HYPOTHALAMUS and NEUROENDOCRINE SYSTEMS

 

FOUNDATIONS (ZABORSZKY, 02-26-02)

 

1. Boundaries and Subdivisions

 

2. Major Fiber Systems of the Hypothalamus

 

3. Connections of the Hypothalamus

 

4. Hypothalamic Nuclei

 

5. Magno- and Parvocellular Neurosecretory System

 

6. Reflex Control of Vasopressin and Oxytocin Secretion

 

7. Brain-Pituitary Gonadal Axis

 

8. Stress

 

9. Behavioral State Control

 

10. Circadian Timing

 

11. Food Intake Regulation

 

12. Drinking

 

13. Temperature Regulation

 

 

 

 

 

 

 

 

 

 

 

 

 


 

1. Boundaries and Subdivisions

 

The hypothalamus forms the ventral part of the diencephalon.  The hypothalamus can be divided longitudinally into periventricular, medial and lateral cell groups.  The medial and periventricular hypothalamus contains most of the neurons concerned with regulation of the pituitary, but also important efferent sources for projections to brainstem and spinal autonomic areas.  The medial hypothalamus has, in addition, extensive reciprocal connections with the medial division of the 'extended amygdala’.  The hippocampus, either directly or via the septum, also sends afferents to medial hypothalamus.  The lateral preoptic-hypothalamic  (LPO-LH) continuum contain numerous cells which are interspersed among fibers of the medial forebrain bundle (MFB). The LPO-LH area shares a wide variety of reciprocal connections with the forebrain, caudal brainstem, and spinal cord.  The physiology of this area is complicated by the fact that many axons traverse this area which may or may not synapse locally.  Fig. 1 and Table 1 and 2 summarizes some of the functions of the hypothalamus.

 

2. Major Fiber Systems of the Hypothalamus

 

Some of the heavily myelinated hypothalamic fiber tracts, e.g. fornix, mamillothalamic tract, stria medullaris, stria terminalis, medial forebrain bundle can be identified by blunt dissections or using myelin staining, however, the direction of fibers within these tracts can be identified only by experimental tract-tracing methods.

 

Fornix. The fornix connects the hippocampal formation with the septal area, anterior thalamus and hypothalamus.

 

Mammillothalamic Tract and Mammillary Peduncle.  The mammillary body in the caudal part of the hypothalamus is surrounded by a capsule of heavily myelinated fibers.  Its function is not well known.  Most of its efferent fibers leave the mammillary body in a dorsal direction as the mammillothalamic tract, which proceeds towards the anterior thalamic nuclei.  Collaterals of the mammillothalamic fibers form the mammillotegmental tract, which projects to tegmental cell groups in mesencephalon.  These cell groups in turn give rise to the mammillary peduncle, which terminates primarily in the lateral mammillary nucleus.

 

Stria Medullaris.  The stria medullaris, which can be easily recognized on the mediodorsal side of the thalamus, connects the lateral preoptic-hypothalamic region with the habenular complex.  However, like most other hypothalamic pathways, the stria medullaris is a complicated bundle that contains many different
fiber components with various origins and terminations.

 

Stria Terminalis.  The stria terminalis reciprocally connects the amygdaloid body and the medial hypothalamus.  Similar to the fornix, the stria terminalis makes a dorsally convex detour behind and above the thalamus.  It can be identified in the floor of the lateral ventricle, where it accompanies the thalamostriate vein in the groove that separates the thalamus from the caudate nucleus.  In the region of the anterior commissure, the stria terminalis divides into different components, which distribute their fibers to the bed nucleus of the stria terminalis, medial hypothalamus and other areas in the basal parts of the forebrain.  The stria terminalis is an important pathway for amygdaloid modulation of hypothalamic functions.  The amygdaloid body is also related to the lateral hypothalamus through a diffuse ventral pathway that spreads out underneath the lentiform nucleus.

 

Dorsal Longitudinal Fasciculus The DLF is a component of an extensive periventricular system of descending and ascending fibers, that connects the hypothalamus with the midbrain gray and other regions in the pons and medulla oblongata including preganglionic autonomic nuclei.

 

Medial Forebrain Bundle.  The MFB is an assemblage of loosely arranged, mostly thin fibres, which extends from the septal area to the tegmentum of the midbrain.  It traverses the lateral preoptico-hypothalamic (LPO-LH) area, the scattered neurons of which are collectively designated as the bed nucleus of the mfb.  THe bundle is highly complex, comprising a variety of short and long ascending and descending links.

 

 

3. Connections of the Hypothalamus

 

Most of the connections of the hypothalamus consist of fine, unmyelinated fiber systems that cannot be traced accurately in normal myelin- or fiber-stained preparations.  As a result, much of what is now known about the connections of the hypothalamus has been learned in the last decade or so, since the introduction of the axonal tracer methods.  These connections are summarized below.

 

Afferents

 

Cortical Inputs.  Cortical inputs to the hypothalamus in the rat arise primarily from insular, lateral frontal, infralimbic, and prelimbic areas.  These afferents principally supply the lateral hypothalamic area.

 

Visceral inputs.  The nucleus of the solitary tract (NTS) projects directly to the hypothalamus in the rat.  In the monkey and human, presumably the visceral afferent influence from the NTS is relayed to the hypothalamus via the projection of the NTS to the parabrachial nucleus.

 

Olfactory inputs.  In rodents, olfactory input arrives via relays in the olfactory tubercle, anterior olfactory nucleus, corticomedial amygdala and olfactory cortex.  From these regions, secondary olfactory afferents terminate throughout the lateral hypothalamus.

 

Visual inputs may reach the hypothalamus via a direct retinal projection.  In all mammalian species, including humans, some retinal fibers leave the optic chiasm and pass dorsally into the hypothalamus, where they innervate the suprachiasmatic nuclei.

 

Somatosensory information may also reach the hypothalamus via a direct route: a projection to the lateral hypothalamic area from wide-dynamic-range mechanoreceptive neurons in the spinal dorsal horn.

 

Auditory input. Despite extensive study, no direct projection to the hypothalamus from the auditory system has been identified.  Recently, however, it has been shown that acoustic stimulation induce LH release in birds (MeiFang et al., 1998). Many hypothalamic neurons respond best to complex sensory stimuli, suggesting that the sensory information that drives them is highly processed.  It is likely, therefore, that much of the sensory information that reaches the hypothalamus travels by polysynaptic routes involving convergence of cortical sensory pathways in the amygdala, hippocampus and cerebral cortex.

 

Monoamine cell groups.  Each of the classes of monoamine cell groups in the rat brainstem provides innervation to the hypothalamus.

 

Projections from limbic regions. Hippocampal efferents via the precommissural fornix-lateral septum innervates all three longitudinally organized columns of the hypothalamus. Several cell groups of the amygdala project via the stria terminalis or the ventral amygdalofugal pathway to the hypothalamus. The ventral subiculum project via the medial corticohypothalamic tract to the medial hypothalamic cell groups.

 

The Circumventricular Organs (CVOs). Chemosensory information from plasma or CSF reach the hypothalamus via input from projections of CVOs. CVOs has specialized fenestrated capillaries, permitting relatively large molecules to leave the vascular bed and enter the extracellular milieu. Two of these regions, the subfornical organ (SFO) and area postrema (AP) have extensive connections with hypothalamic nuclei involved in neuroendocrine and homeostatic regulation. Two other CVOs, the organon vasculosum laminae terminalis (OVLT) and the median eminence (ME), are located within the hypothalamus.

 

 

Efferents

 

The main outflow of hypothalamic nuclei are directed 1) median eminence (parvocellular neurons), 2) posterior pituitary (magnocellular) to influence neuroendocrine responses; 3) sympathetic and parasympathetic pregangionic cell groups in the brainstem and spinal cord to influence autonomic functions (primarily the dorsal, medial and lateral parvocellular division of the PVN); 4) several cell groups in the hypothalamus project to the amygdala, bed nucleus of the stria terminalis, to the basal nucleus of Meynert, periaqueductal gray (PAG), visceral sensory areas of the thalamus (ventroposterior parvocellular nucleus) cerebral cortex (anterior insular cortex, anterior tip of the cingulate cortex), and brainstem (nucleus of the solitary tract= NTS,   parabrachial nucleus) to influence various behavioral responses.

 

 

4. Hypothalamic Nuclei (Fig. 2)

 

The suprachiasmatic nucleus (SCN) has been identified as an endogeneous timekeeper.  This nucleus receive afferents directly and indirectly from the retina in order to synchronize otherwise free-running circadian rhythms with the day-night cycle.  At least some of its actions, particularly on hormonal rhythms, appear to be mediated via projections to the medial hypothalamus.

 

The paraventricular nucleus (PVN), in addition to the magnocellular neurons, contain several subgroups of small (parvicellular) neurons   containing    a variety of putative neurotransmitters.  Some of the parvicellular neurons (e.g. CRF=corticotropin releasing factor) project to the median eminence where they participate in the regulation of the anterior pituitary. Other   neurons   in   the   PVN   project to sympathetic  and parasympathetic    autonomic    areas   in   the   medulla    and   the intermediolateral cell columns of the spinal cord.  The PVN has been implicated in a variety of behaviors including feeding, thirst, cardiovascular mechanisms as well as organization of autonomic and endocrine responses to stress.

 

The arcuate nucleus among others contain dopamine which acts as a prolactin inhibiting factor at the median eminence.  In additions, its neurons are eostrogen sensitive and project to the preoptic LHRH neurons.  This circuit is involved in the regulation of gonadotropin secretion and sexual behavior during female reproductive cycle.

 

The ventromedial nucleus (VMH) in addition to its output to the median eminence, with their other projections is thought to participate in the organization of reproductive behavior, as well as in metabolic regulatory functions.

 

The dorsomedial hypothalamic nucleus (DMH) among others is involved in mediating leptin actions to the PVN. Fibers from the SCN via the DMH towards the locus coerules are suggested to participate in circadian regulation of sleep and walking.

 

The mammillary body is at the caudal border of the hypothalamus.  The lateral and medial mammillary nuclei are the recipient of a massive input from the hippocampus that arrives via the fornix.  These nuclei project via the mammillo-thalamic tract to the anterior nuclei of the thalamus.  These nuclei are frequently damaged in Korsakoff's patients.

 

 

5. Magno- and Parvocellular Neurosecretory System (Fig. 3-6)

 

The  magnocellular neurons of the  supraoptic (SON) and paraventricular  (PVN) nuclei along with scattered clusters of large cells between these two nuclei comprise the hypothalamo-hypophyseal system.  These cells send oxytocin and vasopressin containing fibers to the posterior pituitary.  The latter is the well known antidiuretic hormone (ADH) and is released in response to changes in the osmotic pressure of circulating blood or extracellular space.  ADH controls the water-balance.  In plarticular, it is responsible for the retention of water, which is regulated by the effect of vasopressin on the distal tubules of the kidneys.

 

Oxytocin, through its effect on the uterine smooth muscle and the myoepithelial cells of the mammary glands, promotes uterine contraction during birth and milk ejection after birth.  Potent stimulatory input for uterine contraction reaches the brain via afferents from the vagina or cervix and the nipples.

 

Hypothalmic (parvocellular) neurons originating in the preoptic, arcuate, ventromedial, periventricular, paraventricular nuclei transport a variety of releasing and inhibiting hormones to the portal vessels of the median eminence.  These are then transported to the capillary beds of the anterior pituitary where they regulate the secretion of the pituitary troph hormones: TRH (Thyrotropin-Releasing Hormone) → TSH (Thyrotropin), CRH or CRF (Corticotropin-Relasing Hormone) → ACTH  (Adrenocorticotropin Hormone), GnRH (Gonadotropin-Releasing Hormone) → FSH (Follicle-Stimulating Hormone) and LH (Luteinizing Hormone), GHRH (Growth Hormone-Releasing Hormone) and GHRIH (somatostatin) → GH (Growth Hormone), PRF (Prolactin-Releasing Factor) and PIF (Prolactin Release-Inhibiting Factor) → Prolactin, MRF (Melanocyte-stimulating hormone Releasing Factor) and MIF (Melanocyte-stimulating hormone release Inhibiting Factor) → MSH (Melanocyte-Stimulating Hormone). Figures 3 summarizes the design of the parvo- and magnocellular neurosecretrory system.

 

The magno- and parvocellular cell groups producing the hypothalamic hormones receive a variety of stimuli from different parts of the brain, primarily within the hypothalamus, but also from extrahypothalamic areas including the amygdaloid body, hippocampus and various brainstem areas.  Furthermore, it is well known that monoamines and several neuropeptides serve as modulators of the neuroendocrine system, and both manoaminergic and peptidergic fibers, besides those carrying the specific hypothalamic hormones, can be traced to the periventricular zone and even into the median eminence, where they would have an opportunity to interact with the parvicellular neurosecretory system or even discharge neuroactive substances directly into the portal system. Figures 4-6 summarize the input-output relations of the PVN.

 

The subject of neuroendocrine control mechanism is complicated further by the fact that many neurons in the nervous system, including the hypothalamic magnocellular and parvocellular neurosecretory neurons, contain two or even several neuroactive substances.  A well known example is provided by the parvocellular CRF neurons in the PVN.  They also contain vasopressin and the two substances are released together into the portal vessels, through which they are likely to cooperate in the control of ACTH-release from the adenohypophysis. Hypothalamic neurons, including the neurosecretory neurons, are also subject to hormonal feedback control.  Such feedback mechanisms are often quite complicated in the sense that they involve not only the neurosecretory hypothalamic neurons but also hormone sensitive cells in other brain regions, which in turn are in a position to modulate hypothalamo-hypophysial function. Peripheral hormones (e.g. estrogen, etc) exert their feedback actions also at the level of the median eminence and the  anterior pituitary.

 

 

6. Reflex Control of Vasopressin and Oxytocin Secretion (Figs. 7-10)

 

The nonapeptides, oxytocin (OT) and vasopressin (VP), two major biologically active hormones, are synthesized in separate cell populations in the supraoptic and paraventricular nuclei of the hypothalamus.  These peptides are carried by axoplasmic transport to various areas within the CNS and to the posterior pituitary.  OT and VP are released from nerve endings in the neural lobe of the pituitary to reach the sytemic circulation and influence primarily fluid balance (VP) and milk ejection/uterus contraction (OT).  In addition, by their axonal projections in the CNS, VP and OT also play a role in neurotransmission.

 

 

Vasopressin

 

Periphreal VP functions largely to maintain arteriolar perfusion pressure and intravascular volume.  One of the most potent effective stimuli for VP secretion is a rise in extracellular osmolality.  Although less potent, other indicators of extracellular fluid depletion also stimulate VP release, including decreased plasma volume (hypovolemia, hemorrhage), decreased blood pressure (hypotension), and peripheral hypoxia or hyperkapnia or both. In contrast, drinking fluids, even when they are hypertonic, results in an abrupt fall in systemic VP levels, presumably via stimulation of osmoreceptors in the oropharynx.  In addition, various stressors, fever, pain and nausea and emetic agents such apomorphine  causes VP (and OT) release.

Circulating VP maintains extracellular fluid balance by acting a) at the kidney, where it stimulates increased retention of water and enhanced Na and Cl excretion, b) at arterioles, where it is one of the most potent vasoconstrictors, c) it also modulate sympathetic transmission and d) affect the baroreceptor reflex by a central effect mediated by the area postrema. Peripheral VP has also been found to have effects on hepatic glycogenolysis, platelet aggregation and blood coagulation.

Under emergency conditions, the peptide causes vasoconstriction in skin, gastrointestinal tract and kidney.  It serves to shunt blood from these tissues to the brain, heart and lung.

Peripheral Receptors and Pathways.  Vasopressin is under tonic inhibitory influence both from atrial receptors of the heart (low pressure receptors) and from baroceptors (high pressure) in the aortic arch (X) and carotid sinus (IX).  Reduction in the discharge of these receptors by a decrease in blood volume or blood pressure results in the release of vasopressin.  An additional excitatory influence on vasopressin release is provided by carotid body chemoreceptors and peripheral osmoreceptors.  The signal from arterial baroreceptors, cardiopulmonary receptors and peripheral osmoreceptors from the mesenteric and hepatic vasculature is carried through the IX and X. nerves to the NTS.  From the NTS information though GABAergic neurons or directly may reach Al noradrenergic neurons which project to SON and PVN VP neurons to stimulate VP release.  Bilateral carotid occlusion releases vasopressin and leads to activation of carotid body chemoreceptors.  It is not clear, however, how the excitatory input from the chemoreceptors reach the SON or PVN.  A possibility is that excitatory input to the VP neurons may arise from cholinergic neurons that lie dorsal to SON.  Indeed, ACh injected into the SON results in nicotine mediated vasopressin release.

Central Receptors and Pathways.  Lesion studies established that both the subfornical organ (SFO) and the AV3V (MPO, PPO, Mdn, OVLT) are involved in the neural regulation of salt and water balance.  Both the SFO and the AV3V region are the major target for the dipsogenic action of blood born ANG II.  When blood pressure falls, the kidneys release renin into the bloodstream. Renin triggers a biochemical cascade that produces AII (angiotensin).  ANG II is released from the periphery as a chemical signal of hypovolemia and hypotension.  Additional evidence suggests that the SFO and OVLT and the magnocellular neurons themselves may be sensitive to changes in extracellular osmolality and sodium concentration.  Afferents from the SFO and AV3V reach both PVN and SON neurons. The projection from SFO to PVN and SON use ANG II as transmitters.

 In addition, NTS afferents reach the AV3V region through the parabrachial area.  Afferents from the BST to PVN, SON serve to integrate central cardiovascular and "limbic" information. 

Clinicopathology.  The lack of VP results in a condition known as diabetes insipidus (DI), which is characterized by an increased production of urine (polyuria).  The loss of fluid, in turn results in an excessive thirst (polydipsia).  Often DI is caused by lesions of the base of the brain (e.g. tumors or skull fructures) involving the SON, PVN or the hypothalamohypophyseal tract.

 

Oxytocin

 

In contrast, circulating OT is best known in female reproduction, where it is involved in the maintenance of parturition and the initiation of lactation.  Thus, OT acts on the smooth muscle of the endometrium during labor and delivery to increase the frequency and intensity of uterine contractions, and on the myoepithelial cells surrounding mammillary alveolar glands to cause milk let-down in response to suckling.  An increase in circulating OT also accompanies ejaculation in males.  OT also stimulate the release of both insulin and glucagon from the pancreas, and act on adipocytes, indicating that it plays a role also in metabolic regulation related to feeding.  Vaginal and uterine distension receptors, somatic sensory receptors from the nipple and breast and nociceptive information from much of the body are all relayed initially to the dorsal horn of the spinal cord, from where axons project to the Al cell group and the caudal NTS.  From here, ENK, SS and inhibin B pathways may mediate specific stimuli to OX neurons in the hypothalamus.

 

7. Brain-Pituitary Gonadal Axis (Figs. 11-17)

 

The hypothalamus plays a major integrative role in the control of maternal and reproductive behavior, including sexual development, and differentiation, as well as sexual behavior.  Important stimuli for the various aspects of reproductive functions come 'from a variety of exteroceptive and interoceptive sources including circulating gonadal steroids.

Differences between male and female are not limited to sexual organs and secondary sex characteristics; they are also evident within the CNS.  For example, there is a sexually dimorphic nucleus of the preoptic area, which is considerably larger in males and the same is true for a cell group in the sacral spinal cord known as the nucleus bulbocavernosus.

The sex hormones, i.e. androgens and estrogens, play important roles both in the development and differentiation of the male and female sex organs and sexual behavior.  GnRH producing neurons, responding to sensory input and to circulating gonadal steroids, control the secretion of LH and FSH from the anterior pituitary.  GnRH secreting neurons with projections to the portal system in the median eminence are located in preoptic-anterior periventricular area.  LH and FSH are released from the pituitary into the systemic circulation in response to GnRH, and they travel to the gonads, where they direct gamete production, as well as gonadal (testosterone in male and estrogen and progesterone in female) hormone production.

Sexual behavior involves a number of general (e.g. respiratory and cardiovascular) and specific (e.g. erection, ejacaulation, etc) responses mediated in large part by the autonomic nervous system.  Although several of these specific responses represent involuntary or reflex phenomena, descending pathways from the hypothalamus or basal forebrain regions play a significant modulatory role.  A critical brain area in male copulatory behavior seems to be the medial preoptic area, whereas feminine sexual behavior appears to be more dependent on regions in and around the ventromedial nucleus.

          GnRH neurons are born outside the brain in the olfactory placod and migrate caudally to their final positions in the septal, preoptic and anterior hypothalamic areas. GnRH release into the portal bloodstream occurs in a coordinated fashion, with distinct pulses of GnRH secretion. The pulsatile stimulation of the anterior pituitary by GnRH leads, in turn, to pulsatile release of LH and FSH from the pituitary gonadotropes into the peripheral bloodstream. Multiple-unit recording electrodes placed in the medial basal hypothalamus of rhesus monkeys have measured spikes of electrical activity that correspond in time to pulses of LH release. These bursts of electrical activity may come from GnRH neurons themselves or from neurons that impinge upon the GnRH neural system and thereby govern its firing pattern. The question of how GnRH neurons, distributed diffusely throughout the hypothalamus, coordinate the release of discrete pulses of GnRH into portal bloodstream remains unanswered. GnRH neurons might actually form an interconnected network. Anatomical studies showing synapses between GnRH neurons, and perhaps cytoplasmic bridges between adjacent GnRH neurons.

          Synaptic input from a variety of neuronal types has been reported, including other GnRH neurons; dopaminergic, noradrenergic, serotoninergic; neurons containing GAD, CRF, SP, NT, B-END, etc.

          GnRH release is affected by the negative feedback of steroid hormones at the level of the hypothalamus. Steroid hormone (estrogen, progesterone) negative feedback decreases the frequency of pulsatile GnRH stimulation of the pituitary and thus results in decreased frequency of pulsatile LH release (e.g. luteal phase). At the level of the hypothalamus, steroid hormones likely modulate the firing of neurons that project to LHRH neurons, since the LHRH neurons themselves lack estrogen receptors.

          Pulsatile stimulation of the pituitary by GnRH is necessary to maintain normal function of pituitary gonadotropes. There is a fairly narrow window of acceptable frequency for stimulation of the pituitary by GnRH to achieve normal gonadotropin secretion. Pulse frequency faster or lower than once per hour usually leads to an inhibition of gonadotropin secretion.      The gonadal steroid hormones estradiol and testosterone can negatively feedback at the levels of the hypothalamus and pituitary to decrease the frequency of pulsatile gonadotropin secretion and decrease the amplitude of LH pulses. In women, pulsatile LH secretion in the early follicular phase of the menstrual cycle, when circulating concentrations of ovarian steroid hormones are quite low, occurs at a frequency of approximately one pulse per hour. As the follicular phase progresses, and the developing ovarian follicles begin to secrete increasing amounts of estradiol, slows to one pulse every 90 min. In the luteal phase of the menstrual cycle, when the ovary is secreting large quantities of progesterone, as well as estradiol, LH pulses occur at a frequency of once every 6-12 h. In contrast, in man, LH pulse frequency remains rather stable throughout adulthood, at approximately one pulse every 2-3 hr. However, testosterone levels play a large part in determining this frequency, as seen by the effects of castration.

          Negative feedback of steroid hormones at the level of pituitary decreases the sensitivity of pituitary gonadotropes to GnRH  and thus results in  a decrease in the amplitude of LH pulses but not the frequency of LH pulses.

          Ovarian function is cyclic. In women, the first half of the menstrual cycle is characterized by the growth and development of a cohort of follicles, culminating with selection and ovulation of the ovum from a single dominant follicle at about 14 days from the beginning of the cycle. After ovulation, the cells of the collapsed follicle are reorganized into a transiently functional, steroid producing gland, the corpus luteum, that secretes progesterone and estrogen. Should fertilization of the ovulated ovum occur, these ovarian hormones play a critical role in preparing the uterus for implantation of the developing embryo. If a pregnancy does not occur, the corpus luteum spontaneously becomes dysfunctional after approximately 14 days. Withdrawal of gonadal hormones to the uterus leads to menses and the period of follicular development is reinitiated. In women, and other primates with menstrual bleeding, Day 1 of the menstrual cycle is designated the first day of menstrual bleeding. Species, in which the endometrium is reabsorbed, rather than sloughed, show a behavioral predisposition to mating during the time of ovulation. Cycles in these species are referred to as estrus cycle.

          A key event in the regulation of cyclic ovarian function is the midcycle gonadotropin surge.  The dominant follicle responds to FSH and LH by greatly increasing estradiol synthesis in the last few days of the follicular phase. The sustained high levels of estradiol act at the hypothalamus and pituitary to cause a positive feedback effect such that a large outpouring of LH and FSH is released from the pituitary. This very high levels of LH and FSH released during the surge trigger the final maturation of the ovum within the dominant follicle and trigger ovulation of that follicle. In women, estradiol positive feedback occurs when estradiol levels reach approximately 150 pg ml –1 for at least 36h.  Both the magnitude and the duration of elevated estaradiol are critical for the induction of the gonadotropin surge. Estradiol elicits positive feedback by acting at the level of the hypothalamus to increase GnRH release and at the level of the pituitary to increase gonadotrope sensitivity to GnRH. The mechanism of the estradiol positive feedback action is unknown.  However, measurement of GnRH release during the surge is not pulsatile, suggesting, that the surge may involve a GnRH release mechanism that is distinctly different from the normal, pulsatile GnRH release mechanism. Although the hypothalamopituitary system plays a critical role in producing the midcycle gonadotropin surge, the timing of events in the ovarian cycle clearly is regulated by the ovaries.

          Estrous cycles. The regulation of ovarian cyclicity in the female rat differs from that in primates in several important aspects: 1) the rat has a 4-5 day estrus cycle; 2) in rodents multiple follicles ovulate during each cycle, in contrast to ovulation of a single follicle in primates; 3) in the rat the corpus luteum produces progesterone for only a very short period of time (day of the estrus) during a nonfertile cycle. This lack of a true luteal phase in the rat cycle allows the cycle length to be shortened. A short estrus cycle is advantageous for this relatively short-lived species, because it increases the frequency with which the rat enters periods of fertility (i.e. estrus). If pregnancy occurs, the life span of the corpus luteum is extended to provide steroid hormone support to the uterine lining.

            Estrogen and cognition. It is well established that estrogens affect the brain throughout the life span. Moreover, the effects are not limited to the areas primarily involved in reproduction but also include areas relevant to memory, such as the basal forebrain and hippocampus. For example, there is extensive evidence that estrogen levels are correlated positively with dendritic spine densities within CA1 of the hippocampus and that estrogen administration ameliorates learning deficits and cholinergic abnormalities in ovariectomized rats (McEwen). Also, there are sex differences in the rate of development and the magnitude of age related impairments in spatial reference memory (Markowska, 1999) that is paralleled with altered estrogen levels. Figures 11-17 summarizes various aspects of the reproductive cycle, including LHRH, LH, FSH, estradiol and progestreon regulation.

 

8. Stress (Fig. 18-20)

 

          The brain-pituitary-adrenal axis is a key player in an animal’s response to stressful stimuli. Other participants are the adrenal medulla, which produces NE and A and the autonomic nervous system, which modulates physiologic functions through neurotransmitters.

          Corticotropin-releasing hormone (CRH) is a 41 amino-acid peptide expressed in the hypothalamus. The region with highest expression is the medial parvicellular part of the PVN. CRH neurons in the mpPVN project to the external ME, where peptides are secreted into the portal bloodstream, through which they are transported to the anterior pituitary. In addition to CRH, the same neurons in the PVN also express and release vasopressin, although most of the VP is expressed in neighboring magnocellular elements of the PVN that project to the posterior pituitary. Corticotropes in the anterior pituitary express receptors for CRH and VP.  In response to stimulation, corticoctropes synthesize and release adrenocorticotropic hormone (ACTH). ACTH through the systemic circulation binds and activate its receptors on the surface of cells of the adrenal cortex. In response to receptor activation, adrenocortical cells synthesize glucocorticoids.

          Concentration of circulating glucocorticoids and ACTH show a daily rhythm. In humans, glucocorticoid levels peak around 7 AM and decline steadily throughout the day. The nadir is reached in the late evening at 7PM to midnight after which glucocorticoid levels begin to rise.  The phase of the daily ACTH rhythm precedes that of the glucocorticoids by about 1-2 hrs. CRH expression of the PVN has also a daily rhythm, consistent with a negative feedback of glucocorticoids. Food intake, or the anticipation of eating is a major factor in controlling the CRH cycle, in addition to SCN.

          Basal activity of the brain-pituitary-adrenal axis oscillates, but the system is activated under emergency conditions through neural input. In the hypothalamus, the PVN appears to sum and integrate input from numerous loci. Input to PVN is divided into several broad classes, like brainstem (catecholaminergic fibres via the NTS convey viscerosensory information); hypothalamic or limbic inputs (amygdala, septum, hippocampus, prefrontal cortex reach PVN primarily via the bed nucleus of the stria terminalis). Blood-borne signals through neural projection from SFO and OVLT apparently also reach stress-related PVN parvicellular neurons.

          Removal of the adrenal, and hence of glucocorticoids, removes the negative feedback effects of these steroids. In this situation, concentrations of CRH and VP mRNA in the mp PVN increase.

          Glucocorticoid receptors are members of a superfamily of receptors that act as ligand-regulated transacting receptors. In each case, the receptor protein resides in the cytoplasm in a complex containing heat-shock proteins, which fold the receptor into the appropriate configuration for recognizing corticosteroid ligands. Upon steroid binding, the receptor moves to the nucleus of the cell and interacts with specific hormone recognition (or response) elements on the DNA, thereby changing transcription rate.

          CRH neurons of the PVN contain steroid receptors, and glucocorticoids inhibit transcription of CRH and VP genes through genomic feedback However, numerous other brain regions also express steroid receptors. These regions, including the hippocampus exert negative feedback on the brain-pituitary-adrenal axis through projections to PVN.

          Chronic administration of corticosteroid changes the morphology of field CA3 cell’s dendrites. These changes may lead to cell death. On the other hand, dentate granule cells are damaged by a lack of glucocorticoids: there is a dramatic loss of these cells after adrenalectomy.

          Stress influences learning and memory. Aging and chronic stress can lead to hippocampus dependent learning deficit via alteration of glucocorticoid receptors.

 

 

9. Behavioral State Control (Fig. 21)

 

          The hypothalamus play an important role in sleep regulation. Lesion of the preoptic region produces insomnia in rats, wheras chemical or electrical stimulation of this region causes sleep. In contrast, lesions of the caudal hypothalamus produces somnolence, suggesting that this region is involved in arousal. Recently, Saper and colleagues (Sherin et al; 1996), using the localization of the protein product of the immediate-early gene c-fos, defined the location of hypothalamic neurons that are active during sleep. These, (presumably GABAergic) cells, located in the ventrolateral preoptic region (VLPO), project to the caudal hypothalamic histaminergic cell groups in the tuberomammilary nucleus (TMN), which promotes arousal and diffusely innervate the cerebral cortex.

          Several cell groups in the hypothalamus (see SUBCORTICAL ACTIVATING SYSTEMS) project diffusely to the cerebral cortex and have been postulated to be important in arousal. Recently, a 130 amino acid containing protein, called hypocretin (orexin) was isolated from the hypothalamus (Sutcliffe et al, 1997). In situ hybridization and immunocytochemical studies revealed that neurons expressing this protein are located exclusively in the tuberal region of the hypothalamus around the fornix. Although it is a restricted group of cells, their projection were widely distributed in the brain, including a diffuse projection to the cerebral cortex. Knockout mice that lack the gene encoding this protein show narcolepsy, indicating that this protein is important in arousal. 

          Saper suggests that between sleep and wake-promoting brain regions reciprocal interactions exist which results in stable wakefulness and sleep (Fig. 21). Disruption of wake- or sleep-promoting pathways results in behavioral state instability. For example, after lesion of the VLPO, the animals experience much more wakefulness. Similarly,  hypocertin-knockout mice or narcoleptic persons fall asleep more rapidly than unaffected individuals.

 

 

10. Circadian Timing

 

          The circadian timing system is composed of central and peripheral neural elements. Critical components include the photoreceptors and visual pathways (e.g retinohypothalamic tract to the SCN), circadian clocks or pacemakers (such as the suprachiasmatic nucleus) and output pathways (pineal gland, melatonin) to couple pacemakers to effectors. SCN neurons are spontaneously active circadian oscillator even when deprived of the afferent signals.  With SCN lesions, the sleep-wake rhythm is eliminated, but the amount of time spent asleep and awake and the amount of REM and nonREM sleep is unaffected. Light establishes both the phase and period of the pacemaker, and thus is the dominant entraining stimulus or Zeitgeber (time-giver) of the circadian system. The pacemaker can be viewed as a somewhat inaccurate clock, which must be repetedly reset.

          The circadian system responds to changes in luminance, the total amount of light, but not to color, shape, movement or other visual parameters.  The responsiveness of the circadian system is not altered in mutant mice with retinal degeneration and nearly complete loss of rods or cones. Recently, it has been shown that a small percentage (1-2% in the rodent’s retina) of retinal ganglion cells are light-sensitive, contain a protein melanopsin, register luminance levels and project to the SCN (Hattar et al., 2002; Berson et al., 2002). It is suggested that this, separate visual circuit, running in parallel with the image-forming visual system, encodes the general level of environmental illuminmation and drives certain photic responses, including synchronization of the biological clock with the light-dark cycle, acute suppression of locomotor behavior, control of pupil size, melatonin release, etc.  Only a subpopulation of retinal ganglion cells project to the SCN and the intergeniculate leaflet (IGL), a thalamic component of the circadian timer system. The IGL modulates entrainment by transmitting information about nonphotic (locomotor) events to the SCN.   The ganglion cell-SCN circuit uses Glu as transmitter, the IGL uses GABA and NPY. A 5HT mediated projection from the raphe nuclei can regulate SCN oscillation.

          The SCN project to the subparaventricular zone and other hypothalamic neurons and several other nonhypothalamic structures of the diencephalons. Direct activation of these efferent circuits controls the expression of circadian rhythmicity. SCN projections have peak daytime firing rates that are about twice as fast as night rates. However, transplantation studies suggest that humoral mechanism may play a role in rhythm regulation. In a series of an elegant experiments, Aston-Jones (2001) have shown that an SCN-Dorso Medial Hypothalamic (DMH)-locus coeruleus (LC) pathway is necessary for circadian rythmicity in the LC firing. The transmitter of the DMH-LC may be orexin/hypocretin. This route may be important in circadian regulation of sleep and waking.

          SCN ablation results in loss of estrus cycles. The precise timing of the release of GnRH to produce the LH surge is a function of a series of endocrine and neural events that lead to proestrus, with the SCN providing the crucial temporal signal.

          In habitats with marked seasonal variation of temperature and food supply, survival of species requires seasonal regulation of reproduction. The SCN via the PVN-intermediolateral column-superiocr cervical ganglion –pineal gland route provides a pulse of melatonin secretion that faithfully mirrors the lengths of day and night. Thus, the duration of the melatonin signal allows the animals to track time on an annual basis.

Melatonin receptors were identified in the SCN, the midline thalamic nuclei and the pars tuberalis of the anterior pituitary. Melatonin can affect pacemaker function via its action on SCN cells.

          Circadian function is genetically controlled, and data indicate that function is maintained by the expression of clock genes that code for specific proteins that feed back on the nucleus to control their own production.

 

 

11. Food Intake (figs. 22-28)

 

The hypothalamus integrates a variety of sensory stimuli, both synaptic and hormonal, and modulates autonomic outflow to viscera.

            Although recent work does not support the conclusion that the VMH functions as a satiety center, it remains possible that neurons in this nucleus playing an important role in insulin secretion. It is also clear the SCN influences the temporal organization of feeding. For example, nocturnal rodents normally feed almost exclusively during the dark phase of the photoperiod, but rodents whoose SCN is ablated feed throughout the light-dark cycle.

            Leptin is a circulating hormone produced by white adipose tissue and has potent effects on feeding behavior, thermogenesis and neuroendocrine response. Leptin regulates energy homeostasis, its absence in humans and rodents cause severe obesity. Congenital leptin deficiency resulting from mutations within the leptin gene causes extreme obesity. Moreover, a mutation in the leptin receptor gene results in morbid obesity, failure to undergo puberty and decreased level of growth hormone and thyroid hormone. However, most obese humans do not have mutations in the leptin or leptin-receptor genes but have high levels of circulating leptin that fail to prevent obesity. By analogy with diabetes mellitus, these people may have functional leptin resistance and impaired responsiveness to circulating leptins.

            Within the brain, leptin’s main site of action seems to be the hypothalamus. Leptin receptors localized to several hypothalamic nuclei, including, the ventromedial, arcuate, dorsomedial nuclei. This distribution of leptin receptors is consistent with a large body of literature implicating the ventrobasal hypothalamus in feeding, as shown by large lesions of the VMH that produced increased food intake and obesity. An important target of leptin are also the NPY neurons of the arcuate nucleus. NPY is a potent stimulator of food intake and leptin inhibit NPY neurons. Expression of NPY mRNA in the arcuate nucleus is elevated in response to fasting and in leptin-deficient ob/ob and leptin-resistent db/db mice. In ob/ob mice and fasted rats, exogeneous treatment with leptin suppresses NPY overexpression.

Leptin acts directly through hypothalamic long form leptin receptors.  The leptin receptor have a docking site for janus kinases (JAK), a family of tyrosine kinases involved in intracellular cytokine signaling. Activated JAK phosphorylates members of the signal transduction and transcription  (STAT) family of intracellular proteins. STAT proteins, in turn, stimulate transcription of target genes that mediate some of leptin’s cellular effect. Leptin can affect neuronal firing rate independently of its tarnscriptional effect.  This response is absent in db/db mice which lack the functional long-form leptin receptor, and thus cannot activate STAT proteins. Leptin also activate immediate early genes in specific nuclear groups. Intraveneous leptin activates several regions thought to be involved in regulation of energy balance, including VMH, DMH and PVN nuclei. Leptin administration also activates cells in the superior-lateral parabrachial nucleus that project a CCK containing pathway to the VMH.  Recently a leptin-inducible inhibitor of leptin signal tarnsduction has been described, which is rapidly induced in the hypothalamus following systemic leptin administration.

   Leptin treatment activates neurons in the PVN. The PVN may regulate many of the responses of leptin, as it has chemically and anatomically specific projection to brain control sites involved in the maintenance of autonomic and endocrine homeostasis.    The PVN is uniquely equipped to control activities both in the endocrine, autonomic and somatomotor systems.  It is populated by both magno and parvocellular neurosecretory neurons, that project to the neurphypophysis and to the portal system in the median eminence, thus can affect thyroide hormone, growth hormone and ACTH secretion.  The regulation of the hypothalamo-pituitary adrenal axis, which is controlled by CRF secreting neurons may be of special importance in this context.  For instance, corticosterone affects the carbohydrate metabolism, and it is well known that different forms of stress can influence eating behavior. Leptin regulates both the thyroid and hypothalamo-pituitary-adrenal axis.  Parvocellular neurons in the PVN also project to the brainstem parasympathetic and to the sympathetic preganglionic autonomic nuclei in the medulla and spinal cord.  Through these pathways, the PVN can directly influence the hormone secretion from pancreas and adrenal medulla as well as somatomotor activities, that may be relevant in feeding behavior. Leptin receptors are in the PVN, but the highest density of leptin receptor mRNA is in the ventrobasal hypothalamus, including the arcuate nucleus. Microinjection of leptin into the arcuate nucleus area results in anorexic response and icv injection of leptin no longer reduce food intake after the arcuate nucleus has been destroyed, suggest that leptin effects on PVN include inputs from leptin-responsive neurons  from  arcuate nuclei to the PVN. Leptin signals from the arcuate nucleus are mediated by NPY and POMC (pro-opiomelanocortin)/CART (cocaine- and amphetamine-regulated transcript)-containing efferents. 

   Lesions of the lateral hypothalamus can cause decreased food intake.  Neurons in the LHA, including those that contain melanin-concentrating hormone (MCH) project to the cerebral cortex. Ob/ob (leptin deficient) mice have elevated level of MCH mRNA and this overexpression is normalized by leptin administration.  Another peptide, orexin/hypocretin is found in the perifornical region (PFA) projecting to several areas of the neuraxis, including the cerebral cortex may be important in mediating leptin effect. Indeed, orexin icv injection increase feeding behavior. Figures 22-27 summarizes leptin receptor signaling and the various hypothalamic neuropeptide signaling pathways involved in mediating leptin and insulin action. 

Noradrenaline (NA) is synthesized in brainstem locus coeruleus and NTS region (A1 noradrenergic cell group). These areas project both caudally to the spinal cord and rostrally to the hypothalamus, thalamus and cortex. In some of these neurons, including those projecting to the PVN, noradrenaline is colocalized with NPY. Like NPY, injection of noradrenaline into the PVN increases food intake. The observation of elevated NA levels in the PVN of ob/ob mice may indicate that increased NA signaling in th ePVN may contribute to hyperphagia induced by leptin deficiency.

Dopamine. Feeding effects of dopamine vary with the brain region under study. For example, mesolimbic dopamine pathways, originating from the SN-VTA that project to the nucleus accumbens, striatum and cerebral cortex seem to contribute to the rewarding aspects of palatable food. In contrast, arcuate DA neurons seems to inhibit food intake. In ob/ob mice the reduced arcuate DA level may contribute to hyperphagia induced by leptin deficiency.

Serotonin. Several centrally acting drugs developed for obesity treatment (e.g. dexflenfluramine) increase 5HT receptor signaling and thereby suppress food intake, whereas antagonists have the opposite effect. The 5HT2C subtype is implicated in this process as knockout of this receptor increases food intake. Leptin increases 5HT turnover and thus it is possible that at least some of leptin’s weight-reducing effects are mediated through increased 5HT signaling.

According to a recent integrated model (Fig. 28), the adiposity signals, leptin and insulin stimulate a catabolic pathway (POMC/CART) and inhibit an anabolic pathway (NPY/AGRP) that originates in the arcuate nucleus. These pathways project to the PVN and LHA/PFA, where they make connections with central autonomic pathways that project to brainstem autonomic regions that process satiety signals.  Afferent input related to satiety from the liver, gastrointestinal tract and from peptides such as CCK are transmitted through the vagus nerve to the nucleus of the solitary tract (NTS), where they are integrated with descending hypothalamic input. Net neuronal output from the NTS leads to termination of individual meals, and is potentiated by catabolic projections from the PVN and inhibted by input from LHA/PFA. Reduced input from adiposity signals (for example during diet-induced weight loss), therefore, increases meal size by reducing brainstem responses to satiety signals.

 

 

12. Drinking

 

Fluid homeostasis is regulated by several interdependent mechanisms, one of which, i.e. the retention of water by the kidney mediated by the hypothalamo-hypophysial vasopressin system, was discussed above.  Increased water intake is another mechanism of replenishing body fluids.  Although we often drink spontaneously, drinking can also be activated by water deficit, i.e. deprivation induced drinking.  Deprivation-induced drinking is regulated primarily by osmotic changes in the blood or a change in blood volume, e.g. hemorrhage.  The osmoreceptors for drinking, like the ones regulating vasopressin release, are located in the OVLT and neighboring medial preoptic area.  Drinking in response to reduced blood volume is initiated by two different mechanisms.  One type of input originates in mechanoreceptors in the pulmonary artery and the vena cava, and reaches the hypothalamic integration centers for drinking via the nucleus of the solitary tract.  Another important stimulus is blood-born, i.e. angiotensin II, which activates the neural circuit for drinking behavior through its action on the subfornical organ.  As indicated above, the subfornical organ is also an intermediary structure in another important mechanism for fluid homeostasis, i.e. the place through which blood-borne angiotensin II (which is released in increased amount in response to reduced blood volume) can activate the vasopressin system to reduce the loss of water through the kidney.

 

 

13. Temperature Regulation

 

In order to sense changes in body temperature and in the surrounding temperature, thermoreceptors are located largely in the skin (free nerve endings) and in the brain, where thermosensitive neurons are found primarily in the preoptic-anterior hypothalamic area (PO-AH).  The thermosensitive neurons in the PO-AH sense the temperature of the blood that passes through this richly vascularized region.  Based on this information and that from various peripheral receptors, neural assemblies both in the anterior and posterior hypothalamus coordinate the activity needed to maintain the body temperature within fairly narrow limits.  In response to increased body temperature, hypothalamic neurons initiate a series of processes that result in heat -loss, including peripheral vasodilatation and sweating.  A drop in the surrounding temperature leads to a series of events including peripheral vasoconstriction, piloerection, increased metabolism and shivering in order to preserve heat. Thermosensitive neurons appear to influence arousal mechanisms integral to regulation of the sleep-wake cycle. For example, Dennis McGinty and colleagues have shown that warming the preoptic region during waking suppresses arousal–related neuronal activity in the caudal hypothalamus and in magnocellular basal telecephalon neurons that have diffuse cortical projections. The consequence of this experimental warming include decreased motor activity, reduced metabolic activity and respiratory rate and enhanced peripheral heat loss. All these effects are similar to changes that characterize the onset of sleep.

The preoptic region of the hypothalamus also plays a role in producing fever. Circulating cytokines may act at or near the OVLT to elicit fever, and prostaglandin injection induces fever has been identified in the region surrounding the OVLT.

 

 

 

Further Reading

 

Aston-Jones et al: A neural circuit for circadian regulation of arousal. Nature Neuroscience 4, 732, 2001.

Berson et al: Phototransduction by retinal ganglion cells that set the circadian clock. Science 295, 1070, 2002.

Card et al: The Hypothalamus: An overview of regulatory systems. In: Fundamental Neuroscience (Zigmond et al., eds), pp 1013-1026.

Akil et al: Neuroendocrine Systems I: Overview- Thyroid and adrenal axes. In: Fundamental Neuroscience (Zigmond et al., eds), pp 1127-1150.

Elmquist et al. Unraveling the central nervous system pathways underlying responses to leptin. Nature Neuroscience 1, 445, 1998.

Frohman et al: Neuroendocrine Systems II: Growth, reproduction and lactation. In Fundamental Neuroscience (Zigmond et al., eds) pp 1151-1187.

Hattar et al: Melanopsin-containing retinal ganglion cells: architecture, projections, and intrinsic photosensitivity. Science 295, 1065, 2002.

Markowska: Sex dimorphism in the rate of age-related decline in spatial memory: Relevance to alterations in the estrous cycle. J. Neuroscience 19, 8122, 1999.

Moore: Circadian timing. In: Fundamental Neuroscience (Zigmond et al., eds) pp 1189-1205.

Saper et al: The sleep switch: hypothalamic control of sleep and wakefulness. Trends Neurosci 24, 726, 2001.

Schwartz et al: Central nervous system control of food intake. Nature 404, 661, 2000.