Next Article in Journal
Target Temperature Management Effect on the Clinical Outcome of Patients with Out-of-Hospital Cardiac Arrest Treated with Extracorporeal Cardiopulmonary Resuscitation: A Nationwide Observational Study
Next Article in Special Issue
The Head-Up Tilt Table Test as a Measure of Autonomic Functioning among Patients with Myalgic Encephalomyelitis/Chronic Fatigue Syndrome
Previous Article in Journal
Clinical Outcome of Multiple Platelet-Rich Plasma Injection and Correlation with PDGF-BB in the Treatment of Knee Osteoarthritis
Previous Article in Special Issue
Possible Role of Fibrinaloid Microclots in Postural Orthostatic Tachycardia Syndrome (POTS): Focus on Long COVID
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

How Sex Hormones Affect Migraine: An Interdisciplinary Preclinical Research Panel Review

by
Frederick Godley III
1,*,
John Meitzen
2,
Hadas Nahman-Averbuch
3,
Mary Angela O’Neal
4,
David Yeomans
5,
Nanette Santoro
6,
Nina Riggins
7 and
Lars Edvinsson
8
1
Association of Migraine Disorders, P.O. Box 870, North Kingstown, RI 02852, USA
2
Department of Biological Sciences, NC State University, Raleigh, NC 27695, USA
3
Division of Clinical and Translational Research, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
4
Brigham and Women’s Hospital, 45 Francis St, Boston, MA 02115, USA
5
Department of Anesthesia, Pain and Perioperative Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305, USA
6
Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO 80045, USA
7
Brain Performance Center and Research Institute, San Diego, CA 92122, USA
8
Division of Experimental Vascular Research, Department of Clinical Sciences, Lund University Hospital, 22185 Lund, Sweden
*
Author to whom correspondence should be addressed.
J. Pers. Med. 2024, 14(2), 184; https://doi.org/10.3390/jpm14020184
Submission received: 19 December 2023 / Revised: 26 January 2024 / Accepted: 30 January 2024 / Published: 7 February 2024
(This article belongs to the Special Issue New Challenges and Perspectives in Neurology and Autonomic Disorders)

Abstract

:
Sex hormones and migraine are closely interlinked. Women report higher levels of migraine symptoms during periods of sex hormone fluctuation, particularly during puberty, pregnancy, and perimenopause. Ovarian steroids, such as estrogen and progesterone, exert complex effects on the peripheral and central nervous systems, including pain, a variety of special sensory and autonomic functions, and affective processing. A panel of basic scientists, when challenged to explain what was known about how sex hormones affect the nervous system, focused on two hormones: estrogen and oxytocin. Notably, other hormones, such as progesterone, testosterone, and vasopressin, are less well studied but are also highlighted in this review. When discussing what new therapeutic agent might be an alternative to hormone therapy and menopause replacement therapy for migraine treatment, the panel pointed to oxytocin delivered as a nasal spray. Overall, the conclusion was that progress in the preclinical study of hormones on the nervous system has been challenging and slow, that there remain substantial gaps in our understanding of the complex roles sex hormones play in migraine, and that opportunities remain for improved or novel therapeutic agents. Manipulation of sex hormones, perhaps through biochemical modifications where its positive effects are selected for and side effects are minimized, remains a theoretical goal, one that might have an impact on migraine disease and other symptoms of menopause. This review is a call to action for increased interest and funding for preclinical research on sex hormones, their metabolites, and their receptors. Interdisciplinary research, perhaps facilitated by a collaborative communication network or panel, is a possible strategy to achieve this goal.

1. Introduction

Migraine is a neurological disorder affecting 12% of adults around the world at any one point in time [1]. Migraine symptoms can be different in women than men. Women can have more frequent and intense headaches with a higher risk of chronification [2]. Migraine is now recognized as the number one cause of disability globally for women aged 15–49 [3]. This gender difference in the behavior of migraine as a disease highlights the role of sex hormones in its pathophysiology. This review was the result of a round-table discussion among a panel of basic scientists from different disciplines on the topic of how sex hormones exert their effect on the nervous system, particularly migraine disease. The panelists were asked to discuss what the gaps in our knowledge were, what the barriers were, whether they could identify any new therapeutic agents that would provide an alternative treatment for migraine, and if there was an explanation for the clinical observation that the prevalence of some migraine-related symptoms, such as vestibular migraine and sinus pain and pressure, increase during perimenopause while headaches tend to recede.

2. Sex Hormone Fluctuation as a Trigger of Migraine

Migraine tends to follow a classic temporal pattern throughout a cisgender woman’s life that corresponds with sex hormone fluctuations during reproductive milestones in the female lifespan. Puberty is a key period with significant changes in sex hormone levels. Interestingly, in children and adolescents, the prevalence of migraine headaches is nearly equivalent in boys and girls [4], but during puberty, the prevalence of migraine between men and women diverges and is 3–4 times higher in women compared to men [5,6]. This sex difference corresponds to the onset of menarche and falls after menopause.
Migraine symptoms can be linked to menstrual cycle changes (menstrual migraine) and 18–25% of women with migraine experience migraine or headaches during menstruation [7]. Menstrual migraine can be associated with a higher frequency of migraine-accompanying symptoms and more frequent and severe migraine attacks [8]. A comparison of women with and without migraine shows that those with migraine are characterized by faster late-luteal-phase estrogen decline compared to women without migraine. Thus, the timing and rate of estrogen withdrawal has been proposed to be a marker of vulnerability to migraine in women [9]. Contraceptive pills reduce the number of migraine attacks, migraine days, pain scores, disability scores, and migraine medication use while reducing the frequency of aura, and lowering, but not eliminating, the risks of cardiovascular complications or other side effects [10,11,12]. Another strategy is to use estrogen supplementation with a pill, vaginal gel or patch during the menstrual week.
Migraine is a heterogeneous disease associated with many possible combinations of genetic defects which share a common phenotype of intermittent pain or other hypersensitivities. This accounts for the unpredictable response of migraineurs to medications and the effect of hormones on the nociceptive system is no exception. For some, a drop in estrogen triggers a menstrual migraine attack without aura; for others, high levels of estrogen can trigger an attack with aura [13].
Migraine disease has a complex relationship with pregnancy. For 8% of women with migraine, their headaches worsen during the first trimester. This is especially true for migraine without aura, which is more hormonally driven [14,15,16]. The majority of women with migraine generally experience reduced migraine symptoms by the third trimester [17]. However, many women have the acute onset of headaches during pregnancy. Approximately 60% of these new headaches will be related to migraine but caution must be taken to evaluate pregnant women for secondary headaches [18]. A third of women will have postpartum headaches [19]. For those who continue to have migraine symptoms during their pregnancy and immediately postpartum, treatment options are limited to protect the fetus. There are specific recommendations for safe care of women with migraine headaches during pregnancy and breastfeeding [20].
Perimenopause, the period of two to eight years when menses first become irregular prior to the year after the end of menses, is a time when hormonal fluctuations are still occurring, and pre-existing migraine symptoms can remain unchanged, improve, or worsen [21,22,23]. In total, 8–13% of women report their first migraine during perimenopause [24,25]. However, many women see a decrease in headache prevalence during this period [26,27], most prominently in women who already suffer from migraine with aura [28]. For unexplained reasons, mid-facial pain and pressure and vestibular migraine can become prominent symptoms during perimenopause and menopause [29]. Hormone replacement therapy, or menopause replacement therapy (MRT), usually a continuous dosing of estrogen alone or estrogen plus progestin (ethinyl estradiol 5 µg combined with norethindrone acetate 1 mg, estradiol 1 mg combined with 0.5 mg norethindrone acetate, or transdermal estradiol combined with one-quarter or one-half of a 5 mg norethindrone daily) [30], remains an option, particularly for those women who have not had a hysterectomy because estrogen alone increases the risk of endometrial cancer. Transdermal estrogen patches or gels can be efficacious and less risky than systemic estrogen replacement in treating migraine [7,23,31]. A significant shortcoming of supplemental hormone therapies is that they do not provide migraine relief for all women and, for some, headaches become more severe. But a second major shortcoming of MRT is that, although the dosing of sex hormones is roughly half that of birth control pills, the risks of heart disease, stroke, blood clots, and breast cancer are not eliminated [13,30,32].
The bottom line is that current sex hormone supplements play a valuable role in mitigating the symptoms of migraine, but, because they are still associated with serious complications, especially migraine with aura, and exacerbate migraine symptoms in some, many medical professionals choose not to use hormone supplements in their migraine treatment plan. For example, plant-derived hormones (phytoestrogens) and the derivative bio-identical hormones are effective in reducing menstrual-related migraine headaches [33], but there is no rigorous scientific evidence that these supplements are safer or more natural compared to the current hormonal interventions. Phytoestrogen-containing foods, such as soy, are recommended over supplements, and all phytoestrogens should be avoided if there is a chance of pregnancy because these compounds might adversely affect the endocrine system. It is speculated that they might be safer in older women, such as those suffering from menopausal symptoms, particularly hot flashes [34,35], but currently there is not enough evidence to conclude that the benefits of phytoestrogens outweigh their potential health risks [36], and they do not appear to be ideal migraine preventive agents. Thus, since many women with migraine are unable to find an effective preventive therapy, there remains the challenge to understand how sex hormone supplements work, with the goal that select metabolites or synthetic derivatives might be both efficacious and safer than current hormonal therapies.

3. Which Sex Hormones Should Be the Target?

3.1. Estrogen

Estrogen plays a complicated role in migraine disease. Both drops and fluctuations in estrogen are associated with migraine symptoms, but its effect varies between individuals because of different receptors, metabolites, and interactions with other hormones. The dominant understanding of how crucial estrogen is in protecting individuals from migraine symptoms is what happens when estrogen levels decline: the estrogen withdrawal hypothesis. This hypothesis theorizes that drops in plasma estrogen trigger migraine attacks and neuroinflammation, eventually leading to chronic sensitization [37]. There are several possible mechanisms to explain his theory. One explanation is that estrogen suppresses pain by binding to estrogen receptor alpha (ER alpha) and estrogen receptor beta (ER beta), which are primarily associated with cell nuclei in the trigeminal ganglia. Activation of these nuclear receptors regulates inflammatory genes that ultimately suppresses cell excitability [38]. Also, this hypothesis may be explained by drops in estrogen leading to higher levels of calcitonin gene-related peptide (CGRP) [23].
CGRP is believed to be among the critical neuropeptides responsible for the throbbing pain associated with a migraine attack and the neuroinflammation that causes both pain and that perhaps cause neuroplastic neural changes responsible for chronic central sensitization [39]. Specifically, estrogen may also increase neurogenic vasodilation and gene regulation. For example, in mice, expression of neuropeptide Y and galanin, two neuropeptides which may inhibit or modulate CGRP mechanisms in trigeminal neurons, may play a part in the fluctuations of head pain during the estrus cycle [40].
While the estrogen withdrawal hypothesis focuses primarily on the trigeminal nerves, it is important to recognize the wider-ranging actions of estrogen in other parts of the body and brain [41]. A second mechanism to explain the estrogen withdrawal theory was demonstrated in an animal model where reduced levels of estrogen were shown to increase the frequency of cortical spreading depressions, the electrophysiological event believed to be responsible for triggering the trigeminal system and headaches, as well as auras [42].
There are various mechanisms that might explain how cortical spreading depressions are initiated. For example, estrogen is known to rapidly alter cellular excitability and gene expression in hypothalamic neurons [43,44]. And estrogen affects energy homeostasis via the proopiomelanocortin (POMC) neurons in the hypothalamic arcuate [45], and may play a role in migraine. Other brain regions, such as the mesolimbic cortical reward system, have also been implicated and show profound estrogen sensitivity [46,47,48]. The complexity stems from having three forms of estrogen (estrone, estradiol and estriol), thirteen estradiol metabolites, and two classes of receptors with different isomers which are functionally distinct and differentially distributed throughout the brain. Estrogen has other metabolic functions that might contribute to pain control indirectly, such as its indirect effect on serotonin [49].

3.2. Progesterone

Progesterone, the second major sex hormone, is produced in the ovaries, adrenal glands and placenta, and primarily helps maintain pregnancy. Progesterone with estradiol is found at the onset of menstrual migraines. Nonetheless, it is more likely that the withdrawal of estradiol, rather than progesterone, initiates migraine headaches. Instead, progesterone appears to protect neurons by suppressing neuroinflammation and reducing trigeminal nerve sensitivity. In one study, the receptive field size of facial trigeminal mechanoreceptors was not increased by treatment with progesterone, unlike the effects of estradiol [50].
It may be in the interplay with additional factors where progesterone plays an integral role in pain modulation. In a longitudinal study of fibromyalgia, it was high levels of progesterone and testosterone together that were associated with less pain [51]. Progesterone and testosterone are able to penetrate the blood-brain barrier and function as precursors for neurosteroids. There is an example of a progesterone derivative which enhances GABA function by modulating GABA receptors and, in turn, inhibits neuronal sensitivity [52,53]. Furthermore, both progesterone and allopregnanolone appear to dampen nociception in the trigeminovascular system and to reduce neurogenic inflammation in migraine through neuron-glia interactions [52]. In addition, in animals, progesterone and estradiol affect two CNS pathways that lead to increased neuroprotection [54]. But the role of progesterone in neuroinflammation is complicated by the finding that, during menstruation, prostaglandins rise and promote neuroinflammation through the release of substance P, neurokinins, and CGRP [55].
Currently, synthetic progesterone is used as a form of birth control and a migraine preventive agent in the form of a continuous low dose of progestin. Bio-identical progesterone can be delivered in three formulations: orally, topically, and as a suppository. Progesterone may improve insomnia as a mild sedative, and improve sleep apneas by stimulating respiration [56]. Finally, the progesterone metabolite, allopregnanolone, plays a role in the disproportionate level of mood disorders in susceptible women [57], and may begin to explain the high prevalence of anxiety in those with migraine.

3.3. Testosterone

A popular belief is that testosterone is the male hormone whereas estrogen is the female hormone. However, this is an oversimplification, as both estrogen and testosterone have important roles to play in individuals of either sex [58]. In both males and females, the balance between estrogen and testosterone production throughout life influences the function of both reproductive and nonreproductive organs [58].
Testosterone could be a potential therapeutic target, as it has an antinociceptive effect [59,60,61,62,63]. In animal studies, after gonadectomy or the blocking of testosterone receptors, animals appeared more sensitive to nociceptive stimuli [64,65,66,67,68]. The few human studies performed support an analgesic effect of testosterone, as higher testosterone levels are associated with lower experimental pain sensitivity [69]. Studies on the relationship of testosterone to migraine are few. Testosterone levels are lower in adults with migraine vs. without migraine, and are related to migraine severity. Interestingly, even when similar testosterone levels are found, men with migraine more frequently report symptoms of androgen deficiency compared to men with no migraine. However, one study found that no differences in testosterone levels were found in women with vs. without migraine, and that migraine pain intensity was not correlated with testosterone levels. In addition, transgender subjects who were given androgen-blocking medication and estrogen replacement developed increased levels of migraine with aura, similar to the effect of estrogen replacement therapy in cisgender women [13]. Since men with lower levels of androgen are prone to cluster headaches [70], the androgen deficiency model of migraine is based on the premise that testosterone offers neuroprotection. This theory is complicated by finding that, in contrast to estrogen which promotes neuroinflammation through CGRP and other neuropeptides, testosterone promotes neuroinflammation through microglial pathways. Therefore, while testosterone supplementation in females might protect against progression to chronic migraine, it will not have the same effect due to the gender-specific physiology of males [71].
Testosterone appears to be able to effectively reduce symptoms by suppressing spreading depressions, increasing serotonin, stabilizing cerebral blood flow, and reducing cell excitability and neuroinflammation [72]. These metabolic effects may explain the findings that testosterone treatment can improve clinical pain and experimental pain sensitivity in patients with chronic pain, including in patients with temporomandibular joint pain, fibromyalgia, and migraine [73,74,75,76], and that testosterone treatment delivered by a subcutaneous implant significantly reduces migraine intensity [75]. Thus, although testosterone is not thought to play a causal role in migraine, it likely modulates pain. Nonetheless, limited evidence and complex effects are reasons that testosterone is not included in migraine management guidelines.

3.4. Oxytocin

Oxytocin’s (OT) therapeutic effects in migraine are complex and widespread in the nervous system, including at the level of the primary sensory neuron, spinal cord, and in a variety of brain regions associated with pain processing and modulation [77,78,79]. A recent theory is that menstrual migraines are related to a drop in both estrogen and OT during menstruation. Whether the lower concentrations of OT are secondary to the effect of less available estrogen in the CNS is not yet known.
The effect of OT on migraine has been shown via a case report in which intravenous OT provided analgesia and migraine relief [80]. In addition, double-blind, placebo-controlled clinical studies have shown evidence that intranasal OT sprays are efficacious for treating migraine pain in adult men and women [77,81] and experimental-evoked pain in men [82]. A benefit of oxytocin as a treatment for migraine is that it is routinely administered intranasally for inducing labor, postpartum care, and for enhancing lactation, and its safety profile is well documented. In addition, intranasal oxytocin in humans has no major side effects [83].
OT is a neuropeptide that exerts its pain-inhibitory effects both at the level of the primary afferent fiber and in the central nervous system. The first mechanism is via the descending neural pathway from the paraventricular nucleus (PVN) to the dorsal horn of the spinal cord [84,85]. Signals from the PVN release oxytocin in the spinal dorsal horn that activate GABAergic interneurons in the dorsal horn which secondarily recruit other inhibitory GABAergic interneurons and suppress pain signals carried by ascending A-delta and C-fibers [86,87,88,89]. The second mechanism is where OT released from the supraoptic nucleus (SON) in the hypothalamus, periaqueductal gray (PAG), rostral ventromedial medulla (RVM), and the spinal dorsal horn [90,91] modulates central endogenous pain pathways by raising nociceptive thresholds [92,93]. OT can suppress headache pain by binding to oxytocin receptors (OTRs) specifically in the trigeminal nucleus and trigeminal ganglia [94]. Imaging studies of migraine patients show overlap in the localization of OT/OTR, particularly those in the brainstem, thought to be migraine generators [95].
OTR mRNA and proteins are expressed in nociceptive C-fibers and Aδ-fibers in the adult rat trigeminal ganglia [94], and have a high level of co-expression with CGRP in trigeminal ganglia neurons [77]. OT dose-dependently blocks the release of calcitonin gene-related peptide (CGRP) from trigeminal afferent neurons innervating the dura in vitro [94]. CGRP is critical for the pathogenesis for chronic migraine, meaning that OTR activation on trigeminal nociceptive neurons could be a key mechanism of decreased headache intensity and frequency in migraine.
OT might have a general anti-inflammatory effect in orofacial nociceptive pathways by activating OTR, which can also suppress pro-inflammatory markers IL-1B and TNFa in the trigeminal ganglia (and in the spinal trigeminal nucleus caudalis) by inhibiting upregulation of these cytokines. A secondary effect is that inflammatory pain stimulates increased OTR gene expression [96]. But with less OT, trigeminal ganglia neurons become more sensitive, enhancing the likelihood of a migraine being triggered [97].

3.5. Vasopressin

Arginine vasopressin (AVP) is a neuropeptide hormone that has an antidiuretic effect in low concentrations, but at higher concentrations it causes vasoconstriction. Together, these effects raise blood pressure. AVP also has a role in pain, behavior, platelet aggregation, and blood coagulation functions. Specifically, AVP, in response to stress and pain, may be relevant to migraine pathophysiology [98,99]. Platelets have more AVP receptors in women who experience migraine [100]. It is possible that the AVP secretion has nothing directly to do with migraine, but, since the highest levels of AVP during a migraine attack may be associated with emesis [101] and vomiting, hypovolemia and nausea without vomiting trigger AVP release. Elevated levels of AVP may be responsible for the facial pallor, antidiuresis, and coagulation abnormalities occasionally observed in migraine [102]. In addition, some migraine precipitators (stress, ethanol, etc.) cause decreased AVP secretion and bioavailability, while some migraine-improving factors (tricyclic antidepressants, sleep, etc.) are associated with an increase in AVP [103]. Intranasal delivery of AVP has been described as an effective therapeutic agent for headache control [104].
Much of AVP is synthesized in the SON of the hypothalamus and, while AVP is largely stored in and secreted from the pituitary, AVP-containing hypothalamic fibers are widely distributed in the CNS [105]. These fibers reach different centers in the brainstem and, in particular, the trigemminal nuclei. The AVP receptors (VP1 and VP2) are found in the trigeminal ganglion [94]. Thus, the AVP system has many ways to modulate migraine pathophysiology. Since there are no direct fibers containing AVP in the trigeminovascular system, it is likely that the peptide may diffuse into this system. Overall, there exists sufficient evidence to maintain interest in the use of AVP to moderate the onset of headaches [106].

3.6. Prolactin

Prolactin (PRL) is a hormone that is responsible for lactation, breast development, and hundreds of other actions needed to maintain homeostasis. PRL is chemically related to growth hormones and placental lactogen hormones. In an animal model, high levels of prolactin increased meningeal trigeminal pain sensitivity by only affecting CGRP in female rodents [107]. In humans, serum prolactin levels are higher in those with migraine. Individuals with prolactin-secreting pituitary adenomas were found to have a higher incidence of headaches and migraine attacks [108]. With monoclonal antibodies targeting prolactin receptors, a recent report opens new possibilities to better understand the complex interaction between prolactin and CGRP, but blocking prolactin receptors in humans poses risks of interfering with the other functions of this hormone [109].

4. Limitations of Current Methods

Advances in understanding sex hormones in humans are hampered by the challenges of reliably creating an equivalent model of a migraine attack and measuring responses to interventions in animal models. Additionally, the translational value of preclinical studies can be uncertain due to a predominant use of males or not reporting sex as a biological variable [110,111], a reliance on ovariectomies, and modeling hormonal changes in animals that have an estrous cycle rather than a human-like menstrual cycle [112]. Furthermore, the effect of sex hormones on migraine and pain may vary depending on the pain model, model species, and experimental design in laboratory settings. The expert panel identified the lack of an established migraine animal model as one of the barriers to rapid progress in migraine research. For human research, the design of effective human studies has been challenging. Blood sampling of hormone levels is complicated by fluctuation throughout the day and month. The differential effect of sex hormone interventions might be impacted by the delivery method, timing of delivery, and dose, as well as sex, age and other conditions and medications of the patients.
As migraine is inherently a complex disorder involving different biological systems including the nervous, endocrine, endothelial, and immune systems, an interdisciplinary and collaborative approach among clinical and preclinical researchers is encouraged. Furthermore, given the limited number of basic scientists exploring this subject, it is critical that there is a cross-pollination of knowledge and ideas for research between often isolated fields of study. For example, chronic pain, which includes fibromyalgia, back pain, and TMJ overlaps with research performed in immunology, headache medicine, and other medical specialties [113,114]. It will take a dramatic increase and maintained effort in advocacy and support from patients and medical professionals to advance our knowledge of migraine and hormonal pathophysiology enough to lead to hormonal therapies of greater precision and safety.

5. Conclusions and Future Directions

While a large body of research has established hormonal changes and fluctuations as a driver of migraine symptoms in women and transgender people, the relation to hormonal life events is not definitively known for the full range of migraine symptoms. A new HEADS (headache, ear, auditory, dizziness, and sinus) Registry is now available to record and track many of these symptoms (reference: headsregistry.lumiio.com). Additionally, clarification of the mechanisms behind the emergence and recession of different migraine-related symptoms remains. Hormones may have both a causal role in migraine generation and also contribute to pain propagation.
This panel identified several potential hormones and mechanisms that show promise for improved migraine therapeutics, but the conclusion was that more resources need to be concentrated on this significantly debilitating neurovascular condition. In particular, the gender-specific nature of migraine disease calls for the need to better understand how hormones affect the nervous system. New areas of research are required to better understand the mechanisms by which sex hormones relate to changes in migraine symptoms during the periods of hormonal fluctuation in puberty, menstruation, pregnancy, and perimenopause. Translationally relevant animal models of migraine will play a key role in providing mechanistic insights, especially when coupled with clinical data. We highlight the theoretical opportunity to create novel hormone-based therapeutic molecules that might desensitize the hyperactive migraine nervous system without the potential side effects of contraceptives and hormone replacement therapy. Moreover, progress in understanding how hormones affect the nervous system will lead to innovations in treating not only migraine, but other menopausal symptoms.

Author Contributions

Conceptualization, F.G.III; writing—original draft preparation, F.G.III; writing—review and editing, all authors. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

Fred Schwaller, was a freelance writer who contributed to the manuscript; Leigh Serth was a valuable assistant in the manuscript preparation.

Conflicts of Interest

F.G. is a consultant for Allergen and Pfizer. A.O. is a consultant for Crico and Teladocs N.R. consulted for Gerson Lehrman Group, participated in compensated work with AcademicCME. was a Principal investigator (PI) on research with Electrocore, Theranica, Eli Lilly, was an uncompensated PI on research with products of Theraspecs, Dolor technologies, is an advisor for Theranica, is on the NeurologyLive Advisory board., is on the Board and received compensation for editing NeurologyLive issue, is a Board member of Miles for Migraine, and is a Project Advisor for Clinical Awareness Initiative with Clinical Neurological Society of America Inc. N.S. is a member of the Scientific Advisory Boards for Astellas and Menogenix, Inc, companies involved in clinical trials of non-hormone treatments for hot flashes, a consultant for Amazon Project Ember which is developing home measurements of hormones to be applied to women’s health, and is a consultant for Ansh Laboratories, an immunoassay company that specializes in ovarian peptides. D. Y is an inventor of two oxytocin patents—one covering the use in headache, the other covering a magnesium formulation. These have been licensed by Tonix Pharmaceuticals which is pursuing a chronic migraine trial.

References

  1. Burch, R.C.; Buse, D.C.; Lipton, R.B. Migraine: Epidemiology, Burden, and Comorbidity. Neurol. Clin. 2019, 37, 631–649. [Google Scholar] [CrossRef] [PubMed]
  2. Tsai, C.K.; Tsai, C.L.; Lin, G.Y.; Yang, F.C.; Wang, S.J. Sex Differences in Chronic Migraine: Focusing on Clinical Features, Pathophysiology, and Treatments. Curr. Pain Headache Rep. 2022, 26, 347–355. [Google Scholar] [CrossRef] [PubMed]
  3. Vos, T.; Abajobir, A.A.; Abate, K.H.; Abbafati, C.; Abbas, K.M.; Abd-Allah, F.; Abdulkader, R.S.; Abdulle, A.M.; Abebo, T.A.; Abera, S.F.; et al. Global, regional, and national incidence, prevalence, and years lived with disability for 328 diseases and injuries for 195 countries, 1990–2016: A systematic analysis for the Global Burden of Disease Study 2016. Lancet 2017, 390, 1211–1259. [Google Scholar] [CrossRef] [PubMed]
  4. Szperka, C. Headache in Children and Adolescents. Continuum 2021, 27, 703–731. [Google Scholar] [CrossRef] [PubMed]
  5. Tonini, M.C. Gender differences in migraine. Neurol. Sci. 2018, 39, 77–78. [Google Scholar] [CrossRef] [PubMed]
  6. Vetvik, K.G.; MacGregor, E.A. Sex differences in the epidemiology, clinical features, and pathophysiology of migraine. Lancet Neurol. 2017, 16, 76–87. [Google Scholar] [CrossRef]
  7. MacGregor, E.A.; Frith, A.; Ellis, J.; Aspinall, L.; Hackshaw, A. Incidence of migraine relative to menstrual cycle phases of rising and falling estrogen. Neurology 2006, 67, 2154–2158. [Google Scholar] [CrossRef] [PubMed]
  8. Chalmer, M.A.; Kogelman, L.J.A.; Ullum, H.; Sørensen, E.; Didriksen, M.; Mikkelsen, S.; Dinh, K.M.; Brodersen, T.; Nielsen, K.R.; Bruun, M.T.; et al. Population-Based Characterization of Menstrual Migraine and Proposed Diagnostic Criteria. JAMA Netw. Open 2023, 6, e2313235. [Google Scholar] [CrossRef]
  9. Pavlović, J.M.; Allshouse, A.A.; Santoro, N.F.; Crawford, S.L.; Thurston, R.C.; Neal-Perry, G.S.; Lipton, R.B.; Derby, C.A. Sex hormones in women with and without migraine: Evidence of migraine-specific hormone profiles. Neurology 2016, 87, 49–56. [Google Scholar] [CrossRef]
  10. Calhoun, A.H.; Batur, P. Combined hormonal contraceptives and migraine: An update on the evidence. Clevel. Clin. J. Med. 2017, 84, 631–638. [Google Scholar] [CrossRef]
  11. Warhurst, S.; Rofe, C.J.; Brew, B.J.; Bateson, D.; McGeechan, K.; Merki-Feld, G.S.; Garrick, R.; Tomlinson, S.E. Effectiveness of the progestin-only pill for migraine treatment in women: A systematic review and meta-analysis. Cephalalgia 2018, 38, 754–764. [Google Scholar] [CrossRef] [PubMed]
  12. Lyall, M.; de Oliveira, B.R.; Mody, S.K. Considerations for Contraceptive Use Among Patients with Migraines. Curr. Obstet. Gynecol. Rep. 2023, 12, 57–63. [Google Scholar] [CrossRef]
  13. MacGregor, E.A. Migraine, menopause and hormone replacement therapy. Post Reprod. Health 2018, 24, 11–18. [Google Scholar] [CrossRef]
  14. Aubé, M. Migraine in pregnancy. Neurology 1999, 53 (Suppl. S1), S26–S28. [Google Scholar]
  15. Ertresvåg, J.M.; Zwart, J.A.; Helde, G.; Johnsen, H.J.; Bovim, G. Headache and transient focal neurological symptoms during pregnancy, a prospective cohort. Acta Neurol. Scand. 2005, 111, 233–237. [Google Scholar] [CrossRef]
  16. Kvisvik, E.V.; Stovner, L.J.; Helde, G.; Bovim, G.; Linde, M. Headache and migraine during pregnancy and puerperium: The MIGRA-study. J. Headache Pain 2011, 12, 443–451. [Google Scholar] [CrossRef]
  17. Goadsby, P.J.; Goldberg, J.; Silberstein, S.D. Migraine in pregnancy. BMJ 2008, 336, 1502–1504. [Google Scholar] [CrossRef] [PubMed]
  18. Robbins, M.S.; Farmakidis, C.; Dayal, A.K.; Lipton, R.B. Acute headache diagnosis in pregnant women: A hospital-based study. Neurology 2015, 85, 1024–1030. [Google Scholar] [CrossRef]
  19. Sances, G.; Granella, F.; Nappi, R.E.; Fignon, A.; Ghiotto, N.; Polatti, F.; Nappi, G. Course of migraine during pregnancy and postpartum: A prospective study. Cephalalgia 2003, 23, 197–205. [Google Scholar] [CrossRef]
  20. Wells, R.E.; Turner, D.P.; Lee, M.; Bishop, L.; Strauss, L. Managing Migraine during Pregnancy and Lactation. Curr. Neurol. Neurosci. Rep. 2016, 16, 40. [Google Scholar] [CrossRef]
  21. Mueller, L. Predictability of exogenous hormone effect on subgroups of migraineurs. Headache 2000, 40, 189–193. [Google Scholar] [CrossRef] [PubMed]
  22. Hodson, J.; Thompson, J.; Al-Azzawi, F. Headache at menopause and in hormone replacement therapy users. Climacteric J. Int. Menopause Soc. 2000, 3, 119–124. [Google Scholar] [CrossRef] [PubMed]
  23. Ibrahimi, K.; Couturier, E.G.M.; MaassenVanDenBrink, A. Migraine and perimenopause. Maturitas 2014, 78, 277–280. [Google Scholar] [CrossRef] [PubMed]
  24. Granella, F.; Sances, G.; Zanferrari, C.; Costa, A.; Martignoni, E.; Manzoni, G.C. Migraine without aura and reproductive life events: A clinical epidemiological study in 1300 women. Headache 1993, 33, 385–389. [Google Scholar] [CrossRef] [PubMed]
  25. Cupini, L.M.; Matteis, M.; Troisi, E.; Calabresi, P.; Bernardi, G.; Silvestrini, M. Sex-hormone-related events in migrainous females. A clinical comparative study between migraine with aura and migraine without aura. Cephalalgia 1995, 15, 140–144. [Google Scholar] [CrossRef] [PubMed]
  26. Wang, S.J.; Fuh, J.L.; Lu, S.R.; Juang, K.D.; Wang, P.H. Migraine prevalence during menopausal transition. Headache 2003, 43, 470–478. [Google Scholar] [CrossRef] [PubMed]
  27. Freeman, E.W.; Sammel, M.D.; Lin, H.; Gracia, C.R.; Kapoor, S. Symptoms in the menopausal transition: Hormone and behavioral correlates. Obstet. Gynecol. 2008, 111, 127–136. [Google Scholar] [CrossRef]
  28. Mattsson, P. Hormonal factors in migraine: A population-based study of women aged 40 to 74 years. Headache 2003, 43, 27–35. [Google Scholar] [CrossRef]
  29. Park, J.H.; Viirre, E. Vestibular migraine may be an important cause of dizziness/vertigo in perimenopausal period. Med. Hypotheses 2010, 75, 409–414. [Google Scholar] [CrossRef]
  30. Pavlović, J.M. Evaluation and management of migraine in midlife women. Menopause 2018, 25, 927–929. [Google Scholar] [CrossRef]
  31. Kaiser, H.J.; Meienberg, O. Deterioration or onset of migraine under oestrogen replacement therapy in the menopause. J. Neurol. 1993, 240, 195–196. [Google Scholar] [CrossRef]
  32. Sacco, S.; Ricci, S.; Degan, D.; Carolei, A. Migraine in women: The role of hormones and their impact on vascular diseases. J. Headache Pain 2012, 13, 177–189. [Google Scholar] [CrossRef]
  33. Burke, B.E.; Olson, R.D.; Cusack, B.J. Randomized, controlled trial of phytoestrogen in the prophylactic treatment of menstrual migraine. Biomed. Pharmacother. 2002, 56, 283–288. [Google Scholar] [CrossRef] [PubMed]
  34. Patisaul, H.B.; Jefferson, W. The pros and cons of phytoestrogens. Front. Neuroendocrinol. 2010, 31, 400–419. [Google Scholar] [CrossRef]
  35. Chen, M.N.; Lin, C.C.; Liu, C.F. Efficacy of phytoestrogens for menopausal symptoms: A meta-analysis and systematic review. Climacteric 2015, 18, 260–269. [Google Scholar] [CrossRef]
  36. Rietjens, I.M.C.M.; Louisse, J.; Beekmann, K. The potential health effects of dietary phytoestrogens. Br. J. Pharmacol. 2017, 174, 1263–1280. [Google Scholar] [CrossRef]
  37. Sarajari, S.; Oblinger, M.M. Estrogen Effects on Pain Sensitivity and Neuropeptide Expression in Rat Sensory Neurons. Exp. Neurol. 2010, 224, 163–169. [Google Scholar] [CrossRef] [PubMed]
  38. Welch, K.M.A.; Brandes, J.L.; Berman, N.E.J. Mismatch in how oestrogen modulates molecular and neuronal function may explain menstrual migraine. Neurol. Sci. 2006, 27 (Suppl. S2), S190–S192. [Google Scholar] [CrossRef] [PubMed]
  39. Wattiez, A.S.; Sowers, L.P.; Russo, A.F. Calcitonin gene-related peptide (CGRP): Role in migraine pathophysiology and therapeutic targeting. Expert Opin. Ther. Targets 2020, 24, 91–100. [Google Scholar] [CrossRef] [PubMed]
  40. Puri, V.; Cui, L.; Liverman, C.S.; Roby, K.F.; Klein, R.M.; Welch, K.M.A.; Berman, N.E.J. Ovarian steroids regulate neuropeptides in the trigeminal ganglion. Neuropeptides 2005, 39, 409–417. [Google Scholar] [CrossRef]
  41. Rettberg, J.R.; Yao, J.; Brinton, R.D. Estrogen: A master regulator of bioenergetic systems in the brain and body. Front. Neuroendocrinol. 2014, 35, 8–30. [Google Scholar] [CrossRef] [PubMed]
  42. Kudo, C.; Harriott, A.M.; Moskowitz, M.A.; Waeber, C.; Ayata, C. Estrogen modulation of cortical spreading depression. J. Headache Pain 2023, 24, 62. [Google Scholar] [CrossRef] [PubMed]
  43. Chen, Q.; Zhang, W.; Sadana, N.; Chen, X. Estrogen receptors in pain modulation: Cellular signaling. Biol. Sex Differ. 2021, 12, 22. [Google Scholar] [CrossRef] [PubMed]
  44. Kelly, M.J.; Rønnekleiv, O.K. Minireview: Neural Signaling of Estradiol in the Hypothalamus. Mol. Endocrinol. 2015, 29, 645–657. [Google Scholar] [CrossRef]
  45. Stincic, T.L.; Grachev, P.; Bosch, M.A.; Rønnekleiv, O.K.; Kelly, M.J. Estradiol Drives the Anorexigenic Activity of Proopiomelanocortin Neurons in Female Mice. eNeuro 2018, 5, ENEURO.0103-18.2018. [Google Scholar] [CrossRef]
  46. Cahill, C.M.; Cook, C.; Pickens, S. Migraine and Reward System—Or Is It Aversive? Curr. Pain Headache Rep. 2014, 18, 410. [Google Scholar] [CrossRef]
  47. Krentzel, A.A.; Proaño, S.B.; Dorris, D.M.; Setzer, B.; Meitzen, J. The estrous cycle and 17β-estradiol modulate the electrophysiological properties of rat nucleus accumbens core medium spiny neurons. J. Neuroendocrinol. 2022, 34, e13122. [Google Scholar] [CrossRef]
  48. Yoest, K.E.; Quigley, J.A.; Becker, J.B. Rapid effects of ovarian hormones in dorsal striatum and nucleus accumbens. Horm. Behav. 2018, 104, 119–129. [Google Scholar] [CrossRef]
  49. Aggarwal, M.; Puri, V.; Puri, S. Effects of estrogen on the serotonergic system and calcitonin gene-related peptide in trigeminal ganglia of rats. Ann. Neurosci. 2012, 19, 151–157. [Google Scholar] [CrossRef] [PubMed]
  50. Bereiter, D.A.; Stanford, L.R.; Barker, D.J. Hormone-induced enlargement of receptive fields in trigeminal mechanoreceptive neurons. II. possible mechanisms. Brain Res. 1980, 184, 411–423. [Google Scholar] [CrossRef] [PubMed]
  51. Schertzinger, M.; Wesson-Sides, K.; Parkitny, L.; Younger, J. Daily Fluctuations of Progesterone and Testosterone Are Associated with Fibromyalgia Pain Severity. J. Pain 2018, 19, 410–417. [Google Scholar] [CrossRef]
  52. Chuang, S.H.; Reddy, D.S. 3β-Methyl-Neurosteroid Analogs Are Preferential Positive Allosteric Modulators and Direct Activators of Extrasynaptic δ-Subunit γ-Aminobutyric Acid Type A Receptors in the Hippocampus Dentate Gyrus Subfield. J. Pharmacol. Exp. Ther. 2018, 365, 583–601. [Google Scholar] [CrossRef]
  53. Reddy, D.S. Neurosteroids: Endogenous role in the human brain and therapeutic potentials. Prog. Brain Res. 2010, 186, 113–137. [Google Scholar] [CrossRef] [PubMed]
  54. Singh, M. Ovarian Hormones Elicit Phosphorylation of Akt and Extracellular-Signal Regulated Kinase in Explants of the Cerebral Cortex. ENDO 2001, 14, 407–416. [Google Scholar] [CrossRef] [PubMed]
  55. Jang, Y.; Kim, M.; Hwang, S.W. Molecular mechanisms underlying the actions of arachidonic acid-derived prostaglandins on peripheral nociception. J. Neuroinflamm. 2020, 17, 30. [Google Scholar] [CrossRef] [PubMed]
  56. Andersen, M.L.; Bittencourt, L.R.A.; Antunes, B.I.; Tufik, S. Effects of Progesterone on Sleep: A Possible Pharmacological Treatment for Sleep-Breathing Disorders? CMC 2006, 13, 3575–3582. [Google Scholar] [CrossRef]
  57. Standeven, L.R.; McEvoy, K.O.; Osborne, L.M. Progesterone, reproduction, and psychiatric illness. Best Pract. Res. Clin. Obstet. Gynaecol. 2020, 69, 108–126. [Google Scholar] [CrossRef]
  58. Simpson, E.R. Sources of estrogen and their importance. J. Steroid Biochem. Mol. Biol. 2003, 86, 225–230. [Google Scholar] [CrossRef]
  59. Bartley, E.J.; Palit, S.; Kuhn, B.L.; Kerr, K.L.; Terry, E.L.; DelVentura, J.L.; Rhudy, J.L. Nociceptive processing in women with premenstrual dysphoric disorder (PMDD): The role of menstrual phase and sex hormones. Clin. J. Pain 2015, 31, 304–314. [Google Scholar] [CrossRef]
  60. Bartley, E.J.; Palit, S.; Kuhn, B.L.; Kerr, K.L.; Terry, E.L.; DelVentura, J.L.; Rhudy, J.L. Natural variation in testosterone is associated with hypoalgesia in healthy women. Clin. J. Pain 2015, 31, 730–739. [Google Scholar] [CrossRef]
  61. Choi, J.C.; Park, Y.H.; Park, S.K.; Lee, J.S.; Kim, J.; Choi, J.I.; Yoon, K.B.; Lee, S.; Lim, D.E.; Choi, J.Y.; et al. Testosterone effects on pain and brain activation patterns. Acta Anaesthesiol. Scand. 2017, 61, 668–675. [Google Scholar] [CrossRef]
  62. Choi, J.C.; Chung, M.I.; Lee, Y.D. Modulation of pain sensation by stress-related testosterone and cortisol. Anaesthesia 2012, 67, 1146–1151. [Google Scholar] [CrossRef] [PubMed]
  63. Teepker, M.; Peters, M.; Vedder, H.; Schepelmann, K.; Lautenbacher, S. Menstrual variation in experimental pain: Correlation with gonadal hormones. Neuropsychobiology 2010, 61, 131–140. [Google Scholar] [CrossRef]
  64. Aloisi, A.M.; Ceccarelli, I.; Fiorenzani, P. Gonadectomy affects hormonal and behavioral responses to repetitive nociceptive stimulation in male rats. Ann. N. Y. Acad. Sci. 2003, 1007, 232–237. [Google Scholar] [CrossRef] [PubMed]
  65. Ceccarelli, I.; Scaramuzzino, A.; Massafra, C.; Aloisi, A.M. The behavioral and neuronal effects induced by repetitive nociceptive stimulation are affected by gonadal hormones in male rats. Pain 2003, 104, 35–47. [Google Scholar] [CrossRef] [PubMed]
  66. Gaumond, I.; Arsenault, P.; Marchand, S. Specificity of female and male sex hormones on excitatory and inhibitory phases of formalin-induced nociceptive responses. Brain Res. 2005, 1052, 105–111. [Google Scholar] [CrossRef] [PubMed]
  67. Aloisi, A.M.; Ceccarelli, I.; Fiorenzani, P.; De Padova, A.M.; Massafra, C. Testosterone affects formalin-induced responses differently in male and female rats. Neurosci. Lett. 2004, 361, 262–264. [Google Scholar] [CrossRef] [PubMed]
  68. Stoffel, E.C.; Ulibarri, C.M.; Craft, R.M. Gonadal steroid hormone modulation of nociception, morphine antinociception and reproductive indices in male and female rats. Pain 2003, 103, 285–302. [Google Scholar] [CrossRef]
  69. Basaria, S.; Travison, T.G.; Alford, D.; Knapp, P.E.; Teeter, K.; Cahalan, C.; Eder, R.; Lakshman, K.; Bachman, E.; Mensing, G.; et al. Effects of testosterone replacement in men with opioid-induced androgen deficiency: A randomized controlled trial. Pain 2015, 156, 280–288. [Google Scholar] [CrossRef]
  70. Verhagen, I.E.; Brandt, R.B.; Kruitbosch, C.M.A.; MaassenVanDenBrink, A.; Fronczek, R.; Terwindt, G.M. Clinical symptoms of androgen deficiency in men with migraine or cluster headache: A cross-sectional cohort study. J. Headache Pain 2021, 22, 125. [Google Scholar] [CrossRef]
  71. Martinez, C.I.; Liktor-Busa, E.; Largent-Milnes, T.M. Molecular mechanisms of hormones implicated in migraine and the translational implication for transgender patients. Front. Pain Res. 2023, 4, 1117842. [Google Scholar] [CrossRef]
  72. Ahmad, A.H.; Ismail, Z. c-fos and its Consequences in Pain. Malays. J. Med. Sci. 2002, 9, 3–8. [Google Scholar] [PubMed]
  73. White, H.D.; Brown, L.A.J.; Gyurik, R.J.; Manganiello, P.D.; Robinson, T.D.; Hallock, L.S.; Lewis, L.D.; Yeo, Y.-T.J. Treatment of pain in fibromyalgia patients with testosterone gel: Pharmacokinetics and clinical response. Int. Immunopharmacol. 2015, 27, 249–256. [Google Scholar] [CrossRef] [PubMed]
  74. Fischer, L.; Clemente, J.T.; Tambeli, C.H. The protective role of testosterone in the development of temporomandibular joint pain. J. Pain 2007, 8, 437–442. [Google Scholar] [CrossRef] [PubMed]
  75. Glaser, R.; Dimitrakakis, C.; Trimble, N.; Martin, V. Testosterone pellet implants and migraine headaches: A pilot study. Maturitas 2012, 71, 385–388. [Google Scholar] [CrossRef] [PubMed]
  76. English, K.M.; Steeds, R.P.; Jones, T.H.; Diver, M.J.; Channer, K.S. Low-dose transdermal testosterone therapy improves angina threshold in men with chronic stable angina: A randomized, double-blind, placebo-controlled study. Circulation 2000, 102, 1906–1911. [Google Scholar] [CrossRef] [PubMed]
  77. Tzabazis, A.; Kori, S.; Mechanic, J.; Miller, J.; Pascual, C.; Manering, N.; Carson, D.; Klukinov, M.; Spierings, E.; Jacobs, D.; et al. Oxytocin and Migraine Headache. Headache 2017, 57 (Suppl. S2), 64–75. [Google Scholar] [CrossRef] [PubMed]
  78. Rash, J.A.; Aguirre-Camacho, A.; Campbell, T.S. Oxytocin and pain: A systematic review and synthesis of findings. Clin. J. Pain 2014, 30, 453–462. [Google Scholar] [CrossRef] [PubMed]
  79. Shamay-Tsoory, S.G.; Abu-Akel, A. The Social Salience Hypothesis of Oxytocin. Biol. Psychiatry 2016, 79, 194–202. [Google Scholar] [CrossRef]
  80. Phillips, W.J.; Ostrovsky, O.; Galli, R.L.; Dickey, S. Relief of acute migraine headache with intravenous oxytocin: Report of two cases. J. Pain Palliat. Care Pharmacother. 2006, 20, 25–28. [Google Scholar] [CrossRef]
  81. Wang, Y.L.; Yuan, Y.; Yang, J.; Wang, C.H.; Pan, Y.J.; Lu, L.; Wu, Y.Q.; Wang, D.X.; Lv, L.X.; Li, R.R.; et al. The interaction between the oxytocin and pain modulation in headache patients. Neuropeptides 2013, 47, 93–97. [Google Scholar] [CrossRef]
  82. Paloyelis, Y.; Krahé, C.; Maltezos, S.; Williams, S.C.; Howard, M.A.; Fotopoulou, A. The Analgesic Effect of Oxytocin in Humans: A Double-Blind, Placebo-Controlled Cross-Over Study Using Laser-Evoked Potentials. J. Neuroendocrinol. 2016, 28. [Google Scholar] [CrossRef] [PubMed]
  83. MacDonald, E.; Dadds, M.R.; Brennan, J.L.; Williams, K.; Levy, F.; Cauchi, A.J. A review of safety, side-effects and subjective reactions to intranasal oxytocin in human research. Psychoneuroendocrinology 2011, 36, 1114–1126. [Google Scholar] [CrossRef] [PubMed]
  84. Iwasaki, M.; Lefevre, A.; Althammer, F.; Creusot, E.C.; Łąpieś, O.; Petitjean, H.; Hilfiger, L.; Kerspern, D.; Melchior, M.; Küppers, S.; et al. An analgesic pathway from parvocellular oxytocin neurons to the periaqueductal gray in rats. Nat. Commun. 2023, 14, 1066. [Google Scholar] [CrossRef] [PubMed]
  85. Swanson, L.W.; McKellar, S. The distribution of oxytocin- and neurophysin-stained fibers in the spinal cord of the rat and monkey. J. Comp. Neurol. 1979, 188, 87–106. [Google Scholar] [CrossRef] [PubMed]
  86. Breton, J.D.; Veinante, P.; Uhl-Bronner, S.; Vergnano, A.M.; Freund-Mercier, M.J.; Schlichter, R.; Poisbeau, P. Oxytocin-induced antinociception in the spinal cord is mediated by a subpopulation of glutamatergic neurons in lamina I-II which amplify GABAergic inhibition. Mol. Pain 2008, 4, 19. [Google Scholar] [CrossRef] [PubMed]
  87. Rojas-Piloni, G.; López-Hidalgo, M.; Martínez-Lorenzana, G.; Rodríguez-Jiménez, J.; Condés-Lara, M. GABA-mediated oxytocinergic inhibition in dorsal horn neurons by hypothalamic paraventricular nucleus stimulation. Brain Res. 2007, 1137, 69–77. [Google Scholar] [CrossRef] [PubMed]
  88. Condés-Lara, M.; González, N.M.; Martínez-Lorenzana, G.; Delgado, O.L.; Freund-Mercier, M.J. Actions of oxytocin and interactions with glutamate on spontaneous and evoked dorsal spinal cord neuronal activities. Brain Res. 2003, 976, 75–81. [Google Scholar] [CrossRef]
  89. Miranda-Cardenas, Y.; Rojas-Piloni, G.; Martínez-Lorenzana, G.; Rodríguez-Jiménez, J.; López-Hidalgo, M.; Freund-Mercier, M.J.; Condés-Lara, M. Oxytocin and electrical stimulation of the paraventricular hypothalamic nucleus produce antinociceptive effects that are reversed by an oxytocin antagonist. Pain 2006, 122, 182–189. [Google Scholar] [CrossRef]
  90. Yang, J.; Liang, J.Y.; Li, P.; Pan, Y.J.; Qiu, P.Y.; Zhang, J.; Hao, F.; Wang, D.X. Oxytocin in the periaqueductal gray participates in pain modulation in the rat by influencing endogenous opiate peptides. Peptides 2011, 32, 1255–1261. [Google Scholar] [CrossRef]
  91. Yang, J.; Li, P.; Liang, J.Y.; Pan, Y.J.; Yan, X.Q.; Yan, F.L.; Hao, F.; Zhang, X.Y.; Zhang, J.; Qiu, P.Y.; et al. Oxytocin in the periaqueductal grey regulates nociception in the rat. Regul. Pept. 2011, 169, 39–42. [Google Scholar] [CrossRef]
  92. Yang, J.; Yang, Y.; Chen, J.M.; Liu, W.Y.; Wang, C.H.; Lin, B.C. Central oxytocin enhances antinociception in the rat. Peptides 2007, 28, 1113–1119. [Google Scholar] [CrossRef] [PubMed]
  93. Yang, J.; Yang, Y.; Chen, J.M.; Liu, W.Y.; Lin, B.C. Investigating the role of the hypothalamic supraoptic nucleus in nociception in the rat. Life Sci. 2008, 82, 166–173. [Google Scholar] [CrossRef] [PubMed]
  94. Warfvinge, K.; Krause, D.N.; Maddahi, A.; Grell, A.-S.; Edvinsson, J.C.A.; Haanes, K.A.; Edvinsson, L. Oxytocin as a regulatory neuropeptide in the trigeminovascular system: Localization, expression and function of oxytocin and oxytocin receptors. Cephalalgia 2020, 40, 1283–1295. [Google Scholar] [CrossRef] [PubMed]
  95. Warfvinge, K.; Krause, D.; Edvinsson, L. The distribution of oxytocin and the oxytocin receptor in rat brain: Relation to regions active in migraine. J. Headache Pain 2020, 21, 10. [Google Scholar] [CrossRef] [PubMed]
  96. Huang, C.L.; Liu, F.; Zhang, Y.Y.; Lin, J.; Fu, M.; Li, Y.L.; Zhou, C.; Li, C.J.; Shen, J.F. Activation of oxytocin receptor in the trigeminal ganglion attenuates orofacial ectopic pain attributed to inferior alveolar nerve injury. J. Neurophysiol. 2021, 125, 223–231. [Google Scholar] [CrossRef] [PubMed]
  97. Bharadwaj, V.N.; Porreca, F.; Cowan, R.P.; Kori, S.; Silberstein, S.D.; Yeomans, D.C. A new hypothesis linking oxytocin to menstrual migraine. Headache J. Head Face Pain 2021, 61, 1051–1059. [Google Scholar] [CrossRef] [PubMed]
  98. Dalkara, T.; Nozari, A.; Moskowitz, M.A. Migraine aura pathophysiology: The role of blood vessels and microembolisation. Lancet Neurol. 2010, 9, 309–317. [Google Scholar] [CrossRef] [PubMed]
  99. Gupta, V. Does Vasopressin Serve a Vasomotor Adaptive Function in Migraine? Cephalalgia 1993, 13, 221. [Google Scholar] [CrossRef]
  100. Buschmann, J.; Leppla-Wollsiffer, G.; Nemeth, N.; Nelson, K.; Kirsten, R. Migraine patients show increased platelet vasopressin receptors. Headache 1996, 36, 586–588. [Google Scholar] [CrossRef]
  101. Hampton, K.K.; Esack, A.; Peatfield, R.C.; Grant, P.J. Elevation of plasma vasopressin in spontaneous migraine. Cephalalgia 1991, 11, 249–250. [Google Scholar] [CrossRef]
  102. Peatfield, R.C.; Hampton, K.K.; Grant, P.J. Plasma vasopressin levels in induced migraine attacks. Cephalalgia 1988, 8, 55–57. [Google Scholar] [CrossRef]
  103. Bahadoram, M.; Mahmoudian-Sani, M.R.; Keikhaei, B.; Alikhani, K.; Bahadoram, S. The antimigraine action of arginine-vasopressin: A theoretical basis. Future Neurol. 2020, 15, FNL51. [Google Scholar] [CrossRef]
  104. Yang, J.; Lu, L.; Wang, H.C.; Zhan, H.Q.; Hai, G.F.; Pan, Y.J.; Lv, Q.Q.; Wang, D.X.; Wu, Y.Q.; Li, R.R.; et al. Effect of intranasal arginine vasopressin on human headache. Peptides 2012, 38, 100–104. [Google Scholar] [CrossRef] [PubMed]
  105. Warfvinge, K.; Krause, D.N.; Maddahi, A.; Edvinsson, J.C.A.; Edvinsson, L.; Haanes, K.A. Estrogen receptors α, β and GPER in the CNS and trigeminal system—Molecular and functional aspects. J. Headache Pain 2020, 21, 131. [Google Scholar] [CrossRef]
  106. Maddahi, A.; Edvinsson, L.; Warfvinge, K. Expression of vasopressin and its receptors in migraine-related regions in CNS and the trigeminal system: Influence of sex. J. Headache Pain 2022, 23, 152. [Google Scholar] [CrossRef]
  107. Avona, A.; Mason, B.N.; Burgos-Vega, C.; Hovhannisyan, A.H.; Belugin, S.N.; Mecklenburg, J.; Goffin, V.; Wajahat, N.; Price, T.J.; Akopian, A.N.; et al. Meningeal CGRP-Prolactin Interaction Evokes Female-Specific Migraine Behavior. Ann. Neurol. 2021, 89, 1129–1144. [Google Scholar] [CrossRef] [PubMed]
  108. Al-Karagholi, M.A.M.; Kalatharan, V.; Ghanizada, H.; Gram, C.; Dussor, G.; Ashina, M. Prolactin in headache and migraine: A systematic review of clinical studies. Cephalalgia 2023, 43, 3331024221136286. [Google Scholar] [CrossRef]
  109. Maciuba, S.; Bowden, G.D.; Stratton, H.J.; Wisniewski, K.; Schteingart, C.D.; Almagro, J.C.; Valadon, P.; Lowitz, J.; Glaser, S.M.; Lee, G.; et al. Discovery and characterization of prolactin neutralizing monoclonal antibodies for the treatment of female-prevalent pain disorders. mAbs 2023, 15, 2254676. [Google Scholar] [CrossRef] [PubMed]
  110. Mamlouk, G.M.; Dorris, D.M.; Barrett, L.R.; Meitzen, J. Sex bias and omission in neuroscience research is influenced by research model and journal, but not reported NIH funding. Front. Neuroendocrinol. 2020, 57, 100835. [Google Scholar] [CrossRef]
  111. Will, T.R.; Proaño, S.B.; Thomas, A.M.; Kunz, L.M.; Thompson, K.C.; Ginnari, L.A.; Jones, C.H.; Lucas, S.-C.; Reavis, E.M.; Dorris, D.M.; et al. Problems and Progress regarding Sex Bias and Omission in Neuroscience Research. eNeuro 2017, 4, ENEURO.0278-17.2017. [Google Scholar] [CrossRef] [PubMed]
  112. Labastida-Ramírez, A.; Rubio-Beltrán, E.; Villalón, C.M.; MaassenVanDenBrink, A. Gender aspects of CGRP in migraine. Cephalalgia 2019, 39, 435–444. [Google Scholar] [CrossRef] [PubMed]
  113. Lenert, M.E.; Avona, A.; Garner, K.M.; Barron, L.R.; Burton, M.D. Sensory Neurons, Neuroimmunity, and Pain Modulation by Sex Hormones. Endocrinology 2021, 162, bqab109. [Google Scholar] [CrossRef]
  114. Harvey, M.P.; Dubois, M.C.; Chalaye, P.; Sansoucy, Y.; Marchand, S. Sex-Related Effects of Adrenergic Drugs on Conditioned Pain Modulation: A Randomized Controlled Cross-Over Double-Blind Trial. Pain Res. Manag. 2022, 2022, 2757101. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Godley, F., III; Meitzen, J.; Nahman-Averbuch, H.; O’Neal, M.A.; Yeomans, D.; Santoro, N.; Riggins, N.; Edvinsson, L. How Sex Hormones Affect Migraine: An Interdisciplinary Preclinical Research Panel Review. J. Pers. Med. 2024, 14, 184. https://doi.org/10.3390/jpm14020184

AMA Style

Godley F III, Meitzen J, Nahman-Averbuch H, O’Neal MA, Yeomans D, Santoro N, Riggins N, Edvinsson L. How Sex Hormones Affect Migraine: An Interdisciplinary Preclinical Research Panel Review. Journal of Personalized Medicine. 2024; 14(2):184. https://doi.org/10.3390/jpm14020184

Chicago/Turabian Style

Godley, Frederick, III, John Meitzen, Hadas Nahman-Averbuch, Mary Angela O’Neal, David Yeomans, Nanette Santoro, Nina Riggins, and Lars Edvinsson. 2024. "How Sex Hormones Affect Migraine: An Interdisciplinary Preclinical Research Panel Review" Journal of Personalized Medicine 14, no. 2: 184. https://doi.org/10.3390/jpm14020184

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop