D provide a regulatory signal to increase intestinal calcium absorption.
Describe phosphate absorption.
The small intestine is also a primary site for phosphate absorption. Uptake occurs by both active transport and passive diffusion, but secondary active transport is the primary mechanism. This involves movement of phosphate into epithelial cells via a Na/Pi symporter along with transport via another carrier at the basolateral membrane. Phosphate is efficiently absorbed from the intestine but is not highly regulated. To a minor extent, active transport of phosphate is coupled to calcium transport. Therefore, when active transport of Ca2+ is low, as with vitamin D deficiency, phosphate absorption is also low.
What is the composition of Ca2+ in the body that is filterable?
Filterable calcium comprises about 60% of the total calcium in the plasma and consists of calcium ion and calcium bound to a filterable anion such as bicarbonate or citrate. The remaining 40% of the total calcium circulates bound to proteins and thus is not filterable. The majority of calcium is reabsorbed in the proximal tubule. Ordinarily, only about 1.5% of filtered calcium is eventually excreted in the urine, with the remaining 98.5% reabsorbed and returned to the plasma.
Describe where in the kidneys Ca2+ is absorbed.
Approximately 70% of filtered calcium is reabsorbed in the proximal tubule, 20% from the loop of Henle and 9% from the distal tubule, the remaining 1% is excreted in the urine. Calcium excretion is regulated by the kidney primarily at the distal tubule. Calcium reabsorption by the proximal tubule occurs by two pathways: transcellular and paracellular (see figure). Transcellular calcium reabsorption accounts for 20% of proximal reabsorption.
Describe the mechanism of Ca2+ reabsorption.
Calcium reabsorption through the cell is an active, two step process. Calcium enters the apical membrane into the cell down its electrochemical gradient via a calcium channel (TRPV5). This gradient is exceptionally steep because the ionized calcium concentration in the cell is around 0.1 M while that of tubular fluid is about 6 mg/dl (1.5 mM). The cell interior is electrically negative with respect to the luminal side of the apical membrane and this also favors calcium entry into the cell. Calcium is extruded across the basolateral membrane against its electrochemical gradient via Ca ATPase and a 3Na/1 Ca antiporter. 80% of calcium is reabsorbed between cells across the tight junctions (i.e., paracellular pathway.)
Describe Ca2+ reabsorption by Henle’s loop.
Calcium reabsorption by Henle’s loop is restricted to the thick ascending limb and is similar to the proximal tubule except that less goes via a transcellular route (recall this region has lower water permeability). Calcium reabsorption by the distal tubule is exclusively transcellular, and the mechanism is similar to that in the proximal tubule.
Describe the interaction between Ca2+ and PTH.
Calcium and PTH: Parathyroid hormone (PTH) exerts powerful control on renal calcium excretion and is a key factor in maintaining calcium homeostasis. PTH dramatically stimulates calcium reabsorption by the thick ascending limb of Henle and the distal tubule thus promoting calcium retention and lowering urinary calcium.
What can disturb Ca2+ excretion?
Several factors can disturb calcium excretion. For example, an increase in plasma phosphate elevates PTH levels and thereby decreases calcium excretion. In contrast, a decline in plasma phosphate has the opposite effect.
Describe renal handling of phosphate.
The majority of ingested phosphate is absorbed from the GI tract and the primary route of excretion of this phophate is via the urine. Kidneys therefore play a key role in regulating body phosphate homeostasis. Ordinarily about 90% of filtered phosphate is reabsorbed and 10% is excreted in the urine. Phosphate reabsorption occurs via secondary active transport process at two sites along the nephron; the proximal tubule and the distal tubule. The majority (80%) of phosphate reabsorption occurs in the proximal tubule and 10% is reabsorbed in the distal tubule. Parathyroid hormone inhibits phosphate reabsorption in the proximal tubule by increasing cAMP levels and thus has a major regulatory effect on phosphate homeostasis. As a result of its actions, parathyroid hormone increases urinary phosphate excretion (phosphaturia) with an accompanying decrease in plasma phosphate concentration.
Describe the mechanism of phosphate reabsorption.
Phosphate reabsorption by the proximal tubule occurs mainly, if not exclusively, by a transcellular route. Phosphate uptake cross the apical membrane occurs by a 2Na/Pi symport mechanism. Phosphate exits across the basolateral membrane, most likely by a Pi-anion antiporter. The cellular mechanism of phosphate reabsorption by the distal tubule has not been characterized. The major reason why phosphate is excreted is that the levels in the tubular lumen exceed the transport maximum for reabsorption.
Describe the inorganic composition of bone.
Mature bone can be simply described as inorganic mineral deposited on an organic framework. The mineral portion of bone is composed largely of calcium phosphate in the form of hydroxyapatite crystals (Ca10(PO4)6(OH)2). The mineral portion of bone typically comprises about 25% of its volume and roughly half the weight of bone. Bone also contains considerable amounts of carbonate, magnesium, and sodium in addition to calcium and phosphate.
Describe the organic composition of bone.
The organic matrix of bone on which the bone mineral is deposited is called osteoid. Type I collagen is the primary constituent of osteoid.. Electron microscopic study of bone reveals needle like hydroxyapatite crystals lying alongside collagen fibers.
What is the importance of the interaction of the organic and inorganic composition of bone?
This orderly association of hydroxyapatite crystals with the collagen fibers is responsible for the strength and hardness characteristic of bone. Loss of either bone mineral or organic matrix greatly affects the 85 mechanical properties of bone. Complete demineralization of bone leaves a flexible collagen framework, and complete removal of organic matrix leaves a bone with its original shape, but extremely brittle.
What are the two basic forms of bone?
Cortical (compact) bone is the outer layer of all bones and is generally about 80% of the total bone mass. It is composed of bone mineral and osteoid interrupted by blood vessels and a sparse population of osteocytes nested within the bone. These osteocytes are interconnected with one another and with the osteoblasts on the surface of the bone by canaliculi, through which the osteocytes extend cellular processes. These connections permit the transfer of calcium from the interior of the bone to the surface. Medullary (spongy) bone is composed of thin spicules of bone that extend from the cortex into the medullary cavity. The lacework of bone spicules is lined in many areas by osteoblasts and osteoclasts, the cells involved in bone remodeling. Trabecular bone is constantly being synthesized and resorbed by these cellular elements. Similar bone turnover occurs in cortical bone, but the rate is much lower.
What are the three principal cells types involved in bone formation and resorption?
The three principal cell types involved in bone formation and resorption are osteoblasts, osteocytes and osteoclasts (see figure).
Describe osteoblasts.
Osteoblasts are located on the bone surface and are responsible for osteoid synthesis. Numerous cytoplasmic processes connect adjacent osteoblasts on the bone surface and connect osteoblasts with osteocytes deeper in the bone. Osteoid produced by osteoblasts is secreted into the space adjacent to the bone. Eventually, new osteoid becomes mineralized, and in the process osteoblasts are surrounded by mineralized bone.
Describe osteocytes.
As osteoblasts are progressively engulfed by mineralized bone, they lose much of their bone forming ability and become quiescent. At this point they are called osteocytes. Many of the cytoplasmic connections in the osteoblast stage are maintained into the osteocyte stage. These connections become visible channels, or canaliculi, that provide direct contact for osteocytes deep in bone with other osteocytes and with the bone surface. It is generally believed that these canaliculi provide a mechanism for transfer of nutrients, hormones and waste products between the bone surface and its interior. Osteoblasts arise from primitive cells called osteoprogenitor cells, within the connective tissue of the mesenchyme. A variety of proteins in the bone, known as skeletal growth factors, attract osteoprogenitor cells, direct their differentiation into osteoblasts, and stimulate their growth.
Describe osteoclasts.
Osteoclasts are cells responsible for bone resorption. They are large, multinucleated cells located on bone surfaces. Osteoclasts promote bone resorption by secreting acid and proteolytic enzymes into the space adjacent to the bone surface. Surfaces of osteoclasts facing bone are ruffled to increase their surface area and promote bone resorption. Bone resorption is a two step event. First, osteoclasts create a local acidic environment that increases solubility of surface bone mineral. Second, proteolytic enzymes secreted by osteoclasts degrade organic matrix of the bone.
What role does Ca2+ and phosphate flux have in bone formation and remodeling?
The flux of calcium and phosphate into and out of bone each day reflects a turnover of bone mineral and changes in bone structure generally referred to as remodeling. Bone remodeling occurs along most of the outer surface of the bone, making it either thinner or thicker, as required. In long bones, remodeling can also occur along the inner surface of the bone shaft, next to the marrow cavity. Remodeling is a beneficial adaptive process that allows bone to be reshaped to meet changing mechanical demands placed on the skeleton and to store or mobilize calcium rapidly.
Describe the differences between formation and resorption during different time periods in life.
During the growth years formation exceeds resorption, and skeletal mass increases. A peak in bone mass is reached between ages 20 and 30 years. Thereafter equal rates of formation and resorption stabilize the bone mass until age 35 to 40 years, at which time resorption begins to exceed formation, and the total mass slowly decreases.
Describe the mechanism of bone remodeling.
Bone remodeling begins with endocrine signals to resting osteoblasts (e.g., PTH and calcitriol). These cells then generate local paracrine signals to nearby osteoclasts and osteoclast precursors. The osteoclasts resorb an area of mineralized bone, and local macrophages complete the cleanup of dissolved elements. The process then reverses to formation as osteoblast precursors are recruited to the site and differentiate into active osteoblasts. These lay down new organic matrix and mineralize it. Thus new bone replaces the previously resorbed mature bone. Various paracrine signals released from osteoblasts include macrophage colony stimulating factor (M-CSF), a TNF cytokine family member (RANK ligand) which stimulates osteoclast formation and bone resoprtion, and osteoprotegerin which is a “decoy receptor” which prevents RANK ligand binding to osteoclasts and their precursors. Recent ongoing studies suggest that the balance between the amounts of RANK ligand and osteoprotegerin produced by osteoblasts are important factors in the pathology of both osteoporosis and osteopetrosis.
Describe osteoporosis.
is a state of reduced bone mass per unit volume with a normal ratio of mineral to matrix. It is a disease characterized by bone fragility and an increased susceptibility to fractures of the hip, spine, and wrist. Drugs given to treat osteoporosis include bisphosphonates and calcitonin which inhibit osteoclast function. Estrogen also decreases the formation and activity of osteoclasts probably through changes in signals generated by osteoblasts. In postmenopausal women osteoporosis is sometimes treated with estrogen receptor modulators.
Describe osteopetrosis.
is a rare congenital disorder (present at birth) in which the bones become overly dense.
Describe osteomalacia.
is a skeletal disorder associated with demineralization. In children this disease is called rickets and it is usually due to lack of vitamin D. The etiology of rickets includes hypocalcemia and skeletal abnormalities.
Describe the regulation of plasma Ca2+ and phosphate concentrations.
Regulatory mechanisms for calcium include rapid non-hormonal mechanisms with limited capacity and somewhat slower hormonally regulated mechanisms with much greater capacity.
Describe protein bound Ca2+.
The association of calcium with proteins is a simple reversible, chemical equilibrium process. Protein-bound calcium therefore has the capacity to serve as a buffer of free plasma calcium concentrations. This effect is rapid and does not require complex signaling pathways but its capacity is quite limited and it cannot serve a long term role in calcium homeostasis.
Describe the readily exchangeable pool of Ca2+ in bone.
Recall that approximately 99% of total body calcium is present in bone, and there is roughly 1-2 kg of calcium in a normal adult. The majority of the calcium in bone exists as mature hardened bone mineral that is not readily exchangeable but can be moved into the plasma via hormonal mechanisms. However, approximately 1% (or 10 g) of the calcium in bone is in a simple chemical equilibrium with plasma calcium. This readily exchangeable source of calcium is primarily located on the surface of newly formed bone. Any change in free calcium in the plasma or extracellular fluid therefore, results in a shift of calcium either into or out of bone mineral until a new equilibrium is reached. Although this mechanism, like that described above, provides for a rapid defense against changes in free calcium concentrations, it too, is limited in its capacity and thus can only provide for very short term fine tuning of calcium homeostasis.
Describe the hormonal mechanisms provide high-capacity, long term regulation of plasma Ca2+ and phosphate.
Hormonal mechanisms have a large capacity and the ability to make long term adjustments in calcium and phosphate fluxes, but they do not respond instantaneously. It may take several minutes or hours for the response to occur and adjustments to be made. However, these are the principal mechanisms by which plasma calcium and phosphate concentrations are regulated.
Describe PTH chemistry and its role in regulating Ca2+.
One of the primary regulators of plasma calcium concentrations is parathyroid hormone (PTH). PTH is an 84-amino acid peptide produced by the parathyroid glands. Synthetic peptides containing the first 34 amino terminal residues appear to be as active as the native hormone. There are two pairs of parathyroid glands, located on the dorsal surface of the left and right lobes of the thyroid gland. Because of this close proximity, damage to the parathyroid glands themselves or to their blood supply may occur during surgical removal of the thyroid gland.
What is the stimulus for PTH secretion?
The primary physiologic stimulus for PTH secretion is a decrease in plasma calcium. The relationship between PTH secretion and total plasma calcium is shown in the graph. It is actually a decrease in the ionized calcium concentration that triggers an increase in PTH secretion. Note in this figure that as calcium concentration decreases from a normal value of 10mg/dl to approximately 8.5-9 mg/dl, there is a significant increase in PTH secretion. The net effect of PTH is to increase the flow of calcium into plasma, and thus return concentrations toward normal.
Describe calcitonin chemistry and its role in regulating Ca2+.
(CT) is a 32 amino acid peptide produced by parafollicular cells of the thyroid gland. In contrast to PTH, CT secretion is stimulated by an increase in plasma calcium. Hormones of the GI tract, especially gastrin, also promote CT secretion. In bone, CT opposes the action of PTH on osteoclasts by inhibiting their activity. This leads to decreased bone resorption and an overall net transfer of calcium from plasma into bone. CT plays only a minor role in the ongoing regulation of calcium homeostasis. However, nasal application of CT has proved beneficial to individuals with osteoporosis. The CT which is used clinically is actually salmon CT since this peptide has a potency about 100 fold higher than human CT.
Describe calcitriol chemistry and its role in regulating Ca2+.
The third key hormone involved in regulating plasma calcium is vitamin D3 (cholecalciferol). More precisely, a metabolite of vitamin D3 (i.e., 1,25-dihydroxycholecalciferol or calcitriol) serves as a hormone in calcium homeostasis. Vitamin D3 can be provided by the diet or formed in the skin by the action of ultraviolet light on 7-dehydrocholesterol. In many countries where food is not supplemented with vitamin D, this pathway provides the major source of vitamin D. Typically exposure to moderately bright sunlight for 30-120 min/day provides enough vitamin D to supply the body’s needs without any dietary supplementation.
Describe vitamin D3.
Vitamins D3 is relatively inactive. However, it undergoes a series of transformation in the liver and kidneys that converts it into a powerful calcium-regulatory hormone. The first step occurs in the liver and involves addition of a hydroxyl group to carbon 25, to form 25-hydroxycholecalciferol. This reaction is largely unregulated, although certain drugs and liver diseases may affect this step. 25-hydroxycholecalciferol is released into the blood, and in the kidney it undergoes a second hydroxylation reaction. The product is 1,25-dihydroxycholecalciferol (calcitriol), the principal hormonally active form of the vitamin. The biologic activity of calcitriol is approximately 1000 times greater than that of 25-hydroxycholecalciferol. The formation of calcitriol in the kidneys is catalyzed by the enzyme 1-hydroxylase, located in proximal tubule cells.
Describe the mechanism of action of calcitriol.
The net effect of calcitriol (1,25-dihydroxycholecalciferol) is to increase both calcium and phosphate concentrations in plasma. Calcitriol primarily influences the GI tract, although it has actions in bone and kidney as well. In the kidneys, calcitriol increases tubular reabsorption of calcium and phosphate, promoting retention of both ions in the body. However, this is a weak and probably only minor effect of the hormone. In bone, calcitriol enhances the actions of PTH on osteoclasts, thereby increasing bone resorption. In the GI tract, calcitriol stimulates calcium and phosphate absorption by the small intestine, increasing plasma concentrations of both. This effect is mediated by increased production of calcium transport proteins in the intestinal epithelia (see earlier diagram).
Describe the control of the activity of 1-alpha-hydroxylase.
The activity of 1-hydroxylase is tightly controlled. First, it is enhanced by PTH. Thus, if plasma calcium levels fall, PTH secretion increases and this promotes formation of calcitriol. In contrast, 1-hydroxylase activity is inhibited by an increase in plasma phosphate concentration. This does not appear to involve any intermediate hormonal signals but apparently involves direct effects upon either the enzyme or cells in which the enzyme is located. Finally, the product calcitriol can feed back to inhibit further synthesis.
Describe the mechanism of action of PTH.
is essential for life. Complete absence of PTH causes death from hypocalcemic tetany within just a few days, but this can be avoided with hormone replacement therapy. The net effects of PTH on plasma calcium and phosphate and its sites of action are shown in the next figure. PTH causes an increase in plasma calcium concentration while decreasing plasma phosphate. This decrease in phosphate concentration is important with regard to calcium homeostasis. At normal plasma concentrations calcium and phosphate are at or near chemical saturation levels. If PTH were to increase both calcium and phosphate levels, they would simply crystalize in bone or soft tissues as calcium phosphate, and the necessary increase in plasma calcium concentration would not occur. Thus, the effect of PTH to lower plasma phosphate is an important aspect of its role in regulating plasma calcium.
What actions does PTH have in the kidneys?
PTH has several important actions in the kidneys. It stimulates calcium reabsorption in the thick ascending limb and distal tubule, thereby decreasing calcium loss in the urine and increasing plasma concentrations. It also inhibits phosphate reabsorption in the proximal tubule, leading to increased urinary phosphate excretion and a decrease in plasma phosphate. In bone, PTH has 2 actions, i.e., a fast phase called osteolysis which involves stimulation of osteoclasts to transport calcium from the interior of the bone to the blood stream and a slower phase leading to activation of osteoblasts. Both of these actions increase bone resorption and thus the delivery of calcium from bone into plasma. In addition to stimulating existing osteoclasts, PTH stimulates maturation of preosteoclasts into active osteoclasts. The actions of PTH to promote bone resorption are augmented by calcitriol. PTH does not appear to have any major direct effects on the GI tract. However, because it increases calcitriol formation, it ultimately increases absorption of both calcium and phosphate from the GI tract.
What role does PTH have in osteoporosis?
Interestingly , although PTH enhances bone resorption, in fact administration of PTH has sometimes proved beneficial in patients with osteoporosis since it also stimulates bone remodeling which involves both bone resorption and bone formation. In this way new bone replaces old and the tendency to fracture is reduced.
What stimulates bone formation?
PTH, calcitrol, insulin, androgens, GH, growth factors
What inhibits bone formation?
corticosteroids
What stimulates bone resorption?
PTH, calcitriol, thyroid hormone, corticosteroids, prostaglandins, growth factors
What inhibits bone resorption?
calcitonin, estrogens
Describe primary hyperparathyroidism.
Is excessive PTH secreted by a parathyroid adenoma that is no longer under the negative feedback control of calcium. The excess of PTH leads to alterations in functions of bone cells and renal tubules. Thus, it may result in kidney stones and calcium deposition in kidney tubules; osteoporosis and osteomalacia, and hypercalcemia leading to muscular weakness. It is more prevalent in women than men because pregnancy stimulates hypertrophy of the parathyroid glands due to the increased need for calcium mobilization.
Describe hypoparathyroidism.
These are condition of reduced PTH and defective PTH action, respectively. Hypoparathyroidism can be idiopathic , but most commonly it is of the postsurgical type in which the parathyroids were removed surgically along with a diseased thyroid gland. Effective treatment is calcitriol administration to raise blood calcium levels by promoting intestinal calcium absorption.
Describe pseudohypoparathyroidism.
These are condition of reduced PTH and defective PTH action, respectively. Pseudohypoparathyroidism is a hereditary disorder and it is characterized by failure to respond to PTH. This results in individuals with short stature, short metacarpals and ectopic calcification.
What disorders are classified as hypocalcemia?
hypoparathyroidism, pseudohypoparathyroidism, osteomalacia/rickets, chronic renal failure
What disorders are classified as hypercalcemia?
vitamine D intoxication, primary hyperparathyroidism
What is the differential diagnosis of hypoparathyroidism?
DEC Ca2+, INC phosphate, DEC PTH, no change in alk. phosphate
What is the differential diagnosis of pseudohypoparathyroidism?
DEC Ca2+, INC phosphate, INC PTH, no change in alk. phosphate
What is the differential diagnosis of osteomalacia/rickets?
DEC Ca2+, DEC phosphate, INC PTH, INC alk. phosphate
What is the differential diagnosis of chronic renal failure?
DEC Ca2+, INC phosphate, INC PTH, INC alk. phosphate
What is the differential diagnosis of vitamin D intoxication?
INC Ca2+, INC phosphate, DEC PTH, INC INC INC 24 hr. urine Ca2+
What is the differential diagnosis of primary hyperparathyroidism?
INC Ca2+, DEC phosphate, INC PTH, INC 24 hr. urine Ca2+
Give some general info for the hypocalcemia disorders.
-In the kidneys, PTH stimulates calcium and inhibits phosphate resorption. PTH also enhances calcitriol formation by stimulating 1-hydroxylase. Hypoparathyroidism therefore leads to hypocalcemia and hyperphosphatemia. Hyperphophatemia per se can also lead to reduced plasma calcium levels. In part this is due to increased “ectopic calcification” as calcium phosphate precipitates out of solution. Recall as well that elevated phosphate levels inhibit 1-hydroxylase activity leading to reduced cacitriol formation in the kidneys. Pseudohypoparathyroidism leads to the same pathology as hypoparathyroidism but in this case it is due to lack of response to PTH.
-Osteomalacia is typically due to lack of calcitriol. This reduces calcium uptake by the intestine. PTH increases due to the resulting hypocalcemia. High PTH inhibits phosphate reabsorption by the kidneys.
-Patients with chronic renal failure have increased serum phosphate levels because of declining renal excretory function. Plasma calcium levels decrease, partly because of the increased phosphate serum concentration, and partly because of decreased synthesis of calcitriol by the kidneys (kidney function is declining). PTH levels increase because of the hypocalcemia leading to bone decalcification (osteomalacia).
Give some general info for the hypercalcemia disorders.
-Vitamin D intoxication increases calcium (and to some extent phosphate) absorption by the intestine. The hypercalcemia reduces PTH formation. Since PTH inhibits phosphate reabsorption by the kidneys lowering PTH will increase plasma phosphate concentration. The filtered load of calcium in the kidneys is increased due to the hypercalcemia and reabsorption is reduced due to reduced PTH. These effects greatly increase urine calcium levels.
-The cause of hypercalcemia in the vast majority of cases is hyperparathyroidism. Most commonly, hyperparathyroidism is due to a tumor in the parathyroid gland which enhances the synthesis of PTH. High PTH levels increase calcium absorption from the intestine (via calcitriol), from the kidney and from bone leading to hypercalcemia. Urine calcium is high due to the hypercalcemia even though reabsorption is enhanced. Urine calcium goes up more with vitamin D intoxication than with hyperparathyroidism because PTH is reduced in the former. The high PTH levels of hyperparathyroidism reduce phosphate reabsorption by the kidneys leading to hypophosphatemia. With time osteoporosis and osteomalacia develop because of enhanced bone resorption. “Brown tumors” can develop which consist of collections of osteocytes intermixed with poorly mineralized bone.
What structures are available for the internal temperature control of the body?
1. CNS “thermostat”-hypothalamus
2. cold receptors
3. heat receptors
4. heat loss/conversation mechanisms
5. thermal neutral zone (TNZ)
What regulatory events in the anterior hypothalamus help regulate temperature?
1. heat dissipation events
2. over-warmed blood and peripheral sensation impulses
What regulatory events in the posterior hypothalamus help regulate temperature
1. heat conservation events
2. over-cooled blood and peripheral sensation impulses
Describe the relationship between environmental temperature zones and heat production (HP).
in the thermoneutral zone (from cool -> optimum -> wam) there is a minimal amount of HP and is called the basal metabolic rate (to have muscle tone, CV tone), as the environmental temperature INC pass UCT (heat stress, ~91.45) or DEC pass LCT (cold stress, ~95.5), HP INC linearly, if temperature rises or drops enough can lead to death, death from heat can result more quicker than that for cold stress
What heat loss mechanisms (that occurs during hypothermia) are available to maintain homeothermy?
1. non-evaporative cooling-radiation, convection and conduction
2. evaporating cooling-respiration, skin
What influences heat loss mechanisms that maintain homeothermy?
1. body surface area (surface:mass ratio, for a child it is large and will go into hypothermia quickly to protect the brain, low for adults, takes long time for them to go into hypothermia)
2. body coverings (clothes, animals w/ fur)
3. water exchange
4. blood flow
5. environment (temperature (cold INC heat loss), wind (wind chill factor, changes microclimate around the body and changes heat, humidity (more difficulty losing heat))
What heat gain mechanisms (that occurs during hyperthermia) are available to maintain homeothermy?
the sources of heat gain include
1. food (fatty foods-high metabolic production of heat)
2. body reserves (fat for heat conservation)
3. rumen or cecum fermentation
4. environment (opportunity to stand in shade or sun when hot or cold)
What influences heat gain mechanisms that maintain homeothermy?
calorigenic hormones, production (milk, meat, wool), muscular activity, maintenance
Describe the partitioning of avenues of heat loss from man at 75F.
1. radiation-67%
2. evaporation-23%
3. conduction to air (convection)-10%, if a calm environment have less loss via conduction
What factors INC heat production (over basal metabolic rate)?
1. exercise or shivering
2. imperceptible tensing of muscles
3. chemical INC of metabolic rate
4. specific dynamic action of food
5. disease (fever)
What factors DEC heat loss?
1. shif in blood distribution
2. DEC in tissue conductance
3. counter-current heat exchange
What factors enhance heat loss?
1. sweating
2. panting
3. cooler environment
4. INC skin circulation (vasodilation)
5. DEC clothing or shorter fur insulation
6. INC insensible water loss
7. INC radiating surface
8. INC air movement (convection)
Describe counter-current heat exchange.
the key is that major arteries and veins flow next to each other, in order to conserve heat blood is shunted in a maximum amount from the periphery to the central part, in cold, heat can be directed by diffusion from artery to vein so vein can take it back to the center, in heat, heat can be directed by diffusion from vein to artery, takes heat from core to periphery, current is artery to periphery, counter-current is vein to core
What is the highest fever that a human can have and still survive?
107-108 then start having CNS seizures, can go there under controlled medical conditions
What are the signs and symptoms of hyperthermia?
dizziness, abdominal distress (sometimes with vomiting), sometimes delirium, sometime seizures, loss of consciousness, INC core body temperature
What are some hyperthermia temperatures and what activities are associated with them?
102°F-105°F = febrile disease & hard exercise
104°F- 107°F = therapy
106°F-111°F = heat stroke and brain lesions
>111°F-113°F = death likely
>113°F = death almost a certainty
Describe fever.
Elevated core body temperature caused primarily by leukocyte endogenous pyrogens or bacterial endotoxins. pyrogenic factors released when leukocytes are lysed or killing endotoxins
Describe environmental hyperthermia.
Elevated core body temperature caused by an inability to loose excess body heat in high ambient environments
Describe malignant hyperthermia.
A rapidly progressive reaction to certain muscle relaxant and gaseous anesthetic agents in certain people (inherited autosomal dominant trait), seen in about 1:20,000 surgical cases, may be rapidly fatal. Can be reverse with a certain druge that must be quickly administered, caused by certain anesthetics or muscles relaxants like succinyl-choline
How is core temperature measured?
esophageal temperature can get to level of the heart, gives the core temperature
Describe the relationship between age and temperature.
there is no statistical difference in skin temperature between young and old, in core temperature there is a difference between old and young (old has a higher core temp), due mostly to counter-current heat exchange (old people have a lower ability to transfer heat via this mechanism are not shifting same amount of blood, may be due to compliance of vascular system), there is a significant difference between elderly and young in forearm vascular conductance
What happens with metabolism with INC in age?
high metabolism of childhood DEC with age
Describe the effect of humidity on temperature.
lower humidity makes the temperature feel colder because evaporating more, the degrees given on the chart are not actual temperatures but equivalent temperatures of what the person feels like they are being exposed to
What are the signs and symptoms of hypothermia?
DEC alertness and consciousness, shivering may or may not be present depending upon the severity of the hypothermia, reduced core body temperature, severe hypothermia may mimic death, therefore no hypothermic patient should be pronounced dead until they are warm and dead
What are some hypothermia temperatures and what activities are associated with them?
 98.6°F-95°F = maximum shivering occurs
 »94°F = memory impairment
 »92°F = brain recall <30%
 » 92°F = paradoxical undressing (abrupt vasodilation)
 » 90°F = muscle rigidity
 80°F-85°F = loss of consciousness
 <80°F = cardiac arrest and death
What happens during frostbite?
tissues freeze, the body will do whatever is necessary to conserve the core, will sacrifice limbs, noses to keep the core at a good temperature, when tissues freeze the first thing that happens is that ice crystals are formed in the cells that is bigger in volume than the water in the cell, if have frozen tissue want to thaw it fast (if thaw it slow then ice crystals get bigger and burst leading to even more cell damage)
What affect does wind chill have on temperature?
as wind chill DEC (DEC in temperature of wind chill) the equivalent temperature felt DEC
What effect does depth of submersion and duration of submersion have on decompression?
as depth of submersion INC and duration of submersion INC, the INC chance that decompression is required, divers must choose between going deep for 25 mins or remaining near the surface for a much longer period, at 100 ft. have about 30 mins before having to decompress
Describe what happens during compression (which occurs under hyperbaric conditions).
compression causes N to dissolve in blood, so when pressure is released the dissolved N will come out of solution and causes bubbling, bubbling is bad b/c it can cause embolisms which can cause strokes, heart attacks and serious pulmonary disease
What is barotrauma?
Decompression illness, characterized by hemoptysis and, frequently, stroke; resulting from pulmonary hyper-inflation leading to alveoli and pulmonary tissue rupture and, frequently, cerebral arterial gas embolism. (physical trauma, come up to lower pressure to quickly)
What is bends?
Decompression illness, characterized by severe joint pain and other signs of generalized embolitic problems, resulting gas bubble embolism in arterioles and capillaries. (where N bubbles are coming out) (treatment? Put them back in pressure
What is Caisson’s disease?
For purposes of this course, this is a particular type of decompression illness, characterized by aseptic necrosis of bone, probably resulting from repeated microembolism of bone arterioles.
What is nitrogen narcosis?
Stupor and mental confusion in divers breathing compressed air caused by the amount of nitrogen dissolved in the blood as a result of the depth and duration of the dive., NO is an anesthetic, if enough N in blood then get similar results, reports that some divers enter a euphoria, experts say only do non-decompression dives, always dive with a partner
Describe the relative narcotic potency of gases in deep sea diving relative to N.
1. N-1.00
2. He-0.18
3. Ne-0.32
4. H-0.41
5. Ar-1.46
6. Krypton-8.9
7. Xenon-31.8
8. NO-28.3
For hyperbaric environments, describe the relationship between atmosphere change and sea water depth.
for every 33 feet deeper in sea water get 1 atm atmospheres absolute change
Describe the basic concept of the heart-brain circulation.
develops by selective ischemia in response to the diving reflex, in normal circulation, venous blood enters heart which sends blood to the lungs for oxygentation, blood then reenters heart and distributes blood to all other tissues, under hyperbaric conditions, the diving reflex takes affect and blood from the heart goes only to the brain and does not reach other tissues
What effect does emmersion have on EKG?
emmersion results in bradycardia, blood flow into calf is clearly much reduced during dive, some 12 seconds before and after emmersion there is a lot more blood flow to the calf
What are the signs and symptoms of acute mountain sickness (AMS)?
hypobaric disease, Headache, lightheaded, muscular weakness, anorexia, drowsiness, nausea, vomiting, decreased urine output, difficulty sleeping, memory deficits, skin pallor, and cyanosis of lips and nail beds. Symptoms most severe in 2nd or 3rd day. #1 problem with reduction of O2 (hypobaria)
what is the treatment for AMS?
With mild symptoms, rest, light diet, increased fluid intake, headache remedy. With severe symptoms, descent to lower altitude and/or oxygen therapy.
What are the signs and symptoms of high altitude pulmonary edema (HAPE)?
Any of those seen with AMS; dyspnea; persistant, non-productive cough; severe fatigue; hemoptysis in 20% of cases; coma (6-12 hours prior to death); generalized cyanosis; crepitant rales; rhonchi; hypotension; and tachycardia. leakage of capillaries, high altitude has caused the capillaries to fail
What is the treatment for HAPE?
Bed rest, oxygen administration, descent to lower altitude if possible, diuretics of limited value
What are the signs and symptoms of high altitude encephalopathy (HAP)?
Any seen with AMS and HAPE; severe occipital headaches; cervical pain; tinnitus; anesthesia/paresthesia of arms and legs; hemiplegia; arthralgia; confusion, stupor; coma; cervical rigidity; abnormal Babinski sign; localized areas of cutaneous anesthesia; and dilation of retinal vessels. real serious, affect on the brain, have all the previous signs and symptoms, but now having those above
What is the treatment for HAP?
Oxygen administration and descent to lower altitude (portable hypobaric “chamber”)
What are the signs and symptoms of chronic mountain sickness?
Exceptionally high red cell mass and count, right heart enlargement, and congestive heart failure.
What is the treatment for hypobaria?
Descent to lower altitude!
Describe the relationship between barometric pressure and O2 pressure at high altitude
as altitude INC, air and O2 pressure as % of sea level DEC, Reno is about 80%, 5000 ft above there is also an affect on night vision because pigments used in night vision don’t recover quickly
What is the metnal effect of hypoxia?
a person is put in a pressure chamber and asked to wrist continuously, within 90 seconds start to lose it, make spelling mistakes and duplicate letters, give O2 back at 20% and within 30 seconds he does ok
Describe the effects of acute exposure to low atmospheric pressures on alveolar gas concentrations and arterial O2 saturation.
at altitude of 0, arterial O2 saturation is 97%, as altitude INC, arterial O2 saturation DEC, at altitude of 30,000 ft arterial O2 saturation goes to 24
What is the effect of high altititude on arterial O2 saturation when one is breathing air and when breathing pure O2?
when breathing pure O2, takes higher altitude to start seeing a DEC in arterial O2 saturation, but drop in O2 saturation is much more rapid
What are various responses to high altitude (14,000 ft) and what does the body to do acclimate to it?
1. INC basal rate-basal rate in some subjects returns to normal in 3 days
2. subjects cannot sleep-after 3 days most subjects can sleep
3. all subjects show Cheyne-Stokes respiration during sleep-most subjects cease Cheyne-stokes respiration in sleep, all subjects show INC sensitivity to further INC in altitude
4. resting tidal volume INC conspicuously, the hypoxic drive from chemoreceptors does not change with acclimitizaiton-there is a redistribution of blood, an INC in the chest area, the lung capillary bed is continuously dilated, the functional residual capacity INC
5. uncompensated alkalosis-many subjects achieve normal acid-base equilibrium (time needed varies)
6. O2 delivery to tissues is inadequate-there is an INC in O2 deliverd to tissues, total Hb goes up as much as 90%, the hematocrit rises from a normal of 40-45 to as high as 60-70, Hb goes from 15-22 gm%, blood volumen INC 20-30%, plasma volume stays the same
7. some subjects experience headache and nausea-these symptoms disappear in 3 days
What are the responses to 16,000 feet altitude and what is the acclimation to it?
fatigue, headache, loss of apetite, sleeplessness (all subjects), systolic and diastolic pressure showed uniform elevation-all returned to normal in 14 days
What are the responses to 21,500 feet altitude and what is the acclimation to it?
symptoms above returned, plus progressive exertional dyspnea, irritative coughs, impatience, depression and nocturnal muscle cramps, a persistant INC in heart rate, expected even after acclimatization-all improved in 10 days
Summarize the changes due to microgravity.
1. changes in blood system-DEC immune response, DEC RBC count
2. headward fluid shif-neurovestibular disturbances, INC blood volume in head and trunk (which leads to CV adaptation and INC urine production)
3. DEC weight bearing-muscular decondition and bone demineralization
What is the clinical horizon line?
line that shows values that humans must stay below to avoid clinical disease from space travel, if stay above this line have clinical disease (such as INC in neurovestibular system, fluids and electrolytes and CV system)
Describe the significance of the bone and calcium metabolism line.
goes up and won’t come back down, keeps going up and bone anc Ca2+ metabolism will never acclimate to microgravity
Describe the significane of radiation effects.
still going up too, keeps going up and radiation effects will never acclimate to microgravity
What are the components of the vestibular system?
visual, inner ear (semicircular canals), proprioceptors in legs
What is space adaptation syndrome (SAS, space nausea)?
in microgravity, 2 of the systems go nuts, inner ear and proprioception in legs) visual orientation fine, leads to motion sickness
What vestibular systems components do fish have?
have only vision and semicircular canals, swim in spirals because they are trying to find the gravity vector, swim normally after 3 days because they became visually oriented, ignored the semicircular component, visual input is very powerful
What are the results of the SAS experiments?
1. no predictors for SAS
2. vestibular disturbance lasts for week or more after landing
3. loss of spatial orientation
4. loss of muscle stamina
Describe fluid shift.
1. on earth, blood tends to pool in the lower body
2. promptly upon entering weightlessness, fluids shift toward the head, complain about fullness and the head is stuffy, when going into weightlessness, part is overpressurized and blood goes to head
3. after a time, the body adapts to weightlessness, the kidneys reduce the volume of fluid, relieving pressure in the head and chest
4. the body reacts immediately upon reentering Earth’s gravity, fluids are shifted from the head toward the feet
Describe the tilt test.
heart rate, BP and change in leg volume during a 25-min, 70 degree tilt test protocol, the first and last 5 min being in the horizontal, supine position, pre-flight mean curves are solid lines and the first postflight test values are dashed, heart rate INC, BP stays about the same, leg volume INC as well
Describe the lower body pressure test.
heart rate and change in leg volume during a 25 min LBNP test protocol, preflight mean curves are dashed and the first postflight test date 3 hour after splashing down are solid lines, heart rate INC, change in leg volume also INC, creates pressure on lower body creating a stress on CV system because it sucks blood into lower body
Describe the fluid shift with being bed ridden.
lying down shift 11% of the total blood volume away from the legs to the rest of the body, of the volume that shifts, 78% goes to the thorax, 20% to the head and neck, 2% to the abdominal organs
Describe CV deconditioning.
1. fluid shift
2. DEC heart size
3. reduced exercise capacity
4. orthostatic hypotension (IG)
Describe the changes in serum protein, albumin and total Ca2+ levels as a function of flight duration.
the mean are the values from nine crewmen for the first 28 days, six crewmen until day 56 and three crewmen until 84 d, changes are shown as percentage change from preflight control levels, serum total Ca2+ INC rapidly and remained INC for the duration of the flight, plasma total protein levels were INC in the first crew during the first month of flight, but remain near control levels for the remaining crews, and plasma albumin levels dropped dramatically during the first few weeks of flight and remained depressed for the remainder of the flight, total serum Ca2+ INC then levels off, total serum protein DEC, then goes back to preflight levels, serum albumin DEC then levels off at a lower level, each takes about 20 days to level off
Describe the changes in urine and fecal Ca2+ as a function of flight duration.
both urine Ca2+ and fecal Ca2+ INC (fecal Ca2+ DEC initially then INC), during postflight days, takes about 10 days for the fecal and urine Ca2+ levels to go back to normal, we don’t know where the Ca2+ loss in feces will end, by the time reach Mars probably not able to stand up without pathological fractures due to osteoporosis, urine Ca2+ levels off at about 20 days
WEhat does weightlessness do for bone and Ca2+ metabolism?
have more resorption than formation, if you have too much in the serum it’ll be kicked out in the urine, more Ca2+ is absorbed than secreted from the gut, serum Ca2+ is excreted in urine
Describe the relationship between space flight and bed rest.
in both there is an effect in cephalad fluid shift and redistribution of bone, there is a DEC in bone formation, Ca2+ balance, and 1,25 (OH)2D, there is an INC in fecal Ca2+, urine Ca2+, in bone resorption (there is an INC/NC), there is an INC in serum Ca+ in space flight, in bed rest there is INC/NC, for PTH there is DEC in space flight and NC in bed rest, for serum osteocalcin there is a DEC for space flight and NC in bed rest, for bone strength there is a DEC in space flight and not done for bed rest
Describe the loss of muscle in space.
1. static strength reduced by 10 – 40% in calf
2. all other muscle groups had 10-15% atrophy
3. loss of muscle in animals was from 10-30% of wet weight
4. no loss of muscle cells (nuclei) but in mRNA
What are the spaceflight induced physiological changes? which are of greatest concern fro long duration missions?
CV decondidtioning (*), negative Ca2+ balance (*), space adaptation syndrome, red cell mass reduction (not sure where they go but it really doesn’t cause a problem), immunological changes (*), other mineral changes, hormonal changes, radiation effects (*, high energy cosmic radiation is a large threat due to the magnetic field absence that is normally around the sphere of the earth)
What are some characteristics of gap junctions?
1. message transmission-directly from cell to cell
2. local
3. specificity depends on anatomic location
4. low resistant, transfer of small molecules from one cell cytosol to the next, diffusion
What are some characteristics of synaptic transmission?
1. message transmission-across synaptic cleft
2. local
3. specifically depends on anatomic location and receptors
4. how neurons communicate
What are some characteristics of paracrine and autocrine transmission?
1. message transmission-by diffusion in interstitial fluid
2. locally diffuse
3. specifically depends on receptors
4. works on large EC space
What are some characteristics of endocrine signaling?
1. message transmission-by circulating body fluids
2. general
3. specifically depends on receptors
4. general type of communication, substance released from cells and released into circulation and transported in bloodstream, can come into contact with all cells but depends on specificity of the receptor on the cell
Give some examples of paracrine and autocrine signaling.
Paracrine signaling: Glucose -> insulin INC -> glucagon DEC
Autocrine signaling: Insulin is needed for optimal insulin secretion
Define hormone.
A chemical substance that is:
- released into the blood by specialized secretory cells
- transported to its target by the circulation
- elicits a physiological response in its target cell
What is a target cell?
A cell that expresses receptors for a hormone
What is a hormone receptor?
A protein expressed by a target cell that can bind a hormone with high specificity and affinity; and that, upon binding its ligand, elicits physiological effects by activating intracellular signaling mechanisms.
What is a binding/carrier protein?
A circulating protein to which a hormone can reversibly bind without eliciting physiological effects. Binding proteins protect hormones against loss in the kidney, slow degradation and cellular uptake, and buffer changes in free hormone concentration. are really important because they protect hormones from quick degradation, hormone w/out binding proteins have a very short half life
What is an endocrine gland?
A “ductless gland” that produces and secretes signaling molecules (hormones) into the circulation. Highly vascularized. May develop from any of the germinal layers
What are some examples of classical endocrine glands?
Pituitary, thyroid, parathyroids, pancreatic (Langerhans) islets, adrenals, ovaries, testes, placenta.
What are some examples of nonclassical endocrine glands?
Hypothalamus (neurohormones), heart (atrial natriuretic factor), kidney (renin, erythropoietin, active vitamin D), adipose tissue (leptin, adiponectin, resistin).
Describe the synthesis, secretion and metabolism of oligo/polypeptide, protein and glycoprotein hormones.
1. Synthesized as any protein; stored in secretory vesicles
2. Preprohormone -> prohormone -> hormone
3. Glycosylation may prolong half-life
4. Metabolized in the circulation, in target cells, liver, kidney
Describe the synthesis, secretion and metabolism of steroids.
1. Synthesized from cholesterol as needed – no storage
2. Manufactured in the mitochondria and cytosol via a series of enzymatic reactions
3. Reversibly bound to carrier proteins in the circulation
4. May be converted in the periphery into more potent or even different compounds (eg. testosterone -> dihydrotestosterone)
5. Inactivated by reduction and conjugation (sulfate, glucuronide), products can be detected in urine (used for diagnosis)
Describe the synthesis, secretion and metabolism of amines.
1. Tyrosine derivatives (catecholamines, thyroid hormones)
2. Stored in granules (catecholamines) or as part of a precursor protein (thyroid hormones)
3. Thyroid hormones bind to circulating carrier proteins
4. Metabolized intracellularly into inactive or active products (e.g. thyroxine -> triiodothyronine)
5. Catecholamines are inactivated by conjugation with sulfate or glucuronic acid and can be detected in urine (used for Dx)
What patterns of hormone secretion?
include pulsatile, circadian, episodic, fluctuations may also occur on longer time scales (montly, seasonal (not typical in humans)), multiple serial measurements may be required for diagnosis, ex: cortisol peaks in deep sleep
What are some problems with hormone measurements?
- Concentrations in the circulation are VERY low (~nmol/L)
- Most hormones cannot be detected by specific chemical reactions
- Bioassays (quantification by magnitude of effect) are difficult to standardize and interpret and are labor-intensive; some require the use of animals
Describe immunoassays.
detection is based on antibody-antigeneration, highly specific, very sensitive
Describe radioimmunoassay (RIA).
typical labels include protein hormones (125 I), other hormones 14 C, 3 H, procedure involves:
1. antibody on solid phase + labeled antien + patient’s serum containing antigen
2. bound activity + free activity, wash the supernatant and measure bound activity (antigen in patient’s serum competes with labeled antigen for binding sides on solid phase antibodies)
3. graph bound activity vs. antigen concentration in patients serum, as more antigen is in patients serum the less bound activity there is
Describe Enzyme-linked-Immuno sorbent Assay (ELISA).
use fluorescent antibodies as well, steps include:
1. monocional antibodies on solid phase + patient’s serum containing antigen
2. antigen binds to antibodies
3. add enzyme-linked second antibody
4. chemical reaction generates an optical signal (colorimetric assay)
Describe the mechanism of action for steroids, thyroid, retinoic acid and vitamin D superfamily.
lipophilic hormone diffuses through cell membrane, enters cell and binds to receptor, the receptor may reside in the cytoplasm or bound to DNA, receptors are ligand-activated transcription factors, once hormone is bound to receptor binds to hormone response element and leads to transcription and translation
Describe the mechanism that all other hormones act through?
act through membrane receptors (G-protein coupled or ion-channel linked receptors), both act to INC Ca2+
Describe the courses of hormone action.
1. translocation of proteins into cell membranes (within seconds)
2. phosphorylation of metabolic enzymes -> altered activity (10-15 mins)
3. effects on mRNA translation and DNA transcription (hours-days)
4. ex: insulin receptor (enzyme linked receptor, tyrosine kinase), composed of four subunits, alpha are external and react with the substrate
What determines how well the target tissue responds to hormonal stimulation?
1) Capacity to respond: The magnitude of response to a given concentration of the hormone
2) Sensitivity to stimulation: The concentration of a hormone required for a given effect (e.g. half-maximal response). This is determined by: a) Affinity of receptors: the concentration of a hormone needed to occupy ½ of available receptors, b) Availability of receptors: Up- or down-regulation: the primary mechanisms whereby sensitivity is regulated
Describe regulation of hormone secretion.
Hormone secretion must be turned on/off at precisely the right times
1) Open-loop control
-Signal ON -> hormone increases -> effect ON
-Signal OFF -> hormone decreases -> effect OFF
Example: suckling -> oxytocin (from hypothalamus) -> milk ejection
2) Negative feedback (closed-loop regulation)
Signal ON -> hormone increases -> effect ON à signal OFF -> hormone decreases -> effect OFF à Signal ON, etc
Example: decrease in blood glucose -> glucagon increases -> blood glucose increases -> glucagon decreases, etc
-Closed-loop regulation occurs around a set point (thermostat)
-Precise regulation requires 2 opposing systems (“A/C & heating”)
-Set point may be changed or overridden (e.g. catecholamines increase blood glucose even when insulin secretion is increased)
Describe the negative feedback control of glucose.
integrated control of blood glucose occurs by glucagon and insulin, when blood glucose concentration is low, glucagon is high and insulin is low, when blood glucose is high, glucagon is low and insulin is high
Describe positive feedback regulation of hormone secretion.
Signal ON -> hormone increases -> effect ON -> signal increases -> hormone increases -> effect ON -> signal increases, etc, this eventually leads to a cataclysmic event that terminates the “vicious circle”, Example: fetus’ head -> stretch of uterine isthmus -> oxytocin increases -> contraction of myometrium -> fetus’ head pushed against isthmus -> more oxytoxin produced and released
Describe the pituitary gland.
The pituitary measures ~1 cm in diameter and is located in the sella turcica. It is connected to the hypothalmus by the pituitary stalk (also known as the infundibulum or the hypophysial stalk)
What are the components of the pituitary gland?
anterior (adenohypophysis) and posterior (neurohypophysis) lobes, infundibulum and pars intermedia (avascular, separates the anterior and posterior lobe, not much functional significance, more well developed in animals (makes hormones there in some animals, melanocyte stimulating hormone)
Describe the embryonic development of the pituitary gland.
During embryonic development the two lobes of the pituitary gland originate from different sources:
-ANTERIOR – from an invagination of the pharyngeal epithelium (Rathke’s pouch).
-POSTERIOR – from a neural tissue outgrowth from the hypothalamus
-This explains why the anterior and posterior lobe are so distinct in structure and function
Describe the innervation of the neurohypophysis.
is the storage area for hypothalamic hormones, has connections with the neurons in the ventral hypothalamus and hypothalamic neurons in the paraventricular and supraoptic nuclei, releases oxytocin and ADH
What are the contents of the hypophyseal portal system?
1. primary capillary plexus
2. hypophyseal portal veins
3. secondary capillary plexus
What does the posterior pituitary secrete?
oxytocin and ADH
What does the anterior pituitary secreted?
TSH, FSH, LH, ACTH, PH, PRL
Describe the control of pituitary secretion.
 Secretion by the anterior pituitary is controlled by hormones secreted by neurons within the hypothalamus.
 Secretion from the posterior pituitary is from magnocellular neurons which originate in the hypothalamus and terminate in the posterior lobe.
What are the acidophiles of the anterior pituitary gland?
1. somatotrophes-~40%, include growth hormone (GH, somatotrophin)
2. lactotrophes-AKA mammotrophes, ~20%, include prolactin (PRL, comes from the same protein as GH, just cleaved differently)
What are the basophils of the anterior pituitary gland?
1. corticotrophes-20%, include adrenocrocticotropic hormone (ACTH)
2. thyrotophres-5%, include thyroid stimulating hormone (TSH)
3. gonadotrophes-5%, include follicle-stimulating hormone (FSH) and luteinizing hormone (LH)
What hormones fall in the glycoprotein family and what are their targets and major actions?
comprised of alpha and beta subunits + carb chains
1. TSH-targets the thyroid gland, INC synthesis and secretion of thyroid hormones
2. FSH-targets the ovary and INC folliculogenesis and estrogen synthesis there, also targets the testis (Sertoli cells) and INC sperm maturation
3. LH-targets the ovary (Graafian follicles to INC ovulation and formation of the corpus luteums) and the (corpus luteum to INC estrogen and progesterone synthesis), also targets testis (Leydig cells to INC testosterone synthesis)
What hormones fall in the GH/PRL family polypeptides?
1. GH-target most tissues to INC growth in stature and mass, INC IGF-1 production, INC protein synthesis, DEC glucose utilization, INC fat utilization
2. PRL-targets mammary glands to INC milk secretion and INC growth of mammary glands
oversecretion of GH leads to oversecretion of PRL as well
What hormones fall in the pro-opiomelanocortin (POMC) family (polypeptides that are products of the same POMC gene)?
1. ACTH-target adrenal cortex (zona fasciculate and reticularis) to INC synthesis and secretion of adrenal cortical steroids
2. beta-lipotropin-target fat to Inc fat mobilization, little activity in humans
beta-endorphins and melanocyte-stimulating hormone (MSH) are members of the POMC family released from the intermediate lobe of the pituitary in human embryos and pregnant women
Describe the cleavage of the POMC gene.
cleavage of POMC occurs by endopeptidases (prohormone convertases), cleavage of POMC gene forms beta-lipotropin, ACTH, gama-lipotropin and beta-endorphin, corticotrophe forms JP, ACTH, gama-lipotropin and beta-endorphin, melanotrophe forms JP, alpha-MSH, CLIP, gamma-lipotropin and beta-endorphin, cleavage shown below dashed line occurs in intermediate lobe (non-functional in adult humans)
What enzymes make ACTH?
cleavage of POMC gene by PC1 and PC2
Describe the hypothalamic-hypophysial portal system.
Hypothalamic releasing / inhibitory hormones are secreted in the hypothalamus; enter the portal system and are conducted to the anterior pituitary where they influence secretion from glandular cells. The hypophysial portal blood flow only represents a tiny fraction of the cardiac output. Only minute amounts of neural secretions are needed for effect. this is an example of neuroendocrine regulation (fast and highly specific), transports hormones from hypothalamus to the anterior pituitary
Describe the regulation of the anterior pituitary.
contains long loop negative feedback, ultra-short loop negative feedback and short loop negative feedback, hypothalamus is a collecting center for environmental and sensory information, release of hypothalamic releasing/inhibitory factors can be influenced by pain, emotions, light and dark, sleep rhythms, exercise, concentrations of electrolytes and hormones in the blood
What is the structure and primary action on the anterior pituitary of thyrotropin-releasing hormone (TRH)?
a peptide that INC synthesis and secretion of TSH and PRL
What is the structure and primary action on the anterior pituitary of gonadotropin-releasing hormone (GnRH)?
a peptide that INC synthesis and secretion of LH and FSH
What is the structure and primary action of corticotrophin release hormone (CRH) and argining vasopressing (AVP)?
peptides that INC synthesis and secretion of ACTH
What is the structure and primary action of growth hormone releasing hormone (GHRH)?
a peptide that INC synthesis and secretion of GH
What is the structure and primary action of growth hormone inhibitory hormone (somatostatin)?
a peptide that DEC GH secretion
What is the structure and action of prolactin inhibiting hormone?
a dopamine that DEC synthesis and secretion of PRL
Describe hypothalamic releasing/inhibitory hormones (hypophysiotropic hormones)
• Hypothalamic hormones stimulate pituitary hormone secretion by increasing intracellular [Ca2+] via G-protein-coupled receptors. 2nd messengers include cAMP, IP3 and DAG.
• Several hypothalamic hormones are released in a pulsatile fashion but only pulsatile GnRH release is critical for pituitary (gonadotropin) secretion.
Describe the regulation of LH release by GnRH.
GnRH was sampled in pituitary portal blood.
LH was sampled in peripheral blood.
Note the rapid regulation of pituitary hormone (LH) secretion by the hypothalamic neurohormone (GnRH).
Describe the cells and hormones of the posterior pituitary gland.
secrete ADH (anti-diuretic hormone, vasopressin) and oxytocin
 The posterior pituitary is comprised primarily of glial-like cells called pituicytes – these cells do not secrete hormones.
 Hormones of the posterior pituitary are secreted by neurons.
 The bodies of these neurons are located in the hypothalamus
 The hormones are synthesized in the cell bodies and transported down the nerve axons into the posterior pituitary.
Describe the magnocellular neurosecretory system.
regulates the posterior pituitary via the paraventricular and supraoptic nucleus, oxytocin and vasopressin is produced by the magnocellular system in the paraventricular nucleus into the posterior pituitary
Describe the structure, origin, binding/carrier protein, half-life, functions and mechanism of action of oxytocin.
-Structure: nonapeptide
-Origin: magnocellular neurons in the paraventricular (oxytocin) & supraoptic nuclei (ADH)
-Binding/carrier protein: neurophysin I, bind at the axoplasm
-Half-life (T½): ~2 min
-Functions:
1. Milk ejection – contracts myoepithelial cells that surround the mammary alveoli
2. Hastens delivery, promotes delivery of the placenta, reduces bleeding – contracts uterine smooth muscle
(NOT THE PHYSIOLOGICAL TRIGGER FOR PARTURITION)
-Mechanism of action: via G-Protein coupled receptor; 2nd messengers: IP3, DAG
Describe the regulation of oxytocin secretion.
suckling and cervical stretch receptors stimulate the CNS to communicate with the paraventricular and supraoptic nuclei to tell the posterior pituitary to release oxytocin, oxytocin tells the mammary glands to let-down milk and tells the uterus to contract which has a positive feedback role of stimulating cervical stretch receptors and INC oxytocin release
What is the structure, origin, binding/carrier protein, half-life, functions and mechanism of action for ADH/(AVP)?
-Structure: nonapeptide
-Origin: magnocellular neurons in the supraoptic & paraventricular nuclei
-Binding/carrier protein: neurophysin II
-Half-life (T½): ~2 min
-Functions & mechanisms of action:
1. Reabsorption of water in connecting/collecting tubules via V2 rec. -> cAMP -> insertion of aquaporin-2 into the luminal membrane
2. Vascular smooth muscle contraction via V1 rec. -> IP3, DAG -> INC [Ca2+]i
Describe the regulation of ADH secretion.
volume receptors in the thorax and osmoreceptors in the hypothalamus stimulate the CNS, paraventricular and supraoptic nucleis to tell the posterior pituitary to release ADH, ADH stimulates water reabsorption in the kidney which INC plasma volume which stimulates volume receptors (+ feedback) as well as DEC plasma osmolality which stimulates osmoreceptors (+ feedback)
Describe the problem, causes and principle symptoms of diabetes insipidus.
-Problem: Unable to conserve water due to ↓ADH synthesis (central DI) or insensitivity to ADH at the collecting ducts (nephrogenic DI)
-Causes: trauma, tumors, infection (e.g. meningitis)
-Principle symptoms: -INC water loss from kidneys (polyuria) triggers, INC thirst (polydipsia)
Describe the problem, causes and principle symptoms of syndomre of inappropriate ADH secretion (SIADH)?
-Problem: inappropriate ADH secretion -> INC Water retention -> atria of heart stretched -> -INC ANF, DEC plasma renin activity -> sodium loss -> hyponatremia and DEC plasma osmolality; concentrated urine
-Causes: Ectopic: ADH secreted from lung cancer, etc., Eutopic: Stroke, infection
-Principle symptoms: asymptomatic during early stages especially if serum sodium falls slowly. Rapid fall associated with confusion, drowsiness, convulsions, coma and death.
Describe the morphology of the adrenal glands.
from outermost to innermost, zona glomerulosa (aldosterone) -> zona fasciculate (cortisol and androgens) -> zona reticularis (cortisol and androgens) -> medulla (catecholamines), direction of blood flow is from capsule -> outermost to innermost
Describe the adrenal glands.
 The adrenals are required for maintenance of life: Complete adrenal insufficiency (e.g. Addison’s disease), if untreated, leads to death within 2 weeks. Cause of death: circulatory collapse (shock) or hypoglycemic coma.
 composed of 4 functional zones:
 The adrenals often act as one functional unit to cope with a change in the external or internal environment that threatens HOMEOSTASIS.
What are the four functional zones of the adrenal glands, what hormones are there and what controls it?
1. zona glomerulosa-mineralcorticoids, controlled by kidney (renin-angiotensin)
2. zona fasciculate-glucocorticoids, controlled by hypothalamic-pituitary axis via ACTH
3. zona reticularis-androgens, controlled by hypothalamic-pituitary axis via ACTH
4. medulla-catecholamines, controlled by sympathetic nervous system
Describe the adrenal medulla.
~10% of the adrenal mass
-Origin: neuroectoderm
-Microanatomy: modified postganglionic neurons (chromaffin cells) innervated by cholinergic preganglionic neurons from the thoracolumbar spinal cord.
-Secretory products: epinephrine : norepinephrine = 5:1, The adrenals are the sole source of epinephrine. Norepinephrine is also released from sympathetic terminals and extraadrenal chromaffin cells.
-The adrenal medulla is not essential for survival as long as the sympathetic nervous system is intact.
Describe the biosynthesis of catecholamines.
1. tyrosine -> DOPA (via tyrosine hydroxylase (TH), rate-limiting step (inhibited by catecholamines)
2. DOPA -> dopamine (via aromatic L-amino acid decarboxylase (AAD))
3. dopamine -> norepinephrine (via dopamine beta-hydroxylase (DBH), occurs within secretory granule)
4. norepinephrine -> epinephrine (via phenylethanolamine-N-methyltransferase (PNMT), inducible by glucocorticoids)
Describe the storage, release, metabolism and excretion of adrenal medullary hormones.
-Storage: in granules with ATP, enkephalins & b-endorphin
-Release: ACh -> nicotinic receptors -> Na+ influx -> depolarization -> VSCC -> [Ca2+]i INC -> fusion with membrane (exocytosis)
-Metabolism: Uptake into sympathetic nerve terminals; reused as NE or degraded by MAO (monoamine oxidase), Capillary endothelium, heart, liver, etc.: metabolization by COMT (catecholamine-ortho-methyl-transferase)
-Excretion: inactivated products are conjugated with glucuronic acid & sulfates -> urine (can be exploited for diagnostic purposes)
What is the main function of adrenal medullary hormones?
To make rapid, short-term adjustments to actual or threatened changes in the external or internal environment (“fright, flight, or fight”). In the longer term, actions are supported by glucocorticoids.
What is the target tissues of the adrenal medullary hormones?
Coordinated actions to ensure survival: INC cardiac output, INC perfusion of brain & working muscles, INC supply of energy-rich fuels, INC pulmonary ventilation, INC visual acuity, INC muscular performance, quiescence of the gut to permit diversion of blood flow
What is the mechanism of action for the adrenal medullary hormones?
a1 receptors coupled to Gaq -> IP3/DAGINC
a2 receptors coupled to Gai -> cAMP DEC
b1 & b2 receptors coupled to Gas -> cAMP INC
for beta-E > NE, for alpha-NE > E
What is the regulation of adrenal medullary hormones?
hypothalamus -> sympathetic preganglionic fibers
What do the beta-receptor mediated adrenal medullary hormones do?
1. INC force and rate of heart contractions (beta1), arteriolar dilation (skeletal muscle)
2. INC glycogenolysis (beta2) and INC lipolysis (beta1)
3. relaxation of bronchial muscle (beta2)
4. DEC motility of GI tract (relaxation, beta2)
5. relaxation of urinary bladder (beta2)
6. relaxation of uterus (beta2)
7. INC renin secretion
8. INC tension generation and INC neuromuscular transmission
What do alpha receptor mediated adrenal medullary hormones do?
1. arteriolar constriction (skin, renal, splanchnic, genital, alpha1)
2. INC gluconegogenesis
3. INC sphincter contraction of Gi tract
4. INC sphincter contraction of urinary gladder
5. sweating
6. pupil dilation (contraction of radial muscle)
What is pheochromocytoma?
-What is it?-Chromaffin cell tumor
-Why is it a problem? Causes over secretion of catecholamines
-What are the dominant features? Elevated heart rate, Systemic hypertension, Anxiety, Pallor and sweating, Hyperglycemia
-How is it diagnosed? Detection of increased levels of catecholamines and their metabolites in the blood and urine
What is the origin, microanatomy, and chief secretory products of the adrenal cortex?
-Origin: mesoderm
-Microanatomy: lipid-containing, cuboidal cells arranged in clusters, bundles or tangled networks
-Chief secretory products: steroids (products of the fetal adrenal cortex will be covered elsewhere)
-Z. glomerulosa: aldosterone (mineralocorticoid)
-Z. fasciculata: cortisol (glucocorticoid) corticosterone (glucocorticoid)
-Z. reticularis: after puberty: dehydroepiandrosterone (DHEA) and -sulfate (DHEAS), androstendione (androgens)
Describe the biosyntehsis of adrenal cortical hormones.
starts from cholesterol, the rate limiting step is side chain cleavage by desmolase that forms pregnenolone which is the precursor for all the cortical hormones, progesterone is rapidly converted not secreted, aldosterone synthase is found only in the z. glomerulosa
What is the transport, half-life, normal blood concentration, metabolism and excretion of adrenal cortical hormones?
-Transport in blood: Corticosteroid Binding Globulin (CBG; transcortin), albumin. Approx. 90-95% of cortisol is bound; 60% of aldosterone is bound.
-Half-life: cortisol: 60-90 min; aldosterone: ~20 min
-Normal blood concentration: Aldosterone: 6ng/dL (secretory rate 0.15 mg/day; Cortisol 12µg/dL (secretory rate 15-20 mg/day)
-Metabolism: Liver: reduction + conjugation with glucuronic acid & sulfates.
-Excretion: 25% excreted in bile ® feces. Remaining conjugates are filtered by kidneys ® urine.
What is the cellular mechanism of action of adrenal cortical hormones?
Adrenal cortical hormones are steroids – elicit effects by producing changes in gene transcription. Effects are not immediate - require 45-60 min for proteins to be synthesized; full effect may take hrs or days to develop.
Describe the glucocorticoids.
-Cortisol (also known as hydrocortisone; very potent; accounts for 95% of all glucocorticoid activity)
-Corticosterone (less potent than cortisol; accounts for ~4% of all glucocorticoid activity)
-Main function: Effects on metabolism of proteins, carbohydrates and fats. Glucocorticoids have widespread actions which help restore homeostasis after stress. In the absence of glucocorticoids, even mild physical or mental stress can be deadly.
-Targets: many – see table. Virtually every tissue is affected.
What are the actions of the glucocorticoid hormones?
1. DEC corticotrophin-Releasing hormone (negative feedback)
2. DEC ADH secretion
3. INC gluconeogenesis
4. INC maturation and surfactant production during fetal development
5. DEC ACTH secretion and synthesis (negative feedback)
6. INC GFR (needed to excrete dilute urine)
7. INC protein catabolism, DEC glucose oxidation, DEC insuling sensitivity, DEC protein synthesis
8. DEC blood concentration of eosinophils, basophils and lymphocytes
9. DEC cellular immunity
What happens when glucocorticoids act on proteins?
they INC protein breakdown which leads to INC in blood urea nitrogen, preservation of carb reserves and mobilization of alternative, high energy fuels (protein and fat)
What are the actions of glucocorticoids in injury and stress?
Glucocorticoid secretion increases in stressful situations (i.e. trauma, infection, surgery, intense cold or heat.) Why is this beneficial?...Not fully understood. Mobilization of amino acids and fats may be beneficial as this makes them available for energy and synthesis of other compounds required for maintenance and reproduction of new cells (e.g. glucose, new proteins, purines, etc.), In addition, cortisol has anti-inflammatory effects. Sometimes the inflammation associated with a disease or trauma can be more damaging than the condition itself. e.g. rheumatoid arthritis.
What is the therapeutic utility in allergies, autoimmune diseases and transplantation of glucocorticoids?
• Stabilizes lysosomal membranes – proteolytic enzymes are confined to lysosomes.
• Decreases production of T cells and antibodies (DEC B cell proliferation).
• Decreases formation of prostaglandins and leukotrienes, thus DEC vasodilation DEC capillary permeability DEC mobility of white blood cells (WBCs).
• Reduces fever by decreasing release of interleukin-1 (IL-1) and other inflammatory cytokines from WBCs.
Describe the regulation of glucocorticoid secretion.
-Cortisol secretion controlled by adrenocorticotropic hormone (ACTH) secreted by anterior pituitary. Cortisol exerts negative feedback effects on the hypothalamus and anterior pituitary gland – DEC ACTH.
-ACTH secretion is controlled by corticotrophin-releasing hormone (CRH) secreted into the hypophysial portal system of hypothalamus.
How does ACTH stimulate adrenal steroid synthesis?
¬ 1. INC cholesterol liberation from LDL
¬ 2. INC transcription of LDL receptors and enzymes involved in hormone synthesis
¬ 3. INC transfer of cholesterol over the mitochondrial membrane
¬ [Long term ACTH stimulation also causes hypertrophy and proliferation of adrenocortical cells]
Describe the circadian rhythm of glucocorticoid secretion.
CRH, ACTH and cortisol secretion are high in the early morning but low in the late evening (diurnal rhythm). Release is pulsatile and the diurnal rhythm reflects changes in the pulse amplitude. therefore Measurements of blood cortisol levels must be expressed in terms of the time in the cycle.
describe the mineralcorticoids.
-Aldosterone (very potent; accounts for 90% of all mineraldocorticoid activity)
-Desoxycorticosterone, corticosterone & cortisol (also provide mineralocorticoid activity; cortisol 1/3000 as potent as aldosterone but plasma concentration of cortisol 2000 times higher)
what are the main actions of the mineralcorticoids?
1. Sodium retention Required for the reabsorption of ~2% of filtered Na+. Accompanied by absorption of water (corresponds to ~ 3.5L EC fluid per day). Sodium and water retention can escape from long-lasting stimulation by aldosterone (15-25mm Hg increase in arterial pressure produces pressure natriuresis & diuresis)
2. Potassium excretion
3. Proton excretion & bicarbonate retention
What are the targets of the mineralcorticoids?
principal cells of the distal tubule & collecting duct; intercalated cells in the collecting duct; colon; sweat & salivary glands
What are the genomic and non-genomic effects of aldosterone?
-Genomic effects: Transcription of genes involved in ion transport: Increases reabsorption of Na+ and excretion of K+
-Non-genomic effects: activation of second messenger system: Increases H+ secretion and bicarbonate retention
describe the regulation of aldosterone secretion.
ACTH and angiotensin II stimulate alodsterone secretion, aldosterone secretion stimulates renal K+ excretion which DEC K+ concentration which stimulates aldosterone secretion, aldosterone secretion also stimulates renal Na+ retention which INC blood volume and inhibits the JG apparatus, that secretes renin, renin turns angiotensinogen into angiotensin I which (via angiotensin converting enzyme) turns into angiotensin II
Describe the regulation of aldosterone synthesis by angiotensin II.
angiotensin II binds to receptor which activates PLC which INC Ca2+ levels via DAG and IP3, Ca2+ activates CAM kinase which stimulates cholesterol synthesis into aldosterone
Describe the adrenal androgens.
-Dehydroepiandrosterone (DHEA)
-Androstenedione, dehydroepiandrosterone sulfate (DHEAS)
What are the main actions of the adrenal androgens?
Normally only weak effects in humans. May contribute to early development of male sex organs; growth of the pubic and axillary hair in females. In the testes adrenal androgens converted to testosterone.
What is hypoadrenalism (Addison’s disease)? what causes it?
destruction of the adrenal cortex by autoimmunity (80%), tuberculosis, cancer
What happens with mineralcorticoid deficiency?
Loss of Na+ and water -> DEC EC fluid, DEC plasma volume, hemoconcentration, hyponatremia. Retention of K+ and H+ ions -> hyperkalemia, mild acidosis. DEC cardiac output -> circulatory shock -> death
What happens with glucocorticoid deficiency?
impaired fuel mobilization, DEC gluconeogenesis -> hypoglycemia, weight loss, weakness, Extreme sensitivity to stress -> death
Describe the pigmentation associated with Addison’s disease.
Lack of normal negative feedback to hypothalamus and anterior pituitary by cortisol -> ACTH + MSH release increased -> melanin formation increased.
What causes hyperadrenalism (Cushing’s syndrome)?
(1) Hypersecretion of CRH or ACTH (from the pituitary: Cushing’s disease [70%]); (2) adrenal cortex adenomas [20-25%]; (3) ectopic secretion of ACTH (e.g. in lung cancer); (4) large doses of glucocorticoids for long periods.
What happens with glucocorticoid excess with hyperadrenalism?
 increased gluconeogenesis -> hyperglycemia, reduced sensitivity to insulin -> ‘adrenal diabetes’
 increased fat mobilization from lower body and redistribution to the thoracic and abdomen (buffalo torso); facial edema (moon face)
 protein loss (in muscle, skin, bone, collagen & lymphoid tissue, not in the liver) -> weakness, osteoporosis, striae on skin
 increased susceptibility to infections -> death
What happens with androgen excess with hyperadrenalism?
acne, hirsutism (excess growth of facial hair)
What is the cause of primary aldosteronism (Conn’s syndrome)?
Small, aldosterone-secreting tumor of the zona glomerulosa
What happens with mineralcorticoid excess with Conn’s syndrome?
 hypokalemia -> occasional periods of muscle paralysis
 modest hypervolemia -> hypertension
 Excess aldosterone + INC arterial pressure leads to feedback suppression of renin secretion therefore low plasma renin concentration
What is the cause of adrenogenital syndrome due to adrenocortical tumor?
Adrenocortical tumor secreting excessive quantities of androgens
What happens with androgen excess due to adrenogenital syndrome?
 Female – masculinisation (including beard growth, deepening of voice, body hair, growth of clitoris)
 Male –rapid development of male sexual organs in prepubertal males
How is adrenogential syndrome diagnosed?
levels of 17-ketosteroids (androgen metabolite) in the urine may be 10-15 times normal
What causes congenital adrenaly hyperplasia?
Autosomal recessive disorder – enzyme inefficiencies most commonly leading to deficient production of cortisol and aldosterone together with excessive production of sex steroids. 95% of cases due to P450c21 hydroxylase deficiency
What are the different types of P450c21 hydroxylase deficiencies?
-Severe P450c21 deficiency: Life-threatening vomiting & dehydration in first weeks of life. Ambigious genitalia of genetically female infants due to prenatal virilization.
-moderate P450c21 deficiency: prepubertal virilization
-Mild P450c21 deficiency: infertility in women due to anovulation
Describe the morphology of the thyroid gland.
thyroid gland is composed of follicles (100-300 microM diameter), filled with colloid (major constituent is thyroglobulin) and lined with cuboidal epithelial cells
What are the thyroid hormones?
-Tetraiodothyronine (Thyroxine; T4)-Comprises 93% of total secretion (~80 µg/day)
Most is converted to T3 in the tissues
-Triiodothyronine (T3)-Comprises 7% of total secretion (~4 µg/day) Similar function as thyroxine but 4 times more potent, Concentration in the plasma is much less than T4, Shorter half-life than thyroxine (1 day vs. 6 days)
What are the stages of the biosynthesis of thyroid hormones?
1. iodide trapping-transport of iodide into the thyroid follicles
2. formation and secretion of thyroglobulin
3. oxidation of iodide and coupling of iodine with thyroglobulin
4. endocytosis of thyroglobulin and release of thyroid hormones
5. recycling of iodine for reuse
Describe stage 1 of the biosynthesis of thyroid hormones.
Ingested iodine is absorbed through the small intestine and transported in the plasma to the thyroid.
-Iodide pumps (sodium iodide symporters) located on the basal membrane of the thyroid epithelial cell actively pump iodide ions into the cytosol.
-Concentration of iodide inside the thyroid cells can be 30-250 times higher than in the blood.
Describe stage 2 of the biosynthesis of thyroid hormones.
• Thyroglobulin, a large glycoprotein, is synthesized by the endoplasmic reticulum and Golgi apparatus of the thyroid cells.
• Thyroglobulin is then secreted into the follicular colloid.
• Each molecule of thyroglobulin contains ~70 tyrosine amino acids.
Describe stage 3 of the biosyntehsis of thyroid hormones.
• Iodide exits apical membrane and is oxidized by thyroperoxidase in the presence of hydrogen peroxide.
• Oxidized iodine binds with tyrosine amino acids within the thyroglobulin molecule (organification). Promoted by iodinase – within seconds to minutes 1/6th of the tyrosines become bound.
• Successive iodination and coupling of iodotyrosine residues results in the formation of T3 and T4 (see next slide).
Describe the steps in the formation of T3 and T4.
1. tyrosine -> monoiodotyrosine (MIT) via iodinase
2. MIT -> diiodotyrosine (DIT)
3. MIT + DIT -> 3,5,3 triiodotyrosine (T3)
4. DIT + DIT -> thyroxine (T4)
Describe stage 4 of the biosynthesis of thyroid hormones.
• After coupling, each thyroglobulin contains up to 30 T4 molecules and a few T3 molecules.
• Thyroglobulin is isolated from the EC fluid therefore must renter the follicular cell prior to secretion. Uptake is via endocytosis by pseudopodia.
• Lysosomes in the cell cytoplasm fuse with pinocytotic vesicles. Proteases in lysosomes mix with colloid. T3 and T4 released and diffuse through basal membrane into bloodstream.
Describe stage 5 of the biosynthesis of thyroid hormones.
• ~75% of the iodinated tyrosine in thyroglobulin never becomes T3 or T4, but remains as monoiodotyrosine (MIT) and diiodotyrosine (DIT).
• When thyroglobulin is digested MIT and DIT are freed into the cytosol of the thyroid cell.
• Iodine is cleaved from MIT and DIT by tyrosine deiodinase and recycled.
Describe the use of radioiodide to assess thyroid function.
pics compare uptake of radioactive iodide by normal thyroid gland and a cold nodule that does not take up tracer (is indicative of compromised thyroid function), radioactive iodide can also accumulated in follicular lumen, the urinary excretion of radioiodide can also be used as a test of thyroid function
Describe the storage of thyroid hormones.
-The thyroid gland can store enough thyroid hormone to supply the body with its normal
requirements for 2-3 months.
-Thyroid hormones remain part of the thyroglobulin molecule during storage in the follicular colloid.
-Thyroid hormones are also ‘stored’ in blood and target tissues bound with plasma and intracellular proteins and are used slowly over days/weeks.
Describe the transport of thyroid hormones in the blood.
In the blood over 99% of T3 and T4 combine immediately with thyroxine-binding globulin (>70%), thyroxine-binding prealbumin and albumin - plasma proteins synthesized by the liver. Binding proteins are not saturated and store 3x the amount secreted per day, therefore:
-acute changes in hormone secretion have very little effect on free hormone concentrations thyroid hormones are not minute-to-minute regulators.
-changes in binding protein levels that occur in liver or kidney disease have significant effects on thyroid hormone concentrations.
The binding proteins’ affinity for T4 is 10 times higher than for T3 Therefore the half-life for T4 is longer (6 days) than for T3 (1 day)
Describe thyroid hormone metabolism
93% of thyroid hormone released by the thyroid gland is T4. However ~50% of the secreted T4 is deiodinated in extrathyroidal tissues to T3.
Describe the different types of deiodinase.
• Deiodinase type I: liver, kidney, CNS, ant. pituitary, thyroid, removal of 1 iodine from the outer ring à T3, removal of 1 iodine from the inner ring à reverse T3 (rT3): INACTIVE
• Deiodinase type II: brain, pituitary, T3 for local use.
• Deiodinase type III: in many tissues, Only removes iodine from the inner ring à degradation.
Describe the further degradation of rT3 and T3.
rT3 and T3 can be further deiodinated by type I and type III deiodinases à degradation Thyroid hormones can also be conjugated (to glucuronic acid) and excreted in the bile
Describe the cellular mechanism of thyroid hormone action.
Thyroid hormone receptors have a much higher affinity for T3 than T4. (More than 90% of the thyroid hormone molecules that bind with the receptors is T3), Thyroid hormone receptors (TRs) are nuclear receptors. TRs are located at thyroid response elements of genes. Upon activation, TRs initiate transcription and thus formation of new proteins.
Describe the cellular metabolic activity action of thyroid hormones.
-Thyroid hormones increase metabolic activity of almost all tissues. Basal metabolic rate can be increased 60-100% above normal if large quantities are secreted.
-INC number and SA of mito, INC ATP formation, INC Na+-K+ ATPase activity and INC protein synthesis leads to INC O2 consumption and INC in basal metabolic rate (BMR)
Describe the role of thyroid hormone in carb metabolism.
Increases glucose absorption, glucose uptake by cells and oxidation, glycolysis, gluconeogenesis, insulin secretion and glycogenolysis.
Describe the role of thyroid horomone in lipid metabolism
Enhances lipogenesis in liver and fat cells, mobilization of lipids from fat stores and oxidation of free fatty acids. Decreases plasma cholesterol by increasing hepatic cholesterol excretion into the bile (via ­LDL receptors in liver).
Describe the role of thyroid hormone in nitrogen metabolism.
Increased protein synthesis & degradation. In excess, protein degradation predominates.
What does the role of thyroid hormone in metabolism do to BMR?
Enhanced mobilization of carbohydrate, fat and protein increases INC O2 consumption therefore INC BMR
What role does thyroid hormone have on the skeletal system?
T3 is required for attainment of normal adult stature and bone maturation. T3 is permissive for growth hormone synthesis, secretion & action.
What role does thyroid hormone have on CNS?
Maturation of the central nervous system during the perinatal period is absolutely dependent on T3. Cerebral & cerebellar growth, myelinization, vascularization, axonal & dendritic density, cell migration & differentiation are all affected.
What amount of thyroid hormone is necessary for proper functioning of the body?
Appropriate amounts are required for the normal operation of virtually all organs. T3 acts as a modulator rather than an all-or-none signal.
What role does thyroid hormone have on the CV system?
INC heart rate (via INC b-adrenergic receptors); Increased metabolism causes rapid O2 consumption and production of metabolic end products and need for heat elimination. Result: vasodilation, INC blood flow, INC cardiac output.
What role does thyroid hormone have on temperature regulation?
T3 maintains sensitivity to, and acts in synergism with, sympathetic stimulation to regulate body temperature.
What role does thyroid hormone have on CNS?
Hyperthyroidism increases excitability and nervousness. Hypothyroidism causes lack of energy, slowness of speech, dulled mental capacity. Mechanisms are unclear.
What role does thyroid hormone have on ANS?
Thyroid hormones amplify the effects of sympathetic stimulation by increasing the number of b-adrenergic receptors, upregulating Galphas and downregulating Galphai.
What is the principal regulator of thyroid synthesis and release?
Thyroid Stimulating Hormone (TSH; thyrotropin) [Secreted by thyrotropes of anterior pituitary gland], TSH receptor couple to Galphas -> INC cAMP -> INC PKA
What are the genomic effects of TSH?
promotes gene transcription for :
1. Iodide pump, thyroglobulin, enzymes involved in T3 & T4 synthesis
2. Nitric oxide synthase ® vasodilation therefore INC blood flow
3. local growth factors ® hyperplasia and hypertrophy of gland
What are the non-genomic effects of TSH?
1. INC iodide pump activity ® INC iodide trapping
2. INC iodination of tyrosine ® INC synthesis of T3 & T4
3. INC proteolysis of thyroglobulin ® INC release of T3 & T4
What is the principal regulator of TSH secretion?
Thyrotropin Releasing Hormone (TRH),TRH release can be increased by certain emotional states (i.e. excitement & anxiety)
What are the actions of TRH?
INC gene transcription and glycosylation of TSH, INC release of TSH (via Gaq à INC IP3 & DAG). Stimulation by TRH is required for optimal TSH secretion.
Describe the negative feedback abilities of thyroid hormone.
it inhibits:
(1) TRH synthesis & secretion in the hypothalamus
(2) Gene expression for TSH and for the TRH receptor in the pituitary
(3) TSH release from the pituitary
Describe the causes of autoimmune hyperthyroidism.
AKA (Graves’ disease, Basedow’s disease, Parry’s disease, toxic goiter, thyrotoxicosis)
Cause: Autoantibodies stimulate TSH receptors producing prolonged stimulation of thyroid hormone synthesis. T4 and T3 high, TSH low.
Describe the signs and symptoms of autoimmune hyperthyroidism.
-Excitability, heat intolerance, sweating, weight loss, diarrhea, palpitations, nervousness, muscle weakness, fatigue, inability to sleep, tremor of the hands (largely due to increased sympathetic activity).
-Enlargement of thyroid gland (diffuse goiter) due to hypertrophy and hyperplasia. Colloid volume is greatly reduced (see left panel; right panel shows normal histology)
-Autoimmune reaction causes edematous swelling of retro-orbital tissues and degeneration of extraocular muscles ® exophthalmos (protrusion of the eyeballs), thyroid stare (inability to achieve or maintain convergence, limited superolateral or upward gaze), blurred or double vision, feeling of pressure behind the eyes. Can lead to irritation/ulceration of the cornea as eyelids do not close completely when blinking or asleep.
What is the treatment for autoimmune hyperthyroidism?
thyroid resection, competitive inhibitors of iodide trapping (thiocyanate, percholate and nitrate ions), inhibitors of iodide oxidation and organification (propylthiouracyl), glucocorticoids, b-blockers.
Describe thyroid adenoma.
autonomous hypersecretion of thyroid hormones -> low TSH -> secretory activity of the remainder of the thyroid is almost totally inhibited.
What are the causes of hypothyroidism?
• Autoimmune disease (Hashimoto’s disease): autoantibodies against thyroglobulin, thyroperoxidase, TSH receptor (blocking type) [low T4 and T3, elevated TSH levels]
• Dietary iodine deficiency
• Inherited defects of hormone synthesis
• Defects in TSH and TSH receptor
• Antithyroid substances (e.g. p-aminosalicylic acid, lithium)
• Iatrogenic (treatments for hyperthyroidism)
What are the signs and symptoms of hypothyroidism?
fatigue, muscular sluggishness, slow heart rate, reduced cardiac output, weight gain, constipation, mental sluggishness, myxedema (more generalized than in hyperthyroidism), intolerance to cold, thick tongue, hoarseness. Goiter (often nodular due to TSH stimulation of healthy areas) Hypercholesterolemia due to reduced excretion by liver.
What is the treatment for hypothyroidism?
daily oral thyroxine or iodide replacement.
What causes cretinism?
Extreme hypothyroidism during fetal life and infancy due to congenital lack of thyroid gland or dietary iodine insufficiency
What are the signs and symptoms of cretinism?
The neonate may have normal appearance and function (maternal thyroid hormones). Symptoms appear a few weeks after birth: sluggish movements, retarded physical and mental development. Unless treatment begins immediately, mental growth remains permanently and severely retarded (CRITICAL TIME WINDOW). Skeletal growth is more inhibited than soft tissue growth leading to a stocky, short appearance; the enlarged tongue may even choke the child. Physical growth can be rescued by treatment initiated at any time during childhood.
What is the treatment for cretinism?
daily oral thyroxine or iodide replacement.
Define growth.
organized addition of new tissue that occurs normally in development. With the exception of longitudinal growth, many of the processes involved continue to operate throughout life (e.g. tissue remodeling). Growth involves genetic, nutritional and environmental factors as well as actions of the endocrine system.
Describe growth hormone.
-Other names: Somatotropic hormone or somatotropin
-Nature: Polypeptide
-Secreted from: somatotropes of anterior pituitary
-Mode of secretion: irregular pulses. Largest burst during deep sleep. Pulse amplitude max. at puberty and declines thereafter.
-Binding protein: Only binds weakly to plasma proteins therefore relatively short half-life of 20 min.
-Receptor: GH binding to enzyme-linked receptor produces activation of kinase cascade which ultimately alters gene transcription.
-Metabolism: reabsorbed & destroyed by the kidney; also degraded by target cells.
-Excretion: minimal amounts in urine.
What affect does GH have on body organs?
Unlike other anterior pituitary hormones, Growth hormone does not function through a target gland – it exerts its effects directly on almost all tissues of the body. Increases size and number of cells and causes specific differentiation of certain cell types.
What effect does GH have on skeletal growth?
(1) INC proliferation of epiphyseal cartilage.
(2) INC conversion of cartilage to new bone (INC length of long bones & skeleton - until epiphyses of long bones fuse with shafts)
(3) INC proliferation of periosteal osteoblasts causing bone thickening
(4) INC bone remodeling
What effect does GH have on metabolism?
INC protein synthesis, DEC protein breakdown [DEC BUN], INC fat utilization, DEC glucose uptake & utilization, INC glucose production by liver
Describe somatomedin hypothesis.
‘Somatomedin hypothesis’: Growth effects of GH are mediated by Insulin-like Growth Factors (IGFs). Somatomedin C (IGF-I) is the most important IGF (Pygmies and Laron dwarfs lack IGF-1).
Describe the production of somatomedin.
Produced in the liver and also at local site of action (i.e. bone and tissues). Stimulate chondrocytes & osteoclasts – INC bone elongation, In the blood IGFs bind strongly to binding proteins (IGFBPs) which are produced in response to GH, IGF-1 & insulin. IGF-I released slowly to the tissues therefore half-life ~20 h. Prolongs actions of GH.
What stimulates the secretion of GH?
(1) Growth hormone releasing hormone (GHRH)
(2) starvation, protein deficiency
(3) DEC blood glucose
(4) DEC blood fatty acids
(5) Exercise
(6) Excitement
(7) Trauma
(8) Estrogen & androgens
(9) Sleep
What inhibits secretion of GH?
(1) Somatostatin
(2) obesity
(3) INC blood glucose
(4) INC blood fatty acids
(5) Aging
(6) Somatomedins (IGFs)
(7) Growth hormone (-ve feedback)
What role does growth hormone releasing hormone (GHRH) have on regulation of GH secretion?
released by the arcuate nucleus, INC GH synthesis and secretion
What role does somatostatin (SST) have on GH secretion?
AKA GH inhibitory hormone, released by the peri- and para- ventricular nucleus, has no affect on GH synthesis, but DEC GH secretion
What role does GH-releasing peptide (ghrelin) have on GH secretion?
secreted by the arcuate nucleus, INC GH and GHRH synthesis and secretion, INC GHRH actions
What is the cause, effect and treatment for dwarfism?
-Cause: Congenital defect, pituitary tumor or trauma causing panhypopituitarism (DEC secretion of all anterior pituitary hormones) or isolated GH (or IGF-I) deficiency.
-Effect: Slow and reduced growth (to ~4 ft). Juvenile appearance (small maxilla & mandible). If due to panhypopituitarism will also have hypothyroidism, hypogonadism and low glucocorticoids.
-Treatment: human GH +/- thyroxine, cortisol
What is the cause, effect and treatment for panhypopituitarism in the adult?
-Cause: pituitary tumors, thrombosis of pituitary blood vessels, trauma.
-Effect: lethargy (hypothyroidism), weight gain (DEC fat mobilization due to lack of adrenocortical and thyroid hormones) and loss of sexual function (DEC gonadotropic hormones).
-Treatment: thyroxine, cortisol
What is the cause, effect and treatment for gigantism (GH hypersecretion during childhood)?
-Cause: hyperactivity of otherwise normal cells, pituitary tumor.
-Effect: Rapid growth of all tissues (up to 8 ft tall). Hyperglycemia (leading to diabetes mellitus in 10%).
-Treatment: somatostatin analogs, surgery
What is the cause, effect and treatment for acromegaly (GH hypersecretion in the adult)?
-Cause: pituitary tumor after adolescence.
-Effect: Bones grow in thickness (not length): cranium, nose, supraorbital ridges, mandible, vertebrae (kyphosis). Enlargement of hands and feet, elongation of ribs (barrel-chested appearance). Enlarged tongue, liver, kidneys, heart. Diabetes.
-Treatment: somatostatin analogs, surgery
-In general, excessive growth hormone can cause ketosis & insulin resistance due to metabolic disturbances
What effect does thyroid hormone have on GH?
Permissive for GH synthesis, release (via INC GHRH receptor) & actions (INC GH receptor, INC IGF-I, INC IGF-I receptor).
What effect does insulin have on GH?
Required for optimal effects of growth hormone (IGF-I is low in diabetes). Growth hormone fails to cause growth in an animal that lacks a pancreas or if carbohydrate is excluded from the diet – lack of energy source and attenuated transport of amino acids into cells.
What effect does gonadal hormones have on GH?
Estrogens produce pubertal growth spurt by increasing GH pulse amplitude & frequency; termination of growth (epiphyseal closure) IN BOTH SEXES. Androgens increase muscle mass.
What effect does glucocorticoids have on GH?
Permissive for optimal growth: INC GHRH receptor & GH synthesis. High concentrations inhibit growth (e.g. pharmacological doses to treat asthma & inflammations).
What is the nature and action of epidermal growth factor?
1. nature-polypeptide (produced by many normal cell types and by some tumors)
2. action-growth, proliferation and differentiation of mesenchymal and epithelial cells
What is the nature and action of nerve growth factor?
1. nature-polypeptide (secreted by neuron’s target)
2. action-growth and differentiation of sympathetic and sensory neurons
What is the nature and action of fibroblast growth factor?
1. nature-polypeptide (heparin-binding proteins)
2. action-wound healing angiogenesis, limb formation
what is the nature and action of platelet-derived growth factor?
1. nature-polypeptide (carried in the alpha-granulesj of platelets)
2. action-initiates replication of CT cells in response to trauma
Describe the morphology of the pancreas.
Richly vascularized and innervated by sympathetic and parasympathetic fibers.
Composed of: (1) acini: secrete digestive juices into duodenum and (2) Islets of Langerhans: secrete insulin and glucagon into blood
What doe the Islets of Langerhans contain?
Alpha (a) cells (25%): secrete glucagon, periphery
Beta (b) cells (60%): secrete insulin and amylin, center
Delta (d) cells (10%): secrete somatostatin, between alpha and beta
PP cells (<5%): secrete pancreatic polypeptide
What role do secreted hormones have on feedback?
Secreted hormones can ‘feedback’ on adjacent cells, directly regulating secretion. (e.g. insulin: DEC glucagon, amylin: DEC insulin, somatostatin: DEC insulin & glucagon)
Describe glucagon.
-Origin: alpha (a) cells of the islets of Langerhans
-Nature: simple, large polypeptide (29 amino acids)
-Synthesis: preproglucagonà proglucagonà glucagon
-Carrier protein: none
-Half-life: 5 min
-Targets: liver, β cells (?)
-Sites of degradation: liver, kidney, plasma
How does glucagon INC blood glucose concentration?
glucagon stimulates glycogenolysis, gluconeogenesis
Describe glycogenolysis.
(breakdown of liver glycogen to glucose), Mechanism of action: activates adenylyl cyclase ® cAMP formation ® protein kinase activation ® activates phosphorylase b kinase ® converts phosphorylase b into phosphorylase a ® INC degradation of glycogen to glucose-1-phosphate ® dephosphorylated & glucose released
Describe gluconeogenesis.
(formation of glucose from non-carbs) Mechanism of action: cAMP/PKA second messenger system activates enzymes required for gluconeogenesis (secs to mins). Also enhances gene transcription of enzymes involved in gluconeogenesis (mins to hrs)
What are some other actions of glucagon?
[When the concentration of glucagon increases above normal], Mobilization of fatty acids from adipose tissue by activating adipose cell lipase (INC lipolysis), Decreases uptake and storage of triglycerides by the liver
What is the most potent factor which controls glucagon secretion?
glucose concentration
What stimulates alpha cells to release glucagon?
amino acids, acetylcholine, epinephrine, NE, VIP, CCK
What inhibits alpha cells to release glucagon?
glucose (via feedback signals), insulin, somatostatin, ketones (via feedback signals), FFA (via feedback signals)
Describe insulin.
-Origin: b cells of the pancreatic islets
-Nature: small, 2-chain protein
-Synthesis & processing: preproinsulin ® proinsulin ® insulin + C-peptide. C-peptide and proinsulin are also secreted but are inactive.
-Carrier protein: none
-Half-life: ~6 min
-Degradation: by insulinase (in liver, kidney, muscle)
-Targets: ~80% of the body’s cells (most brain cells are NOT targets)
What is the cellular mechanism of action for insulin?
1. Insulin receptor is an enzyme-linked receptor.
2. Insulin binds with a-subunits of insulin receptor.
3. b-subunits of receptor become auto-phosphorylated.
4. Activation of tyrosine kinase causes phosphorylation of many intracellular enzymes (including Insulin Receptor Substrates (IRS), PI 3-kinase, Ser/Thr kinases) which are either activated or inactivated producing the cellular response.
What are the effects of insulin stimulation and time course of responses?
1. within seconds-Increases translocation of glucose transporter proteins (e.g.GLUT4) to cell membranes, facilitating glucose uptake. (This occurs in ~80 % of the body’s cells, especially muscle and adipose, but NOT neurons in the brain), Increases uptake of amino acids, PO43- and K+ into cells (by INC Na+/K+ ATPase activity).
2. 10-15 mins-Phosphorylation of intracellular metabolic enzymes, altering their activity (incl. INC PDE leading to DEC cAMP).
3. hours-days-Alters rates of DNA transcription and mRNA translation therefore changing gene expression & protein synthesis
Describe the effects of insulin on muscle.
Resting muscle membrane is only slightly permeable to glucose (fatty acids used as energy source). Glucose usage by muscles increases with exercise (muscle contraction increases permeability to glucose) and stimulation by insulin.
What does insulin stimulation cause?
• Rapid increase in glucose uptake.
• Storage of excess glucose as glycogen.
• Increased glucose metabolism due to decreased release of fatty acids from adipose and decreased utilization of fatty acids by muscle.
• Increased protein storage by increasing amino acid uptake and protein synthesis while decreasing protein degradation (conserves protein stores + acts in synergism with growth hormone)
What are the effects of insulin on the liver?
-Increases glucose uptake (by INC glucokinase activity; phosphorylated glucose cannot diffuse back through the cell membrane) Note: Glucose transporter (GLUT-2) is NOT regulated by insulin in the liver!
-Increases glycogenesis and reduces glycogenolysis. Insulin promotes conversion of excess glucose into glycogen by INC glycogen synthase activity. Breakdown of glycogen inhibited by inactivating liver phosphorylase.
-Inhibits gluconeogenesis by decreasing activity of enzymes (plus less amino acids available as release from muscle decreased)
-Stimulates conversion of excess glucose* into fatty acids. (*glucose not used for metabolism or storage as glycogen).
What are the effects of insulin on adipose?
-Increases uptake of fatty acids from dietary fat & triglycerides released by the liver. It does this by INC lipoprotein lipase in capillaries of adipose tissue. (DEC circulating lipoproteins).
-Increases glucose uptake (via GLUT4) into fat cells – Glucose used to make glycerol which combines with fatty acids to form triglycerides (storage form of fat in adipose).
-Inhibits breakdown of triglycerides already stored in adipose. It does this by DEC hormone-sensitive lipase. (DEC circulating lipoproteins).
-net effect-preservation of fat stores
What other effects does insulin have on adipose (not direct on adipose but has a net effect is to preserve fat stores)?
-Increases glucose uptake and metabolism in most of the body’s tissues (saving fat stores).
-Increases uptake of glucose into liver cells leading to INC fatty acid synthesis. [When liver glucagon concentration reaches ~6% all further glucose used to make fat]. Glucose ® pyruvate ® acetyl-CoA ® fatty acids.
-Increased glucose metabolism leads to excess citrate ions which INC acetyl-CoA carboxylase – INC fatty acid synthesis
What stimulates the release of insulin from beta cells?
1. those that do not stimulate secretion much if glucose is low-glucose, amino acids, fatty acids, ketones, acetyl choline, GI hormones, glucagon
2. excessive amounts can cause insulin resistance + secondary diabetes-GH and cortisol
3. insulin can act back and inhibit glucose, amino acids, fatty acids and ketones
What inhibits the release of insulin form beta cells?
somatostatin, epinephrine, NE
Describe the mechanism for the stimulation of insulin secretion by glucose.
Glucose entry à oxidative metabolism à INC ATP/ADP à inhibition of KATP channels à depolarization à opening of voltage-sensitive Ca2+ channels à INC [Ca2+]i à insulin secretion
Describe the regulation of insulin secretion by glucose.
Insulin levels are low at fasting blood glucose levels and increase substantially in response to hyperglycemia to restore normal blood glucose concentration.
What are the phases of insulin secretion?
1. rapid dumping of preformed insulin
2. release of preformed + freshly synthesized insulin: glucose à INC insulin gene transcription, INC processing of proinsulin
Describe somatostatin as a regulator of glucagon and insulin secretion.
-Origin: delta (d) cells of the islets of Langerhans (& hypothalamus [role of growth hormone inhibitory hormone])
-Nature: polypeptide (14 amino acids)
-Half-life: 3 min
-Targets: pancreatic a and b cells, gallbladder, GI tract (& anterior pituitary [inhibits growth hormone secretion])
What are the actions of somatostatin?
-Action: Prevents rapid exhaustion of food substrates by:
• Decreasing insulin and glucagon secretion from the pancreas
• Decreasing motility of the stomach, duodenum and gallbladder
• Decreasing secretion and absorption in the gastrointestinal tract
Describe amylin.
-Origin: beta (b) cells of the islets of Langerhans
-Nature: polypeptide
-Half-life: ~6 min
-Targets: Autonomic brain centers incl. dorsal vagal nuclei
What does amylin do?
Action: Reduces glucose influx by inhibiting food intake, gastric emptying, and glucagon secretion
What is diabetes mellitus (DM)?
-a syndrome of impaired metabolism caused by either: lack of insulin secretion (Type I) or insulin resistance AND impaired insulin secretion (Type II).
-Note: Classification based on insulin dependence (Insulin-Dependent DM vs. Non-Insulin-Dependent DM) is now less common.
-(Type I is always IDDM; Type II is usually NIDDM but can be IDDM.)
Describe the epidemiology of DM.
~5.1% of US population (~15 million). 800,000 new cases/year (~90% type II). Type II-like or type II-like DM may also develop as a consequence of other conditions (e.g. fibrosis, alcohol consumption, genetic & endocrine disease, pregnancy, etc.).
What are the principal metabolic changes that occur during DM?
(1) increased blood glucose concentration, (2) decreased utilization of glucose, (3) increased utilization of fat and proteins.
In diabetes, what happens in metabolic disturbances due to DM?
-DEC glucose uptake and DEC glucose utilization
-INC hepatic glucose production (even during hyperglycemia)
-INC fat utilization (oxidation of free fatty acids can lead to ketonemia in type I DM)
-INC release of free fatty acids from adipose and DEC lipogenesis in adipose
What is the etiology of type I DM?
injury to β cells causing impaired insulin secretion; due to autoimmune disorder or viral infection. Genetic tendency for susceptibility to b cell destruction from these insults or for b cell degeneration. Usual age of onset ~14 years.
What is the pathophysioloogy of Type I DM?
1. DEC glucose utilization and production
2. INC fat utilization
3. protein depletion
Describe #1 on the pathophysiology of type I DM.
• Hyperglycemia 300–1200 mg/dL
• Glycosuria when blood glucose >180 mg/dL
• Polyuria due to osmotic diuresis à thirst (polydipsia)
• Dehydration due to INCosmotic pressure in extracellular fluid (H2O leaves cells) + osmotic diuresis; in extreme cases: circulatory failure à hypotension à decreased renal blood flow à coma, death.
• Tissue damage (long-term): high glucose concentrations + metabolic abnormalities lead to cellular disruption à vasculopathies, neuropathies, retinopathy, nephropathy.
Describe #2 on the pathophysiology of type I DM.
-Ketonemia due to formation of keto acids at a speed that exceeds their utilization à ketonuria, acetone breath
-Metabolic acidosis due to ketonemia and dehydration à hyperventillation (rapid & deep breathing): compensation through CO2 loss but depletion of bicarbonate reserves à coma, death
-Hypercholesterolemia (due to excess fat utilization; long term) à arteriosclerosis
Describe #3 on the pathophysiology of type I DM.
-Aminoacidemia due to increased protein catabolism in muscle à glucose production by gluconeogenesis
-Asthenia (Lack of energy), weight loss (loss of protein & fat) despite increased appetite and polyphagia à severe wasting
-Note similarities to starvation.
What is the etiology of Type II DM?
impaired β cell function AND insulin resistance. The ultimate causes are unknown but genetic factors probably play a role. Obesity, decreased activity and aging contribute to the insulin resistance. Usual age of onset 50-60 years.
What is the traditional view for the pathogenesis of type II DM?
insulin resistance in muscle and adipose tissue à compensatory INC hepatic glucose production to provide fuel + compensatory INC insulin production to overcome insulin resistance. DM manifests when increased insulin secretion can no longer compensate for the increased glucose output.
What does recent studies suggest for the pathogenesis of type II DM?
indicate that impaired glucose-induced insulin secretion (mitochondrial defect) may be the primary metabolic abnormality. Once acquired, insulin resistance further worsens condition.
What is a third idea for the pathogenesis of type II DM?
insulin resistance in liver à INC hepatic glucose production plus insulin resistance in β cells à insufficient INC insulin secretion in response to high BG
What is the pathophysiology of type II DM?
1. DEC insulin sensitivity to insulin
2. DEC meal-induced insulin secretion
Describe #1 pathophysiology of type II DM?
• Insulin resistance in liver & muscle (DEC translocation of GLUT4) – however, contraction and IGF-I signaling in muscle can stimulate GLUT4 translocation
• Insulin resistance in adipose tissue may INC insulin resistance in other tissues via the release of free fatty acids and the hormones resistin and adiponectin
• Insulin resistance in b cells (prevents compensatory increase in β cell mass)
• result-DEC glucose utilization and storage, hyperglycemia and compensatory hyperinsulinemia
Describe #2 pathophysiology of type II DM.
• Hepatic glucose production doesn’t stop promptly in response to elevated blood glucose
• RESULT: postprandial hyperglycemia and compensatory hyperinsulinemia
Describe glucotoxicity.
(1) Chronic hyperglycemia may further desensitize β cells to glucose stimulation.
(2) Chronic hyperglycemia may block GLUT4 mobilization and worsen insulin resistance.
Describe lipotoxicity.
(1) High levels of circulating free fatty acids can inhibit insulin secretion and insulin signaling.
(2) Excess fatty acids inhibit glucose utilization and may stimulate gluconeogenesis in liver.
Describe the roles of genetic factors in DM.
Studies suggest that genetic factors may determine whether an individuals pancreas can sustain the high output of insulin that is necessary to avoid clinically significant diabetes mellitus
Describe the clinical characteristics of type I DM.
1. usual age at onset-<20 years
2. onset-rapid
3. body mass-low (wasted)
4. plasma insulin-low or absent
5. plasma glucagon-high, can be suppressed
6. plasma glucose-INC
7. insulin sensitivity-normal
8. therapy-insulin
Describe the clinical characteristics of type II DM.
1. usual age at onset->40 years
2. onset-slow
3. body mass-obese
4. plasma insulin-variable
5. plasma glucagon-high, resistant to suppression
6. plasma glucose-INC
7. insuline sensitivity-reduced
8. therapy-weight loss, drugs, insulin
Describe micro- and macrovascular complications.
INC risk for heart attack, stroke, nephropathy and end-stage kidney disease, retinopathy and blindness, ischemia and gangrene of limbs, Hypertension (secondary to renal injury) Atherosclerosis (due to abnormal lipid metabolism)
Describe peripheral neuropathies and ANS dysfunction.
- impaired cardiovascular reflexes
- impaired bladder control
- distal sensory neuropathy (reduced sensation in extremities)
- gastroenteropathy (gastroparesis, constipation with episodes of diarrhea)
-onset-1-25 years after diagnosis, prevalence-up to 50-60% in type I, less common in type II
Describe the complications of DM.
tubby triglyceride, hungry muscle, impaired kidney, upset stomach, depressed HDL, hyperactive liver, burned-out pancreas
How is diabetes diagnosed?
1. Urinary glucose: normal person glucose undetectable; diabetic loses glucose in small to large amounts
2. Fasting blood glucose: normal fasting blood glucose 80-100 mg/dL; values above this indicative of DM.
3. Fasting insulin levels: Type I – insulin levels very low or undetectable; Type II – severalfold higher than normal
4. Glucose tolerance test: assesses time course of blood glucose after ingestion of 1g glucose per kg body weight. Diabetics show greater rise in BG and it takes much longer to return to control levels
5. Acetone breath: due to conversion of keto acids to acetone – can be diagnostic of type I and severe type II DM
What is the therapy for DM?
1. Exercise and dieting: in an attempt to induce weight loss and improve insulin sensitivity.
2. Antidiabetic drugs: Metformin inhibits gluconeogenesis, Thiazolidinediones improve insulin sensitivity, Sulfonylureas (KATP channel blockers) stimulate insulin secretion
3. Insulin: Human insulin produced by recombinanant DNA process. Precipitated with zinc or protein derivatives to extend duration of action to 10-48 hrs.
However, current forms of treatment do not mimic physiological secretion and can’t fully prevent chronic complications.
4. Emerging therapies: aim at strict glycemic control, orally active analogs, pancreas/islet transplantation or the transplantation of β cells derived from adult or embryonic stem cells.
What causes hyperinsulinism?
Excessive levels of insulin due to adenoma of an islet of Langerhans (Insulinoma;10-15% malignant; metastases can also secrete insulin) OR insulin overdose in diabetic patient
What are the symptoms and signs of hyperinsulinism?
‘Insulin shock’ & hypoglycemia
Brain cells almost exclusively use glucose for energy therefore symptoms reflect CNS dysfunction:
50-70 mg/dL: hunger, extreme nervousness, trembling, hallucinations, cold sweat
20-50 mg/dL: seizures, loss of consciousness
<20 mg/dL: coma (unlike hyperglycemic coma: no acetone breath, no hyperventillation), permanent brain damage, death.
What is the treatment for hyperinsulinism?
Immediate IV of large quantities of glucose. Administration of glucagon or epinephrine (INC glycogenolysis in the liver). Permanent damage to the CNS can result if blood glucose levels are not restored rapidly.
-----------------------------------------------------------------------------------------------------------------
Lecture 12 Endocrine hormones and metabolism
-----------------------------------------------------------------------------------------------------------------
Lecture 12 Endocrine hormones and metabolism
Describe homeostasis, metabolic substrates and their stores.
-Availability of metabolic fuels must be constant in spite of intermittent food intake
-Homeostasis requires storage of fuels whenever intake is possible and their mobilization in need
-Homeostatic regulation is provided by the endocrine system and the autonomic nervous system
Describe glucose and glycogen and their storage.
Glucose-all cells can oxidize it, small molecule à only small amounts can be stored total EC glucose = 20 g= energy for 1 hour
Glycogen-polymer form of glucose,large molecule à 300g can be stored= energy for <1 day
Describe protein storage.
-More concentrated than glycogen
-By-products are “expensive” to remove (urea, sulfur-containing amino acids)
-Practically no inert depot à utilization leads to functional deficits -> death (respiratory muscle failure); max. 50% (5-6 kg) can be used
Describe fat storage.
-Triglycerides can be stored in an almost limitless capacity in adipose tissue
- The most concentrated energy store: 250g = energy for 1 day. Normal stores are sufficient for 30-40 days of starvation
-Energetically expensive to build; only the glycerol portion can be converted to glucose
-Limited solubility in water -> transport requires proteins or conversion into small molecules (ketone bodies)
Describe transport of fat to brain.
-Protein-bound lipids cannot enter the BRAIN. Only after 4-5 days of total starvation does the concentration of ketone bodies become high enough that the brain starts to use them – but can only cover 2/3 of its needs. The rest of the brain’s energy must come from blood glucose (no glycogen, no gluconeogenesis).
- Cannot be utilized anaerobically
Describe the metabolic action of catecholamines (in seconds).
Mobilization of fuels in response to, or in preparation for, “fright, flight or fight”
• INC glycogenolysis & gluconeogenesis à hyperglycemia
• INC lipolysis à INC free fatty acids
• INC lactate and pyruvate production in muscle
• INC glucagon and DEC insulin secretion
Describe the metabolic actions of glucocorticoids. (long term actions: hours).
Preservation of carbohydrate reserves and mobilization of alternative fuels
• DEC glucose utilization, INC gluconeogenesis, DEC sensitivity to insulin à hyperglycemia (long term stimulation can lead to diabetes)
• INC lipolysis and fatty acid utilization
• INC protein degradation and amino acid release
Describe the metabolic actions of GH (hours).
-Shift from utilization of glucose to free fatty acids without increased protein breakdown
-DEC glycogenolysis (muscle)
-INC release of free fatty acids from adipose tissue
-INC protein synthesis & DEC degradation.
-DEC insulin sensitivity à hyperinsulinemia à diabetes
Describe the metabolic actions of thyroid hormone (in days)
-Overall increase in metabolism
-INC glucose absorption, oxidation, gluconeogenesis, glycogenolysis
¬ -INC lipogenesis & lipolysis
¬ -INC protein synthesis & degradation.
-(In excess, lipolysis and protein degradation predominate)
Describe the integration of energy balance.
substrate fluxes are regulated by hormones
-fuel storage-insulin
-fuel mobilization-glucagon, catecholamines, glucocorticoids, GH, thyroid hormone
What are normal levels of blood glucose?
Normal fasting BG = 80-90 mg/100ml
Normal postpriandial = 120-140/100ml
Why is tight control of blood glucose so important? What happens if too low?
-Brain cells almost exclusively use glucose for energy therefore symptoms of hypoglycemia reflect CNS dysfunction (see notes on hyperinsulinism)
-Glucose is also the only nutrient used by the retina, germinal epithelium and red blood cells.
-Hypoglycemia can be due to insulin overdose, benign or malignant adenoma of the Langerhans islets (insulinoma), Addison’s disease, hypopituitarism, severe liver disease.
Why is tight control of blood glucose so important? What happens if too high?
Complications of hyperglycemia (see notes on diabetes; effects of cellular dehydration and damage)
Describe hyperglycemia.
INC Insulin
• Acts rapidly to inhibit glucose output and to replenish glycogen stores in the liver.
• Stimulates glucose uptake and glycogen synthesis in other tissues.
• Stimulates glucose utilization in liver and other tissues (slower action).
Describe hypoglycemia.
INC Glucagon, INC Catecholamines
• Help to restore blood glucose levels via glycogenolysis and gluconeogenesis
• Catecholamines decrease demand for glucose by stimulating utilization of alternative fuels in muscle and adipose
• Catecholamines also inhibit insulin and stimulate glucagon secretion
What happens during sustained hypoglycemia (hours)?
In addition to glucagon and catecholamines, glucocorticoids and growth hormone become important, ensuring that:
• The peripheral drain on glucose reserves is minimized by decreasing glucose consumption by muscle and replacing with alternative fuels (free fatty acids, ketones)
• The liver contains enough glycogen to satisfy minute-to-minute needs of glucose-dependent cells by stimulating gluconeogenesis
Describe the integrative hormonal response to hypoglycemia.
This integrative response is launched to ensure blood glucose levels are high enough to satisfy the needs of glucose-dependent cells.
-INC in plasma epinephrine, NE, glucagon, cortisonl and plasma growth hormone
Describe the integrative control of blood glucose concentration.
1. glucose production-stimulated by epinephrine, NE, glucagon and glucocorticoids, inhibited by insulin, occurs in liver
2. glucose consumption-stimulated by insulin, inhibited by glucocorticoids, GH, occurs in muscle and adipose tissue
Describe hormone secretion during postpandial period.
-Increased secretion of insulin, sympathoadrenal activity is low
-Insulin dominates:
• glucose, amino acids, fatty acids are transferred to stores
• dietary glucose is used as fuel
• glucose & fatty acid mobilization from stores is inhibited
Describe hormone secretion during postabsorptive period (3 to < 24 h after meal; BG ~90)
-Insulin, growth hormone and cortisol at low basal levels; glucagon is relatively low
Glucagon dominates:
• Glycogen and gluconeogenesis provide source of blood glucose for glucose-dependent cells (brain, RBCs)
• Increased utilization of free fatty acids by muscle
Describe hormone secretion during fastring for 1-3 days (BG ~70-80)
DEC insulin, INC glucagon, INC GH, cortisol levels unchanged but diurnal pattern of release plays a permissive role in reducing glucose utilization
Conversion of T4 to T3 is attenuated to reduce BMR
• glucose is only used by the brain & blood cells
• glucose is from gluconeogenesis (muscle protein, glycerol from TG)
• free fatty acids provide primary fuel in muscle, kidney, etc.
Describe hormone secretion during fasting for >3 days.
As above, however now exaggerated oscillations in GH but no longer stimulates IGF-I à anabolic effect inhibited
• diminished glucose utilization
• mobilization and utilization of fat continues to increase producing ketones (ketosis): fuel for brain
• gluconeogenesis -> INC NH4+ excretion
Describe hormone secretion during terminal starvation.
When fat stores become depleted, protein stores are consumed rapidly for energy (several hundred grams per day, instead of the normal 30-50g per day)
• Increased gluconeogenesis from protein until respiratory muscle failure à death
Describe hormone secretion during fasting.
glucose DEC, insulin DEC, glucagon INC, cortisol, stays pretty constant, GH INC, T3 DEC
Describe hormone secretion during short-term maximal effort (100-yd dash) exercise.
Energy source for muscle activity: creatine phosphate; glycogen (producing lactate). Reinforced by INC sympathoadrenal activity
Role of the endocrine system: regeneration of stores after exercise: dietary intake -> INC insulin -> replenishment of glycogen stores
Describe hormone secretion during sustained (moderate) aerobic exercise.
-Fuel: 1-2 h: glucose from blood glucose + liver and muscle glycogen, >3 h: increased use of fats to a maximum of 70% total energy consumed
-Hormones: INC catecholamines -> DEC insulin; INC glucagon; INC cortisol; INC GH
à glycogenolysis, gluconeogenesis, mobilization of fat
-Increased glucose uptake by working muscles - independent of insulin
Describe hormone secretion during sustained exercise.
insulin DEC, INC in epinephrine, NE, glucagon, cortisol, GH
Describe appetite control via hypothalamic feeding and satiety centers.
-Lateral Hypothalamic Area (LHA) ‘hunger center’-Stimulation à hyperphagia; Lesion -> hypophagia, Melanin-Concentrating Hormone: INC food intake, Orexins: arousal, INC food-seeking behavior à INC food intake
-Ventromedial nucleus (VMN) ‘satiety center’-Stimulation à satiety, aphagia; Lesion -> hyperphagia, obesity, Brain-derived neurotrophic factor: DEC food intake
-Paraventricular Nucleus (PVN)-Integrates signals from arcuate nucleus, brain stem and periphery Initiates changes in endocrine systems (via CRH & TRH)
-Dorsomedial nucleus (DMN)-Lesions of this area normally depress eating behavior
What role does the arcuate nucleus have on appetite control?
-The principal integration site where signals from the gastrointestinal tract and adipose tissue converge to regulate appetite and energy expenditure. It is accessible to peripheral signals (no blood-brain barrier)
-Neurotransmitters and hormones acting on the hypothalamic feeding and satiety centers can be classified as being either:(1) Anorexigenic (inhibit feeding) or (2) Orexigenic (stimulate feeding)
What is the anorexigenic circuit?
nueonrs of the arcuate nuclei releasing a-MSH and CART
What does alpha-melanocyte-stimulating hormone (a-MSH) do?
• Derived from proopiomelanocortin (POMC)
• act on melanocortin receptors on neurons of PVN
• Release increased by feeding, reduced by fasting
• Activation of MCR-3/4 receptors à DEC food intake and INC energy expenditure by INC sympathetic activity
• Mutations or lack of MCR-4 cause over-eating (hyperphagia) and obesity; excessive activation has been linked to anorexia
What does cocaine- and amphetamine-regulated transcript (CART) do?
• Co-localized with a-MSH
• Release reduced by fasting
• Inhibits food intake
what is the orexigenic circuit?
neurons of the arcuate nuclei releasing neuropeptide Y and agouti-related peptide, Both NPY & AGRP stimulate feeding by activating neurons of the PVN, by inhibiting a-MSH neurons, and by antagonizing effects of a-MSH
Describe neuropeptide Y (NPY).
• Release increased by fasting, reduced after feeding
• Stimulation increases appetite à hyperphagia, obesity, DEC energy expenditure and DEC heat production (DEC sympathetic activity)
Describe agouti-related peptide (AGRP).
• 90% co-localized with NPY
• competitive inhibitor of a-MSH
• Increased in fasting, reduced after feeding
• INC food intake, DEC energy expenditure, DEC heat production
• Gene mutations causing excess AGRP à hyperphagia & obesity
Describe the role of the brain stem on appetite control.
• The actual mechanics of feeding are controlled by the brain stem
• The nucleus tractus solitarius (NTS) makes reciprocal connections with the hypothalamus
• Accessible to peripheral signals (GI & fat hormones): area postrema: no blood-brain barrier
Describe the role of higher centers (including amygdala and prefrontal cortex) on appetite control.
• Play important roles in the control of appetite and feeding
• Lesions of amygdala alters feeding behavior
• Destruction of amygdala à loss of appetite control that determines the type and quality of food eaten
What role does GI distension have on regulating food intake?
Stretch of the stomach and duodenum activates inhibitory signals which are sent via the vagus nerve to suppress the hypothalamic feeding center
What role does GI hormones have on regulating food intake?
-Cholecystokinin (CCK): anorexigenic; released in response to fat entering duodenum.
-Peptide YY: anorexigenic; delays gastric emptying. Secretion is proportional to calories ingested
-Glucagon-like peptide: anorexigenic; INC insulin (which further; Reduced levels may contribute to obesity
-Ghrelin: orexigenic; secretion from stomach and intestine increased during fasting and in anticipation of food.
What factor help maintain energy stores?
• Circulating concentrations of blood glucose, amino acids and lipids: Glucostatic, aminostatic, lipostatic theories: when substrate levels decrease the desire for feeding increases to restore homeostasis – Detected via glucosensitive neurons of the hypothalmus
• Feedback signals from adipose tissue: Leptin: Peptide hormone released from adipocytes that acts to reduce fat storage via various pathways (see next slide)
Describe leptin.
-Protein product of the ob gene expressed in adipose tissue
-Leptin receptors expressed in hypothalamic nuclei; activation causes: DEC orexigenic substances NPY & AgRP, INC a-MSH (anorexigenic) à DEC food intake, INC Sympathetic activity à INC metabolic rate, DEC Insulin à DEC energy storage
-Leptin levels positively correlate with food intake and adiposity
-Mutations in leptin or its receptor cause hyperphagia, obesity, diabetes
-However, no leptin deficiency in most obese humans and peripherally administered leptin has only very modest effects on body weight – may be linked to leptin resistance
What is the definition of obesity?
-Body mass index (BMI) = weight (lbs) × 703 / height2 (in.)
-Overweight: Body weight > 110% of ideal OR BMI > 25
-Obesity: Body weight > 130% of ideal OR BMI > 30
What is the epidemiology of obesity?
~127×106 (~65%) adults in the U.S. are overweight, 60×106 (~31%) are obese and 9×106 are extremely obese (BMI > 40). ~15% of children and adolescents are overweight.
What is the significance of obesity?
Increased risk for a host of diseases, particularly when combined with other risk factors (“metabolic syndrome X”: insulin resistance, excessive abdominal fat, high blood glucose, high blood pressure and high cholesterol)
What is the cause of obesity?
-Food intake > expenditure
-Contributing factors: Genetics (25-75%), endocrine/regulatory imbalance, environment, culture.
Describe anorexia nervosa.
• Psychiatric disorder mainly in adolescent/young women due to distorted body image
• Compulsive dieting, self-imposed starvation à profoundly low body weight
• Delayed gastric emptying, endocrine problems including infertility
Describe bulimia nervosa.
• Recurrent episodes of binge eating with feeling of lack of control over eating. Often followed by self-induced vomiting; use of laxatives or diuretics
• Weight usually does not fall below normal
• Not accompanied by endocrine or GI pathology (aside from dyspepsia)
What is the definition of sleep?
unconsciousness from which the person can be aroused by sensory or other stimuli
What are the two types of sleep?
slow-wave sleep, REM sleep (paradoxical sleep, desynchronized sleep)
Describe slow-wave sleep.
brain waves are very large but very slow, characterizes most sleep each night, deep, restful sleep, associated with a DEC in both peripheral vascular tone and many other vegetative functions of the body, also 10 to 30 percent DEC in BP, respiratory rate, and basal metabolic rate, consolidation of the dreams in memory does not occur here
Describe REM sleep.
rapid eye movement, in this sleep eyes undergo rapid movements despite the fact that the person is still asleep, occurs in episodes during sleep, ~25% of sleep, recurs about every 90 minutes, associated with vivid dreaming, lasts 5-30 minutes, INC with more sleep
What are the characteristics of REM sleep?
1. it is usually associated with active dreaming and active bodily muscle movements
2. even more difficult to arouse, but people usually awaken spontaneously in the morning during an episode of REM sleep
3. depressed muscle tone, indicates strong inhibition of the spinal muscle control areas
4. irregular heart and respiratory rate
5. irregular muscle movements
6. highly active brain in REM sleep, brain patterns similar to wakefulness
What is thought to cause sleep?
believed to be caused by an active inhibitory process, transecting the brain stem at the level of the midpons creates a brain whose cortex never goes to sleep, there seems to be some center located below the midpontile level of the brain stem that is required to cause sleep by inhibiting other parts of the brain
What are some of the areas through to cause sleep?
1. raphe nuclei in the lower half of the pons and in the medulla-these neurons secrete serotonin (if serotonin formation is blocked, animals can’t sleep for several days)
2. nucleus of the tractus solitaries-the sensory region of the medulla and pons also promotes sleep, these signals perhaps act by exciting the raphe nuclei and the serotonin system
3. stimulation of regions in the diencephalon promote sleep (include rostral part of hypothalamus and diffuse nuclei of the thalamus)
What is the major problem with the serotonin theory of sleep?
the blood concentration of serotonin are lower during sleep than during wakefulness
What happens with lesion in sleep-promoting centers?
can cause intense wakefulness with lesions in raphe nuclei
What role does muramyl peptide have on sleep?
accumulates in the CSF and urine in animals kept awake for several days, when add micrograms of this into 3rd ventricle, almost natural sleep occurs in minutes, also nonapeptide from blood and neuronal subsntaces in brain can cause sleep, thought that prolongede wakefulness causes progressive accumulation of sleep factors
What are some possible causes of REM sleep?
drugs that mimic the action of ACh INC the occurrence of REM sleep, thought that large ACh-secreting neurons in the upper brainstem reticular formation might activate many portions of the brain
Describe the effects that sleep has on the nervous system.
more important, any person who has a transected spinal cord in the neck shows no harmful effects in the body beneath the level of transaction that can be attributed to a sleep-wakefulness cycle, prolonged wakefulness is associated with progressive malfunction of the throught processes and sometimes even causes abnormal behavioral activites (sleep is through to restore normal levels of brain activity and normal balance among the different parts of the CNS), sleep restores the principal value of sleep is to restore the natural balance among the neuronal centers
Describe the effects that sleep has on other functional systems of the body.
during slow-wave sleep, sympathetic activity DEC while parasympathetic INC, leads to arterial BP falling, pulse rate DEC, skin vessels dilate, activity of GI tract INC, skeletal muscles fall into a mainly relaxed state and the overall BMR falls by 10 to 30%