fbpx
Wikipedia

Hypothalamic–pituitary–adrenal axis

The hypothalamic–pituitary–adrenal axis (HPA axis or HTPA axis) is a complex set of direct influences and feedback interactions among three components: the hypothalamus (a part of the brain located below the thalamus), the pituitary gland (a pea-shaped structure located below the hypothalamus), and the adrenal (also called "suprarenal") glands (small, conical organs on top of the kidneys). These organs and their interactions constitute the HPA axis.

Schematic of the HPA axis (CRH, corticotropin-releasing hormone; ACTH, adrenocorticotropic hormone)
Hypothalamus, pituitary gland, and adrenal cortex

The HPA axis is a major neuroendocrine system[1] that controls reactions to stress and regulates many body processes, including digestion, immune responses, mood and emotions, sexual activity, and energy storage and expenditure. It is the common mechanism for interactions among glands, hormones, and parts of the midbrain that mediate the general adaptation syndrome (GAS).[2]

While steroid hormones are produced mainly in vertebrates, the physiological role of the HPA axis and corticosteroids in stress response is so fundamental that analogous systems can be found in invertebrates and monocellular organisms as well.

The HPA axis, hypothalamic–pituitary–gonadal (HPG) axis, hypothalamic–pituitary–thyroid (HPT) axis, and the hypothalamic–neurohypophyseal system are the four major neuroendocrine systems through which the hypothalamus and pituitary direct neuroendocrine function.[1]

Anatomy edit

The key elements of the HPA axis are:[3]

CRH and vasopressin are released from neurosecretory nerve terminals at the median eminence. CRH is transported to the anterior pituitary through the portal blood vessel system of the hypophyseal stalk and vasopressin is transported by axonal transport to the posterior pituitary gland. There, CRH and vasopressin act synergistically to stimulate the secretion of stored ACTH from corticotrope cells. ACTH is transported by the blood to the adrenal cortex of the adrenal gland, where it rapidly stimulates the biosynthesis of corticosteroids such as cortisol from cholesterol. Cortisol is a major stress hormone and has effects on many tissues in the body, including the brain. In the brain, cortisol acts on two types of receptors: mineralocorticoid receptors and glucocorticoid receptors, and these are expressed by many different types of neurons. One important target of glucocorticoids is the hypothalamus, which is a major controlling centre of the HPA axis.[4]

Vasopressin can be thought of as "water conservation hormone" and is also known as "antidiuretic hormone(ADH)". It is released when the body is dehydrated and has potent water-conserving effects on the kidney. It is also a potent vasoconstrictor.[5]

Important to the function of the HPA axis are some of the following feedback loops:

  • Cortisol produced in the adrenal cortex will negatively feedback to inhibit both the hypothalamus and the pituitary gland. This reduces the secretion of CRH and vasopressin, and also directly reduces the cleavage of proopiomelanocortin (POMC) into ACTH and β-endorphins.
  • Epinephrine and norepinephrine (E/NE) are produced by the adrenal medulla through sympathetic stimulation and the local effects of cortisol (upregulation enzymes to make E/NE). E/NE will positively feedback to the pituitary and increase the breakdown of POMCs into ACTH and β-endorphins.

Function edit

Release of corticotropin-releasing hormone (CRH) from the hypothalamus is influenced by stress, physical activity, illness, by blood levels of cortisol and by the sleep/wake cycle (circadian rhythm). In healthy individuals, cortisol rises rapidly after wakening, reaching a peak within 30–45 minutes. It then gradually falls over the day, rising again in late afternoon. Cortisol levels then fall in late evening, reaching a trough during the middle of the night. This corresponds to the rest-activity cycle of the organism.[6] An abnormally flattened circadian cortisol cycle has been linked with chronic fatigue syndrome,[7] insomnia[8] and burnout.[9]

The HPA axis has a central role in regulating many homeostatic systems in the body, including the metabolic system, cardiovascular system, immune system, reproductive system and central nervous system. The HPA axis integrates physical and psychosocial influences in order to allow an organism to adapt effectively to its environment, use resources, and optimize survival.[6]

Anatomical connections between brain areas such as the amygdala, hippocampus, prefrontal cortex and hypothalamus facilitate activation of the HPA axis.[10] Sensory information arriving at the lateral aspect of the amygdala is processed and conveyed to the amygdala's central nucleus, which then projects out to several parts of the brain involved in responses to fear. At the hypothalamus, fear-signaling impulses activate both the sympathetic nervous system and the modulating systems of the HPA axis.

Increased production of cortisol during stress results in an increased availability of glucose in order to facilitate fighting or fleeing. As well as directly increasing glucose availability, cortisol also suppresses the highly demanding metabolic processes of the immune system, resulting in further availability of glucose.[6]

Glucocorticoids have many important functions, including modulation of stress reactions, but in excess they can be damaging. Atrophy of the hippocampus in humans and animals exposed to severe stress is believed to be caused by prolonged exposure to high concentrations of glucocorticoids. Deficiencies of the hippocampus may reduce the memory resources available to help a body formulate appropriate reactions to stress.[11]

Immune system edit

There is bi-directional communication and feedback between the HPA axis and the immune system. A number of cytokines, such as IL-1, IL-6, IL-10 and TNF-alpha can activate the HPA axis, although IL-1 is the most potent. The HPA axis in turn modulates the immune response, with high levels of cortisol resulting in a suppression of immune and inflammatory reactions. This helps to protect the organism from a lethal overactivation of the immune system, and minimizes tissue damage from inflammation.[6]

In many ways, the CNS is "immune privileged", but it plays an important role in the immune system and is affected by it in turn. The CNS regulates the immune system through neuroendocrine pathways, such as the HPA axis. The HPA axis is responsible for modulating inflammatory responses that occur throughout the body.[12][13]

During an immune response, proinflammatory cytokines (e.g. IL-1) are released into the peripheral circulation system and can pass through the blood–brain barrier where they can interact with the brain and activate the HPA axis.[13][14][15] Interactions between the proinflammatory cytokines and the brain can alter the metabolic activity of neurotransmitters and cause symptoms such as fatigue, depression, and mood changes.[13][14] Deficiencies in the HPA axis may play a role in allergies and inflammatory/ autoimmune diseases, such as rheumatoid arthritis and multiple sclerosis.[12][13][16]

When the HPA axis is activated by stressors, such as an immune response, high levels of glucocorticoids are released into the body and suppress immune response by inhibiting the expression of proinflammatory cytokines (e.g. IL-1, TNF alpha, and IFN gamma) and increasing the levels of anti-inflammatory cytokines (e.g. IL-4, IL-10, and IL-13) in immune cells, such as monocytes and neutrophils.[13][14][16][17]

The relationship between chronic stress and its concomitant activation of the HPA axis, and dysfunction of the immune system is unclear; studies have found both immunosuppression and hyperactivation of the immune response.[17]

Stress edit

 
Schematic overview of the hypothalamic-pituitary-adrenal (HPA) axis. Stress activates the HPA-axis and thereby enhances the secretion of glucocorticoids from the adrenals.

Stress and disease edit

The HPA axis is involved in the neurobiology and pathophysiology of mood disorders and functional illnesses, including anxiety disorder, bipolar disorder, insomnia, posttraumatic stress disorder, borderline personality disorder, ADHD, major depressive disorder, burnout, chronic fatigue syndrome, fibromyalgia, irritable bowel syndrome, and alcoholism.[18][19] Antidepressants, which are routinely prescribed for many of these illnesses, serve to regulate HPA axis function.[20]

Sex differences are prevalent in humans with respect to psychiatric stress-related disorders such as anxiety and depression, where women are diagnosed with these disorders more often than men.[21] One rodents study found that females may lack the ability to tolerate as well as process stress (particularly for chronic stress) due to possible down regulation of glucocorticoid receptor expression as well as a deficiency of FKBP51 binding protein in the cytosol. By constantly activating the HPA axis, this could lead to higher instances of stress and disorders that would only get worse with chronic stress.[22] Specifically in this rodent study, females showed greater activation of the HPA axis following stress than males. These differences also likely arise due to the opposing actions that certain sex steroids have, such as testosterone and oestrogen. Oestrogen functions to enhance stress-activated ACTH and CORT secretion while testosterone functions to decrease HPA axis activation and works to inhibit both ACTH and CORT responses to stress.[23] However, more studies are required to better understand the underlying basis of these sex differences.

Experimental studies have investigated many different types of stress, and their effects on the HPA axis in many different circumstances.[24] Stressors can be of many different types—in experimental studies in rats, a distinction is often made between "social stress" and "physical stress", but both types activate the HPA axis, though via different pathways.[25] Several monoamine neurotransmitters are important in regulating the HPA axis, especially dopamine, serotonin and norepinephrine (noradrenaline). There is evidence that an increase in oxytocin, resulting for instance from positive social interactions, acts to suppress the HPA axis and thereby counteracts stress, promoting positive health effects such as wound healing.[26]

The HPA axis is a feature of mammals and other vertebrates. For example, biologists studying stress in fish showed that social subordination leads to chronic stress, related to reduced aggressive interactions, to lack of control, and to the constant threat imposed by dominant fish. Serotonin (5-HT) appeared to be the active neurotransmitter involved in mediating stress responses, and increases in serotonin are related to increased plasma α-MSH levels, which causes skin darkening (a social signal in salmonoid fish), activation of the HPA axis, and inhibition of aggression. Inclusion of the amino acid L-tryptophan, a precursor of 5-HT, in the feed of rainbow trout made the trout less aggressive and less responsive to stress.[27] However, the study mentions that plasma cortisol was not affected by dietary L-tryptophan. The drug LY354740 (also known as Eglumegad, an agonist of the metabotropic glutamate receptors 2 and 3) has been shown to interfere in the HPA axis, with chronic oral administration of this drug leading to markedly reduced baseline cortisol levels in bonnet macaques (Macaca radiata); acute infusion of LY354740 resulted in a marked diminution of yohimbine-induced stress response in those animals.[28]

Studies on people show that the HPA axis is activated in different ways during chronic stress depending on the type of stressor, the person's response to the stressor and other factors. Stressors that are uncontrollable, threaten physical integrity, or involve trauma tend to have a high, flat diurnal profile of cortisol release (with lower-than-normal levels of cortisol in the morning and higher-than-normal levels in the evening) resulting in a high overall level of daily cortisol release. On the other hand, controllable stressors tend to produce higher-than-normal morning cortisol. Stress hormone release tends to decline gradually after a stressor occurs. In post-traumatic stress disorder there appears to be lower-than-normal cortisol release, and it is thought that a blunted hormonal response to stress may predispose a person to develop PTSD.[29]

It is also known that HPA axis hormones are related to certain skin diseases and skin homeostasis. There is evidence shown that the HPA axis hormones can be linked to certain stress related skin diseases and skin tumors. This happens when HPA axis hormones become hyperactive in the brain.[30]

Stress and development edit

Prenatal stress edit

There is evidence that prenatal stress can influence HPA regulation. In animal experiments, exposure to prenatal stress has been shown to cause a hyper-reactive HPA stress response. Rats that have been prenatally stressed have elevated basal levels and abnormal circadian rhythm of corticosterone as adults.[31] Additionally, they require a longer time for their stress hormone levels to return to baseline following exposure to both acute and prolonged stressors. Prenatally stressed animals also show abnormally high blood glucose levels and have fewer glucocorticoid receptors in the hippocampus.[32] In humans, prolonged maternal stress during gestation is associated with mild impairment of intellectual activity and language development in their children, and with behavior disorders such as attention deficits, schizophrenia, anxiety and depression; self-reported maternal stress is associated with a higher irritability, emotional and attentional problems.[33]

There is growing evidence that prenatal stress can affect HPA regulation in humans. Children who were stressed prenatally may show altered cortisol rhythms. For example, several studies have found an association between maternal depression during pregnancy and childhood cortisol levels.[34] Prenatal stress has also been implicated in a tendency toward depression and short attention span in childhood.[35]

Early life stress edit

The role of early life stress in programming the HPA axis has been well-studied in animal models. Exposure to mild or moderate stressors early in life has been shown to enhance HPA regulation and promote a lifelong resilience to stress. In contrast, early-life exposure to extreme or prolonged stress can induce a hyper-reactive HPA axis and may contribute to lifelong vulnerability to stress.[36] In one widely replicated experiment, rats subjected to the moderate stress of frequent human handling during the first two weeks of life had reduced hormonal and behavioral HPA-mediated stress responses as adults, whereas rats subjected to the extreme stress of prolonged periods of maternal separation showed heightened physiological and behavioral stress responses as adults.[37]

Several mechanisms have been proposed to explain these findings in rat models of early-life stress exposure. There may be a critical period during development during which the level of stress hormones in the bloodstream contribute to the permanent calibration of the HPA axis. One experiment has shown that, even in the absence of any environmental stressors, early-life exposure to moderate levels of corticosterone was associated with stress resilience in adult rats, whereas exposure to high doses was associated with stress vulnerability.[38]

Another possibility is that the effects of early-life stress on HPA functioning are mediated by maternal care. Frequent human handling of the rat pups may cause their mother to exhibit more nurturant behavior, such as licking and grooming. Nurturant maternal care, in turn, may enhance HPA functioning in at least two ways. First, maternal care is crucial in maintaining the normal stress hypo responsive period (SHRP), which in rodents, is the first two weeks of life during which the HPA axis is generally non-reactive to stress. Maintenance of the SHRP period may be critical for HPA development, and the extreme stress of maternal separation, which disrupts the SHRP, may lead to permanent HPA dysregulation.[39] Another way that maternal care might influence HPA regulation is by causing epigenetic changes in the offspring. For example, increased maternal licking and grooming has been shown to alter expression of the glucocorticoid receptor gene implicated in adaptive stress response.[36] At least one human study has identified maternal neural activity patterns in response to video stimuli of mother-infant separation as being associated with decreased glucocorticoid receptor gene methylation in the context of post-traumatic stress disorder stemming from early life stress.[40] Yet clearly, more research is needed to determine if the results seen in cross-generational animal models can be extended to humans.

Though animal models allow for more control of experimental manipulation, the effects of early life stress on HPA axis function in humans has also been studied. One population that is often studied in this type of research is adult survivors of childhood abuse. Adult survivors of childhood abuse have exhibited increased ACTH concentrations in response to a psychosocial stress task compared to unaffected controls and subjects with depression but not childhood abuse.[41] In one study, adult survivors of childhood abuse that are not depressed show increased ACTH response to both exogenous CRF and normal cortisol release. Adult survivors of childhood abuse that are depressed show a blunted ACTH response to exogenous CRH.[42] A blunted ACTH response is common in depression, so the authors of this work posit that this pattern is likely to be due to the participant's depression and not their exposure to early life stress.

Heim and colleagues have proposed that early life stress, such as childhood abuse, can induce a sensitization of the HPA axis, resulting in particular heightened neuronal activity in response to stress-induced CRH release.[42] With repeated exposure to stress, the sensitized HPA axis may continue to hypersecrete CRH from the hypothalamus. Over time, CRH receptors in the anterior pituitary will become down-regulated, producing depression and anxiety symptoms.[42] This research in human subjects is consistent with the animal literature discussed above.

The HPA axis was present in the earliest vertebrate species, and has remained highly conserved by strong positive selection due to its critical adaptive roles.[43] The programming of the HPA axis is strongly influenced by the perinatal and early juvenile environment, or "early-life environment".[44][45][46] Maternal stress and differential degrees of caregiving may constitute early life adversity, which has been shown to profoundly influence, if not permanently alter, the offspring's stress and emotional regulating systems.[44][45] Widely studied in animal models (e.g. licking and grooming/LG in rat pups),[47] the consistency of maternal care has been shown to have a powerful influence on the offspring's neurobiology, physiology, and behavior. Whereas maternal care improves cardiac response, sleep/wake rhythm, and growth hormone secretion in the neonate, it also suppresses HPA axis activity. In this manner, maternal care negatively regulates stress response in the neonate,[47] thereby shaping his/her susceptibility to stress in later life. These programming effects are not deterministic, as the environment in which the individual develops can either match or mismatch with the former's "programmed" and genetically predisposed HPA axis reactivity. Although the primary mediators of the HPA axis are known, the exact mechanism by which its programming can be modulated during early life remains to be elucidated. Furthermore, evolutionary biologists contest the exact adaptive value of such programming, i.e. whether heightened HPA axis reactivity may confer greater evolutionary fitness.

Various hypotheses have been proposed, in attempts to explain why early life adversity can produce outcomes ranging from extreme vulnerability to resilience in the face of later stress. Glucocorticoids produced by the HPA axis have been proposed to confer either a protective or harmful role, depending on an individual's genetic predispositions, programming effects of early-life environment, and match or mismatch with one's postnatal environment. The predictive adaptation hypothesis (1), the three-hit concept of vulnerability and resilience (2) and the maternal mediation hypothesis (3) attempt to elucidate how early life adversity can differentially predict vulnerability or resilience in the face of significant stress in later life.[48] These hypotheses are not mutually exclusive but rather are highly interrelated and unique to the individual.

(1) The predictive adaptation hypothesis:[48] This hypothesis is in direct contrast with the diathesis stress model, which posits that the accumulation of stressors across a lifespan can enhance the development of psychopathology once a threshold is crossed. Predictive adaptation asserts that early life experience induces epigenetic change; these changes predict or "set the stage" for adaptive responses that will be required in their environment. Thus, if a developing child (i.e., fetus to neonate) is exposed to ongoing maternal stress and low levels of maternal care (i.e., early life adversity), this will program their HPA axis to be more reactive to stress. This programming will have predicted, and potentially be adaptive in a highly stressful, precarious environment during childhood and later life. The predictability of these epigenetic changes is not definitive, however – depending primarily on the degree to which the individual's genetic and epigenetically modulated phenotype "matches" or "mismatches" with their environment (See: Hypothesis (2)).

(2) Three-Hit Concept of vulnerability and resilience:[48] this hypothesis states that within a specific life context, vulnerability may be enhanced with chronic failure to cope with ongoing adversity. It fundamentally seeks to explicate why, under seemingly indistinguishable circumstances, one individual may cope resiliently with stress, whereas another may not only cope poorly, but consequently develop a stress-related mental illness. The three "hits" – chronological and synergistic – are as follows: genetic predisposition (which predispose higher/lower HPA axis reactivity), early-life environment (perinatal – i.e. maternal stress, and postnatal – i.e. maternal care), and later-life environment (which determines match/mismatch, as well as a window for neuroplastic changes in early programming).[49] The concept of match/mismatch is central to this evolutionary hypothesis. In this context, it elucidates why early life programming in the perinatal and postnatal period may have been evolutionarily selected for. Specifically, by instating specific patterns of HPA axis activation, the individual may be more well-equipped to cope with adversity in a high-stress environment. Conversely, if an individual is exposed to significant early life adversity, heightened HPA axis reactivity may "mismatch" them in an environment characterized by low stress. The latter scenario may represent maladaptation due to early programming, genetic predisposition, and mismatch. This mismatch may then predict negative developmental outcomes such as psychopathologies in later life.

(2) Maternal mediation hypothesis:[38] This hypothesis states that maternal care is the primary factor in developing stress resistance later in life.

Ultimately, the conservation of the HPA axis has underscored its critical adaptive roles in vertebrates, so, too, various invertebrate species over time. The HPA axis plays a clear role in the production of corticosteroids, which govern many facets of brain development and responses to ongoing environmental stress. With these findings, animal model research has served to identify what these roles are – with regards to animal development and evolutionary adaptation. In more precarious, primitive times, a heightened HPA axis may have served to protect organisms from predators and extreme environmental conditions, such as weather and natural disasters, by encouraging migration (i.e. fleeing), the mobilization of energy, learning (in the face of novel, dangerous stimuli) as well as increased appetite for biochemical energy storage. In contemporary society, the endurance of the HPA axis and early life programming will have important implications for counseling expecting and new mothers, as well as individuals who may have experienced significant early life adversity.[49]

See also edit

Other major neuroendocrine systems
Related topics
Conditions

References edit

  1. ^ a b Malenka RC, Nestler EJ, Hyman SE (2009). "Chapter 10: Neural and Neuroendocrine Control of the Internal Milieu". In Sydor A, Brown RY (ed.). Molecular Neuropharmacology: A Foundation for Clinical Neuroscience (2nd ed.). New York: McGraw-Hill Medical. pp. 246, 248–259. ISBN 9780071481274.
    •The hypothalamic–neurohypophyseal system secretes two peptide hormones directly into the blood, vasopressin and oxytocin. ...
    •The hypothalamic–pituitary–adrenal (HPA) axis. It comprises corticotropin-releasing factor (CRF), released by the hypothalamus; adrenocorticotropic hormone (ACTH), released by the anterior pituitary; and glucocorticoids, released by the adrenal cortex.
    •The hypothalamic–pituitary–thyroid axis consists of hypothalamic thyrotropin-releasing hormone (TRH); the anterior pituitary hormone thyroid–stimulating hormone (TSH); and the thyroid hormones T3 and T4.
    •The hypothalamic–pituitary–gonadal axis comprises hypothalamic gonadotropin–releasing hormone (GnRH), the anterior pituitary luteinizing hormone (LH) and follicle-stimulating hormone (FSH), and the gonadal steroids.
  2. ^ Selye, Hans (1974). Stress without distress. Philadelphia: Lippincott. ISBN 978-0-397-01026-4.[page needed]
  3. ^ "Getting to know the HPA axis". www.nrdc.org. 2015-05-18. from the original on 2023-08-10. Retrieved 2023-08-08.
  4. ^ Tasker, Jeffrey G.; Herman, James P. (14 July 2011). "Mechanisms of rapid glucocorticoid feedback inhibition of the hypothalamic–pituitary–adrenal axis". Stress. 14 (4): 398–406. doi:10.3109/10253890.2011.586446. PMC 4675656. PMID 21663538.
  5. ^ Cuzzo, Brian; Lappin, Sarah L. (2019), "Vasopressin (Antidiuretic Hormone, ADH)", StatPearls, StatPearls Publishing, PMID 30252325, from the original on 2021-03-25, retrieved 2019-10-19
  6. ^ a b c d del Rey, A.; Chrousos, G. P.; Besedovsky, H. O., eds. (2008). The Hypothalamus-Pituitary-Adrenal Axis. NeuroImmune Biology. Vol. 7. Berczi, I.; Szentivanyi A., series EICs. Amsterdam London: Elsevier Science. ISBN 978-0-08-055936-0. OCLC 272388790. from the original on 10 August 2023. Retrieved 27 February 2022.
  7. ^ MacHale SM, Cavanagh JT, Bennie J, Carroll S, Goodwin GM, Lawrie SM (November 1998). "Diurnal variation of adrenocortical activity in chronic fatigue syndrome". Neuropsychobiology. 38 (4): 213–7. doi:10.1159/000026543. PMID 9813459. S2CID 46856991.
  8. ^ Backhaus J, Junghanns K, Hohagen F (October 2004). "Sleep disturbances are correlated with decreased morning awakening salivary cortisol". Psychoneuroendocrinology. 29 (9): 1184–91. doi:10.1016/j.psyneuen.2004.01.010. PMID 15219642. S2CID 14756991.
  9. ^ Pruessner JC, Hellhammer DH, Kirschbaum C (1999). "Burnout, perceived stress, and cortisol responses to awakening". Psychosom Med. 61 (2): 197–204. doi:10.1097/00006842-199903000-00012. PMID 10204973.
  10. ^ Laura, Freberg (2015-01-01). Discovering behavioral neuroscience : an introduction to biological psychology. Freberg, Laura,, Container of (work): Freberg, Laura. (Third ed.). Boston, MA. p. 504. ISBN 9781305088702. OCLC 905734838.{{cite book}}: CS1 maint: location missing publisher (link)
  11. ^ Frankiensztajn, Linoy Mia; Elliott, Evan; Koren, Omry (June 2020). "The microbiota and the hypothalamus-pituitary-adrenocortical (HPA) axis, implications for anxiety and stress disorders". Current Opinion in Neurobiology. 62: 76–82. doi:10.1016/j.conb.2019.12.003. PMID 31972462. S2CID 210836469.
  12. ^ a b Marques-Deak, A; Cizza, G; Sternberg, E (February 2005). "Brain-immune interactions and disease susceptibility". Molecular Psychiatry. 10 (3): 239–250. doi:10.1038/sj.mp.4001643. PMID 15685252. S2CID 17978810.
  13. ^ a b c d e Otmishi, Peyman; Gordon, Josiah; El-Oshar, Seraj; Li, Huafeng; Guardiola, Juan; Saad, Mohamed; Proctor, Mary; Yu, Jerry (2008). "Neuroimmune Interaction in Inflammatory Diseases". Clinical Medicine: Circulatory, Respiratory, and Pulmonary Medicine. 2: 35–44. doi:10.4137/ccrpm.s547. PMC 2990232. PMID 21157520.
  14. ^ a b c Tian, Rui; Hou, Gonglin; Li, Dan; Yuan, Ti-Fei (June 2014). "A Possible Change Process of Inflammatory Cytokines in the prolonged Chronic Stress and its Ultimate Implications for Health". The Scientific World Journal. 2014: 780616. doi:10.1155/2014/780616. PMC 4065693. PMID 24995360.
  15. ^ Hall, Jessica; Cruser, desAgnes; Podawiltz, Alan; Mummert, Diana; Jones, Harlan; Mummert, Mark (August 2012). "Psychological Stress and the Cutaneous Immune Response: Roles of the HPA Axis and the Sympathetic Nervous System in Atopic Dermatitis and Psoriasis". Dermatology Research and Practice. 2012: 403908. doi:10.1155/2012/403908. PMC 3437281. PMID 22969795.
  16. ^ a b Bellavance, Marc-Andre; Rivest, Serge (March 2014). "The HPA-immune axis and the immunomodulatory actions of glucocorticoids in the brain". Frontiers in Immunology. 5: 136. doi:10.3389/fimmu.2014.00136. PMC 3978367. PMID 24744759.
  17. ^ a b Padgett, David; Glaser, Ronald (August 2003). (PDF). Trends in Immunology. 24 (8): 444–448. doi:10.1016/S1471-4906(03)00173-X. PMID 12909458. Archived from the original (PDF) on 2016-03-27. Retrieved 12 February 2016.
  18. ^ Spencer RL, Hutchison KE (1999). "Alcohol, aging, and the stress response". Alcohol Research & Health. 23 (4): 272–83. PMC 6760387. PMID 10890824.
  19. ^ Smith, Carli J.; Emge, Jacob R.; Berzins, Katrina; Lung, Lydia; Khamishon, Rebecca; Shah, Paarth; Rodrigues, David M.; Sousa, Andrew J.; Reardon, Colin; Sherman, Philip M.; Barrett, Kim E.; Gareau, Mélanie G. (2014-10-15). "Probiotics normalize the gut-brain-microbiota axis in immunodeficient mice". American Journal of Physiology. Gastrointestinal and Liver Physiology. 307 (8): G793–G802. doi:10.1152/ajpgi.00238.2014. ISSN 0193-1857. PMC 4200314. PMID 25190473.
  20. ^ Pariante CM (August 2003). "Depression, stress and the adrenal axis". Journal of Neuroendocrinology. 15 (8): 811–2. doi:10.1046/j.1365-2826.2003.01058.x. PMID 12834443. S2CID 1359479.
  21. ^ Rosinger, Zachary; Jacobskind, Jason; Park, Shannon; Justice, Nicholas; Zuloaga, Damian (2017). "Distribution of Corticotropin-releasing factor receptor 1 in the developing mouse forebrain: A novel sex difference revealed in the rostral periventricular hypothalamus". Neuroscience. 361: 167–178. doi:10.1016/j.neuroscience.2017.08.016. PMC 7173945. PMID 28823817.
  22. ^ Palumbo, Michelle C.; Dominguez, Sky; Dong, Hongxin (2020). "Sex differences in hypothalamic–pituitary–adrenal axis regulation after chronic unpredictable stress". Brain and Behavior. 10 (4): e01586. doi:10.1002/brb3.1586. PMC 7177572. PMID 32154650.
  23. ^ Handa, R. J.; Weiser, M. J.; Zuloaga, D. G. (2009). "A Role for the Androgen Metabolite, 5α-Androstane-3β,17β-Diol, in Modulating Oestrogen Receptor β-Mediated Regulation of Hormonal Stress Reactivity". Journal of Neuroendocrinology. 21 (4): 351–358. doi:10.1111/j.1365-2826.2009.01840.x. PMC 2727750. PMID 19207807.
  24. ^ Douglas AJ (March 2005). "Central noradrenergic mechanisms underlying acute stress responses of the Hypothalamo–pituitary–adrenal axis: adaptations through pregnancy and lactation". Stress. 8 (1): 5–18. doi:10.1080/10253890500044380. PMID 16019594. S2CID 24654645.
  25. ^ Engelmann M, Landgraf R, Wotjak CT (2004). "The hypothalamic-neurohypophysial system regulates the hypothalamic–pituitary–adrenal axis under stress: an old concept revisited". Frontiers in Neuroendocrinology. 25 (3–4): 132–49. doi:10.1016/j.yfrne.2004.09.001. PMID 15589266. S2CID 41983825.
  26. ^ Detillion CE, Craft TK, Glasper ER, Prendergast BJ, DeVries AC (September 2004). "Social facilitation of wound healing". Psychoneuroendocrinology. 29 (8): 1004–11. doi:10.1016/j.psyneuen.2003.10.003. PMID 15219651. S2CID 5986340.
  27. ^ Winberg S, Øverli Ø, Lepage O (November 2001). "Suppression of aggression in rainbow trout (Oncorhynchus mykiss) by dietary L-tryptophan". The Journal of Experimental Biology. 204 (Pt 22): 3867–76. doi:10.1242/jeb.204.22.3867. PMID 11807104.
  28. ^ Coplan JD, Mathew SJ, Smith EL, et al. (July 2001). "Effects of LY354740, a novel glutamatergic metabotropic agonist, on nonhuman primate hypothalamic–pituitary–adrenal axis and noradrenergic function". CNS Spectrums. 6 (7): 607–12, 617. doi:10.1017/S1092852900002157. PMID 15573025. S2CID 6029856.
  29. ^ Miller GE, Chen E, Zhou ES (January 2007). "If it goes up, must it come down? Chronic stress and the hypothalamic–pituitary–adrenocortical axis in humans". Psychological Bulletin. 133 (1): 25–45. doi:10.1037/0033-2909.133.1.25. PMID 17201569.
  30. ^ Kim JE, Cho BK, Cho DH, Park HJ (July 2013). "Expression of hypothalamic–pituitary–adrenal axis in common skin diseases: evidence of its association with stress-related disease activity". Acta Dermato-Venereologica. 93 (4): 387–93. doi:10.2340/00015555-1557. PMID 23462974.
  31. ^ Koehl M, Darnaudéry M, Dulluc J, Van Reeth O, Le Moal M, Maccari S (September 1999). "Prenatal stress alters circadian activity of hypothalamo–pituitary–adrenal axis and hippocampal corticosteroid receptors in adult rats of both gender". Journal of Neurobiology. 40 (3): 302–15. doi:10.1002/(SICI)1097-4695(19990905)40:3<302::AID-NEU3>3.0.CO;2-7. PMID 10440731.
  32. ^ Weinstock M, Matlina E, Maor GI, Rosen H, McEwen BS (November 1992). "Prenatal stress selectively alters the reactivity of the hypothalamic-pituitary adrenal system in the female rat". Brain Research. 595 (2): 195–200. doi:10.1016/0006-8993(92)91049-K. PMID 1467966. S2CID 44382315.
  33. ^ Weinstock M (August 2008). "The long-term behavioural consequences of prenatal stress" (PDF). Neuroscience and Biobehavioral Reviews. 32 (6): 1073–86. doi:10.1016/j.neubiorev.2008.03.002. PMID 18423592. S2CID 3717977. (PDF) from the original on 2023-08-10. Retrieved 2014-05-04.
  34. ^ Gutteling BM, de Weerth C, Buitelaar JK (December 2004). "Maternal prenatal stress and 4-6 year old children's salivary cortisol concentrations pre- and post-vaccination". Stress. 7 (4): 257–60. doi:10.1080/10253890500044521. hdl:2066/54819. PMID 16019591. S2CID 38412082.
  35. ^ Buitelaar JK, Huizink AC, Mulder EJ, de Medina PG, Visser GH (2003). "Prenatal stress and cognitive development and temperament in infants". Neurobiology of Aging. 24 (Suppl 1): S53–60, discussion S67–8. doi:10.1016/S0197-4580(03)00050-2. PMID 12829109. S2CID 3008063.
  36. ^ a b Flinn MV, Nepomnaschy PA, Muehlenbein MP, Ponzi D (June 2011). "Evolutionary functions of early social modulation of hypothalamic–pituitary–adrenal axis development in humans". Neurosci Biobehav Rev. 35 (7): 1611–29. doi:10.1016/j.neubiorev.2011.01.005. PMID 21251923. S2CID 16950714.
  37. ^ Liu D, Diorio J, Tannenbaum B, et al. (September 1997). "Maternal care, hippocampal glucocorticoid receptors, and hypothalamic–pituitary–adrenal responses to stress". Science. 277 (5332): 1659–62. doi:10.1126/science.277.5332.1659. PMID 9287218. S2CID 2753764.
  38. ^ a b Macrì S, Würbel H (December 2006). "Developmental plasticity of HPA and fear responses in rats: a critical review of the maternal mediation hypothesis". Hormones and Behavior. 50 (5): 667–80. doi:10.1016/j.yhbeh.2006.06.015. PMID 16890940. S2CID 36877565.
  39. ^ de Kloet ER, Sibug RM, Helmerhorst FM, Schmidt MV, Schmidt M (April 2005). "Stress, genes and the mechanism of programming the brain for later life". Neuroscience and Biobehavioral Reviews. 29 (2): 271–81. doi:10.1016/j.neubiorev.2004.10.008. PMID 15811498. S2CID 24943895.
  40. ^ Schechter, Daniel S.; Moser, Dominik A.; Paoloni-Giacobino, Ariane; Stenz, Ludwig; Gex-Fabry, Marianne; Aue, Tatjana; Adouan, Wafae; Cordero, María I.; Suardi, Francesca; Manini, Aurelia; Sancho Rossignol, Ana (2015). "Methylation of NR3C1 is related to maternal PTSD, parenting stress and maternal medial prefrontal cortical activity in response to child separation among mothers with histories of violence exposure". Frontiers in Psychology. 6: 690. doi:10.3389/fpsyg.2015.00690. ISSN 1664-1078. PMC 4447998. PMID 26074844.
  41. ^ Heim C.; Newport D. J.; Heit S.; Graham Y. P.; Wilcox M.; Bonsall R.; Nemeroff C. B. (2000). "Pituitary-adrenal and autonomic responses to stress in women after sexual and physical abuse in childhood". JAMA. 284 (5): 592–597. doi:10.1001/jama.284.5.592. PMID 10918705.
  42. ^ a b c Heim C.; Newport D.J.; Bonsall R.; Miller A.H.; Nemeroff C.B. (2001). "Altered Pituitary-Adrenal Axis Responses to Provocative Challenge Tests in Adult Survivors of Childhood Abuse". Am J Psychiatry. 158 (4): 575–581. doi:10.1176/appi.ajp.158.4.575. PMID 11282691.
  43. ^ Denver RJ (Apr 2009). "Structural and functional evolution of vertebrate neuroendocrine stress systems" (PDF). Ann N Y Acad Sci. 1163 (1): 1–16. Bibcode:2009NYASA1163....1D. doi:10.1111/j.1749-6632.2009.04433.x. hdl:2027.42/74370. PMID 19456324. S2CID 18786346. from the original on 2023-08-10. Retrieved 2019-09-01.
  44. ^ a b Oitzl MS, Champagne DL, van der Veen R, de Kloet ER (May 2010). "Brain development under stress: hypotheses of glucocorticoid actions revisited". Neurosci Biobehav Rev. 34 (6): 853–66. doi:10.1016/j.neubiorev.2009.07.006. PMID 19631685. S2CID 25898149.
  45. ^ a b Horton TH (Jan 2005). "Fetal origins of developmental plasticity: animal models of induced life history variation". Am. J. Hum. Biol. 17 (1): 34–43. doi:10.1002/ajhb.20092. PMID 15611963. S2CID 23466894.
  46. ^ Matthews SG (Mar 2000). "Antenatal glucocorticoids and programming of the developing CNS". Pediatr Res. 47 (3): 291–300. doi:10.1203/00006450-200003000-00003. PMID 10709726.
  47. ^ a b Champagne FA, Francis DD, Mar A, Meaney MJ (Aug 2003). "Variations in maternal care in the rat as a mediating influence for the effects of environment on development". Physiol Behav. 79 (3): 359–71. CiteSeerX 10.1.1.335.3199. doi:10.1016/s0031-9384(03)00149-5. PMID 12954431. S2CID 18599019.
  48. ^ a b c Daskalakis NP, Bagot RC, Parker KJ, Vinkers CH, de Kloet ER (Sep 2013). "The three-hit concept of vulnerability and resilience: toward understanding adaptation to early-life adversity outcome". Psychoneuroendocrinology. 38 (9): 1858–73. doi:10.1016/j.psyneuen.2013.06.008. PMC 3773020. PMID 23838101.
  49. ^ a b Roth TL, Matt S, Chen K, Blaze J (Dec 2014). "Bdnf DNA methylation modifications in the hippocampus and amygdala of male and female rats exposed to different caregiving environments outside the homecage". Dev Psychobiol. 56 (8): 1755–63. doi:10.1002/dev.21218. PMC 4205217. PMID 24752649.

External links edit

  • Mind-Body-Health.net page on HPA axis
  • HPA Axis: Explanation of the Body's Central Stress Response System w/Diagram

hypothalamic, pituitary, adrenal, axis, hypothalamic, pituitary, adrenal, axis, axis, htpa, axis, complex, direct, influences, feedback, interactions, among, three, components, hypothalamus, part, brain, located, below, thalamus, pituitary, gland, shaped, stru. The hypothalamic pituitary adrenal axis HPA axis or HTPA axis is a complex set of direct influences and feedback interactions among three components the hypothalamus a part of the brain located below the thalamus the pituitary gland a pea shaped structure located below the hypothalamus and the adrenal also called suprarenal glands small conical organs on top of the kidneys These organs and their interactions constitute the HPA axis Schematic of the HPA axis CRH corticotropin releasing hormone ACTH adrenocorticotropic hormone Hypothalamus pituitary gland and adrenal cortexThe HPA axis is a major neuroendocrine system 1 that controls reactions to stress and regulates many body processes including digestion immune responses mood and emotions sexual activity and energy storage and expenditure It is the common mechanism for interactions among glands hormones and parts of the midbrain that mediate the general adaptation syndrome GAS 2 While steroid hormones are produced mainly in vertebrates the physiological role of the HPA axis and corticosteroids in stress response is so fundamental that analogous systems can be found in invertebrates and monocellular organisms as well The HPA axis hypothalamic pituitary gonadal HPG axis hypothalamic pituitary thyroid HPT axis and the hypothalamic neurohypophyseal system are the four major neuroendocrine systems through which the hypothalamus and pituitary direct neuroendocrine function 1 Contents 1 Anatomy 2 Function 3 Immune system 4 Stress 4 1 Stress and disease 4 2 Stress and development 4 2 1 Prenatal stress 4 2 2 Early life stress 5 See also 6 References 7 External linksAnatomy editThe key elements of the HPA axis are 3 The paraventricular nucleus of the hypothalamus It contains neuroendocrine neurons which synthesize and secrete vasopressin and corticotropin releasing hormone CRH The anterior lobe of the pituitary gland CRH and vasopressin stimulate the anterior lobe of pituitary gland to secrete adrenocorticotropic hormone ACTH once known as corticotropin The adrenal cortex It produces glucocorticoid hormones mainly cortisol in humans in response to stimulation by ACTH Glucocorticoids in turn act back on the hypothalamus and pituitary to suppress CRH and ACTH production in a negative feedback cycle CRH and vasopressin are released from neurosecretory nerve terminals at the median eminence CRH is transported to the anterior pituitary through the portal blood vessel system of the hypophyseal stalk and vasopressin is transported by axonal transport to the posterior pituitary gland There CRH and vasopressin act synergistically to stimulate the secretion of stored ACTH from corticotrope cells ACTH is transported by the blood to the adrenal cortex of the adrenal gland where it rapidly stimulates the biosynthesis of corticosteroids such as cortisol from cholesterol Cortisol is a major stress hormone and has effects on many tissues in the body including the brain In the brain cortisol acts on two types of receptors mineralocorticoid receptors and glucocorticoid receptors and these are expressed by many different types of neurons One important target of glucocorticoids is the hypothalamus which is a major controlling centre of the HPA axis 4 Vasopressin can be thought of as water conservation hormone and is also known as antidiuretic hormone ADH It is released when the body is dehydrated and has potent water conserving effects on the kidney It is also a potent vasoconstrictor 5 Important to the function of the HPA axis are some of the following feedback loops Cortisol produced in the adrenal cortex will negatively feedback to inhibit both the hypothalamus and the pituitary gland This reduces the secretion of CRH and vasopressin and also directly reduces the cleavage of proopiomelanocortin POMC into ACTH and b endorphins Epinephrine and norepinephrine E NE are produced by the adrenal medulla through sympathetic stimulation and the local effects of cortisol upregulation enzymes to make E NE E NE will positively feedback to the pituitary and increase the breakdown of POMCs into ACTH and b endorphins Function editRelease of corticotropin releasing hormone CRH from the hypothalamus is influenced by stress physical activity illness by blood levels of cortisol and by the sleep wake cycle circadian rhythm In healthy individuals cortisol rises rapidly after wakening reaching a peak within 30 45 minutes It then gradually falls over the day rising again in late afternoon Cortisol levels then fall in late evening reaching a trough during the middle of the night This corresponds to the rest activity cycle of the organism 6 An abnormally flattened circadian cortisol cycle has been linked with chronic fatigue syndrome 7 insomnia 8 and burnout 9 The HPA axis has a central role in regulating many homeostatic systems in the body including the metabolic system cardiovascular system immune system reproductive system and central nervous system The HPA axis integrates physical and psychosocial influences in order to allow an organism to adapt effectively to its environment use resources and optimize survival 6 Anatomical connections between brain areas such as the amygdala hippocampus prefrontal cortex and hypothalamus facilitate activation of the HPA axis 10 Sensory information arriving at the lateral aspect of the amygdala is processed and conveyed to the amygdala s central nucleus which then projects out to several parts of the brain involved in responses to fear At the hypothalamus fear signaling impulses activate both the sympathetic nervous system and the modulating systems of the HPA axis Increased production of cortisol during stress results in an increased availability of glucose in order to facilitate fighting or fleeing As well as directly increasing glucose availability cortisol also suppresses the highly demanding metabolic processes of the immune system resulting in further availability of glucose 6 Glucocorticoids have many important functions including modulation of stress reactions but in excess they can be damaging Atrophy of the hippocampus in humans and animals exposed to severe stress is believed to be caused by prolonged exposure to high concentrations of glucocorticoids Deficiencies of the hippocampus may reduce the memory resources available to help a body formulate appropriate reactions to stress 11 Immune system editThere is bi directional communication and feedback between the HPA axis and the immune system A number of cytokines such as IL 1 IL 6 IL 10 and TNF alpha can activate the HPA axis although IL 1 is the most potent The HPA axis in turn modulates the immune response with high levels of cortisol resulting in a suppression of immune and inflammatory reactions This helps to protect the organism from a lethal overactivation of the immune system and minimizes tissue damage from inflammation 6 In many ways the CNS is immune privileged but it plays an important role in the immune system and is affected by it in turn The CNS regulates the immune system through neuroendocrine pathways such as the HPA axis The HPA axis is responsible for modulating inflammatory responses that occur throughout the body 12 13 During an immune response proinflammatory cytokines e g IL 1 are released into the peripheral circulation system and can pass through the blood brain barrier where they can interact with the brain and activate the HPA axis 13 14 15 Interactions between the proinflammatory cytokines and the brain can alter the metabolic activity of neurotransmitters and cause symptoms such as fatigue depression and mood changes 13 14 Deficiencies in the HPA axis may play a role in allergies and inflammatory autoimmune diseases such as rheumatoid arthritis and multiple sclerosis 12 13 16 When the HPA axis is activated by stressors such as an immune response high levels of glucocorticoids are released into the body and suppress immune response by inhibiting the expression of proinflammatory cytokines e g IL 1 TNF alpha and IFN gamma and increasing the levels of anti inflammatory cytokines e g IL 4 IL 10 and IL 13 in immune cells such as monocytes and neutrophils 13 14 16 17 The relationship between chronic stress and its concomitant activation of the HPA axis and dysfunction of the immune system is unclear studies have found both immunosuppression and hyperactivation of the immune response 17 Stress edit nbsp Schematic overview of the hypothalamic pituitary adrenal HPA axis Stress activates the HPA axis and thereby enhances the secretion of glucocorticoids from the adrenals Stress and disease edit The HPA axis is involved in the neurobiology and pathophysiology of mood disorders and functional illnesses including anxiety disorder bipolar disorder insomnia posttraumatic stress disorder borderline personality disorder ADHD major depressive disorder burnout chronic fatigue syndrome fibromyalgia irritable bowel syndrome and alcoholism 18 19 Antidepressants which are routinely prescribed for many of these illnesses serve to regulate HPA axis function 20 Sex differences are prevalent in humans with respect to psychiatric stress related disorders such as anxiety and depression where women are diagnosed with these disorders more often than men 21 One rodents study found that females may lack the ability to tolerate as well as process stress particularly for chronic stress due to possible down regulation of glucocorticoid receptor expression as well as a deficiency of FKBP51 binding protein in the cytosol By constantly activating the HPA axis this could lead to higher instances of stress and disorders that would only get worse with chronic stress 22 Specifically in this rodent study females showed greater activation of the HPA axis following stress than males These differences also likely arise due to the opposing actions that certain sex steroids have such as testosterone and oestrogen Oestrogen functions to enhance stress activated ACTH and CORT secretion while testosterone functions to decrease HPA axis activation and works to inhibit both ACTH and CORT responses to stress 23 However more studies are required to better understand the underlying basis of these sex differences Experimental studies have investigated many different types of stress and their effects on the HPA axis in many different circumstances 24 Stressors can be of many different types in experimental studies in rats a distinction is often made between social stress and physical stress but both types activate the HPA axis though via different pathways 25 Several monoamine neurotransmitters are important in regulating the HPA axis especially dopamine serotonin and norepinephrine noradrenaline There is evidence that an increase in oxytocin resulting for instance from positive social interactions acts to suppress the HPA axis and thereby counteracts stress promoting positive health effects such as wound healing 26 The HPA axis is a feature of mammals and other vertebrates For example biologists studying stress in fish showed that social subordination leads to chronic stress related to reduced aggressive interactions to lack of control and to the constant threat imposed by dominant fish Serotonin 5 HT appeared to be the active neurotransmitter involved in mediating stress responses and increases in serotonin are related to increased plasma a MSH levels which causes skin darkening a social signal in salmonoid fish activation of the HPA axis and inhibition of aggression Inclusion of the amino acid L tryptophan a precursor of 5 HT in the feed of rainbow trout made the trout less aggressive and less responsive to stress 27 However the study mentions that plasma cortisol was not affected by dietary L tryptophan The drug LY354740 also known as Eglumegad an agonist of the metabotropic glutamate receptors 2 and 3 has been shown to interfere in the HPA axis with chronic oral administration of this drug leading to markedly reduced baseline cortisol levels in bonnet macaques Macaca radiata acute infusion of LY354740 resulted in a marked diminution of yohimbine induced stress response in those animals 28 Studies on people show that the HPA axis is activated in different ways during chronic stress depending on the type of stressor the person s response to the stressor and other factors Stressors that are uncontrollable threaten physical integrity or involve trauma tend to have a high flat diurnal profile of cortisol release with lower than normal levels of cortisol in the morning and higher than normal levels in the evening resulting in a high overall level of daily cortisol release On the other hand controllable stressors tend to produce higher than normal morning cortisol Stress hormone release tends to decline gradually after a stressor occurs In post traumatic stress disorder there appears to be lower than normal cortisol release and it is thought that a blunted hormonal response to stress may predispose a person to develop PTSD 29 It is also known that HPA axis hormones are related to certain skin diseases and skin homeostasis There is evidence shown that the HPA axis hormones can be linked to certain stress related skin diseases and skin tumors This happens when HPA axis hormones become hyperactive in the brain 30 Stress and development edit Prenatal stress edit There is evidence that prenatal stress can influence HPA regulation In animal experiments exposure to prenatal stress has been shown to cause a hyper reactive HPA stress response Rats that have been prenatally stressed have elevated basal levels and abnormal circadian rhythm of corticosterone as adults 31 Additionally they require a longer time for their stress hormone levels to return to baseline following exposure to both acute and prolonged stressors Prenatally stressed animals also show abnormally high blood glucose levels and have fewer glucocorticoid receptors in the hippocampus 32 In humans prolonged maternal stress during gestation is associated with mild impairment of intellectual activity and language development in their children and with behavior disorders such as attention deficits schizophrenia anxiety and depression self reported maternal stress is associated with a higher irritability emotional and attentional problems 33 There is growing evidence that prenatal stress can affect HPA regulation in humans Children who were stressed prenatally may show altered cortisol rhythms For example several studies have found an association between maternal depression during pregnancy and childhood cortisol levels 34 Prenatal stress has also been implicated in a tendency toward depression and short attention span in childhood 35 Early life stress edit This article needs editing to comply with Wikipedia s Manual of Style Please help improve the content August 2023 Learn how and when to remove this template message The role of early life stress in programming the HPA axis has been well studied in animal models Exposure to mild or moderate stressors early in life has been shown to enhance HPA regulation and promote a lifelong resilience to stress In contrast early life exposure to extreme or prolonged stress can induce a hyper reactive HPA axis and may contribute to lifelong vulnerability to stress 36 In one widely replicated experiment rats subjected to the moderate stress of frequent human handling during the first two weeks of life had reduced hormonal and behavioral HPA mediated stress responses as adults whereas rats subjected to the extreme stress of prolonged periods of maternal separation showed heightened physiological and behavioral stress responses as adults 37 Several mechanisms have been proposed to explain these findings in rat models of early life stress exposure There may be a critical period during development during which the level of stress hormones in the bloodstream contribute to the permanent calibration of the HPA axis One experiment has shown that even in the absence of any environmental stressors early life exposure to moderate levels of corticosterone was associated with stress resilience in adult rats whereas exposure to high doses was associated with stress vulnerability 38 Another possibility is that the effects of early life stress on HPA functioning are mediated by maternal care Frequent human handling of the rat pups may cause their mother to exhibit more nurturant behavior such as licking and grooming Nurturant maternal care in turn may enhance HPA functioning in at least two ways First maternal care is crucial in maintaining the normal stress hypo responsive period SHRP which in rodents is the first two weeks of life during which the HPA axis is generally non reactive to stress Maintenance of the SHRP period may be critical for HPA development and the extreme stress of maternal separation which disrupts the SHRP may lead to permanent HPA dysregulation 39 Another way that maternal care might influence HPA regulation is by causing epigenetic changes in the offspring For example increased maternal licking and grooming has been shown to alter expression of the glucocorticoid receptor gene implicated in adaptive stress response 36 At least one human study has identified maternal neural activity patterns in response to video stimuli of mother infant separation as being associated with decreased glucocorticoid receptor gene methylation in the context of post traumatic stress disorder stemming from early life stress 40 Yet clearly more research is needed to determine if the results seen in cross generational animal models can be extended to humans Though animal models allow for more control of experimental manipulation the effects of early life stress on HPA axis function in humans has also been studied One population that is often studied in this type of research is adult survivors of childhood abuse Adult survivors of childhood abuse have exhibited increased ACTH concentrations in response to a psychosocial stress task compared to unaffected controls and subjects with depression but not childhood abuse 41 In one study adult survivors of childhood abuse that are not depressed show increased ACTH response to both exogenous CRF and normal cortisol release Adult survivors of childhood abuse that are depressed show a blunted ACTH response to exogenous CRH 42 A blunted ACTH response is common in depression so the authors of this work posit that this pattern is likely to be due to the participant s depression and not their exposure to early life stress Heim and colleagues have proposed that early life stress such as childhood abuse can induce a sensitization of the HPA axis resulting in particular heightened neuronal activity in response to stress induced CRH release 42 With repeated exposure to stress the sensitized HPA axis may continue to hypersecrete CRH from the hypothalamus Over time CRH receptors in the anterior pituitary will become down regulated producing depression and anxiety symptoms 42 This research in human subjects is consistent with the animal literature discussed above The HPA axis was present in the earliest vertebrate species and has remained highly conserved by strong positive selection due to its critical adaptive roles 43 The programming of the HPA axis is strongly influenced by the perinatal and early juvenile environment or early life environment 44 45 46 Maternal stress and differential degrees of caregiving may constitute early life adversity which has been shown to profoundly influence if not permanently alter the offspring s stress and emotional regulating systems 44 45 Widely studied in animal models e g licking and grooming LG in rat pups 47 the consistency of maternal care has been shown to have a powerful influence on the offspring s neurobiology physiology and behavior Whereas maternal care improves cardiac response sleep wake rhythm and growth hormone secretion in the neonate it also suppresses HPA axis activity In this manner maternal care negatively regulates stress response in the neonate 47 thereby shaping his her susceptibility to stress in later life These programming effects are not deterministic as the environment in which the individual develops can either match or mismatch with the former s programmed and genetically predisposed HPA axis reactivity Although the primary mediators of the HPA axis are known the exact mechanism by which its programming can be modulated during early life remains to be elucidated Furthermore evolutionary biologists contest the exact adaptive value of such programming i e whether heightened HPA axis reactivity may confer greater evolutionary fitness Various hypotheses have been proposed in attempts to explain why early life adversity can produce outcomes ranging from extreme vulnerability to resilience in the face of later stress Glucocorticoids produced by the HPA axis have been proposed to confer either a protective or harmful role depending on an individual s genetic predispositions programming effects of early life environment and match or mismatch with one s postnatal environment The predictive adaptation hypothesis 1 the three hit concept of vulnerability and resilience 2 and the maternal mediation hypothesis 3 attempt to elucidate how early life adversity can differentially predict vulnerability or resilience in the face of significant stress in later life 48 These hypotheses are not mutually exclusive but rather are highly interrelated and unique to the individual 1 The predictive adaptation hypothesis 48 This hypothesis is in direct contrast with the diathesis stress model which posits that the accumulation of stressors across a lifespan can enhance the development of psychopathology once a threshold is crossed Predictive adaptation asserts that early life experience induces epigenetic change these changes predict or set the stage for adaptive responses that will be required in their environment Thus if a developing child i e fetus to neonate is exposed to ongoing maternal stress and low levels of maternal care i e early life adversity this will program their HPA axis to be more reactive to stress This programming will have predicted and potentially be adaptive in a highly stressful precarious environment during childhood and later life The predictability of these epigenetic changes is not definitive however depending primarily on the degree to which the individual s genetic and epigenetically modulated phenotype matches or mismatches with their environment See Hypothesis 2 2 Three Hit Concept of vulnerability and resilience 48 this hypothesis states that within a specific life context vulnerability may be enhanced with chronic failure to cope with ongoing adversity It fundamentally seeks to explicate why under seemingly indistinguishable circumstances one individual may cope resiliently with stress whereas another may not only cope poorly but consequently develop a stress related mental illness The three hits chronological and synergistic are as follows genetic predisposition which predispose higher lower HPA axis reactivity early life environment perinatal i e maternal stress and postnatal i e maternal care and later life environment which determines match mismatch as well as a window for neuroplastic changes in early programming 49 The concept of match mismatch is central to this evolutionary hypothesis In this context it elucidates why early life programming in the perinatal and postnatal period may have been evolutionarily selected for Specifically by instating specific patterns of HPA axis activation the individual may be more well equipped to cope with adversity in a high stress environment Conversely if an individual is exposed to significant early life adversity heightened HPA axis reactivity may mismatch them in an environment characterized by low stress The latter scenario may represent maladaptation due to early programming genetic predisposition and mismatch This mismatch may then predict negative developmental outcomes such as psychopathologies in later life 2 Maternal mediation hypothesis 38 This hypothesis states that maternal care is the primary factor in developing stress resistance later in life Ultimately the conservation of the HPA axis has underscored its critical adaptive roles in vertebrates so too various invertebrate species over time The HPA axis plays a clear role in the production of corticosteroids which govern many facets of brain development and responses to ongoing environmental stress With these findings animal model research has served to identify what these roles are with regards to animal development and evolutionary adaptation In more precarious primitive times a heightened HPA axis may have served to protect organisms from predators and extreme environmental conditions such as weather and natural disasters by encouraging migration i e fleeing the mobilization of energy learning in the face of novel dangerous stimuli as well as increased appetite for biochemical energy storage In contemporary society the endurance of the HPA axis and early life programming will have important implications for counseling expecting and new mothers as well as individuals who may have experienced significant early life adversity 49 See also editOther major neuroendocrine systemsHypothalamic neurohypophyseal system Hypothalamic pituitary gonadal axis Hypothalamic pituitary thyroid axisRelated topicsACTH stimulation test Allostatic load Antidepressants Cortisol awakening response Dexamethasone Dexamethasone suppression test Fight or flight response Hydrocortisone Major depressive disorder Psychoneuroendocrinology Sickness behavior ConditionsAddison s disease Adrenal insufficiency Cushing s syndromeReferences edit a b Malenka RC Nestler EJ Hyman SE 2009 Chapter 10 Neural and Neuroendocrine Control of the Internal Milieu In Sydor A Brown RY ed Molecular Neuropharmacology A Foundation for Clinical Neuroscience 2nd ed New York McGraw Hill Medical pp 246 248 259 ISBN 9780071481274 The hypothalamic neurohypophyseal system secretes two peptide hormones directly into the blood vasopressin and oxytocin The hypothalamic pituitary adrenal HPA axis It comprises corticotropin releasing factor CRF released by the hypothalamus adrenocorticotropic hormone ACTH released by the anterior pituitary and glucocorticoids released by the adrenal cortex The hypothalamic pituitary thyroid axis consists of hypothalamic thyrotropin releasing hormone TRH the anterior pituitary hormone thyroid stimulating hormone TSH and the thyroid hormones T3 and T4 The hypothalamic pituitary gonadal axis comprises hypothalamic gonadotropin releasing hormone GnRH the anterior pituitary luteinizing hormone LH and follicle stimulating hormone FSH and the gonadal steroids Selye Hans 1974 Stress without distress Philadelphia Lippincott ISBN 978 0 397 01026 4 page needed Getting to know the HPA axis www nrdc org 2015 05 18 Archived from the original on 2023 08 10 Retrieved 2023 08 08 Tasker Jeffrey G Herman James P 14 July 2011 Mechanisms of rapid glucocorticoid feedback inhibition of the hypothalamic pituitary adrenal axis Stress 14 4 398 406 doi 10 3109 10253890 2011 586446 PMC 4675656 PMID 21663538 Cuzzo Brian Lappin Sarah L 2019 Vasopressin Antidiuretic Hormone ADH StatPearls StatPearls Publishing PMID 30252325 archived from the original on 2021 03 25 retrieved 2019 10 19 a b c d del Rey A Chrousos G P Besedovsky H O eds 2008 The Hypothalamus Pituitary Adrenal Axis NeuroImmune Biology Vol 7 Berczi I Szentivanyi A series EICs Amsterdam London Elsevier Science ISBN 978 0 08 055936 0 OCLC 272388790 Archived from the original on 10 August 2023 Retrieved 27 February 2022 MacHale SM Cavanagh JT Bennie J Carroll S Goodwin GM Lawrie SM November 1998 Diurnal variation of adrenocortical activity in chronic fatigue syndrome Neuropsychobiology 38 4 213 7 doi 10 1159 000026543 PMID 9813459 S2CID 46856991 Backhaus J Junghanns K Hohagen F October 2004 Sleep disturbances are correlated with decreased morning awakening salivary cortisol Psychoneuroendocrinology 29 9 1184 91 doi 10 1016 j psyneuen 2004 01 010 PMID 15219642 S2CID 14756991 Pruessner JC Hellhammer DH Kirschbaum C 1999 Burnout perceived stress and cortisol responses to awakening Psychosom Med 61 2 197 204 doi 10 1097 00006842 199903000 00012 PMID 10204973 Laura Freberg 2015 01 01 Discovering behavioral neuroscience an introduction to biological psychology Freberg Laura Container of work Freberg Laura Third ed Boston MA p 504 ISBN 9781305088702 OCLC 905734838 a href Template Cite book html title Template Cite book cite book a CS1 maint location missing publisher link Frankiensztajn Linoy Mia Elliott Evan Koren Omry June 2020 The microbiota and the hypothalamus pituitary adrenocortical HPA axis implications for anxiety and stress disorders Current Opinion in Neurobiology 62 76 82 doi 10 1016 j conb 2019 12 003 PMID 31972462 S2CID 210836469 a b Marques Deak A Cizza G Sternberg E February 2005 Brain immune interactions and disease susceptibility Molecular Psychiatry 10 3 239 250 doi 10 1038 sj mp 4001643 PMID 15685252 S2CID 17978810 a b c d e Otmishi Peyman Gordon Josiah El Oshar Seraj Li Huafeng Guardiola Juan Saad Mohamed Proctor Mary Yu Jerry 2008 Neuroimmune Interaction in Inflammatory Diseases Clinical Medicine Circulatory Respiratory and Pulmonary Medicine 2 35 44 doi 10 4137 ccrpm s547 PMC 2990232 PMID 21157520 a b c Tian Rui Hou Gonglin Li Dan Yuan Ti Fei June 2014 A Possible Change Process of Inflammatory Cytokines in the prolonged Chronic Stress and its Ultimate Implications for Health The Scientific World Journal 2014 780616 doi 10 1155 2014 780616 PMC 4065693 PMID 24995360 Hall Jessica Cruser desAgnes Podawiltz Alan Mummert Diana Jones Harlan Mummert Mark August 2012 Psychological Stress and the Cutaneous Immune Response Roles of the HPA Axis and the Sympathetic Nervous System in Atopic Dermatitis and Psoriasis Dermatology Research and Practice 2012 403908 doi 10 1155 2012 403908 PMC 3437281 PMID 22969795 a b Bellavance Marc Andre Rivest Serge March 2014 The HPA immune axis and the immunomodulatory actions of glucocorticoids in the brain Frontiers in Immunology 5 136 doi 10 3389 fimmu 2014 00136 PMC 3978367 PMID 24744759 a b Padgett David Glaser Ronald August 2003 How stress influences the immune response PDF Trends in Immunology 24 8 444 448 doi 10 1016 S1471 4906 03 00173 X PMID 12909458 Archived from the original PDF on 2016 03 27 Retrieved 12 February 2016 Spencer RL Hutchison KE 1999 Alcohol aging and the stress response Alcohol Research amp Health 23 4 272 83 PMC 6760387 PMID 10890824 Smith Carli J Emge Jacob R Berzins Katrina Lung Lydia Khamishon Rebecca Shah Paarth Rodrigues David M Sousa Andrew J Reardon Colin Sherman Philip M Barrett Kim E Gareau Melanie G 2014 10 15 Probiotics normalize the gut brain microbiota axis in immunodeficient mice American Journal of Physiology Gastrointestinal and Liver Physiology 307 8 G793 G802 doi 10 1152 ajpgi 00238 2014 ISSN 0193 1857 PMC 4200314 PMID 25190473 Pariante CM August 2003 Depression stress and the adrenal axis Journal of Neuroendocrinology 15 8 811 2 doi 10 1046 j 1365 2826 2003 01058 x PMID 12834443 S2CID 1359479 Rosinger Zachary Jacobskind Jason Park Shannon Justice Nicholas Zuloaga Damian 2017 Distribution of Corticotropin releasing factor receptor 1 in the developing mouse forebrain A novel sex difference revealed in the rostral periventricular hypothalamus Neuroscience 361 167 178 doi 10 1016 j neuroscience 2017 08 016 PMC 7173945 PMID 28823817 Palumbo Michelle C Dominguez Sky Dong Hongxin 2020 Sex differences in hypothalamic pituitary adrenal axis regulation after chronic unpredictable stress Brain and Behavior 10 4 e01586 doi 10 1002 brb3 1586 PMC 7177572 PMID 32154650 Handa R J Weiser M J Zuloaga D G 2009 A Role for the Androgen Metabolite 5a Androstane 3b 17b Diol in Modulating Oestrogen Receptor b Mediated Regulation of Hormonal Stress Reactivity Journal of Neuroendocrinology 21 4 351 358 doi 10 1111 j 1365 2826 2009 01840 x PMC 2727750 PMID 19207807 Douglas AJ March 2005 Central noradrenergic mechanisms underlying acute stress responses of the Hypothalamo pituitary adrenal axis adaptations through pregnancy and lactation Stress 8 1 5 18 doi 10 1080 10253890500044380 PMID 16019594 S2CID 24654645 Engelmann M Landgraf R Wotjak CT 2004 The hypothalamic neurohypophysial system regulates the hypothalamic pituitary adrenal axis under stress an old concept revisited Frontiers in Neuroendocrinology 25 3 4 132 49 doi 10 1016 j yfrne 2004 09 001 PMID 15589266 S2CID 41983825 Detillion CE Craft TK Glasper ER Prendergast BJ DeVries AC September 2004 Social facilitation of wound healing Psychoneuroendocrinology 29 8 1004 11 doi 10 1016 j psyneuen 2003 10 003 PMID 15219651 S2CID 5986340 Winberg S Overli O Lepage O November 2001 Suppression of aggression in rainbow trout Oncorhynchus mykiss by dietary L tryptophan The Journal of Experimental Biology 204 Pt 22 3867 76 doi 10 1242 jeb 204 22 3867 PMID 11807104 Coplan JD Mathew SJ Smith EL et al July 2001 Effects of LY354740 a novel glutamatergic metabotropic agonist on nonhuman primate hypothalamic pituitary adrenal axis and noradrenergic function CNS Spectrums 6 7 607 12 617 doi 10 1017 S1092852900002157 PMID 15573025 S2CID 6029856 Miller GE Chen E Zhou ES January 2007 If it goes up must it come down Chronic stress and the hypothalamic pituitary adrenocortical axis in humans Psychological Bulletin 133 1 25 45 doi 10 1037 0033 2909 133 1 25 PMID 17201569 Kim JE Cho BK Cho DH Park HJ July 2013 Expression of hypothalamic pituitary adrenal axis in common skin diseases evidence of its association with stress related disease activity Acta Dermato Venereologica 93 4 387 93 doi 10 2340 00015555 1557 PMID 23462974 Koehl M Darnaudery M Dulluc J Van Reeth O Le Moal M Maccari S September 1999 Prenatal stress alters circadian activity of hypothalamo pituitary adrenal axis and hippocampal corticosteroid receptors in adult rats of both gender Journal of Neurobiology 40 3 302 15 doi 10 1002 SICI 1097 4695 19990905 40 3 lt 302 AID NEU3 gt 3 0 CO 2 7 PMID 10440731 Weinstock M Matlina E Maor GI Rosen H McEwen BS November 1992 Prenatal stress selectively alters the reactivity of the hypothalamic pituitary adrenal system in the female rat Brain Research 595 2 195 200 doi 10 1016 0006 8993 92 91049 K PMID 1467966 S2CID 44382315 Weinstock M August 2008 The long term behavioural consequences of prenatal stress PDF Neuroscience and Biobehavioral Reviews 32 6 1073 86 doi 10 1016 j neubiorev 2008 03 002 PMID 18423592 S2CID 3717977 Archived PDF from the original on 2023 08 10 Retrieved 2014 05 04 Gutteling BM de Weerth C Buitelaar JK December 2004 Maternal prenatal stress and 4 6 year old children s salivary cortisol concentrations pre and post vaccination Stress 7 4 257 60 doi 10 1080 10253890500044521 hdl 2066 54819 PMID 16019591 S2CID 38412082 Buitelaar JK Huizink AC Mulder EJ de Medina PG Visser GH 2003 Prenatal stress and cognitive development and temperament in infants Neurobiology of Aging 24 Suppl 1 S53 60 discussion S67 8 doi 10 1016 S0197 4580 03 00050 2 PMID 12829109 S2CID 3008063 a b Flinn MV Nepomnaschy PA Muehlenbein MP Ponzi D June 2011 Evolutionary functions of early social modulation of hypothalamic pituitary adrenal axis development in humans Neurosci Biobehav Rev 35 7 1611 29 doi 10 1016 j neubiorev 2011 01 005 PMID 21251923 S2CID 16950714 Liu D Diorio J Tannenbaum B et al September 1997 Maternal care hippocampal glucocorticoid receptors and hypothalamic pituitary adrenal responses to stress Science 277 5332 1659 62 doi 10 1126 science 277 5332 1659 PMID 9287218 S2CID 2753764 a b Macri S Wurbel H December 2006 Developmental plasticity of HPA and fear responses in rats a critical review of the maternal mediation hypothesis Hormones and Behavior 50 5 667 80 doi 10 1016 j yhbeh 2006 06 015 PMID 16890940 S2CID 36877565 de Kloet ER Sibug RM Helmerhorst FM Schmidt MV Schmidt M April 2005 Stress genes and the mechanism of programming the brain for later life Neuroscience and Biobehavioral Reviews 29 2 271 81 doi 10 1016 j neubiorev 2004 10 008 PMID 15811498 S2CID 24943895 Schechter Daniel S Moser Dominik A Paoloni Giacobino Ariane Stenz Ludwig Gex Fabry Marianne Aue Tatjana Adouan Wafae Cordero Maria I Suardi Francesca Manini Aurelia Sancho Rossignol Ana 2015 Methylation of NR3C1 is related to maternal PTSD parenting stress and maternal medial prefrontal cortical activity in response to child separation among mothers with histories of violence exposure Frontiers in Psychology 6 690 doi 10 3389 fpsyg 2015 00690 ISSN 1664 1078 PMC 4447998 PMID 26074844 Heim C Newport D J Heit S Graham Y P Wilcox M Bonsall R Nemeroff C B 2000 Pituitary adrenal and autonomic responses to stress in women after sexual and physical abuse in childhood JAMA 284 5 592 597 doi 10 1001 jama 284 5 592 PMID 10918705 a b c Heim C Newport D J Bonsall R Miller A H Nemeroff C B 2001 Altered Pituitary Adrenal Axis Responses to Provocative Challenge Tests in Adult Survivors of Childhood Abuse Am J Psychiatry 158 4 575 581 doi 10 1176 appi ajp 158 4 575 PMID 11282691 Denver RJ Apr 2009 Structural and functional evolution of vertebrate neuroendocrine stress systems PDF Ann N Y Acad Sci 1163 1 1 16 Bibcode 2009NYASA1163 1D doi 10 1111 j 1749 6632 2009 04433 x hdl 2027 42 74370 PMID 19456324 S2CID 18786346 Archived from the original on 2023 08 10 Retrieved 2019 09 01 a b Oitzl MS Champagne DL van der Veen R de Kloet ER May 2010 Brain development under stress hypotheses of glucocorticoid actions revisited Neurosci Biobehav Rev 34 6 853 66 doi 10 1016 j neubiorev 2009 07 006 PMID 19631685 S2CID 25898149 a b Horton TH Jan 2005 Fetal origins of developmental plasticity animal models of induced life history variation Am J Hum Biol 17 1 34 43 doi 10 1002 ajhb 20092 PMID 15611963 S2CID 23466894 Matthews SG Mar 2000 Antenatal glucocorticoids and programming of the developing CNS Pediatr Res 47 3 291 300 doi 10 1203 00006450 200003000 00003 PMID 10709726 a b Champagne FA Francis DD Mar A Meaney MJ Aug 2003 Variations in maternal care in the rat as a mediating influence for the effects of environment on development Physiol Behav 79 3 359 71 CiteSeerX 10 1 1 335 3199 doi 10 1016 s0031 9384 03 00149 5 PMID 12954431 S2CID 18599019 a b c Daskalakis NP Bagot RC Parker KJ Vinkers CH de Kloet ER Sep 2013 The three hit concept of vulnerability and resilience toward understanding adaptation to early life adversity outcome Psychoneuroendocrinology 38 9 1858 73 doi 10 1016 j psyneuen 2013 06 008 PMC 3773020 PMID 23838101 a b Roth TL Matt S Chen K Blaze J Dec 2014 Bdnf DNA methylation modifications in the hippocampus and amygdala of male and female rats exposed to different caregiving environments outside the homecage Dev Psychobiol 56 8 1755 63 doi 10 1002 dev 21218 PMC 4205217 PMID 24752649 External links edit nbsp Wikimedia Commons has media related to Hypothalamic pituitary adrenal axis Mind Body Health net page on HPA axis HPA Axis Explanation of the Body s Central Stress Response System w Diagram Retrieved from https en wikipedia org w index php title Hypothalamic pituitary adrenal axis amp oldid 1186062537, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.