Review Article
More Information

Submitted: February 14, 2024 | Approved: March 19, 2024 | Published: March 20, 2024

How to cite this article: Gupta VK. Laws of Pathophysiology of Migraine in the Third Millennium. J Neurosci Neurol Disord. 2024; 8: 024-032.

DOI: 10.29328/journal.jnnd.1001094

Copyright License: © 2024 Gupta VK. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Keywords: Migraine; Cortical spreading depression; Scintillating Scotoma (SS); Trigeminal nerve; Ophthalmic division of the trigeminal nerve; Autonomic nervous system; Intraocular pressure; Migraine-glaucoma-nexus; Digital ocular massage

 FullText PDF

Laws of Pathophysiology of Migraine in the Third Millennium

Vinod Kumar Gupta*

Medical Director, Physician, Gupta Medical Clinic, S-407, Greater Kailash-Part Two, New Delhi-110048, India

*Address for Correspondence: Dr. Vinod Kumar Gupta, M.B.; B, S; M.D. (General Medicine), Medical Director, Physician, Gupta Medical Clinic, S-407, Greater Kailash-Part Two, New Delhi-110048, India, Email: dr_vkgupta@yahoo.com

Science is the art of systematic and reproducible measurements, ultimately leading to knowledge supported by a holistic logic. Besides serendipity, there are 6 ways in general to obtain knowledge: authoritarianism; mysticism; rationalism, empiricism; pragmatism; and scepticism. Over the last 100 years, a canonical mythology – cortical spreading depression (CSD) – has prevailed in migraine pathophysiology. Conversely, a well-defined adaptive/protective role has evolved for CSD in locusts, Drosophila, and mammals. Additionally, an elaborate but entirely symptomatic nosologic system has arbitrarily evolved in migraine / primary headache. While the so-called systematic but symptomatic classification system of migraine / primary headache keeps on advancing the data-bank exponentially, the cause-effect nexus continues to obscure the most important systematic and insightful components of the knowledge of primary headache. The first step in advancing the cause-effect mystery of migraine / primary headache is to create a conceptual, consistent, and important adaptive-pathogenetic divide in the massive and disparate data-linked pathophysiology of the disorder. Once certain definitive principles (not laboratory/neuroimaging / genetic/epidemiologic data) emerge in the science of migraine / primary headache, we become empowered to understand the complex but key phenotypic blueprint as well as the neuro-pathophysiology / neuropsychiatry of the entity, including the visual (nasal visual-field sparing digitally-displaceable and eyeball-movement-synchronous scintillating scotomata), the lateralizing fronto-temporal-nuchal headache exclusively involving the ophthalmic division of the trigeminal nerve, and the associated features such as ‘stress’, ‘post-stress’, ‘autonomic storm’, ‘protean’ and ‘spontaneous’ onset and offset, and headache-aborting nausea-vomiting. In this manner, we have also evolved principles to begin to understand the most complex female predominance of migraine patients in adults [F:M=3:1] as well as the decline of prevalence in migraine attacks following menopause and advancing age. The Laws of the Pathophysiology of Migraine encompass the invaluable neurological / neuro-ophthalmological shift in pathophysiology from the brain to the eye.

Cortical Spreading Depression (CSD) is implicated in a diverse set of intracranial pathologies, and, in the early 21st Century, with over 100 years of experimentation and deduction, is the generally accepted pathogenetic mechanism linked to neurogenic inflammation (NI) that is widely accepted to underlie migraine [1-6]. With the exception of traumatic brain injury in humans, CSD has been recorded by several methods only in experimental animals. Besides this limitation in human migraine patients, there is a well-known protective/adaptive role for Spreading Depression (SD)-like events in lower vertebrates, such as locusts, for surviving extreme environmental conditions through a highly conserved pathway that may modulate SD in other organisms, including Drosophila, and, other mammals [7-12] as well as an exhaustively investigated neurovascular pre-conditioning dependent neuroprotective effect by increasing genome stability, and, neurovascular pre-conditioning effect in rodents [13-17]. These neuronal/neurovascular biologically adaptive/protective effects of CSD / SD that are well-known over 3 decades are systematically excluded in attempted syntheses by a large school of scientists that maintain a pathogenetic hyper-to-exclusive focus for CSD with a potentially therapeutic role for suppression of CSD/SD, particularly for migraine, epilepsy, transient global amnesia, epilepsy or cerebrovascular disorders in the neurocritical care unit [6,14]. Brain neuronal death in CSD-linked otherwise normal brains is not associated with immediate early genes, growth factors/neurotransmitters/neuromodulatory and/or inflammatory mediator systems. Primary/spontaneous or periodic “self-cycling” onset and “self-limited” offset of heterogenous headache as well as nasal visual-field sparing non-homonymous scintillating scotomata/hallucinations/visual experiences that are digitally displaceable by physical pressure and synchronously mobile with eye movements are absolutely inexplicable through CSD / NI or through any other form of primary CNS dysfunction (see below) [18]. Several other forms of proposed or potential systemic CNS dysfunction/aberration are inconsistent with the primary and inviolable phenotypic principles of migraine [19-24].

Migraine is the most common primary headache, and in the last 100 years, migraine and CSD / SD – have been exhaustively and exponentially investigated. Pain is the most prominent component of migraine, while CSD is typically a painless hypestheic physiologic principle that does not induce aversion but rather promotes eating, drinking, and exploratory behavior [14,25]. CSD in cats, additionally, is not associated with the release of calcitonin-gene-related peptide (CGRP) [26]. CGRP has been the most prominent neurotransmitter/neuropeptide over the last 2 decades in migraine research.

The magnesium-CSD-migraine nexus is untenable and is the outcome of a series of carefully nurtured canonical myths over decades [27]. Primary potential / presumed “periodic/episodic” intrinsic brain magnesium ionic (Mg2+) depletion is central to the variation of CNS hyperexcitability, migraine threshold, and occurrence of the migraine attack [28,29]. Intrinsic brain Mg2+ depletion (hypomagnesemia), however, has a significant and fundamentally adaptive function in cardiovascular and cerebrovascular tissues, as well as in many other organ structures with well-guarded supplementation in the Intensive Care Unit (ICU) [30,31]. The brain neurovascular therapeutic effect of exogenous supplementation of magnesium is limited by crossing the Blood-Brain Barrier (BBB) as well as the sustained higher levels in CSF and in the mural level of Mg2+ in brain arteries [32]. Across different tissues, the percentage and entry of Mg2+ are considerably delayed—variably up to hours [33]. Another reason for the limited exchange of Mg2+ with tissues is its binding with water, restricting the bare ion entry through the membrane. Common to all ions, this situation is greater for Mg2+ because the radius of its hydrated moiety combined with water is 400 times larger than its ionic radius [33-35]. As such, it is almost impossible for magnesium to pass through narrow channels in biological membranes, opposite to calcium, because of its ‘hydration cover’ [36,37].

Laws of pathophysiology of migraine – phenotypic principles and evolution of the cause-effect divide

Key phenotypic features of migraine attacks indicate the operation of certain pathophysiologic principles that, in turn, define the role of certain neurologic absolutes. These neurologic absolutes are not otherwise apparent despite the availability of advanced / advancing technology in the laboratory and/or neuroimaging as well as an exponentially increasing data bank. The limitations of the methodology of science of migraine / primary headache have been discussed: there is no physical sign or set of signs or organic aberration to create a robust, generalizable, predictable, or logically-sound pathophysiological blueprint or overview, whereas the sophisticated nosology is purely arbitrary and symptomatic (see above) [38]. On the contrary, and converse to the science of medicine in general, it is the symptoms of migraine, beginning from stress-linked Autonomic Nervous System (ANS) activation / ‘storm’ along with the noradrenergic-vasopressinergic-serotonergic ‘system’ that provides the basis for definitive laws of pathophysiology of the disorder. The pathophysiologic role of vasopressin (AVP) in migraine has evolved gradually from systemic vasoconstriction, anti-nociception, and behavior control to [39] to control of migraine-attack related ocular choroidal circulation, control of rise of (or fall in) Intraocular Pressure (IOP) (in contrast to rising in systemic blood pressure), control of the expansion of anterior ocular segment and corneosceral envelope, and spontaneous onset and offset of migraine headache-generating ophthalmic nerve (V1) nociceptive traffic [18, 30,39,40].

Stress, the post-stress phase, missing a meal, alcohol imbibition (weekend migraine headache attack), or insufficient sleep are the commonest precipitants of migraine but have no link to CSD. Post-stress migraine remains enigmatic and involves an adaptive/protective ‘stress-related’ pre-ictal combined ‘pre-prodromal / pre-premonitory’ phase that prevents the genesis of the migraine attack for a limited duration (several hours to a few days), through a collective or concatenation of anti-migraine physiological forces. While the role of ‘stress / ‘pre-stress’ / ‘occult’ stress in migraine attacks cannot itself be studied, the typical delay in stress-linked headache offers definitive clues to its role in the pathogenesis. Lateralization of headache (unilateral, bilateral, side-fixed, or side-shifting) with spontaneous migraine attacks is the most typical stress-alcohol-sleep-linked feature of migraine that has not yet been rationalized (see below). Additionally, experimentally-induced Nitroglycerine (NTG) triggered migraine is unlikely to generate neurologically consistently non-protean lateralizing pathognomonic Scintillating Scotoma (SS) or typical headache of migraine, with consistent onset, duration, or offset; besides, its link to CSD-BBB disruption is also uncertain [41-47]. NTG, in low doses, lowers IOP in varied circumstances (Mishra, et al. 2014). Sustained relatively high-dose intravenous administration of NTG with subtle oculo-sympathetic/parasympathetic – pupil, and by inference, the ocular choroid [48,49] as well as cardiac migrainous ANS deficiency [50-52], as in experimental use of NTG to precipitate migraine, with changes in systemic blood pressure, cardiac pre-load reduction, left-ventricular stroke volume, and ocular vasodilatation can raise IOP but such a study has never been done. There is no brain/brain vascular lesion induced by NTG that might induce recurrent, reproducible yet protean and persistent lateralizing headache of migraine as well as recurrent or persistent pathognomonic SS.

Migraine typically and selectively involves the ophthalmic trigeminal (V1) rather than all three divisions of the trigeminal nerve, a feature inconsistent with diffuse and/or consistent or repetitive and spontaneous primary propagation of CSD or any other form of CNS modulation / sensory dysmodulation in the trigeminal nerve, but neuro-anatomically consistent with characteristically selective lateralization (unilateral, bilateral, side-shift, or side-fixed) of a headache to the peri-ocular, the fronto-temporal, and the nuchal region, thus formulating the first law of migraine pathophysiology. Osmophobic migraine (involving odours, including scents, foods, and cigarette smoke) and ice-cream headache also clearly reflect a neural circuit exclusively involving V1. No systemic influence (from serotonin to CGRP) can rationalize the lateralizing headache of migraine [53].

A typically protean or unpredictable and delayed onset, duration, offset, and degree/severity of pain and its accompaniments, absenteeism, or presenteeism is a highly characteristic feature of migraine attacks. Experimental (nitroglycerine-induced)/or spontaneous (non-experimental) onset of migraine attacks is not always predictable but commonly indeterminate and with the pre-ictal ‘pre-prodromal/pre-premonitory’ or ‘prodromal/premonitory’ phase largely inaccessible or occult to systematic examination or clinical evaluation. Experimentally, not all rodents or humans exposed to NTG develop migraine or migraine-like features. Similarly, while almost all humans (~80%) suffer psychodynamic stress (stress-is-life, life-is-stress, Hans Selye) [54], only approximately one/fifth of humans suffer migraine attacks. The concatenation or orchestration of cross-disciplinary pathophysiologic systems that variably manage migraine-related pre-stress/stress/post-stress phases constitutes the second law of migraine pathophysiology. A complex multifocal adaptive mechanism involving intrinsic vasopressinergic-noradrenergic-serotonergic activation, that is, in turn, affected by a very large number of clinical circumstances and situations (endogenous or exogenous or both) likely offers a “protective” effect that significantly delays migraine attacks for several hours or days, prevents experimental migraine in some rodents/four-fifths of humans under protracted stress, or controls the severity and duration of migraine attacks [27]. The multiple end-organ (ocular) effects of this complex adaptive-protective system have been detailed and are briefly mentioned below [18,40,55]. Vasopressin (AVP) surges in the early phases of migraine attacks can generate early nausea/vomiting in migraineurs and abort migraine attacks, a classic adaptive/emotional mechanism that constitutes the third law of migraine pathophysiology. Nausea and/or vomiting are accompanied by intense and rapid AVP release with hyponatremia. Virtually instantaneous increases in plasma AVP from 100 to 1000 times commonly occur with transient nausea, stress, or pain unaccompanied by vomiting or changes in Systemic Blood Pressure (SBP). All adaptive functions have a protean and/or variably complex function across organs in the human body acting to preserve or challenge physiologic body function, e.g. fluid retention has renal and extra-renal manifestations in migraine, whereby it can increase cardiac right/left ventricular stroke volume but create renal overload with urea/creatinine elevation and electrolyte disturbances. Not all migraine attacks are accompanied by nausea-vomiting or the abortion of migraine headache attacks by nausea/vomiting, a classically adaptive manifestation. Amitriptyline is a first-line prophylactic drug for the management of both migraine as well as Cyclic Vomiting Syndrome (CVS), with or without migraine, that can cause hyponatremia and AVP excess, commonly linked to low SBP (and rarely high SBP) with anti-stress mechanisms, choroidal arterial vasoconstriction, ocular hypotension, ocular vasoconstriction, and antinociception [56-58]. Oxytocin may play a role in nausea-based disgust or anticipatory nausea in rats [59]. Oxytocin has no known vasoconstrictive influence, which effect is paramount in keeping migraine in remission or prophylaxis.

Photophobia is not necessarily greater on the side of the headache, and CSD or cortical central visual dysfunction is unlikely to have a major or central role in its genesis [27]. Phonophobia (including rustling of newspaper) and other forms of hyper-reactions or hyper-sensitizations are part of a generalized hyper-reactivity that is also a prominent part of a generalized hyper-sensitivity characteristic of migraine. Further experimental evidence, the surge of Randomized Controlled Clinical Trials (RCCT) with neuropeptides (such as calcium-gene related neuropeptide-antagonists and -receptor antagonists), neuroimaging including cerebral perfusion studies, genotyping, and animal experiments) have made the migraine phenotype increasingly complex while keeping intact the cause-effect conundrum (brain vs. eye).

Ocular pathophysiology & fundamentals of migraine

Recently, the primary role of the eye in the pathogenesis of migraine has gained much traction in the Third Millennium, validating the cross-disciplinary integrative KISS principle with both breadth and depth of knowledge of migraine towards a comprehensive and gestaltic synthesis that has defied scientific thinking over 25 centuries. There exist several carefully nurtured myths in migraine pathophysiology that have stymied the creation of such an overarching hypothesis [14,27,60], with the elaborate symptom-based classification system and its variants [38] prominently in the forefront. The study of the fragmented but exponentially expansive data bank of migraine / primary headache and the speculations surrounding its pathophysiology is the best example of the ability of an elaborate classification system in medicine to prevent the emergence of a comprehensive synthesis or fundamental understanding. No systemic belief or canonical myth – including nitric oxide, neuropeptide/neurotransmitter, dietary, platelet, or cardiac involvement including patent foramen ovale, atrial septal defect, Ebstein’s anomaly, coarctation of aorta, or obesity – can rationalize the first law of neurological lateralization of migraine headache [31]. Sparing of the nasal visual field in Scintillating Scotoma (SS) is an equally if not more demanding neurologically lateralizing pathognomonic phenomenon (see below). The first truly robust, predictable, defensible, and progressive hypothesis regarding migraine was published in the early 21st century with a synopsis earlier in 1990 [30,61].

Pathognomonic-to-classical features of migraine are not reported after enucleation of one or both eyes or following sightlessness [62-66]. Even more importantly, the SS is not distributed homonymously and SS does not expand into the nasal visual field, two absolute features that make brain / central visual field origin of the pathognomonic phenomenon impossible. After enucleation of either or both eyes, the typical paracentral horse-shoe-shaped expanding positive SS as well as the typical migraine headache is not reported [63,64]. Sightlessness markedly attenuates migrainous SS as well as photophobia [66]. The Presence of the eye is essential to manifest typical-to-pathognomonic lateralizing clinical features of migraine, constituting the fourth, and perhaps, the most important clinically absolute law of migraine pathophysiology [27].

Mechanical or physical displacement of the SS is the single most important clinical absolute to determine the site of origin of the positive hallucination or visual pathognomonic phenomenon of migraine [67]. The pathognomonic positive SS of migraine is physically displaceable by digital / finger pressure, and, is thus retinal in origin [18]. Clinical study of SS of migraine on these lines constitutes the fifth law of migraine pathophysiology and forms the crux for understanding this otherwise formidably complex disorder [18].

The crani-ocular pathophysiological system involved in migraine is largely protected during stress, with the post-stress phase manifesting as the commonest cause of migraine attacks. There are several components of this important protective feature of migraine, which can be collectively summed as the sixth law of migraine pathophysiology. Much data has been accumulated about the Autonomic Nervous System (ANS) and migraine at the levels of the pupil and the heart, without any synthesis / co-synthesis emerging across these and several other exponentially enlarging evidentiary banks. Succinctly, migraine is an autonomic storm, that begins often much before the prodrome – a phase most aptly termed the ‘pre-prodromal / pre-premonitory’ phase, in other words, the truly occult pre-ictal phase of migraine [18]. Migraine philology and philosophy have completely ignored both the generally accepted and scientifically robust adaptive-protective alarm function of the ANS in the animal kingdom as well as its speculative neurological link to CSD / central brain neuronal dysmodulation / sensory dysfunction. Furthermore, speculative pathogenetic involvement of the “intrinsic CNS sympathomimetic system” was proposed as the basis of the biobehavioral model of migraine, a theory that held overriding sway in the last quarter of the last century [18,27,28,40].

Key issues involving stress-protective function of the eye include control of choroidal ocular circulation (adrenergic and vasopressinergic), Intraocular Pressure (IOP), and generation of algogenic/nociceptive impulses in the ophthalmic nerve from mechanical deformation of the corneoscleral junction [18,40]. Maintenance of relative intraocular hypotension delays the onset of migraine attacks. Vasopressin (AVP) raises systemic blood pressure but lowers IOP, a prominent component of the ocular protective influence of stress on the development of migraine attacks. The rate of change of systemic blood pressure rather than an absolute value determines the migraine threshold [30].

The limited duration of the headache of migraine (4 hours - 72 hours) is in itself a clinically typical-to-pathognomonic feature. The components of the tissues of the eye that provide effective ocular tamponade in the face of relative ocular hypertension generating algogenic/nociceptive impulses during migraine headaches are responsible for terminating the migraine attack spontaneously. These ocular features along with external (digital) self-compression of the eye constitute the seventh law of migraine [40].

A combination of these seven pathophysiologic laws underscores the propensity of pubertal/post-pubertal children/adolescents, and adults (F:M = 3:1) to develop migraine, ophthalmoplegic migraine, menstrual migraine, and migraine in the first trimester of pregnancy as well as the tendency of migraine attacks to decline with later trimesters of pregnancy, ageing, and menopause [18]. The migraine-glaucoma nexus (primary open-angle glaucoma/normal tension glaucoma) is gradually becoming more apparent. The protean nature of migraine – even in the same individual on different occasions – does not allow biologically valid conclusions to emerge from statistics (p - values) related to randomized controlled clinical trials. Variations of deformability of the corneoscleral envelope in response to mechanical IOP pressure fluctuations with protean/variable generation of nociceptive impulses in V1 is linked to the connective-tissue component of migraine, a key feature that will be discussed elsewhere.

These seven laws detail the biophysics and biophysiology of migraine, keeping mathematical/statistical/nosologic/speculative conclusions or guidelines at bay. Neuropeptides/neurotransmitters do not compose the biology of migraine. In the absence of such basic fundamentals, RCCT and the use of a placebo increase theoretical and therapeutic confusion in migraine/primary headaches [27].

“True progress in science requires the ability to grapple with the immeasurable, the daring to view phenomenology differently, to give birth to and maintain a disquieting but penetrating dys-synchrony, to confront and not be overawed by history or consensus, to sustain searing self-criticism, and to carry curiosity and imagination to a defensible and generalizable conclusion. Canonical authoritarianism, empiricism, mythicalism, serendipity, cynicism, and irrational skepticism and mysticism over 25 centuries compel the need to refine, redefine, advance, overturn, and crystallize scientific thinking about oculo-cephalic / cephalic pain to a new dimension” [27].

  • Cortical Spreading Depression (CSD) has well-defined neuronal/neurovascular ‘pre-conditioning’ adaptive/protective features in vertebrates (locusts, Drosophila) and mammals (rodents).
  • The suppressive therapeutic role of CSD in the neuro-critical unit is speculative.
  • CSD is absolutely non-nociceptive, promotes eating/drinking behaviour in rodents, and is not relevant to the prominently painful disorder of migraine or its brain-magnesium linked pathophysiology or purely symptomatic nosology.
  • There is no pharmacotherapeutic difference between migraine with aura (MWA) and migraine without aura (MWoA).
  • CSD in MWA and ‘silent’ CSD in MWoA are purely speculative.
  • Scintillating scotoma is not homonymous, spares nasal-visual field, is digitally/physically displaceable, and moves synchronously with eye movements, all features absolutely against cerebral-visual origin.
  • CSD and several other diverse proposed forms of ‘primary’ involvement of brain cannot rationalize adaptation/protection during pre-stress / stress phase in ~80% of migraine patients; delay in onset of headache following stress/nitroglycerine challenge/alcohol imbibition (several hours to a few days); stress-related migraine; spontaneous but uncertain onset; spontaneous, variable, but limited offset (4 hours - 72 hours); lateralizing headache; and vomiting-induced self-abortion of migraine attacks in a fraction of cases.
  • Lateralizing fronto-temporal-nuchal headache (unilateral/bilateral/side-fixed/side-shifting) does not involve all three divisions of the trigeminal nerve.
  • Migraine selectively / exclusively involves ophthalmic division of the trigeminal nerve (V1).
  • Noradrenergic-vasopressin-serotonergic ‘system’ (extrinsic and/or ocular) offers vasoconstriction, anti-nociception, behaviour control, while the ocular autonomic nervous system, ocular choroidal circulation, Intraocular Pressure (IOP), anterior ocular segment and corneoscleral expansion, generate and decrease nociceptive V1 neural impulses, thereby advancing the migraine-glaucoma nexus.
  • Vasopressin has opposite effects on systemic blood pressure (rise) and IOP (fall).
  • Tamponade effect of eye/s that irregularly (4 h - 72 h) but spontaneously stops migraine headache attacks occurs through a combined tissue and neurotransmitter effect.
  • Release of Calcitonin-Gene-Related Peptide (CGRP) and/or other neuropeptides/neurotransmitters have no definitive role in ‘stress’, ‘autonomic storm’, or genesis/onset/offset of protean unpredictable migraine attacks.
  • The ocular tissue/neurotransmitter basis of occurrence of migraine attacks predominantly in females and with declining frequency of attacks in post-menopausal and in advancing age is outlined (Tables 1,2)
  • Randomized Controlled Clinical Trials (RCCT), the major methodology of medical science over 50 years, heavily rely on cohort randomization and distribution of variables, mathematics/statistics, and the p-value, thereby creating biological confusion. Given the characteristically protean nature of migraine and the use/misuse of placebo, positive RCCT has not been able to offer clear and insightful therapeutic or pathophysiologic guidelines.
  • Table 1: Clinical Modulation of Migraine Attacks. ↑ Arginine Vasopressin (AVP) Bioavailability with Abortion/Remittance of Migraine.
    Nausea / Vomiting Can immediately abort or reduce the clinical impact of migraine attacks commonly in the early phases. Some patients self-induce vomiting by gag reflex to abort migraine attacks, manage/ameliorate alcohol intoxication, or reduce weight in school wrestling, besides established issues in dentistry and bulimia [68]. Alcohol is a depressant that slows the functions of the nervous system, thus diminishing involuntary processes such as breathing, heart rate, and the gag reflex. Even with the blunted gag reflex with the risk of choking, self-induced vomiting, to a degree, is one method of eliminating alcohol from the system. Eating disorders (anorexia nervosa, bulimia nervosa, and binge-eating) are probably linked with migraine and self-induced vomiting as a weight-control strategy [69]. Common to all these disorders is a stress-management strategy that has gone awry or has become overwhelmed.
    https://www.quora.com › Why-dont-I-have-a-gag-reflex.. https://health.cornell.edu/sites/health/files/pdf-library/self-induced-vomiting.pdf
    Sleep Well-defined therapeutic effects on migraine, are generally and increasingly regarded as anecdotal [70-74]. While RCCT cannot yield the undisputed scientific evidence required (see below), there is a bidirectional link between sleep and neuropeptides. Sleep stimulates AVP secretion regardless of electroencephalographic sleep state [75,76]. AVP stimulates sleep by reducing Rapid-Eye Movement [REM] sleep, which regulatory central influence, independent of vasopressor effect, is confirmed in humans [77]. Vasopressin neurons in the suprachiasmatic nucleus significantly control the functional output of the biological clock that is contained within it.
    Tobacco smoking/nicotine There is no definite or mechanistic pathophysiologic link between tobacco smoking and migraine. The bias of the investigator(s) is generally reflected in genetic/statistical large-scale evidence. Smoking cessation is not part of any migraine-preventive strategy (Weinberger and Seng, 2023). Nicotine readily crosses the BBB, and releases AVP, β-endorphin, acetylcholine, norepinephrine, dopamine, serotonin, and adrenocorticotropic hormone; nicotine has definitive antinociceptive action [78,79]. Migraine patients may seek relief by smoking tobacco; others smoke to reduce dysphoria and anxiety. Tobacco-released AVP, in turn, can prevent or protect from migraine. Some migraineurs seek relief by smoking tobacco, which evidence is regarded as anecdotal. Severe migraine headaches can develop after the sudden discontinuation of nicotine gum [80]. Almost 2 decades previously, in a prolonged and meticulous 1 single N-of-1 trial case report in a female allopathic medical doctor with severe life-modulating migraine over 20 years, the theoretical basis and potential clinical application of nicotine gum chewing to prevent migraine was fully elucidated [39]. Imbibition of alcohol, the most common intoxicant that precipitates migraine worldwide, in combination with tobacco smoking, as well as nasal nerve-ending irritation by smoke-emission of tobacco-smoking migraineurs as well as non-smoking migraineurs hopelessly complicates the scientific medical issue. Again, RCCT cannot provide clarity of scientific thinking in this field by undisputed evidence due to additional potential carcinogenicity, particularly of lung, protean nature of migraine, and variations in tobacco/alcohol consumption, while a robust synthesis is invaluable (see below).
    Physical Exercise Regular exercise can alter migraine-triggering threshold [81-83]. The frequency, intensity, and modality of physical exercise along with musculoskeletal and balance dysfunctions that can generate and overcome the autonomic ‘storm’ to prevent or trigger migraine attacks, respectively, (tachycardia, stroke volume, cardiac output, physical conditioning, physical fatigue, ocular autonomic function, ocular blood flow, ocular choroidal function, and IOP) cannot be predicted with precision and can change with time, occasion, training and competition. Individualized osmotic and non-osmotic regulation of AVP may change in competitive events to generate exercise-associated hyponatremia [84]. In such clinical situations, challenges may vary from migraine to death.
    Pregnancy Migraine attacks that begin during pregnancy usually subside within 3 months and a great majority of female migraine patients progressively improve during pregnancy. Fluid overload and relative hyponatremia of pregnancy might restore or enhance AVP efficacy. Elevated estrogen and endogenous opioid levels raise the pain threshold but do not fluctuate. Hypertension raises (13-fold) the risk of worsening of migraine during pregnancy Puerperium (first month) increases the risk of migraine attacks. While breastfeeding lowers the risk of migraine, hypercoagulability is common to both [85].
    Estrogen No definitive vasoconstrictive or anti-nociceptive effect. Anti-stress effects cannot be excluded.
    Oxytocin The antinociceptive effect is probably mediated by AVP; the independent role of oxytocin in pain control is less likely [86,87]. No vasoconstrictive effect that is central to an anti-migraine prophylactic effect. Anti-stress effects cannot be excluded. Elevates IOP when applied to the supraoptic nucleus [88].
    Menopause / Ageing Water retention due to augmented osmotic AVP secretion and renal sodium reabsorption [89,90].
    Caffeine withdrawal Weakly lowers IOP, except in genetically predisposed individuals [91]. At high concentrations, caffeine can increase AVP release [92].
    Table 2: Clinical Modulation of Migraine Attacks.  Arginine Vasopressin (AVP) Bioavailability with Precipitation / Triggering of Migraine.
    Alcohol
    (Morning hangover headache)
    Exhaustion of ANS through vasodilatory effect in the external carotid artery decreases peripheral resistance and increases cutaneous blood flow, and tachycardia during exercise / submaximal exercise; increasing initial rate of urine production with no difference in total cumulative urine output [93]. Combined stimulation (osmotic and non-osmotic) and suppression of AVP.
    Awakening from sleep with startle / early morning migraine Exhaustion of ANS ‘storm’ with startle-induced rise of intraocular pressure.
    Psychologic Trauma Exhaustion of ANS ‘storm’ with rise of intraocular pressure.
    Physical trauma Exhaustion of ANS ‘storm’ with rise of intraocular pressure.
    Weight-lifting headache Elevates IOP by Valsalva manoeuver [94].
    Constipation / Cough Elevates IOP by Valsalva manoeuver [95].
    Systemic blood pressure (SBP) Rate of change of SBP [96-98].
    1. Mathew AA, Panonnummal R. Cortical spreading depression: culprits and mechanisms. Exp Brain Res. 2022 Mar;240(3):733-749. doi: 10.1007/s00221-022-06307-9. Epub 2022 Jan 22. PMID: 35064796.
    2. Takizawa T, Ayata C, Chen SP. Therapeutic implications of cortical spreading depression models in migraine. Prog Brain Res. 2020;255:29-67. doi: 10.1016/bs.pbr.2020.05.009. Epub 2020 Jun 27. PMID: 33008510.                
    3. Soldozy S, Sharifi KA, Desai B, Giraldo D, Yeghyayan M, Liu L, Norat P, Sokolowski JD, Yağmurlu K, Park MS, Tvrdik P, Kalani MYS. Cortical Spreading Depression in the Setting of Traumatic Brain Injury. World Neurosurg. 2020 Feb;134:50-57. doi: 10.1016/j.wneu.2019.10.048. Epub 2019 Oct 23. PMID: 31655239.
    4. Close LN, Eftekhari S, Wang M, Charles AC, Russo AF. Cortical spreading depression as a site of origin for migraine: Role of CGRP. Cephalalgia. 2019 Mar;39(3):428-434. doi: 10.1177/0333102418774299. Epub 2018 Apr 25. PMID: 29695168; PMCID: PMC7007998.
    5. Ashina M, Hansen JM, Do TP, Melo-Carrillo A, Burstein R, Moskowitz MA. Migraine and the trigeminovascular system-40 years and counting. Lancet Neurol. 2019 Aug;18(8):795-804. doi: 10.1016/S1474-4422(19)30185-1. Epub 2019 May 31. PMID: 31160203; PMCID: PMC7164539.
    6. Hartings JA, Shuttleworth CW, Kirov SA, Ayata C, Hinzman JM, Foreman B, Andrew RD, Boutelle MG, Brennan KC, Carlson AP, Dahlem MA, Drenckhahn C, Dohmen C, Fabricius M, Farkas E, Feuerstein D, Graf R, Helbok R, Lauritzen M, Major S, Oliveira-Ferreira AI, Richter F, Rosenthal ES, Sakowitz OW, Sánchez-Porras R, Santos E, Schöll M, Strong AJ, Urbach A, Westover MB, Winkler MK, Witte OW, Woitzik J, Dreier JP. The continuum of spreading depolarizations in acute cortical lesion development: Examining Leão's legacy. J Cereb Blood Flow Metab. 2017 May;37(5):1571-1594. doi: 10.1177/0271678X16654495. Epub 2016 Jan 1. PMID: 27328690; PMCID: PMC5435288.
    7. Robertson RM, MacMillan HA, Andersen MK. A cold and quiet brain: mechanisms of insect CNS arrest at low temperatures. Curr Opin Insect Sci. 2023 Aug;58:101055. doi: 10.1016/j.cois.2023.101055. Epub 2023 May 16. PMID: 37201631.
    8. Andersen MK, Jensen NJS, Robertson RM, Overgaard J. Central nervous system shutdown underlies acute cold tolerance in tropical and temperate Drosophila species. J Exp Biol. 2018 Jun 15;221(Pt 12):jeb179598. doi: 10.1242/jeb.179598. PMID: 29739833.                                                                                                                                
    9. Robertson RM, Spong KE, Srithiphaphirom P. Chill coma in the locust, Locusta migratoria, is initiated by spreading depolarization in the central nervous system. Sci Rep. 2017 Aug 31;7(1):10297. doi: 10.1038/s41598-017-10586-6. PMID: 28860653; PMCID: PMC5579280.
    10. Rodgers CI, Armstrong GA, Robertson RM. Coma in response to environmental stress in the locust: a model for cortical spreading depression. J Insect Physiol. 2010 Aug;56(8):980-90. doi: 10.1016/j.jinsphys.2010.03.030. Epub 2010 Apr 7. PMID: 20361971.
    11. Armstrong GA, Rodgers CI, Money TG, Robertson RM. Suppression of spreading depression-like events in locusts by inhibition of the NO/cGMP/PKG pathway. J Neurosci. 2009 Jun 24;29(25):8225-35. doi: 10.1523/JNEUROSCI.1652-09.2009. PMID: 19553462; PMCID: PMC6666049.
    12. Rodgers CI, Armstrong GA, Shoemaker KL, LaBrie JD, Moyes CD, Robertson RM. Stress preconditioning of spreading depression in the locust CNS. PLoS One. 2007 Dec 26;2(12):e1366. doi: 10.1371/journal.pone.0001366. PMID: 18159249; PMCID: PMC2137934.
    13. Drongitis D, Rainone S, Piscopo M, Viggiano E, Viggiano A, De Luca B, Fucci L, Donizetti A. Epigenetics and cortical spreading depression: changes of DNA methylation level at retrotransposon sequences. Mol Biol Rep. 2016 Aug;43(8):755-60. doi: 10.1007/s11033-016-4000-4. Epub 2016 May 12. PMID: 27169424.
    14. Gupta VK. CSD, BBB and MMP-9 elevations: animal experiments versus clinical phenomena in migraine. Expert Rev Neurother. 2009 Nov;9(11):1595-614. doi: 10.1586/ern.09.103. PMID: 19903020.
    15. Shen PJ, Gundlach AL. Prolonged induction of neuronal NOS expression and activity following cortical spreading depression (SD): implications for SD- and NO-mediated neuroprotection. Exp Neurol. 1999 Dec;160(2):317-32. doi: 10.1006/exnr.1999.7218. PMID: 10619550.
    16. Yanamoto H, Xue JH, Miyamoto S, Nagata I, Nakano Y, Murao K, Kikuchi H. Spreading depression induces long-lasting brain protection against infarcted lesion development via BDNF gene-dependent mechanism. Brain Res. 2004 Sep 3;1019(1-2):178-88. doi: 10.1016/j.brainres.2004.05.105. PMID: 15306252.
    17. Kobayashi S, Harris VA, Welsh FA. Spreading depression induces tolerance of cortical neurons to ischemia in rat brain. J Cereb Blood Flow Metab. 1995 Sep;15(5):721-7. doi: 10.1038/jcbfm.1995.92. PMID: 7673367.
    18. Gupta VK. Bimatoprost ophthalmic solution (BOS) 0.3 mg w/v for 1 open trial of long-term preventive therapy of migraine in 3 patients with pathophysiologic shift from brain to eye. Journal of Neuroscience and Neurological Disorders. 2023; 7:2; 134-154.
    19. Gawde P, Shah H, Patel H, Bharathi KS, Patel N, Sethi Y, Kaka N. Revisiting Migraine: The Evolving Pathophysiology and the Expanding Management Armamentarium. Cureus. 2023 Feb 2;15(2):e34553. doi: 10.7759/cureus.34553. PMID: 36879707; PMCID: PMC9985459.
    20. Wang M, Tutt JO, Dorricott NO, Parker KL, Russo AF, Sowers LP. Involvement of the cerebellum in migraine. Front Syst Neurosci. 2022 Oct 13;16:984406. doi: 10.3389/fnsys.2022.984406. PMID: 36313527; PMCID: PMC9608746.
    21. Lin YK, Tsai CL, Lin GY, Chou CH, Yang FC. Pathophysiology of Chronic Migraine: Insights from Recent Neuroimaging Research. Curr Pain Headache Rep. 2022 Nov;26(11):843-854. doi: 10.1007/s11916-022-01087-x. Epub 2022 Oct 7. PMID: 36207509.
    22. Bolay H. Thalamocortical network interruption: A fresh view for migraine symptoms. Turk J Med Sci. 2020 Nov 3;50(SI-2):1651-1654. doi: 10.3906/sag-2005-21. PMID: 32421284; PMCID: PMC7672341.
    23. Kros L, Angueyra Aristizábal CA, Khodakhah K. Cerebellar involvement in migraine. Cephalalgia. 2018 Oct;38(11):1782-1791. doi: 10.1177/0333102417752120. Epub 2018 Jan 22. PMID: 29357683.
    24. Goadsby PJ, Holland PR, Martins-Oliveira M, Hoffmann J, Schankin C, Akerman S. Pathophysiology of Migraine: A Disorder of Sensory Processing. Physiol Rev. 2017 Apr;97(2):553-622. doi: 10.1152/physrev.00034.2015. PMID: 28179394; PMCID: PMC5539409.
    25. Pietrobon D, Moskowitz MA. Chaos and commotion in the wake of cortical spreading depression and spreading depolarizations. Nat Rev Neurosci. 2014 Jun;15(6):379-93. doi: 10.1038/nrn3770. PMID: 24857965.
    26. Piper RD, Edvinsson L, Ekman R, Lambert GA. Cortical spreading depression does not result in the release of calcitonin gene-related peptide into the external jugular vein of the cat: relevance to human migraine. Cephalalgia. 1993 Jun;13(3):180-3; discussion 149. doi: 10.1046/j.1468-2982.1993.1303180.x. PMID: 8395344.
    27. Gupta VK. Pathophysiology of migraine: an increasingly complex narrative to 2020. Future Neurology 2020; 14: 2. Published Online: 24 May 2019. https://doi.org/10.2217/fnl-2019-0003
    28. Welch KM. Migraine. A biobehavioral disorder. Arch Neurol. 1987 Mar;44(3):323-7. doi: 10.1001/archneur.1987.00520150063024. PMID: 3827684.     
    29. Welch KM, D'Andrea G, Tepley N, Barkley G, Ramadan NM. The concept of migraine as a state of central neuronal hyperexcitability. Neurol Clin. 1990 Nov;8(4):817-28. PMID: 1979655.
    30. Gupta VK. Migrainous scintillating scotoma and headache is ocular in origin: A new hypothesis. Med Hypotheses. 2006;66(3):454-60. doi: 10.1016/j.mehy.2005.11.010. Epub 2005 Dec 13. PMID: 16356654.
    31. Gupta VK. Does magnesium supplementation have any role in acute myocardial infarction? No. Cardiovasc Drugs Ther. 1996 Jul;10(3):303-5. doi: 10.1007/BF02627952. PMID: 8877071.
    32. Altura BT, Altura BM. The role of magnesium in etiology of strokes and cerebrovasospasm. Magnesium. 1982; 1: 277-291.
    33. Mathew AA, Panonnummal R. 'Magnesium'-the master cation-as a drug-possibilities and evidences. Biometals. 2021 Oct;34(5):955-986. doi: 10.1007/s10534-021-00328-7. Epub 2021 Jul 2. PMID: 34213669; PMCID: PMC8249833.
    34. Dominguez LJ, Veronese N, Barbagallo M. Magnesium and hypertension in old age. Nutrients. 2021; 13(1): 139. Doi: 10.3390/nu13010139
    35. Maguire ME, Cowan JA. Magnesium chemistry and biochemistry. Biometals. 2002 Sep;15(3):203-10. doi: 10.1023/a:1016058229972. PMID: 12206387.
    36. Gupta VK. Eclampsia in the 21st Century: Paradigm shift from empirical therapy with magnesium sulfate. Basic science synthesis vs. current WHO-recommended pharmacotherapeutic practice. J. Brain and Neurological Disorders. 2023; 6(2). DOI:10.31579/2692 9422/045.
    37. Jahnen-Dechent W, Ketteler M. Magnesium basics. Clin Kidney J. 2012 Feb;5(Suppl 1):i3-i14. doi: 10.1093/ndtplus/sfr163. PMID: 26069819; PMCID: PMC4455825.
    38. Schytz HW, Amin FM, Jensen RH, Carlsen L, Maarbjerg S, Lund N, Aegidius K, Thomsen LL, Bach FW, Beier D, Johansen H, Hansen JM, Kasch H, Munksgaard SB, Poulsen L, Sørensen PS, Schmidt-Hansen PT, Cvetkovic VV, Ashina M, Bendtsen L. Reference programme: diagnosis and treatment of headache disorders and facial pain. Danish Headache Society, 3rd edition, 2020. J Headache Pain. 2021 Apr 8;22(1):22. doi: 10.1186/s10194-021-01228-4. PMID: 33832438; PMCID: PMC8034101.
    39. Gupta VK. Antimigraine action of nicotine: theoretical basis and potential clinical application. Eur J Emerg Med. 2007; 14(4): 243-244.
    40. Gupta VK. Once-weekly nocturnal ocular choroidal vascular decongestion by topical long-acting timolol solution applied to headache-ipsilateral eye to prevent weekend-migraine attacks: Smartphone-app assisted case report of 2 patients for 3-years and a brief mechanistic review. J. Neuroscience and Neurological Surgery. 2021; 9(1). DOI:10.31579/2578-8868/179.
    41. Karsan N, Bose RP, O'Daly O, Zelaya F, Goadsby PJ. Regional cerebral perfusion during the premonitory phase of triggered migraine: A double-blind randomized placebo-controlled functional imaging study using pseudo-continuous arterial spin labeling. Headache. 2023 Jun;63(6):771-787. doi: 10.1111/head.14538. Erratum in: Headache. 2023 Sep;63(8):1199. PMID: 37337681.
    42. Karsan N, Goadsby PJ. Neuroimaging in the pre-ictal or premonitory phase of migraine: a narrative review. J Headache Pain. 2023 Aug 11;24(1):106. doi: 10.1186/s10194-023-01617-x. PMID: 37563570; PMCID: PMC10416375.
    43. Alpay B, Cimen B, Akaydin E, Bolay H, Sara Y. Levcromakalim provokes an acute rapid-onset migraine-like phenotype without inducing cortical spreading depolarization. J Headache Pain. 2023 Jul 24;24(1):93. doi: 10.1186/s10194-023-01627-9. PMID: 37488480; PMCID: PMC10367339.
    44. Wu S, Ren X, Zhu C, Wang W, Zhang K, Li Z, Liu X, Wang Y. A c-Fos activation map in nitroglycerin/levcromakalim-induced models of migraine. J Headache Pain. 2022 Sep 30;23(1):128. doi: 10.1186/s10194-022-01496-8. PMID: 36180824; PMCID: PMC9524028.
    45. He W, Long T, Pan Q, Zhang S, Zhang Y, Zhang D, Qin G, Chen L, Zhou J. Microglial NLRP3 inflammasome activation mediates IL-1β release and contributes to central sensitization in a recurrent nitroglycerin-induced migraine model. J Neuroinflammation. 2019 Apr 10;16(1):78. doi: 10.1186/s12974-019-1459-7. PMID: 30971286; PMCID: PMC6456991.
    46. Schankin CJ, Maniyar FH, Seo Y, Kori S, Eller M, Chou DE, Blecha J, Murphy ST, Hawkins RA, Sprenger T, VanBrocklin HF, Goadsby PJ. Ictal lack of binding to brain parenchyma suggests integrity of the blood–brain barrier for 11 C-dihydroergotamine during glyceryl trinitrate-induced migraine. Brain. 2016l; 139(7): 1994–2001. Doi: 10.1093/brain/aww096. PMCID: PMC4939703. PMID: 27234268.
    47. Dreier JP. Is the blood-brain barrier differentially affected by different variants of migraine? Brain. 2016 Jul;139(Pt 7):1872-4. doi: 10.1093/brain/aww112. PMID: 27343220.
    48. Yildiz MB, Yildiz E, Balci S, Hasirci Bayir BR, Çetinkaya Y. Effect of migraine attack on pupil size, accommodation and ocular aberrations. Eur J Ophthalmol. 2021 Nov;31(6):3450-3455. doi: 10.1177/1120672120975334. Epub 2020 Nov 27. PMID: 33246366.
    49. Eren OE, Ruscheweyh R, Schankin C, Schöberl F, Straube A. The cold pressor test in interictal migraine patients - different parasympathetic pupillary response indicates dysbalance of the cranial autonomic nervous system. BMC Neurol. 2018 Apr 16;18(1):41. doi: 10.1186/s12883-018-1043-2. PMID: 29661162; PMCID: PMC5901875.
    50. Blitshteyn S. Dysautonomia, hypermobility spectrum disorders and mast cell activation syndrome as migraine comorbidities. Curr Neurol Neurosci Rep. 2023 Nov;23(11):769-776. Doi: 10.1007/s11910-023-01307-w. Epub 2023 Oct 17.
    51. Tiwari A, Maurya PK, Qavi A, Kulshreshtha D, Thacker AK, Singh AK. Cranial Autonomic Symptoms in Migraine: An Observational Study. Ann Indian Acad Neurol. 2022 Jul-Aug;25(4):654-659. doi: 10.4103/aian.aian_948_21. Epub 2022 May 12. PMID: 36211151; PMCID: PMC9540922.
    52. Yoshida S, Tanaka H, Mizutani M, Nakao R, Okamoto N, Kajiura M, Kanbara Y, Tamai H. Autonomic nervous system function in adolescent migraineurs. Pediatr Int. 2017 Sep;59(9):991-995. doi: 10.1111/ped.13342. PMID: 28612516.
    53. Gupta VK. Nitric oxide and migraine: Another systemic influence postulated to explain a lateralizing disorder. Eur J Neurol. 1996; 3(2):172-173. 10.1111/j.1468-1331.1996.tb00215.x.
    54. Chrousos GP, Gold PW. Introduction. In: Eds: Chrousos GP, McCarty R, Pacák K, Cizza G, Sternberg E, Gold PW, Kvetňanský R. Stress. Basic Mechanisms and Clinical Implications. ANN NY ACAD SCI. 1995; 771:xv-xviii.
    55. Gupta VK. Self-ocular compression maneuver immediately relieves migraine headache attacks: Case report of managing 100 attacks over 9 years and a mechanistic review. Acta Scientific Neurology. 2021a; 4.8: 72-77.
    56. Frazier R, Li BUK, Venkatesan T. Diagnosis and Management of Cyclic Vomiting Syndrome: A Critical Review. Am J Gastroenterol. 2023 Jul 1;118(7):1157-1167. doi: 10.14309/ajg.0000000000002216. Epub 2023 Feb 15. PMID: 36791365.                                                                                
    57. Pinkhasov A, Xiong G, Bourgeois JA, Heinrich TW, Huang H, Coriolan S, Annamalai A, Mangal JP, Frankel S, Lang M, Raj YP, Dandois M, Barth K, Stewart AL, Rado J, Pesek J, Sanders A, Spearman-McCarthy EV, Gagliardi J, Fiedorowicz JG. Management of SIADH-related hyponatremia due to psychotropic medications - An expert consensus from the Association of Medicine and Psychiatry. J Psychosom Res. 2021 Dec;151:110654. doi: 10.1016/j.jpsychores.2021.110654. Epub 2021 Oct 28. PMID: 34739943; PMCID: PMC10911096.
    58. Venkatesan T, Levinthal DJ, Tarbell SE, Jaradeh SS, Hasler WL, Issenman RM, Adams KA, Sarosiek I, Stave CD, Sharaf RN, Sultan S, Li BUK. Guidelines on management of cyclic vomiting syndrome in adults by the American Neurogastroenterology and Motility Society and the Cyclic Vomiting Syndrome Association. Neurogastroenterol Motil. 2019 Jun;31 Suppl 2(Suppl 2):e13604. doi: 10.1111/nmo.13604. PMID: 31241819; PMCID: PMC6899751.
    59. Boulet NP, Cloutier CJ, Ossenkopp KP, Kavaliers M. Oxytocin, social factors, and the expression of conditioned disgust (anticipatory nausea) in male rats. Behav Pharmacol. 2016 Dec;27(8):718-725. doi: 10.1097/FBP.0000000000000271. PMID: 27740965.
    60. Gupta V. Constipation-related migraine is linked to the effect pf the Valsalva maneuver on the eye: A case report and a mechanistic review. WebmedCentral NEUROLOGY 2010;1(12):WMC001433.
    61. Gupta VK. A hypothesis for the migraine headache syndrome. In: SPIRIT OF ENTERPRISE. THE 1990 ROLEX AWARDS. Section 1 - Applied Sciences and Invention. Buri International, Bern, Switzerland. ISBN 3 7169 2103 3. 1990; 155.
    62. Peatfield RC, Rose FC. Migrainous visual symptoms in a woman without eyes. Arch Neurol. 1981 Jul;38(7):466. doi: 10.1001/archneur.1981.00510070100024. PMID: 7247775.
    63. Nicolodi M, Frezzotti R, Diadori A, Nuti A, Sicuteri F. Phantom eye: features and prevalence. The predisposing role of headache. Cephalalgia. 1997 Jun;17(4):501-4. doi: 10.1046/j.1468-2982.1997.1704501.x. PMID: 9209770.
    64. Sörös P, Vo O, Husstedt IW, Evers S, Gerding H. Phantom eye syndrome: Its prevalence, phenomenology, and putative mechanisms. Neurology. 2003 May 13;60(9):1542-3. doi: 10.1212/01.wnl.0000059547.68899.f5. PMID: 12743251.
    65. Noseda R, Kainz V, Jakubowski M, Gooley JJ, Saper CB, Digre K, Burstein R. A neural mechanism for exacerbation of headache by light. Nat Neurosci. 2010 Feb;13(2):239-45. doi: 10.1038/nn.2475. Epub 2010 Jan 10. PMID: 20062053; PMCID: PMC2818758.
    66. Silva GC, Góes CP, Vincent MB. Aura-like features and photophobia in sightless migraine patients. Arq Neuropsiquiatr. 2014 Dec;72(12):949-53. doi: 10.1590/0004-282X20140200. PMID: 25517643.
    67. Hupp SL, Kline LB, Corbett JJ. Visual disturbances of migraine. Surv Ophthalmol. 1989 Jan-Feb;33(4):221-36. doi: 10.1016/0039-6257(82)90149-7. PMID: 2652358.
    68. Blackmore NPI, Gleaves DH. Self-induced vomiting after drinking alcohol. Int J Ment Health Addiction. 2013; 11: 453–457. https://doi.org/10.1007/s11469-013-9430-9
    69. Łangowska-Grodzka B, Grodzka O, Czarnecki D, Domitrz I. Is there a correlation between migraine and eating disorders? A systematic literature review. Neurol Neurochir Pol. 2023;57(6):457-464. doi: 10.5603/pjnns.97307. Epub 2023 Dec 1. PMID: 38037683.
    70. Tiseo C, Vacca A, Felbush A, Filimonova T, Gai A, Glazyrina T, Hubalek IA, Marchenko Y, Overeem LH, Piroso S, Tkachev A, Martelletti P, Sacco S; European Headache Federation School of Advanced Studies (EHF-SAS). Migraine and sleep disorders: a systematic review. J Headache Pain. 2020 Oct 27;21(1):126. doi: 10.1186/s10194-020-01192-5. PMID: 33109076; PMCID: PMC7590682.
    71. Vgontzas A, Pavlović JM. Sleep Disorders and Migraine: Review of Literature and Potential Pathophysiology Mechanisms. Headache. 2018 Jul;58(7):1030-1039. doi: 10.1111/head.13358. Epub 2018 Aug 8. PMID: 30091160; PMCID: PMC6527324.
    72. Kelman L, Rains JC. Headache and sleep: examination of sleep patterns and complaints in a large clinical sample of migraineurs. Headache. 2005 Jul-Aug;45(7):904-10. doi: 10.1111/j.1526-4610.2005.05159.x. PMID: 15985108.
    73. Sahota PK, Dexter JD. Sleep and headache syndromes: a clinical review. Headache. 1990 Jan;30(2):80-4. doi: 10.1111/j.1526-4610.1990.hed3002080.x. PMID: 2406223.
    74. Blau JN. Resolution of migraine attacks: sleep and the recovery phase. J Neurol Neurosurg Psychiatry. 1982 Mar;45(3):223-6. doi: 10.1136/jnnp.45.3.223. PMID: 7086442; PMCID: PMC491341.
    75. Nadal M. Secretory rhythm of vasopressin in healthy subjects with inversed sleep--wake cycle: evidence for the existence of an intrinsic regulation. Eur J Endocrinol. 1996 Feb;134(2):174-6. doi: 10.1530/eje.0.1340174. PMID: 8630515.
    76. Valiquette G. The neurohypophysis. Neurol Clin. 1987 May;5(2):291-331, vi-vii. PMID: 11681404.
    77. Born J, Kellner C, Uthgenannt D, Kern W, Fehm HL. Vasopressin regulates human sleep by reducing rapid-eye-movement sleep. Am J Physiol. 1992 Mar;262(3 Pt 1):E295-300. doi: 10.1152/ajpendo.1992.262.3.E295. PMID: 1550223.
    78. Sasone L, Milani F, Fabrizi R, Belli M, Cristina M, Zagà V. Nicotine: From discovery to biological effects. Int J Mol Sci. 2023;24(19):14570. Doi: 10.3390/ijms241914570.
    79. Custodio L, Malone S, Bardo MT, Turner JR. Nicotine and opioid co-dependence: Findings from bench research to clinical trials. Neurosci Biobehav Rev. 2022 Mar;134:104507. doi: 10.1016/j.neubiorev.2021.12.030. Epub 2021 Dec 27. PMID: 34968525.
    80. Stalnikowicz R. Nicotine gum withdrawal and migraine headaches. Eur J Emerg Med. 2006 Aug;13(4):247-8. doi: 10.1097/01.mej.0000209066.13697.15. PMID: 16816594.
    81. Varangot-Reille C, Suso-Martí L, Romero-Palau M, Suárez-Pastor P, Cuenca-Martínez F. Effects of Different Therapeutic Exercise Modalities on Migraine or Tension-Type Headache: A Systematic Review and Meta-Analysis with a Replicability Analysis. J Pain. 2022 Jul;23(7):1099-1122. doi: 10.1016/j.jpain.2021.12.003. Epub 2021 Dec 18. PMID: 34929374.
    82. Amin FM, Aristeidou S, Baraldi C, Czapinska-Ciepiela EK, Ariadni DD, Di Lenola D, Fenech C, Kampouris K, Karagiorgis G, Braschinsky M, Linde M; European Headache Federation School of Advanced Studies (EHF-SAS). The association between migraine and physical exercise. J Headache Pain. 2018 Sep 10;19(1):83. doi: 10.1186/s10194-018-0902-y. PMID: 30203180; PMCID: PMC6134860.
    83. Lippi G, Mattiuzzi C, Sanchis-Gomar F. Physical exercise and migraine: for or against? Ann Transl Med. 2018 May;6(10):181. doi: 10.21037/atm.2018.04.15. PMID: 29951503; PMCID: PMC5994516.
    84. Hew-Butler T. Arginine vasopressin, fluid balance and exercise: is exercise-associated hyponatraemia a disorder of arginine vasopressin secretion? Sports Med. 2010 Jun 1;40(6):459-79. doi: 10.2165/11532070-000000000-00000. PMID: 20524712.
    85. Allais G, Chiarle G, Sinigaglia S, Mana O, Benedetto C. Migraine during pregnancy and in the puerperium. Neurol Sci. 2019 May;40(Suppl 1):81-91. doi: 10.1007/s10072-019-03792-9. PMID: 30880362.
    86. Szewczyk AK, Ulutas S, Aktürk T, Al-Hassany L, Börner C, Cernigliaro F, Kodounis M, Lo Cascio S, Mikolajek D, Onan D, Ragaglini C, Ratti S, Rivera-Mancilla E, Tsanoula S, Villino R, Messlinger K, Maassen Van Den Brink A, de Vries T; European Headache Federation School of Advanced Studies (EHF-SAS). Prolactin and oxytocin: potential targets for migraine treatment. J Headache Pain. 2023 Mar 27;24(1):31. doi: 10.1186/s10194-023-01557-6. PMID: 36967387; PMCID: PMC10041814.
    87. Stachenfeld NS, DiPietro L, Palter SF, Nadel ER. Estrogen influences osmotic secretion of AVP and body water balance in postmenopausal women. Am J Physiol. 1998 Jan;274(1):R187-95. doi: 10.1152/ajpregu.1998.274.1.R187. PMID: 9458917.
    88. Zheng H, Lim JY, Kim Y, Jung ST, Hwang SW. The role of oxytocin, vasopressin, and their receptors at nociceptors in peripheral pain modulation. Front Neuroendocrinol. 2021 Oct;63:100942. doi: 10.1016/j.yfrne.2021.100942. Epub 2021 Aug 23. PMID: 34437871.
    89. Yoshizawa T. New experimental model system to study central regulation of intraocular pressure. Jpn J Ophthalmol. 1993;37(1):9-15. PMID: 8320871.
    90. Johnson AG, Crawford GA, Kelly D, Nguyen TV, Gyory AZ. Arginine vasopressin and osmolality in the elderly. J Am Geriatr Soc. 1994 Apr;42(4):399-404. doi: 10.1111/j.1532-5415.1994.tb07488.x. PMID: 8144825.
    91. Kim J, Aschard H, Kang JH, Lentjes MAH, Do R, Wiggs JL, Khawaja AP, Pasquale LR; Modifiable Risk Factors for Glaucoma Collaboration. Intraocular Pressure, Glaucoma, and Dietary Caffeine Consumption: A Gene-Diet Interaction Study from the UK Biobank. Ophthalmology. 2021 Jun;128(6):866-876. doi: 10.1016/j.ophtha.2020.12.009. Epub 2020 Dec 14. PMID: 33333105; PMCID: PMC8154631.
    92. Nicholson SA. Stimulatory effect of caffeine on the hypothalamo-pituitary-adrenocortical axis in the rat. J Endocrinol. 1989 Aug;122(2):535-43. doi: 10.1677/joe.0.1220535. PMID: 2549162.
    93. Marley A, Bakali M, Simpson C. Effect of a moderate alcohol dose on physiological responses during rest and prolonged cycling. Alcohol Alcohol. 2024 Jan 17;59(2):agad079. doi: 10.1093/alcalc/agad079. PMID: 37981293; PMCID: PMC10794168.
    94. Vera J, Jiménez R, Redondo B, Torrejón A, Koulieris GA, De Moraes CG, García-Ramos A. Investigating the Immediate and Cumulative Effects of Isometric Squat Exercise for Different Weight Loads on Intraocular Pressure: A Pilot Study. Sports Health. 2019 May/Jun;11(3):247-253. doi: 10.1177/1941738119834985. Epub 2019 Apr 15. PMID: 30986115; PMCID: PMC6537322.
    95. Gupta VK. Systemic hypertension, headache, and ocular hemodynamics: a new hypothesis. MedGenMed. 2006 Sep 12;8(3):63. PMID: 17406187; PMCID: PMC1781314.
    96. Gupta VK. Patent foramen ovale closure and migraine: science and sensibility. Expert Rev Neurother. 2010 Sep;10(9):1409-22. doi: 10.1586/ern.10.125. PMID: 20819012.
    97. Akerman S, Romero-Reyes M, Karsan N, Bose P, Hoffmann JR, Holland PR, Goadsby PJ. Therapeutic targeting of nitroglycerin-mediated trigeminovascular neuronal hypersensitivity predicts clinical outcomes of migraine abortives. Pain. 2021 May 1;162(5):1567-1577. doi: 10.1097/j.pain.0000000000002142. PMID: 33181579.
    98. Gupta VK. Magnesium therapy for migraine: do we need more trials or more reflection? Headache. 2004 May;44(5):445-6. doi: 10.1111/j.1526-4610.2004.04098_2.x. PMID: 15147256.