Mast cells and neuroinflammation in pathogenesis of neurologic and psychiatric diseases

Cover Page


Cite item

Full Text

Open Access Open Access
Restricted Access Access granted
Restricted Access Subscription or Fee Access

Abstract

The review summarizes current data on the role of neuroinflammation and mast cells in the pathogenesis of nervous and mental diseases, such as multiple sclerosis, Alzheimer’s disease, Parkinson’s disease, amyotrophic lateral sclerosis, depression, autism, migraine, schizophrenia and some others. The contribution of neuroinflammation to the pathogenesis of many of these diseases has been demonstrated. The involvement of mast cells in the development of the neuroinflammatory process has with varying degrees of evidence been shown for multiple sclerosis, amyotrophic lateral sclerosis, Alzheimer’s disease and migraine. There is still no convincing evidence that mast cells contribute to neuroinflammation in Parkinson’s disease, depression, schizophrenia and autism spectrum disorder, although it is possible that they play a role in the pathogenesis of these diseases. Data on the causal role of neuroinflammation and mast cells in the development of neuropsychiatric diseases may become the basis for the development of new approaches to their pharmacological treatment. The review provides data on the first clinical trials of anti-inflammatory and mast cell activity-modulating drugs for the treatment of migraine, Alzheimer’s disease, multiple sclerosis and amyotrophic lateral sclerosis.

Full Text

Abbreviations

CNS, central nervous system; IL, interleukins.

In the last two decades, massive evidence has accumulated, which indicated an important, if not decisive, role of inflammatory processes in the pathogenesis of many nervous and mental diseases. In this regard, interest in the cells of the first line of defense against pathogens ­acting in the nervous system, which regulate the inflammatory process in the brain, has sharply increased. Microglia is the focus, while mast cells, or mastocytes, another group of immunocompetent resident brain cells, are in the background. This review presents the information obtained to date on the role of mast cells and neuroinflammatory response in the pathogenesis of some socially significant nervous and mental diseases.

Overview of mast cells

Mast cells are found in all organs and are derived from hematopoietic stem cells in the bone marrow. They are characterized by numerous secretory granules in the cytoplasm. Granules contain many (over two hundred) different components, including biogenic amines (such as histamine, catecholamines, and polyamines), proteases (tryptase, chymase, and many other proteases specific and nonspecific for mast cells), lysosomal and other enzymes (cathepsins, â-hexosaminidase, â-glucuronidase, heparanase, peroxidase, etc.), cytokines (interleukins [IL]-1â, IL-2, IL-4, IL-6, IL-10, IL-12, IL-13, IL-15, IL-16, tumor necrosis factor (TNF)-á, etc.), growth factors (such as nerve growth factor, stem cell factor, and vascular endothelial growth factor), proteoglycans (serglycin), mucopolysaccharides (heparin and chondroitin sulfate), peptides and hormones (such as vasoactive intestinal peptide, substance P, endorphin, antimicrobial peptide cathelicidin LL-37, and vasoconstricting peptide endothelin-1), and many other substances [1, 2]. Mast cell mediators are grouped into those permanently stored and synthesized de novo after mast cell activation. Mast cell mediators include both proinflammatory cytokines (such as IL-1, IL-2, IL-6, IL-12, and TNF-á), and anti-inflammatory cytokines (IL-4, IL-10, and IL-13). The presence and release of these modulators of inflammation allows mast cells, after stimulation with certain substances, to initiate and control inflammation in the central nervous system (CNS), which in this case is called neuroinflammation.

Distribution of mast cells in the CNS

Histochemical staining, primarily with aniline dyes, is used to stain selectively and detect mast cells on histological specimens. These cells are detected immunohistochemically even more efficiently, using antibodies to enzymes tryptase and chymase, which are found almost exclusively in mast cells. These imaging techniques have detected mast cells in various organs, including the CNS of humans and animals. In the human brain, mast cells were discovered at the end of the nineteenth century in the affected area in stroke, progressive paralysis, and multiple sclerosis [3]. Over the past several decades, mast cells have been identified in the area postrema, pineal gland, choroid plexus, as well as dura mater and pia mater [4]. Some data indicate the presence of mast cells also in the subfornical organ, vascular organ of the terminal plate, midbrain (without specifying the specific structure), pituitary gland, spinal cord, and neoencephalon parietal cortex [5, 6].

In all publications, attention is drawn to the predominant location of mast cells near the blood vessels and brain structures washed by the cerebrospinal fluid of the ventricles, that is, at the boundaries of the brain (and, apparently, the spinal medulla) and fluids washing it, namely, blood and cerebrospinal fluid. The presence of mast cells in the structures of the blood–brain, cerebrospinal fluid–brain, and blood–cerebrospinal fluid barriers indicates their importance for the normal functioning of brain barriers and corresponds to their role as first line cells of the CNS immune defense.

Relationship of mast cells with cells of the nervous system

Numerous receptors are located on mast cells, including those that determine their sensitivity to pathogenic microorganisms, helminths, and allergens. These include receptors for immunoglobulins E and G (FcåRI and FcãRI), various complement receptors, Toll-like and Nod-like receptors, as well as receptors for some type C lectins. Activation of mast cells by pathogens, mediated by these receptors, causes the release of a certain set of mediators that affect the surrounding nerve, glial cells, and endothelial cells in the CNS.

Mast cell tryptase binds to the protease-activated receptor 2 and the Toll-like receptor localized on neurons, astroglial cells, and microglial cells and triggers the MAPK, AKT, and NF-êB signaling pathways on these cells [6–8]. Mast cells also influence astroglial and microglial cells through histamine, activating histamine receptors H1 and H4 [7, 8].

Mast cells are located near vessels, often in direct contact with them and have various vasoactive compounds, such as histamine, serotonin, nitric oxide, endothelin-1, vascular endothelial growth factor, tryptase, proinflammatory cytokines IL-6, IL-8, TNF-á, etc. Consequently, the activation of mast cells has a pronounced effect on the functional activity of the endothelium of cerebral vessels, changing their permeability and, consequently, the permeability of the blood–brain barrier [9, 10]. Since the impairment of the blood–brain barrier is characteristic of almost all nervous and mental diseases, mast cells are probably, and in some cases proven, involved in this process.

By activating astroglia and microglia, mast cells, together with them, regulate the neuroinflammatory process in response to incoming signals about pathogens and organize later the interrelated work of brain cells to restore the nerve tissue [11, 12]. Inflammation is a complex process caused by infection or mechanical damage to counteract antigens and repair subsequently the damaged structures. However, if the inflammatory process in the CNS becomes out of control and chronic, it can become a damaging factor and cause the death of nerve cells. Data obtained in recent years indicate that neuroinflammation plays a significant role in the pathogenesis of several CNS diseases.

Multiple sclerosis is one of the most common neurodegenerative diseases and has an autoimmune nature. Although its etiology is unknown, multiple sclerosis is believed to be initiated by myelin-specific CD4+ T lymphocytes activated at the periphery. When entering the CNS, these cells cause a neuroinflammatory response aimed at myelin-producing oligodendrocytes, as well as neurons, resulting in myelin breakdown and local impairment of nerve conduction, which leads to neurological disorders. In patients with multiple sclerosis, mast cell count in the brain and spinal cord increases significantly (several times), and they were found in the foci of demyelination (so-called plaques of multiple sclerosis) [3, 13, 14]. Concentrations of histamine and tryptase (compounds characteristic of mast cells) in the cerebrospinal fluid of patients with multiple sclerosis also increase [15, 16]. Increased count and activity of mast cells were also revealed in the brain and spinal cord of rats with experimental allergic encephalomyelitis (a model of multiple sclerosis) [17, 18]. In patients with mastocytosis (a disease characterized by activation and increased mast cell count in body tissues), the incidence of multiple sclerosis increased [19, 20]. By contrast, experimental allergic encephalomyelitis develops in a milder form in transgenic mice without mast cells (KitW/Wv) [21]. Opposite results were also obtained in mice without mast cells of a different line (Kit W-sh/W-sh) [22], which may be associated with various responses of animals of two different lines to the induction of experimental allergic encephalomyelitis.

The abovementioned findings reveal the participation of mast cells in the development of multiple sclerosis; however, research results do not give an unambiguous answer whether mast cells are a pathogenic factor or they counteract the disease development. Animal experiments have shown that mast cells can induce apoptosis of oligodendrocytes and demyelination because of proteases they secreted [23, 24]. Multiple sclerosis is also accompanied by infiltration of type 1 T-helpers into the brain through the blood–brain barrier, which leads to inflammation and neurodegeneration [25]. Impairment of the blood–brain barrier integrity is characteristic of multiple sclerosis and even precedes its clinical manifestations [21, 26], and mast cell mediators can disrupt (increase) the permeability of the blood–brain barrier and thereby facilitate infiltration of type 1 T-helpers and other immunocompetent cells into the brain [27]. These data indicate the possible involvement of mast cells as a pathogenic factor. Based on this, drugs were tested, which inhibit (through various mechanisms) the activity of mast cells for the treatment of multiple sclerosis. Preclinical and clinical trials of this kind of drugs, namely, imatinib, masitinib, cladribine, or tyrphostin AG126, gave moderately positive results [28–31]; evobrutinib had no effect on improvement [32]. In addition, a model of experimental allergic encephalomyelitis demonstrated that antagonists of histamine, one of the main products of mast cells, slow down disease development and severity (but do not prevent it) [11, 33], and in patients taking antihistamines, multiple sclerosis is less common [34].

A study of the brain of patients with multiple sclerosis revealed that mast cells in demyelination foci appear because of inflammation, and not vice versa [13]. However, this finding does not indicate that mast cells migrate to the inflammation focus to perform a protective function, and they can also contribute to neurodegeneration.

Thus, by now, mast cells are clearly involved in the development of multiple sclerosis, and most of the evidence confirms that they are a pathogenic factor causing demyelination and neurodegeneration. Thus, the blockade of their activity (particularly inhibition of the action of their mediators) can contribute to the treatment of multiple sclerosis. However, the original reason why mast cells migrate to the focus of inflammation are activated and become a pathogenic factor has not been established yet.

Massive evidence suggests that Alzheimer’s disease, another neurodegenerative disease, is also associated with neuroinflammation. Alzhei­mer’s disease, which leads to profound memory impairment, is morphologically characterized by the appearance of amyloid plaques and neurofibrillary tangles in brain structures, as well as the development of an inflammatory reaction. The neuroinflammatory process is accompanied by the infiltration of immune cells from the blood into the brain and the activation of intracerebral mast, microglial, and astroglial cells.

The role of mast cells in the neuroinflammatory process in Alzheimer’s disease is poorly investigated. Studies have revealed that the mast cell count increases significantly in the brain and spinal cord of patients with Alzheimer’s disease [5, 35]; moreover, in the autopsy material, if their confinement to places of accumulation of amyloid plaques was not noted, in the mouse model of Alzheimer’s disease, mast cells were detected near the amyloid plaques [36]. In addition, the concentrations of histamine, IL-1â, and IL-6 as proinflammatory mediators’ characteristic of mast cells, increase significantly in the blood serum and cerebrospinal fluid of the patients [37]. Some mast cell mediators (IL-1â, IL-6, TNF-á, and prostaglandins) are known to interfere with the process of memory consolidation [38, 39], which indicates a possible mechanism by which mast cell dysfunction may contribute to memory impairment in Alzheimer’s disease.

Based on data on the involvement of neuroinflammation and mast cells in the development of Alzheimer’s disease, researchers have attempted to use nonsteroidal and other anti-inflammatory drugs to treat and prevent this disease; however, studies have yielded conflicting results [40, 41]. Since 2015, clinical trials of cromolyn, which inhibits the activity of mast cells, have been performed in the USA for the treatment of Alzheimer’s disease [42]. By inhibiting mast cells, this drug was believed to stop the neuro­inflammation-induced neurodegeneration and thereby slow down or even stop the progression of Alzheimer’s disease, especially in the early stages. Similarly, a preclinical and clinical study of masitinib, which is another drug that inhibits mast cell activity, has noted some improvement in the condition and the slowdown in the deve­lopment of Alzheimer’s disease [29]. Thus, the data indicate the participation of mast cells in the pathogenesis of Alzheimer’s disease, as they can contribute to the development of neuroinflammation and disruption of the blood–brain barrier in this disease and possibly affect the metabolism of amyloid proteins.

Amyotrophic lateral sclerosis is a neurodegenerative disease that is accompanied by the degeneration of the spinal and cortical motor neurons. The causes of this disease have not been established, but obtained evidence revealed that the inflammatory response in different areas of the CNS, which is implemented by immunocompetent cells, including mast cells, plays an important role in disease development [43, 44]. In this disease, the blood–brain barrier is disrupted, including at the spinal cord level [45]; as already noted, the permeability and integrity of the barrier can change significantly under the influence of vasoactive mediators of mast cells. Consequently, mast cells (or their precursors) can enter the spinal cord from the bloodstream and release various mediators there, some of which can cause local inflammation and impairment of neuronal function [46]. This assumption is based on the detection of mast cell infiltration into the gray matter of the spinal cord and muscle tissue (including around neuromuscular synapses) in both patients with amyotrophic lateral sclerosis and experimental disease models [5, 43, 47]. In addition, a significant increase was found in the concentration of proinflammatory cytokines IL-6, IL-8, IL-12, and IL-15 in the cerebrospinal fluid, at least some of which originated from mast cells [48–50]. In this regard, TNF-á and specific mast cell proteases can participate in demyelination [23, 24], and this indicates directly the possible involvement of mast cells in the degeneration of motor neuron axons, neuromuscular synapses, and muscle fibers.

With the above background information, we studied the effect of mast cell blockade on neurological status and pathomorphological and biochemical parameters in experimental models of amyotrophic lateral sclerosis and in clinical practice. A model of amyotrophic lateral sclerosis demonstrated that inhibition of mast cells with cromolyn or the flavonoid tetramethoxy luteolin led to a slight but significant improvement in neurological manifestations and a decrease in the expression of proinflammatory cytokines and chemokines (particularly IL-6 and TNF-á) in the spinal cord and blood plasma of mice [51, 52]. A similar rat model demonstrated that tyrosine kinase inhibition by masitinib blocked the tyrosine kinase receptor c-kit and present abundantly on mast cells as well as on neutrophils, thereby preventing mast cell and neutrophil infiltration into the muscles, axonal damage, demyelination, and loss of myofibrils in rats [47]. The first two phases of clinical trials of masitinib (in combination with riluzole) in the treatment of patients with amyotrophic lateral sclerosis revealed ­slowing down of the progression of motor symptoms and an elongation of the life of patients [53]. Thus, the data indicate the involvement of mast cells in the pathogenesis of amyotrophic lateral sclerosis, even if their role in disease development has yet to be established more precisely.

Parkinson’s disease ranks first as the most common movement disorder worldwide and ranks second among the most common neurodegenerative diseases. Its development is associated with the degeneration of the substantia nigra dopaminergic neurons. Recent studies have indicated that the pathogenesis of this disease is also associated with a neuroinflammatory process [16, 54, 55].

Data on the possible involvement of mast cells in neuroinflammation in Parkinson’s ­disease are limited. To our knowledge, only one study reported increased amount and activity of mast cells in the midbrain of patients with Parkinson’s disease [6]. Nowadays, no other data on the possible localization of mast cells in the human substantia nigra in norm or patho­logy. Moreover, we managed to find only one work in which mast cells were demonstrated in the substantia nigra of experimental animals (mice) in the norm [56].

Under the action of the selective dopamin­ergic neurotoxin 1-methyl-4-phenylpyridinium (MPP+), which is widely used to model Parkinson’s disease, the mast cell count in the substantia nigra of mice increased several times simultaneously with increased concentration of inflammatory histamine mediators in the substantia nigra and blood serum, as well as leukotrienes, various cytokines, including IL-1â, IL-2, IL-6, CCL2, TNF-á and transforming growth factor-â, and synthesized in mast cells [6, 57, 58]. This reveals the participation of mast cells in the inflammation process.

Moreover, some of the mediators secreted by mast cells, for example, histamine, CCL2 chemokine, transglutaminase-2, matrix metalloproteinase-3, and murine mast cell protease-6 and -7, can induce directly selective degeneration of nigral dopaminergic neurons or contribute to its development [56, 57]. Accordingly, the experimentally induced decrease in mast cell count leads to a decrease in the number of neurons degenerating under the influence of the MPP+ neurotoxin [59]. These data indicate that the increased activity of mast cells can enhance the neurodegeneration process in the substantia nigra, caused by selective dopaminergic neurotoxin in the experiment, or even directly cause neuronal degeneration. However, so far, this is only a hypothesis that is based on a few experimental data and has serious controversial points, the main one of which is that mast cells were not detected in the substantia nigra of humans and were extremely rarely found in expe­rimental animals. As a result, the possible role of mast cells in the pathogenesis of Parkinson’s disease requires further investigation.

The minimal amount of data indicates the probable involvement of neuroinflammation in the pathogenesis of Huntington’s chorea and the involvement of mast cells in this process [60, 61].

In 1993, it was first shown and later confirmed that depression is accompanied by activation of the immune system. This is manifested as increased levels of inflammatory markers [62]. Then, a study hypothesized that inflammation underlies the pathogenesis of depressive disorders, including major depressive disorder (according to the DSM-5 classification), and their immediate cause is an increase in the amount of proinflammatory cytokines. These claims are based on several arrays of data. First, with depression, the concentration of inflammatory markers (IL-1, IL-6, IL-10, IL-17A, TNF-á, as well as C-reactive protein) increases significantly in the blood and cerebrospinal fluid [63–68], and in one-third of patients with major depressive disorder, abnormalities in the chemical and cellular composition of the cerebrospinal fluid characteristic of inflammation are observed [69]. Increased level of TNF was also noted in the prefrontal cortex of patients with depression [70]. These data are consistent with reports that the intake of anti-inflammatory drugs leads to a decrease in the risk of depression and can be used for its treatment [71–73], while antidepressant drugs are effective in the treatment of atopic dermatitis, which development is closely associated with increased activity of mast cells [74].

Second, many chronic inflammatory diseases are accompanied by a depressive state [75, 76], and activation of the human immune system by injecting a low dose of Salmonella endotoxin increased the blood concentrations of proinflammatory cytokines and is accompanied by symptoms of depression such as depressed mood, anxiety, and cognitive impairment [77].

Third, symptoms of depression are noted in patients after the administration of interferon-á or IL-2 for the treatment of hepatitis C and cancer [78, 79]. Typically, depression caused by proinflammatory cytokines in humans is arrested by antidepressants [80], just as in “classical” depression, which suggests that both types of depression have a common pathogenic mechanism associated with the inflammatory process in the body as a whole and in particular in the CNS. Antidepressants used in clinical practice have a pronounced anti-inflammatory effect; as a result, when remission is achieved, the blood concentration of proinflammatory cytokines decreases [66], while in the absence of a therapeutic effect the level of inflammatory markers in the blood does not decrease [81].

These findings indicate the involvement of neuroinflammation caused by proinflammatory cytokines in the pathogenesis of depression. Mast cells, along with other immunocompetent cells, produce and secrete proinflammatory cytokines that cause depression. However, the real contribution of mast cells in general and in particular that of intracerebral mast cells to the induction of neuroinflammation and development of depression is not clear yet. The involvement of mast cells in the pathogenesis of depression is revealed by clinical data on two diseases associated with mast cells, namely, mastocytosis, characterized by activation and increased count of mast cells in body tissues, and mast cell activation syndrome, in which the count of mast cells does not increase; however, their functional activity (release of mediators) increases sharply. Both diseases, in addition to somatic disorders, are accompanied by depression, which is noted in up to 70% of patients, while the same biochemical changes are registered in the blood as in major depressive disorder, and magnetic resonance imaging identifies structural abnormalities in the brain characteristic of depression [82, 83].

Some other circumstantial evidence also points to the possible involvement of intracerebral mast cells in the pathogenesis of depression. In the brain of patients with depression, the binding of ligands to histamine receptors is reduced, and histamine is mainly involved in the regulation of neuroinflammation. In addition, endogenous histamine has antidepressant properties (in experimental models), and the effect of antidepressants is partially implemented by ­binding to histamine H1 and H2 receptors [84, 85]. These data indicate the involvement of the brain histaminergic system in both neuroinflammation and development of depression. Since brain mast cells contain a significant portion of histamine, it is reasonable to assume that intracerebral mast cell histamine is significant in the neuroinflammatory process and development of depression.

Thus, the pathogenesis of depression is closely related or, which is quite possibly, caused by body and brain inflammation with the direct participation of immunocompetent cells, including intracerebral mast cells.

Similar data can be cited about the role of neuroinflammation and mast cells in the pathogenesis of schizophrenia. Schizophrenia is known to be based on an inflammatory process, which is accompanied by increased concentrations of proinflammatory cytokines in the blood serum. In the acute phase of schizophrenia (as in depression), blood concentrations of proinflammatory cytokines IL-1â, IL-6, IL-8, as well as TNF-á, C-reactive protein, and transforming growth factor-â are increased [67, 86], and successful antipsychotic therapy normalizes the concentrations of at least IL-1â, IL-6, and transforming growth factor-â [65]. In addition, increased concentrations of IL-1â, IL-6, and IL-8 were revealed in the cerebrospinal fluid [68]. Based on these data, attempts were made to treat schizophrenia with anti-inflammatory (primarily nonsteroidal anti-inflammatory) drugs, which gave moderately beneficial effects [87].

These findings indicate that the inflammatory process is characteristic of schizophrenia, and changes are noted not only in the blood but also in the cerebrospinal fluid, which indicates a functional impairment of immunocompetent cells in the CNS. A set of proinflammatory cytokines, which concentration is altered in this disease, is characteristic of mast cells. They are assumed to be involved in neuroinflammation that occurs in schizophrenia.

Autism (DSM-5 autistic spectrum disorder) is associated with impaired functioning of the immune system and is characterized by pronounced signs of neuroinflammation. It is reasonable to suppose that autistic spectrum disorder deve­lops during the embryonic period and is associated with the immune system hyper function of the expectant mother. For example, women suffering from diseases associated with impaired immune system, such as asthma, atopic dermatitis, hay fever, mastocytosis, and others, during pregnancy, have a significantly increased risk of having a child with autism [88]. In the blood serum of pregnant women whose children were later diagnosed with autistic disorder, increased concentrations of IL-4 and IL-5 were noted at weeks 15–19 of gestation, and the levels of IL-4, IL-10, and TNF-á and -â were increased in the amniotic fluid [89, 90]. These findings are consistent with the hypotheses about the possible role of perinatal stress in the pathogenesis of autism. Since maternal cytokines pass through the blood–placental barrier, they can enter the fetal brain in excess amounts and have both a direct effect on the development of the fetal nervous system and an indirect effect by changing the activity of microglia and mast cells and stimulating the release of proinflammatory and neurotoxic mediators, which increased concentration leads to the abnormal development of the fetal nervous system and may contribute to the development of autism [88, 89, 91].

Children with autistic spectrum disorder also have elevated serum levels of IL-1â, IL-4, IL-6, IL-8, IL-16, and IL-17A, and their concentration correlates with disease severity [92, 93]. Levels of proinflammatory cytokines IL-6, IL-8, and TNF-á are increased not only in the blood but also in the cerebrospinal fluid of pediatric patients [94, 95], which indicates the activation of immune cells in the CNS. Moreover, levels of IL-6, IL-8, and TNF-á were increased in the frontal cortex [96] and those of IL-6 and IL-10 were increased in the anterior callosal gyrus [94]. The expression of IL-6 was also increased found in the cerebellum [97], which indicates the presence of neuroinflammation in autistic disorder and suggests its involvement in the pathogenesis of this disease.

The majority of proteins, which are markers of inflammation and which concentration is increased in autistic disorder, is synthesized in mast cells (as well as in some other cells involved in immune reactions) and indicates their possible involvement in disease pathogenesis. This is supported by the finding that autism occurs many times more often in children with mastocytosis [98]; conversely, autistic children suffer from allergies much more often, and mast cells were found to play a primary role in the pathogenesis [99]. These findings, together with data on the changes in the levels of proinflammatory cytokines that can be synthesized by mast cells, suggested that “autism is a brain allergy,” which was caused by (like any allergy) excessive activation of mast cells [100, 101]. However, this hypothesis has not been sufficiently substantiated yet to become a generally accepted theory.

Few data support the finding that attention-deficit hyperactivity disorder is also associated with the development of neuroinflammation with the participation of mast cells [102].

Migraine is manifested as periodic attacks of severe throbbing, usually unilateral headache, and refers to primary headaches according to the international classification of headache. For more than a century of research, many hypotheses have been put forward to explain the occurrence of migraine, but its pathogenesis remains unclear. Nevertheless, to date, researchers have created a general scheme of the occurrence of migraine, which is associated with the excitation (for an unknown reason) of neurons of the trigeminal nerve, extending their processes to various parts of the head, including the dura mater, and activation of mast cells, which are predominantly located in the dura mater of the human brain [103]. In the dura mater, the terminals of trigeminal fibers (C-fibers) secrete several neuropeptides, namely, the substance P, CGRP, neurokinin A, vasointestinal peptide, pituitary adenylate cyclase-activating peptide, as well as histamine, nitric oxide, and adenosine triphosphate, which lead to local vasodilation and activation of numerous dural mast cells [103, 104]. Activated mast cells degranulate, releasing many mediators that, together with peptides released from the terminals of trigeminal fibers, cause inflammation of the dura mater and its vessels and excite the terminals of nociceptive afferent fibers (Aä fibers) [105, 106]. This general pattern is supported and supplemented by the results of different studies conducted using various approaches and methods. Thus, study of brain biopsies of patients with headaches showed that, on the side of pain, degranulating mast cells accumulated around the temporal artery [107], and degranulation of mast cells increased the activity and sensitivity of meningeal nociceptors and caused migraine-type headache [108]. A characteristic symptom of mast cell dysfunction is headaches, as up to 2/3 of patients with mastocytosis and mast cell activation syndrome suffer from them, and in every third case, the pain can be defined as migraine [20, 83]. Pharmacological inhibition of dural mast cell degranulation prevents migraine development [109].

Migraine attacks and primary headaches are also known to be accompanied by a significant increase in the level of proinflammatory cytokines IL-1â, IL-10, and TNF-á, as well as histamine and endothelin-1 in the blood and cerebrospinal fluid [110–112], which indicates the presence of an inflammatory process in migraine. The information that nonsteroidal anti-inflammatory drugs help combat migraine is also consistent with these data [113, 114]. If these cytokines are characteristic not only for mast cells but also for other immune competent and protective cells, then histamine is the main mediator of mast cells. Moreover, in pediatric patients with migraine, the level of tryptase, an enzyme that is a marker of mast cells, was increased in the urine. Therapeutic relaxation, which soothes the headache, also lowered the urinary tryptase concentration [115]. These data, which are in good agreement with each other, indicate the activation of meningeal mast cells in migraine and primary headaches, and the intensity of the headaches correlates with the level of mast cell activity.

Which mediators of mast cells can cause algogenic effects? Histamine is well known to cause headache [112, 116, 117], and its action is mediated through histamine H1 receptors; therefore, H1 antagonists can arrest migraine attacks [118]. IL-1â and TNF also increase the activity and sensitivity of meningeal nociceptors [119]. Degranulation of dural mast cells can be induced by various factors such as stress hormones, certain foods, nitric oxide donors, oxidative stress, and strong sensory stimuli, all of which are well known to induce migraine attacks [120]. A recent study revealed that the algogenic effect of mast cell degranulation of the dura mater is mediated by PAR2, a receptor expressed on dural afferents of neurons of the trigeminal nerve nucleus and specifically activated by serine proteases ­tryptase, trypsin, and elastase, which are secreted by mast cells [108]. These findings open up new perspectives for the pharmacological suppression of migraine headaches.

Conclusion

Increasing evidence indicates that CNS inflammation is a key pathophysiological component in the development of nervous and mental diseases. Brain mast cells are the most important group of cells that regulate neuroinflammation. They perceive signals about pathogens through various receptors and, together with microgliocytes, initiate an inflammatory response in the brain and spinal cord. In some cases, for an unknown reason, neuroinflammation gets out of control, becomes chronic, and can cause neurodegeneration. This general pathogenic mechanism may be characteristic of multiple and amyotrophic lateral sclerosis, Alzheimer’s disease, migraine, depression, schizophrenia, autistic spectrum disorder, and Parkinson’s disease. Mast cells are proven to play a role in neuroinflammation in multiple sclerosis, Alzheimer’s disease, amyotrophic lateral sclerosis, and migraine. Data on the importance of neuroinflammation and mast cells in the development of neuropsychic diseases provide reference for the development of new approaches to their pharmacological treatment and prevention.

Additional information

Funding. The study was conducted within the state assignment of the Institute of Experimental Medicine.

Conflict of interest. The authors declare no conflict of interest.

Author contributions. I.P. Grigorev selected and analyzed the literature and wrote and edited the text. D.E. Korzhevskii analyzed the literature and wrote and edited the article.

×

About the authors

Igor P. Grigorev

Institute of Experimental Medicine

Author for correspondence.
Email: ipg-iem@yandex.ru
ORCID iD: 0000-0002-3535-7638

PhD (Biology), Senior Researcher of the Department of General and Special Morphology

Russian Federation, Saint Petersburg

Dmitrii E. Korzhevskii

Institute of Experimental Medicine

Email: DEK2@yandex.ru
ORCID iD: 0000-0002-2456-8165
SPIN-code: 3252-3029
Scopus Author ID: 12770589000

MD, PhD, DSc (Medicine), Professor of the RAS, Head of the Laboratory of Functional Morphology of the Central and Peripheral Nervous System, Department of General and Special Morphology

Russian Federation, Saint Petersburg

References

  1. Komi EAD, Wohrl S, Bielory L. Mast cell biology at molecular level: a comprehensive review. Clin Rev Allergy Immunol. 2020;58(3):342–365. doi: 10.1007/s12016-019-08769-2
  2. Mukai K, Tsai M, Saito H, Galli SJ. Mast cells as sources of cytokines, chemokines, and growth factors. Immunol Rev. 2018;282(1):121–150. doi: 10.1111/imr.12634
  3. Neumann J. Ueber das Vorkommen der sogenannten “Mastzellen” bei pathologischen Veranderungen des Gehirns. Archiv f. Pathol. Anat. 1890;122:378–380. doi: 10.1007/bf01884453
  4. Grigorev IP, Korzhevskii DE. Mast cells in the vertebrate brain: localization and functions. Journal of Evolutionary Biochemistry and Physiology. 2021;57(1):16–33. doi: 10.1134/S0022093021010026
  5. Fiala M, Chattopadhay M, La Cava A, et al. IL-17A is increased in the serum and in spinal cord CD8 and mast cells of ALS patients. J Neuroinflammation. 2010;7:76. doi: 10.1186/1742-2094-7-76
  6. Kempuraj D, Thangavel R, Selvakumar GP, et al. Mast cell proteases activate astrocytes and glia-neurons and release interleukin-33 by activating p38 and ERK1/2 MAPKs and NF-B. Mol Neurobiol. 2019;56(3):1681–1693. doi: 10.1007/s12035-018-1177-7
  7. Dong H, Zhang X, Wang Y, et al. Suppression of brain mast cells degranulation inhibits microglial activation and central nervous system inflammation. Mol Neurobiol. 2017;54(2):997–1007. doi: 10.1007/s12035-016-9720-x
  8. Zhang X, Wang Y, Dong H, et al. Induction of microglial activation by mediators released from mast cells. Cell Physiol Biochem. 2016;38(4):1520–1531. doi: 10.1159/000443093
  9. Kempuraj D, Mentor S, Thangavel R, et al. Mast cells in stress, pain, blood-brain barrier, neuroinflammation and Alzheimer’s disease. Front Cell Neurosci. 2019;13:54. doi: 10.3389/fncel.2019.00054
  10. Ribatti D. The crucial role of mast cells in blood-brain barrier alterations. Exp Cell Res. 2015;338(1):119–125. doi: 10.1016/j.yexcr.2015.05.013
  11. Pinke KH, Zorzella-Pezavento SFG, Lara VS, Sartori A. Should mast cells be considered therapeutic targets in multiple sclerosis? Neural Regen Res. 2020;15(11):1995–2007. doi: 10.4103/1673-5374.282238
  12. Sandhu JK, Kulka M. Decoding mast cell-microglia communication in neurodegenerative diseases. Int J Mol Sci. 2021;22(3):1093. doi: 10.3390/ijms22031093
  13. Ibrahim MZM, Reder AT, Lawand R, et al. The mast cells of the multiple sclerosis brain. J Neuroimmunol. 1996;70(2):131–138. doi: 10.1016/S0165-5728(96)00102-6
  14. Krüger PG. Multiple sclerosis: a mast cell mediated psycho-somatic disease? World J Neurosci. 2018;8(4):444–453. doi: 10.4236/wjns.2018.84035
  15. Conti P, Kempuraj D. Important role of mast cells in multiple sclerosis. Mult Scler Relat Disord. 2016;5:77–80. doi: 10.1016/j.msard.2015.11.005
  16. Skaper SD, Facci L, Zusso M, Giusti P. An inflammation-centric view of neurological disease: beyond the neuron. Front Cell Neurosci. 2018;12:72. doi: 10.3389/fncel.2018.00072
  17. Kim DY, Jeoung D, Ro JY. Signaling pathways in the activation of mast cells cocultured with astrocytes and colocalization of both cells in experimental allergic encephalomyelitis. J Immunol. 2010;185(1):273–283. doi: 10.4049/jimmunol.1000991
  18. Letourneau R, Rozniecki JJ, Dimitriadou V, Theoharides TC. Ultrastructural evidence of brain mast cell activation without degranulation in monkey experimental allergic encephalomyelitis. J Neuroimmunol. 2003;145(1–2):18–26. doi: 10.1016/j.jneuroim.2003.09.004
  19. Rodrigues F, Edjlali M, Georgin-Lavialle S, et al. Neuroinflammatory disorders and mastocytosis: A possible association? J Allergy Clin Immunol Pract. 2019;7(8):2878–2881.e1. doi: 10.1016/j.jaip.2019.04.033
  20. Smith JH, Butterfield JH, Pardanani A, et al. Neurologic symptoms and diagnosis in adults with mast cell disease. Clin Neurol Neurosurg. 2011;113(7):570–574. doi: 10.1016/j.clineuro.2011.05.002
  21. Brown MA, Weinberg RB. Mast cells and innate lymphoid cells: underappreciated players in CNS autoimmune demyelinating disease. Front Immunol. 2018;9:514. doi: 10.3389/fimmu.2018.00514
  22. Li H, Nourbakhsh B, Safavi F, et al. Kit (W-sh) mice develop earlier and more severe experimental autoimmune encephalomyelitis due to absence of immune suppression. J Immunol. 2011;187(1):274–282. doi: 10.4049/jimmunol.1003603
  23. Medic N, Lorenzon P, Vita F, et al. Mast cell adhesion induces cytoskeletal modifications and programmed cell death in oligodendrocytes. J Neuroimmunol. 2009;218(1–2):57–66. doi: 10.1016/j.jneuroim.2009.10.011
  24. Russi AE, Walker-Caulfield ME, Brown MA. Mast cell inflammasome activity in the meninges regulates EAE disease severity. Clin Immunol. 2018;189:14–22. doi: 10.1016/j.clim.2016.04.009
  25. Batoulis H, Addicks K, Kuerten S. Emerging concepts in autoimmune encephalomyelitis beyond the CD4/T(H)1 paradigm. Ann Anat. 2010;192(4):179–193. doi: 10.1016/j.aanat.2010.06.006
  26. Holman DW, Klein RS, Ransohoff RM. The blood-brain barrier, chemokines and multiple sclerosis. Biochim Biophys Acta. 2011;1812(2):220–230. doi: 10.1016/j.bbadis.2010.07.019
  27. Russi AE, Walker-Caulfield ME, Guo Y, et al. Meningeal mast cell-T cell crosstalk regulates T cell encephalitogenicity. J Autoimmun. 2016;73:100–110. doi: 10.1016/j.jaut.2016.06.015
  28. Adzemovic MV, Zeitelhofer M, Eriksson U, et al. Imatinib ameliorates neuroinflammation in a rat model of multiple sclerosis by enhancing blood-brain barrier integrity and by modulating the peripheral immune response. PLoS One. 2013;8(2):e56586. doi: 10.1371/journal.pone.0056586
  29. Folch J, Petrov D, Ettcheto M, et al. Masitinib for the treatment of mild to moderate Alzheimer’s disease. Expert Rev Neurother. 2015;15(6):587–596. doi: 10.1586/14737175.2015.1045419
  30. Giovannoni G, Comi G, Cook S, et al. A placebo-controlled trial of oral cladribine for relapsing multiple sclerosis. N Engl J Med. 2010;362(5):416–426. doi: 10.1056/NEJMoa0902533
  31. Menzfeld C, John M, van Rossum D, et al. Tyrphostin AG126 exerts neuroprotection in CNS inflammation by a dual mechanism. Glia. 2015;63(6):1083–1099. doi: 10.1002/glia.22803
  32. Montalban X, Arnold DL, Weber MS, et al. Placebo-controlled trial of an oral BTK inhibitor in multiple sclerosis. N Engl J Med. 2019;380(25):2406–2417. doi: 10.1056/NEJMoa1901981
  33. Pinke KH, Zorzella-Pezavento SFG, de Campos Fraga-Silva TF, et al. Calming down mast cells with ketotifen: a potential strategy for multiple sclerosis therapy? Neurotherapeutics. 2020;17(1):218–234. doi: 10.1007/s13311-019-00775-8
  34. Yong HY, McKay KA, Daley CGJ, Tremlett H. Drug exposure and the risk of multiple sclerosis: A systematic review. Pharmacoepidemiol Drug Saf. 2018;27(7):133–139. doi: 10.1002/pds.4357
  35. Maslinska D, Laure-Kamionowska M, Maslinski KT, et al. Distribution of tryptase-containing mast cells and metallothionein reactive astrocytes in human brains with amyloid deposits. Inflamm. Res. 2007;56 Suppl 1:S17–S18. doi: 10.1007/s00011-006-0508-8
  36. Harcha PA, Vargas A, Yi C, et al. Hemichannels are required for amyloid -peptide-induced degranulation and are activated in brain mast cells of APPswe/PS1dE9 mice. J Neurosci. 2015;35(25):9526–9538. doi: 10.1523/JNEUROSCI.3686-14.2015
  37. Swardfager W, Lanctot K, Rothenburg L, et al. A meta-analysis of cytokines in Alzheimer’s disease. Biol Psychiatry. 2010;68(10):930–941. doi: 10.1016/j.biopsych.2010.06.012
  38. Malashenkova IK, Krynskiy SA, Khailov NA, et al. The role of cytokines in memory consolidation. Biol Bull Rev. 2016;6(2):126–140. doi: 10.1134/S2079086416020055
  39. Zhang X, Yao H, Qian Q, et al. Cerebral mast cells participate in postoperative cognitive dysfunction by promoting astrocyte activation. Cell Physiol Biochem. 2016;40(1–2): 104–116. doi: 10.1159/000452528
  40. Gupta PP, Pandey RD, Jha D, et al. Role of traditional nonsteroidal anti-inflammatory drugs in Alzheimer’s disease: a meta-analysis of randomized clinical trials. Am J Alzheimers Dis Other Demen. 2015;30(2):178–182. doi: 10.1177/1533317514542644
  41. McGeer PL, Guo JP, Lee M, et al. Alzheimer’s disease can be spared by nonsteroidal anti-inflammatory drugs. J Alzheimers Dis. 2018;62(3):1219–1222. doi: 10.3233/JAD-170706
  42. Safety and efficacy study of ALZT-OP1 in subjects with evidence of early Alzheimer’s disease (COGNITE). Available from: https://clinicaltrials.gov/ct2/show/NCT02547818. Accessed: June 21, 2021.
  43. Graves MC, Fiala M, Dinglasan LA, et al. Inflammation in amyotrophic lateral sclerosis spinal cord and brain is mediated by activated macrophages, mast cells and T cells. Amyotroph Lateral Scler Other Motor Neuron Disord. 2004;5(4):213–219. doi: 10.1080/14660820410020286
  44. Jones MK, Nair A, Gupta M. Mast cells in neurodegenerative disease. Front Cell Neurosci. 2019;13:171. doi: 10.3389/fncel.2019.00171
  45. Rodrigues MC, Hernandez-Ontiveros DG, Louis MK, et al. Neurovascular aspects of amyotrophic lateral sclerosis. Int Rev Neurobiol. 2012;102:91–106. doi: 10.1016/B978-0-12-386986-9.00004-1
  46. Kempuraj D, Thangavel R, Selvakumar GP, et al. Brain and peripheral atypical inflammatory mediators potentiate neuroinflammation and neurodegeneration. Front Cell Neurosci. 2017;11:216. doi: 10.3389/fncel.2017.00216
  47. Trias E, King PH, Si Y, et al. Mast cells and neutrophils mediate peripheral motor pathway degeneration in ALS. JCI Insight. 2018;3(19):e123249. doi: 10.1172/jci.insight.123249
  48. Kuhle J, Lindberg RL, Regeniter A, et al. Increased levels of inflammatory chemokines in amyotrophic lateral sclerosis. Eur J Neurol. 2009;16(6):771–774. doi: 10.1111/j.1468-1331.2009.02560.x
  49. Mitchell RM, Freeman WM, Randazzo WT, et al. A CSF biomarker panel for identification of patients with amyotrophic lateral sclerosis. Neurology. 2009;72(1):14–19. doi: 10.1212/01.wnl.0000333251.36681.a5
  50. Rentzos M, Rombos A, Nikolaou C, et al. Interleukin-15 and interleukin-12 are elevated in serum and cerebrospinal fluid of patients with amyotrophic lateral sclerosis. Eur Neurol. 2010;63(5):285–290. doi: 10.1159/000287582
  51. Granucci EJ, Griciuc A, Mueller KA, et al. Cromolyn sodium delays disease onset and is neuroprotective in the SOD1(G93A) Mouse Model of amyotrophic lateral sclerosis. Sci Rep. 2019;9(1):17728. doi: 10.1038/s41598-019-53982-w
  52. Theoharides TC, Tsilioni I. Amyotrophic lateral sclerosis, neuroinflammation, and cromolyn. Clin Ther. 2020;42(3):546–549. doi: 10.1016/j.clinthera.2020.01.010
  53. Mora JS, Genge A, Chio A, et al. Masitinib as an add-on therapy to riluzole in patients with amyotrophic lateral sclerosis: a randomized clinical trial. Amyotroph Lateral Scler Frontotemporal Degener. 2020;21(1–2):5–14. doi: 10.1080/21678421.2019.1632346
  54. Guzman-Martinez L, Maccioni RB, Andrade V, et al. Neuroinflammation as a common feature of neurodegenerative disorders. Front Pharmacol. 2019;10:1008. doi: 10.3389/fphar.2019.01008
  55. Schwab AD, Thurston MJ, Machhi J, et al. Immunotherapy for Parkinson’s disease. Neurobiol Dis. 2020;137:104760. doi: 10.1016/j.nbd.2020.104760
  56. Hong GU, Cho JW, Kim SY, et al. Inflammatory mediators resulting from transglutaminase 2 expressed in mast cells contribute to the development of Parkinson’s disease in a mouse model. Toxicol Appl Pharmacol. 2018;358:10–22. doi: 10.1016/j.taap.2018.09.003
  57. Kempuraj D, Thangavel R, Fattal R, et al. Mast cells release chemokine CCL2 in response to parkinsonian toxin 1-methyl-4-phenyl-pyridinium (MPP(+)). Neurochem Res. 2016;41(5):1042–1049. doi: 10.1007/s11064-015-1790-z
  58. Liu JQ, Chu SF, Zhou X, et al. Role of chemokines in Parkinson’s disease. Brain Res Bull. 2019;152:11–18. doi: 10.1016/j.brainresbull.2019.05.020
  59. Selvakumar GP, Ahmed ME, Thangavel R, et al. A role for glia maturation factor dependent activation of mast cells and microglia in MPTP induced dopamine loss and behavioural deficits in mice. Brain Behav Immun. 2020;87:429–443. doi: 10.1016/j.bbi.2020.01.013
  60. Jones MK, Nair A, Gupta M. Mast cells in neurodegenerative disease. Front Cell Neurosci. 2019;13:171. doi: 10.3389/fncel.2019.00171
  61. Moller T. Neuroinflammation in Huntington’s disease. J Neural Transm (Vienna). 2010;117(8):1001–1008. doi: 10.1007/s00702-010-0430-7
  62. Maes M. A review on the acute phase response in major depression. Rev Neurosci. 1993;4(4):407–416. doi: 10.1515/REVNEURO.1993.4.4.407
  63. Enache D, Pariante CM, Mondelli V. Markers of central inflammation in major depressive disorder: A systematic review and meta-analysis of studies examining cerebrospinal fluid, positron emission tomography and post-mortem brain tissue. Brain Behav Immun. 2019;81:24–40. doi: 10.1016/j.bbi.2019.06.015
  64. Eswarappa M, Neylan TC, Whooley MA, et al. Inflammation as a predictor of disease course in posttraumatic stress disorder and depression: A prospective analysis from the Mind Your Heart Study. Brain Behav Immun. 2019;75:220–227. doi: 10.1016/j.bbi.2018.10.012
  65. Goldsmith DR, Rapaport MH, Miller BJ. A meta-analysis of blood cytokine network alterations in psychiatric patients: comparisons between schizophrenia, bipolar disorder and depression. Mol Psychiatry. 2016;21(12):1696–1709. doi: 10.1038/mp.2016.3
  66. Hiles SA, Baker AL, de Malmanche T, Attia J. Interleukin-6, C-reactive protein and interleukin-10 after antidepressant treatment in people with depression: a meta-analysis. Psychol Med. 2012;42(10):2015–2026. doi: 10.1017/S0033291712000128
  67. Milenkovic VM, Stanton EH, Nothdurfter C, et al. The role of chemokines in the pathophysiology of major depressive disorder. Int J Mol Sci. 2019;20(9):2283. doi: 10.3390/ijms20092283
  68. Wang AK, Miller BJ. Meta-analysis of cerebrospinal fluid cytokine and tryptophan catabolite alterations in psychiatric patients: comparisons between schizophrenia, bipolar disorder, and depression. Schizophr Bull. 2018;44(1):75–83. doi: 10.1093/schbul/sbx035
  69. Müller N. Immunology of major depression. Neuroimmunomodulation. 2014;21(2–3):123–130. doi: 10.1159/000356540
  70. Dean B, Tawadros N, Scarr E, Gibbons AS. Regionally-specific changes in levels of tumour necrosis factor in the dorsolateral prefrontal cortex obtained postmortem from subjects with major depressive disorder. J Affect Disord. 2010;120(1–3):245–248. doi: 10.1016/j.jad.2009.04.027
  71. Kessing LV, Rytgaard HC, Gerds TA, et al. New drug candidates for depression – a nationwide population-based study. Acta Psychiatr Scand. 2019;139(1):68–77. doi: 10.1111/acps.12957
  72. Fourrier C, Sampson E, Mills NT, Baune BT. Anti-inflammatory treatment of depression: study protocol for a randomised controlled trial of vortioxetine augmented with celecoxib or placebo. Trials. 2018;19(1):447. doi: 10.1186/s13063-018-2829-7
  73. Quinn AL, Dean OM, Davey CG, et al. Youth Depression Alleviation-Augmentation with an anti-inflammatory agent (YoDA-A): protocol and rationale for a placebo-controlled randomized trial of rosuvastatin and aspirin. Early Interv Psychiatry. 2018;12(1):45–54. doi: 10.1111/eip.12280
  74. Suarez AL, Feramisco JD, Koo J, Steinhoff M. Psychoneuroimmunology of psychological stress and atopic dermatitis: pathophysiologic and therapeutic updates. Acta Derm Venereol. 2012;92(1):7–15. doi: 10.2340/00015555-1188
  75. Häuser W, Janke KH, Klump B, Hinz A. Anxiety and depression in patients with inflammatory bowel disease: comparisons with chronic liver disease patients and the general population. Inflamm Bowel Dis. 2011;17(2):621–632. doi: 10.1002/ibd.21346
  76. Maes M, Kubera M, Obuchowiczwa E, et al. Depression’s multiple comorbidities explained by (neuro)inflammatory and oxidative and nitrosative stress pathways. Neuro Endocrinol Lett. 2011;32(1):7–24.
  77. DellaGioia N, Hannestad J. A critical review of human endotoxin administration as an experimental paradigm of depression. Neurosci Biobehav Rev. 2010;34(1):130–143. doi: 10.1016/j.neubiorev.2009.07.014
  78. Borsini A, Pariante CM, Zunszain PA, et al. The role of circulatory systemic environment in predicting interferon-alpha-induced depression: The neurogenic process as a potential mechanism. Brain Behav Immun. 2019;81:220–227. doi: 10.1016/j.bbi.2019.06.018
  79. Capuron L, Ravaud A, Miller AH, Dantzer R. Baseline mood and psychosocial characteristics of patients developing depressive symptoms during interleukin-2 and/or interferon-alpha cancer therapy. Brain Behav Immun. 2004;18(3):205–213. doi: 10.1016/j.bbi.2003.11.004
  80. Capuron L, Hauser P, Hinze-Selch D, et al. Treatment of cytokine-induced depression. Brain Behav Immun. 2002;16(5):575–580. doi: 10.1016/s0889-1591(02)00007-7
  81. Eller T, Vasar V, Shlik J, Maron E. Pro-inflammatory cytokines and treatment response to escitalopram in major depressive disorder. Prog Neuropsychopharmacol Biol Psychiatry. 2008;32(2):445–450. doi: 10.1016/j.pnpbp.2007.09.015
  82. Boddaert N, Salvador A, Chandesris MO, et al. Neuroimaging evidence of brain abnormalities in mastocytosis. Transl Psychiatry. 2017;7(8):e1197. doi: 10.1038/tp.2017.137
  83. Georgin-Lavialle S, Gaillard R, Moura D, Hermine O. Mastocytosis in adulthood and neuropsychiatric disorders. Transl Res. 2016;174:77–85.e1. doi: 10.1016/j.trsl.2016.03.013
  84. Kano M, Fukudo S, Tashiro A, et al. Decreased histamine H1 receptor binding in the brain of depressed patients. Eur J Neurosci. 2004;20(3):803–810. doi: 10.1111/j.1460-9568.2004.03540.x
  85. Lamberti C, Ipponi A, Bartolini A, et al. Antidepressant-like effects of endogenous histamine and of two histamine H1 receptor agonists in the mouse forced swim test. Br J Pharmacol. 1998;123(7):1331–1336. doi: 10.1038/sj.bjp.0701740
  86. Ushakov VL, Malashenkova IK, Krynskiy SA, et al. Basic cognitive architecture, systemic inflammation, and immune dysfunction in schizophrenia. Modern Technologies in Medicine. 2019;11(3):32–40. doi: 10.17691/stm2019.11.3.04
  87. Pandurangi AK, Buckley PF. Inflammation, antipsychotic drugs, and evidence for effectiveness of anti-inflammatory agents in schizophrenia. Curr Top Behav Neurosci. 2020;44:227–244. doi: 10.1007/7854_2019_91
  88. Angelidou A, Asadi S, Alysandratos KD, et al. Perinatal stress, brain inflammation and risk of autism-review and proposal. BMC Pediatr. 2012;12:89. doi: 10.1186/1471-2431-12-89
  89. Abdallah MW, Larsen N, Grove J, et al. Amniotic fluid inflammatory cytokines: potential markers of immunologic dysfunction in autism spectrum disorders. World J Biol Psychiatry. 2013;14(7):528–538. doi: 10.3109/15622975.2011.639803
  90. Goines PE, Croen LA, Braunschweig D, et al. Increased midgestational IFN-, IL-4 and IL-5 in women bearing a child with autism: A case-control study. Mol Autism. 2011;2:13. doi: 10.1186/2040-2392-2-13
  91. Ferretti CJ, Hollander E. The role of inflammation in autism spectrum disorder. In: Müller N, Myint AM, Schwarz M (eds). Immunology and Psychiatry. Current topics in neurotoxicity. Cham: Springer; 2015;8:275–312. doi: 10.1007/978-3-319-13602-8_14
  92. Ahmad SF, Ansari MA, Nadeem A, et al. Elevated IL-16 expression is associated with development of immune dysfunction in children with autism. Psychopharmacology (Berl). 2019;236(2):831–838. doi: 10.1007/s00213-018-5120-4
  93. Siniscalco D, Schultz S, Brigida AL, Antonucci N. Inflammation and neuro-immune dysregulations in autism spectrum disorders. Pharmaceuticals (Basel). 2018;11(2):56. doi: 10.3390/ph11020056
  94. Chez MG, Dowling T, Patel PB, et al. Elevation of tumor necrosis factor-alpha in cerebrospinal fluid of autistic children. Pediatr Neurol. 2007;36(6):361–365. doi: 10.1016/j.pediatrneurol.2007.01.012
  95. Vargas DL, Nascimbene C, Krishnan C, et al. Neuroglial activation and neuroinflammation in the brain of patients with autism. Ann Neurol. 2005;57(1):67–81. doi: 10.1002/ana.20315
  96. Li X, Chauhan A, Sheikh AM, et al. Elevated immune response in the brain of autistic patients. J Neuroimmunol. 2009;207(1–2): 111–116. doi: 10.1016/j.jneuroim.2008.12.002
  97. Wei H, Zou H, Sheikh AM, et al. IL-6 is increased in the cerebellum of autistic brain and alters neural cell adhesion, migration and synaptic formation. J Neuroinflammation. 2011;8:52. doi: 10.1186/1742-2094-8-52
  98. Theoharides TC. Autism spectrum disorders and mastocytosis. Int J Immunopathol Pharmacol. 2009;22(4):859–865. doi: 10.1177/039463200902200401
  99. Theoharides TC, Tsilioni I, Patel AB, Doyle R. Atopic diseases and inflammation of the brain in the pathogenesis of autism spectrum disorders. Transl Psychiatry. 2016;6(6):e844. doi: 10.1038/tp.2016.77
  100. Theoharides TC. Is a subtype of autism an ‘allergy of the brain’? Clin Ther. 2013;35(5):584–591. doi: 10.1016/j.clinthera.2013.04.009
  101. Theoharides TC, Stewart JM, Panagiotidou S, Melamed I. Mast cells, brain inflammation and autism. Eur J Pharmacol. 2016;778:96–102. doi: 10.1016/j.ejphar.2015.03.086
  102. Song Y, Lu M, Yuan H, et al. Mast cell-mediated neuroinflammation may have a role in attention deficit hyperactivity disorder (Review). Exp Ther Med. 2020;20(2):714–726. doi: 10.3892/etm.2020.8789
  103. Rozniecki JJ, Dimitriadou V, Lambracht-Hall M, et al. Morphological and functional demonstration of rat dura mater mast cell–neuron interactions in vitro and in vivo. Brain Res. 1999;849(1–2):1–15. doi: 10.1016/S0006-8993(99)01855-7
  104. Koroleva K, Gafurov O, Guselnikova V, et al. Meningeal mast cells contribute to ATP-induced nociceptive firing in trigeminal nerve terminals: direct and indirect purinergic mechanisms triggering migraine pain. Front Cell Neurosci. 2019;13:195. doi: 10.3389/fncel.2019.00195
  105. Green DP, Limjunyawong N, Gour N, et al. A Mast-cell-specific receptor mediates neurogenic inflammation and pain. Neuron. 2019;101(3):412–420.e3. doi: 10.1016/j.neuron.2019.01.012
  106. Ramachandran R. Neurogenic inflammation and its role in migraine. Semin Immunopathol. 2018;40(3):301–314. doi: 10.1007/s00281-018-0676-y
  107. Dimitriadou V, Henry P, Brochet B, et al. Cluster headache: ultrastructural evidence for mast cell degranulation and interaction with nerve fibres in the human temporal artery. Cephalalgia. 1990;10(5):221–228. doi: 10.1046/j.1468-2982.1990.1005221.x
  108. Hassler SN, Ahmad FB, Burgos-Vega CC, et al. Protease activated receptor 2 (PAR2) activation causes migraine-like pain behaviors in mice. Cephalalgia. 2019;39(1):111–122. doi: 10.1177/0333102418779548
  109. Monro J, Carini C, Brostoff J. Migraine is a food-allergic disease. Lancet. 1984;2(8405):719–721. doi: 10.1016/s0140-6736(84)92626-6
  110. Karpova MI, Simbirtsev AS, Shamurov YS. State of immune system in patients with primary headaches. Medical Immunology. 2010;12(6):529–536. (In Russ.). doi: 10.15789/1563-0625-2010-6-529-536
  111. Iljazi A, Ayata C, Ashina M, Hougaard A. The role of endothelin in the pathophysiology of migraine – a systematic review. Curr Pain Headache Rep. 2018;22(4):27. doi: 10.1007/s11916-018-0682-8
  112. Yuan H, Silberstein SD. Histamine and migraine. Headache. 2018;58(1):184–193. doi: 10.1111/head.13164
  113. Karatygina NV. Non-steroidal anti-inflammatory medicines in complex therapy of migraine. Russian Medical Journal. 2015;23(30):12–15. (In Russ.)
  114. Goldstein J, Hagen M, Gold M. Results of a multicenter, double-blind, randomized, parallel-group, placebo-controlled, single-dose study comparing the fixed combination of acetaminophen, acetylsalicylic acid, and caffeine with ibuprofen for acute treatment of patients with severe migraine. Cephalalgia. 2014;34(13):1070–1078. doi: 10.1177/0333102414530527
  115. Olness K, Hall H, Rozniecki JJ, et al. Mast cell activation in children with migraine before and after training in self-regulation. Headache. 1999;39(2):101–107. doi: 10.1046/j.1526-4610.1999.3902101.x
  116. Worm J, Falkenberg K, Olesen J. Histamine and migraine revisited: mechanisms and possible drug targets. J Headache Pain. 2019;20(1):30. doi: 10.1186/s10194-019-0984-1
  117. Nurkhametova DF, Koroleva KS, Gafurov OS, et al. Mast cell mediators as pain triggers in migraine: comparison of histamine and serotonin in the activation of primary afferents in the meninges in rats. Neurosci Behav Physiol. 2020;50(7):900–906. doi: 10.1007/s11055-020-00983-2
  118. Togha M, Malamiri RA, Rashidi-Ranjbar N, et al. Efficacy and safety of cinnarizine in the prophylaxis of migraine headaches in children: an open, randomized comparative trial with propranolol. Acta Neurol Belg. 2012;112(1):51–55. doi: 10.1007/s13760-012-0011-7
  119. Levy D. Endogenous mechanisms underlying the activation and sensitization of meningeal nociceptors: the role of immuno-vascular interactions and cortical spreading depression. Curr Pain Headache Rep. 2012;16(3):270–277. doi: 10.1007/s11916-012-0255-1
  120. Borkum JM. Migraine triggers and oxidative stress: a narrative review and synthesis. Headache. 2016;56(1):12–35. doi: 10.1111/head.12725

Supplementary files

Supplementary Files
Action
1. JATS XML

Copyright (c) 2021 Grigorev I.P., Korzhevskii D.E.

Creative Commons License
This work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.

СМИ зарегистрировано Федеральной службой по надзору в сфере связи, информационных технологий и массовых коммуникаций (Роскомнадзор).
Регистрационный номер и дата принятия решения о регистрации СМИ: серия ПИ № ФС 77 - 74760 от 29.12.2018 г.


This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies