Journal of Pediatric Neurology and Neuroscience

ISSN: 2642-4797

REVIEW ARTICLE | VOLUME 2 | ISSUE 1 | DOI: 10.36959/595/400 OPEN ACCESS

The Role of Interleukins in Epileptic Encephalopathies and Immunomodulation with Adrenocorticotropic Hormone

Diana C Benitez Ramirez and Angelica M Uscategui Daccaret

  • Diana C Benitez Ramirez 1*
  • Angelica M Uscategui Daccaret 1
  • Pediatric Neurology Program, Department of Pediatrics, National University of Colombia, Liga Central Contra la Epilepsia, Colombia

Ramirez DCB, Daccaret AMU (2018) The Role of Interleukins in Epileptic Encephalopathies and Immunomodulation with Adrenocorticotropic Hormone. J Pediatr Neurol Neurosci 2(1):23-29.

Accepted: June 30, 2018 | Published Online: July 02, 2018

The Role of Interleukins in Epileptic Encephalopathies and Immunomodulation with Adrenocorticotropic Hormone

Abstract


Epileptic encephalopathies make up a group of electroclinical syndromes that are characterized by to have an established presenting age, clinical neurological deterioration secondary to persistent epileptic activity and an irregular response to classical pharmacological treatments. Its etiology is variable, and its precipitating factors are controversial. Since the 90's, the role of inflammation in epilepsy has been described, through to the findings like the presence of inflammatory cells and molecules in cerebrospinal fluid and surgical specimen from affected patients. Among the molecules that actively participate in this process we can find interleukins, which may be pro-inflammatory or anti-inflammatory and that may alter the permeability of the blood-brain barrier, modify the liberation of neurotransmitters and induce permanent neuroexcitation. The persistence of the neuronal excitability promotes the recurrence of seizure and refractariety to the conventional treatments. This is why we propose that the effectiveness of the immunomodulation therapies in patients with epileptic encephalopathies might be due to control the subjacent inflammatory state.

Keywords


Epileptic encephalopathy, Inflammation, Interleukins, Immunomodulation, ACTH, Biomarker

Introduction


Epileptic encephalopathy is a condition where "the electric activity by itself contributes to a severe cognitive and behavioral deterioration of the patient beyond the expected for the subjacent pathology and susceptible to worsening in the future" [1]. It may present at any age, more severely during early ages considering its interference with the process of brain maturing. For diagnosis, it is necessary to demonstrate the alteration in the development and the loss of acquired abilities associated to epileptiform activity, except in patients whose syndromic diagnosis already includes the word encephalopathy [1]. The first case was reported in 1841 by Dr. West, who described the case of his son, which was characterized by spasms and setbacks in his neurodevelopment [2].

At the moment, several forms of epileptic encephalopathies are recognized, and some of them are mentioned in Table 1.

In the last few years research has provided findings that support the hypothesis that relates a poor regulation of immunological responses (innate and acquired) with the physiopathology of various central nervous system (CNS) disorders [3,4] such as neurodegenerative diseases (Alzheimer's disease), autoimmune diseases (antibody encephalitis), cerebrovascular events and epilepsy [3-6].

CNS Inflammation


Immunological activation in the brain is a physiological process that, during normal conditions, favors the modifications of synapses, neuronal plasticity and recovery from lesions, but a prolonged, exacerbated and uncontrolled response may be the cause of multiple disorders by the aforementioned mechanisms [5-7]. In these cases, the use of the term "maladaptive inflammation" has been proposed [7].

The blood-brain barrier (BBB) in a state of integrity limits the entrance of peripheral defense cells that may damage the nervous system, except activated T cells, against various aggressions [7].

Extrinsic events such as traumas, infection, and cerebrovascular events or intrinsic events like degenerative diseases or frequent epileptic activity may trigger the inflammatory cascade in the central nervous system. The immune response has some particularities here: since it does not have the same defense cells circulating in the periphery, others such as vascular pericytes and perivascular macrophages assume that role [7].

Microglia and astrocytes, in addition to their support, communication and nutrition functions in the neuronal microenvironment play a fundamental role in that cascade since they have the ability of activating after injury [7] and release chemokines, and to modify the expression of neurotransmitters and ionic channels in the cell membrane [8-10].

These aggressions on the CNS activate the innate and acquired inflammatory response [9] the first cells to participate are endothelial and glial cells with the synthesis of inflammatory molecules that increase the permeability of the BBB, favor the expression of adhesion molecules and act as chemotactics, allowing lymphocytes and neutrophils to pass through a barrier that has already been injured, besides the activation of the acquired response increasing cell damage [8,11-14]. The activated complement system induces the formation of membrane attack complexes (MAC), which form pores in the cell membranes of the neurons, microglia, oligodendrocytes and astrocytes favoring cell destruction [6,15].

While these phenomena may be common to various neurological pathologies and induced with the same mechanisms in animal models for different entities, each one has its own characteristics that explain its presentation and open the way for considering different immunomodulation therapies.

Inflammation in Epilepsy


In epilepsy, various findings show the existing relation between inflammation and epileptic seizure. In the first place, the demonstration through surgical specimens of patients with Rasmussen syndrome from inflammatory infiltrates in the BBB. Additionally, it has been proven that the damage to the barrier secondary to any injury, including recurrent epileptic seizures, allow cells to pass from the peripheral immune system to the brain tissue, with the resulting activation of the microglia and the astrocytes, which is a key phase in the generation of a seizures [9,12,16].

Furthermore, in highly epileptogenic tissues like cortical dysplasias, the perilesional tissue of the tubers in the tuberous sclerosis complex and hippocampal sclerosis in temporal lobe epilepsy, the presence of different inflammatory mediators that participate in the innate and adaptive immune response such as chemokines (mainly CCL2, CCL3, CCL4, CXCR4, CXCL12), adhesion molecules (VCAM-1), activation of the COX2 way, NFkB, complement, acute phase proteins like HMGB1, activation of TRL-4 and interleukins has been proven and will be delved into below [12,14,16-18]. Chemokines type CCL2 and CCL3 are produced by astrocytes, perivascular microglia and infiltrated leukocytes. These cells facilitate the passage of monocytes, polymorphonuclears, T cells and dendritic cells through the BBB [12].

Once the primary aggression takes place and the activation of CNS cells occurs, the excitotoxicity cascade happens, leading to an increase in the glutamatergic activity and a decrease of the GABAergic inhibition, an alteration in the function of the ionic channels responsible of attaining neuronal homeostasis, such as those of potassium, and a limitation for the glutamate reuptake by the astrocytes, which is the neuronal mechanism for the auto-regulation of the synaptic microenvironment [14].

Studies in murine models for epilepsy have confirmed the existence of active inflammation preceding the onset of the seizures and persistent with time, evidencing the double role as cause and consequence of epilepsy. In this inflammatory microenvironment, molecules like interleukins, interferon, TNF-α, COX2 derivates and growth factors are found, and they initially cause an alteration of the neuronal function and, chronically, neuron, microglia, oligodendrocyte and astrocyte damage and death, which perpetuates inflammation and epileptic seizure [5,9,19].

Interleukins in Epilepsy


In the physiopathology of epileptic seizures, interleukins also participate, proteins produced mainly by leukocytes, which act as second messengers and intervene in the activation and functioning of the immune system. In the CNS, they have been seen to interact in a paracrine and/or endocrine manner with the glia, modifying glioneural communication, increasing glutamate availability, promoting the transcription of genes related to the glutamate receptors and activating astrocytes, which favors a permanent neuronal excitability state [5,20,21]. Along with various inflammatory factors like TNF-, IL-1 and IL-6, they stimulate the secretion of the corticotropin releasing hormone (CRH), which has been related to the increase of epileptic activity, mainly in West syndrome [22,23].

Aside from their role in favoring neuronal hypersynchrony, interleukins are related to the refractoriness to treatment by different mechanisms that favor excitability and reduce inhibition. In the first case, an increase in the release of excitatory neurotransmitters and a decrease in their uptake, an increase in the expression of alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors in the synaptic membrane due to an increase in the transcription of the corresponding genes are produced. They also induce the activation of the G protein associated to ionic channels, which modifies potassium and calcium currents in the neurons and glia, increase some neurotoxic metabolites like quinolinic acid, which is a tryptophan degradation product and act as an antagonist for NMDA receptors [4,21,24,25]. Additionally, there is a reduction in GABAergic receptor currents in the hippocampus and GABA receptor endocytosis is favored, [26,27] promoting a state of hyperexcitability. In murine and porcine models, prolonged exposure (> 6 hours) to inflammatory IL, such as TNF-α, have been confirmed to favor the expression of glycoprotein 2 in endothelial cells of the BBB, an element in charge of preventing the passage of various drugs like antiepileptics. This explains the difficulty that may occur with time of achieving an adequate response to anticonvulsant drugs [28,29].

Another important action of IL is the inhibition of long-term potentiation, which affects learning, memory and neuronal plasticity processes. While neuroinflammation is required for synaptic remodeling in the learning process during normal conditions, when there are pathological events like epilepsy, it interferes with the creation of new synapses, the production of neurotrophic factors, and neuronal excitability required to consolidate memory. In this type of affectations, it has a fundamental role in the overproduction of IL-1 and IL-6 [30,31].

Among the interleukins that have been found to be highly related to the presence of epilepsy in animal models and human studies, as facilitators or modulators of the crisis, we have IL-1, IL-1RA, IL-6, IL-7, IL-10 and IL-17, which have been reported in surgical pieces of patients with temporal lobe epilepsy, in dysplasias and cortical tubers, febrile seizure and some cases of epileptic encephalopathies, mainly in West syndrome [32-34].

Below we mention relevant aspects of these molecules.

Interleukin 1 (IL-1)

Even though it is majorly produced in the periphery and spread to periventricular organs to exert its action in the CNS, it is also produced in neurons and glial cells. It acts in the interleukin 1 receptor type I (IL-1R1), which has a close relation with N-methyl-D-aspartate receptors (NMDA) in hippocampus pyramidal neurons, modulating glutamatergic transmission. It increases neuronal excitability and decreases the convulsive threshold by means of the phosphorylation of channel subunits, which favors the entrance of calcium [5,25,35]. In the astrocytes, IL-1 increases the release of glutamate and inhibits its reuptake, it restricts the inhibition mediated by gamma-aminobutyric acid (GABA) and stimulates the release of IL-6, which has a convulsive effect [5,24,35].

It also acts in the glia and the astrocytes, activating the expression of genes that favor gliosis and modify the permeability of the BBB. It has also been proven that it induces the expression of high-mobility group protein B1 (HMGB1), which plays an important role in the permanent activation of the inflammatory response in epilepsy [6,20,26].

It has been found to be increased in focal epilepsy, mainly in temporal lobe epilepsy, and to be related with the presence of febrile seizure [20].

Interleukin-1 receptor antagonist (IL-1RA)

Interleukin-1 receptor antagonist is a molecule that prevents all the effects caused by the activation and action of IL-1 [9,26,36]. It is considered that its production is related to seizures regulation mechanisms given the fact that it is produced hours after the appearance of IL-1 in a proportion even 100 times higher [20]. In the study by Yamanaka, et al. [36] an increase in the levels of IL-1RA in relation to clinical and encephalographic improvement of the seizures in children with West syndrome after the immunomodulating treatment was observed.

Interleukin 6 (IL-6)

In animal models, a relation between the increase of seizures susceptibility and the increase of the antagonist effects of glutamate has been found. Additionally, a relation between astrogliosis and a decrease in the number of inhibiting interneurons and the proliferation of astrocytes has been reported. At the same time, it stimulates the release of corticotropin hormone from the pituitary, which has an independent convulsive effect. During clinical practice, an increase in the values after tonic-clonic seizures in patients with epileptic encephalopathies has been reported [9,27,37,38].

Interleukin 7 (IL-7)

It intervenes in maintaining the inflammatory response by favoring proliferation, survival and differentiation of proB cells and regulating the growth of T cells [37,39,40]. Due to this, an important role in epilepsy has been stated for it. In a study where samples from adult patients with temporal lobe epilepsy where evaluated, elevated values of this interleukin were reported [41].

Interleukin 10 (IL-10)

Its anti-inflammatory role is widely known due to its inhibition of the synthesis of the cytokines and proinflammatory agents like IFN-γ, IL-2, IL-3, IL12, TNF-α, granulocyte-macrophage colony-stimulating factor (GM-CSF) and reducing the expression of the major histocompatibility complex class II [22,42]. This has led to postulate this IL is a neuroprotector because it inhibits the inflammatory response produced during seizures [20].

Interleukin 17 (IL-17)

It is part of a big interleukin family (IL-17A, IL-17B, IL-17C, IL-17D, IL-17E, IL-17F) and has a proinflammatory action. Type A deserves some interest due to its action on the CNS by activating the glia and stimulating the release of proinflammatory molecules. It also induces neuron death and damage to the BBB on its own [43-45].

All of the above shows that these are key elements in the physiopathogenic process of epilepsy and that they may be potential biomarkers given the fact that they are molecules that are constantly produced during the process of the disease and are modified with the response to treatment. On the other hand, they could be used for monitoring the epileptogenesis process, the severity of the disease, the risk of complications or deterioration and as a follow-up element in the evaluation of the response to treatment and prognosis [46-48]. There is special emphasis on IL-1, IL-1Ra, IL-10 and IL-17, which have been found altered more frequently and with reproducible results in cases of epileptic status. This opens the expectation for tests that predict the response to treatment and the severity in this condition [3,5,6,8,9,12-14,19].

Moreover, the most studied epileptic encephalopathy from this point of view is the West syndrome where there is strong evidence indicating that inflammation plays a fundamental role in the presence and perpetuation of crises. This may be evidenced by means of high proinflammatory interleukin values and decreased anti-inflammatory values, reporting elevated values mainly for IL-1β [33], IL-12 [33,49], IL-6 [33,50] and a decrease in IL-1RA [51], This justifies the need to consider treatment with immunomodulators like ACTH early in this epileptic syndrome.

ACTH and Immunomodulation


Nowadays, inflammation is one the studied therapeutic objectives for the control of epilepsy, especially in epileptic encephalopathies, where clinical deterioration in the patient is progressive and its management is difficult and frequently poorly effective [4,48,52-54].

Among the proposed immunomodulation strategies that have been widely studied and that have shown efficacy in seizures control at experimental and clinical level we may find immunoglobulin G, methylprednisolone, prednisolone and adrenocorticotropic hormone (ACTH).

ACTH is a hormone that is released naturally by the pituitary through the stimulus of the corticotropin releasing hormone (CRH) during a stressful event. ACTH in its plasma state is in charge of stimulating the release of glucocorticoids by the adrenal gland, with the capacity of modifying the function of the receptors of the neurotransmitters and the neuropeptides in the CNS. As such, it fulfills a neuromodulating and immunomodulating action intervening in the inflammatory process [32,55,56].

The mechanisms why it fulfills these functions are stated as follows:

From the neuroendocrinological point of view:

I. It induces the synthesis of peripheral glucocorticoids, which are capable of passing through the BBB and acting on its receptors in the CNS, modifying the voltage dependent calcium channels currents [57].

II. It stimulates the synthesis of neurosteroids by the neurons and the glia, which act as positive allosteric modulators of GABA receptors [9,55,57].

III. They inhibit the release of CRH in the hippocampus, which has a proconvulsing action mainly in the immature brain, favoring glutamatergic transmission, reducing the periods of synaptic hyperpolarization and inducing the expression of proinflammatory IL receptors [9,54,55,57].

And as a neuroimmunomodulator it has the ability to:

I. Regulate the proliferation, apoptosis and cell differentiation by means of the induction of enzymatic acetylation [55,57,58].

II. Control the expression and release of neurotransmitters and neuromodulators.

III. Diminish the susceptibility to seizure through the maturing of the myelin and dendritic formations [9,55,58].

IV. Diminish the production of IL and cellular lymphocyte activation [9].

In infantile spasms, the application of ACTH is considered one of the preferred treatments [59] and has shown clinical improvement in approximately 50% of cases, with spasm remission, normalization of electroencephalic findings and neurodevelopment improvement. The relapse ratio is variable and depends on the etiological diagnosis, the dosing and duration of the treatment [51-55,57,60-66,]. Furthermore, there are reports of effectiveness of the treatment in other type of epileptic encephalopathies like the Landau-Kleffner and the Lennox-Gastaut syndromes and other encephalopathies with different causes and characteristics [67-71].

The correlation between clinical improvement in patients receiving ACTH and changes in the inflammatory response has been made through the measurement of interleukins given their constant expression in some epileptic syndromes like the West syndrome. In patients with good clinical and electrical response to pharmacological treatment, a decrease in proinflammatory IL, such as IL-1, IL-6 and IL-12, and an increase in anti-inflammatory interleukins like IL-1RA have been found [33,36,49,50,71]. In some publications, the response to ACTH has been higher that with other corticoids, which can be explained due to its multiple mechanisms at immunological and endocrine levels.

Specifically, in the West syndrome, the use of ACTH has shown not only a real control of the seizures at short and medium-term, but also improvement in the neurodevelopment process in these children. The proposed mechanisms range from improvement of neuron function when controlling hyperexcitability to considering that the regulation of high proinflammatory activity that hinders development and synaptic connectivity returns to base conditions, which provides an optimal environment for various learning processes [31].

Acknowledgements and Disclosures


Inflammation has a fundamental role in the processes that affect the CNS, particularly in epilepsy, promoting the generation and persistence of crises after an initial trigger. This inflammatory response that is initially produced to delimit the damage and recover function in the affected tissue can be established indefinitely due to the constant production of neurotoxic and pro-inflammatory substances and a failure in endogenous control mechanisms (anti-inflammatories).

The treatments that delimit this process and reduce inflammation leading to the restoration of immunological balance, have shown to be effective in the control of seizure, especially in epileptic encephalopathies, where usually traditional pharmacological treatments show low effectiveness and clinical and functional deterioration of the patient are progressive.

Having follow-up and vigilance tools in patients with treated epilepsy gives us the opportunity of predicting and detecting early complications, generating early therapeutic strategies that may prevent clinical deterioration of the patients and also have individualized management. Thus, further research dealing with biomarkers and the use of available knowledge in daily clinical practice, are necessary.

Conflict of Interest


The authors declare no conflicts of interest.

Financing Sources


Economic support from the Universidad Nacional de Colombia.

References


  1. Berg AT, Berkovic SF, Brodie MJ, et al. (2010) Revised terminology and concepts for organization of seizures and epilepsies: Report of the ILAE Commission on Classification and Terminology, 2005-2009. Epilepsia 51: 676-685.
  2. West WJ (1841) On a peculiar form of infantile convulsions. The Lancet 1: 724-725.
  3. Vezzani A, Granata T (2005) Brain inflammation in epilepsy: Experimental and clinical evidence. Epilepsia 46: 1724-1743.
  4. Yu N, Liu H, Di Q (2013) Modulation of immunity and the inflammatory response: A new target for treating drug-resistant epilepsy. Curr Neuropharmacol 11: 114-127.
  5. Vezzani A, Aronica E, Mazarati A, et al. (2013) Epilepsy and brain inflammation. Exp Neurol 244: 11-21.
  6. Lucas SM, Rothwell NJ, Gibson RM (2006) The role of inflammation in CNS injury and disease. Br J Pharmacol 147: S232-S240.
  7. Xanthos DN, Sandkühler J (2014) Neurogenic neuroinflammation: Inflammatory CNS reactions in response to neuronal activity. Nat Rev Neurosci 15: 43-53.
  8. Devinsky O, Vezzani A, Najjar S, et al. (2013) Glia and epilepsy: Excitability and inflammation. Trends Neurosci 36: 174-184.
  9. Choi J, Koh S (2008) Role of brain inflammation in epileptogenesis. Yonsei Med J 49: 1-18.
  10. Gibbons MB, Smeal RM, Takahashi DK, et al. (2013) Contributions of astrocytes to epileptogenesis following status epilepticus: Opportunities for preventive therapy? Neurochem Int 63: 660-669.
  11. Xu D, Miller SD, Koh S (2013) Immune mechanisms in epileptogenesis. Front Cell Neurosci 7: 1-8.
  12. Fabene PF, Bramanti P, Constantin G (2010) The emerging role for chemokines in epilepsy. J Neuroimmunol 224: 22-27.
  13. Verrotti, Alberto, Latini, et al. (2007) The role of inflammation in epilepsy. Current Pediatric Reviews 3: 312-316.
  14. Marchi N, Granata T, Janigro D (2014) Inflammatory pathways of seizure disorders. Trends Neurosci 37: 55-65.
  15. Allan S, Rothwell NJ (2003) Inflammation in central nervous system injury. Philos Trans R Soc Lond B Biol Sci 358: 1669-1677.
  16. Vezzani A, Friedman A, Dingledine RJ (2013) The role of inflammation in epileptogenesis. Neuropharmacology 69: 16-24.
  17. Specchio N, Fusco L, Claps D, et al. (2010) Epileptic encephalopathy in children possibly related to immune-mediated pathogenesis. Brain Dev 32: 51-56.
  18. Vezzani A (2014) Epilepsy and inflammation in the brain: Overview and pathophysiology. Epilepsy Curr 14: 3-7.
  19. Vezzani A, French J, Bartfai T, et al. (2011) The role of inflammation in epilepsy. Nat Rev Neurol 7: 31-40.
  20. Youngah Y, In Kyung S, In Goo L (2013) The role of cytokines in seizures: Interleukin (IL)-1β, IL-1Ra, IL-8, and IL-10. Korean J Pediatr 56: 271-274.
  21. Vezzani A, Balosso S, Ravizza T (2008) The role of cytokines in the pathophysiology of epilepsy. Brain Behav Immun 22: 797-803.
  22. Modzikowska-Albrecht J, Steinborn B, Zarowski M (2007) Cytokines, epilepsy and antiepileptic drugs--is there a mutual influence ? Pharmacol Rep 59: 129-138.
  23. Frost D, Hrachovy RA (2005) Pathogenesis of infantile spasms: A model based on developmental desynchronization. J Clin Neurophysiol 22: 25-36.
  24. Friedman A, Dingledine R (2011) Molecular cascades that mediate the influence of inflammation on epilepsy. Epilepsia 52: 33-39.
  25. Rijkers K, Majoie HJ, Hoogland G, et al. (2009) The role of interleukin-1 in seizures and epilepsy: A critical review. Exp Neurol 216: 258-271.
  26. Viviani B, Gardoni F, Marinovich M (2007) Cytokines and neuronal ion channels in health and disease. Int Rev Neurobiol 82: 247-263.
  27. Silveira G, de Oliveira ACP, Teixeira AL (2012) Insights into inflammation and epilepsy from the basic and clinical sciences. J Clin Neurosci 19: 1071-1075.
  28. Von Wedel Parlow M, Wölte P, Galla HJ (2009) Regulation of major efflux transporters under inflammatory conditions at the blood-brain barrier in vitro. J Neurochem 111: 111-118.
  29. Barrier B, Hartz AM, Miller DS (2007) Tumor Necrosis Factor Alpha and Endothelin-1 Increase P-Glycoprotein Expression and Transport Activity at the. Mol Pharmacol 71: 667-675.
  30. Galic M, Riazi K, Pittman QJ (2012) Cytokines and brain excitability. Front Neuroendocrinol 33: 116-125.
  31. Yirmiya R, Goshen I (2011) Immune modulation of learning, memory, neural plasticity and neurogenesis. Brain Behav Immun 25: 181-213.
  32. Billiau AD, Wouters CH, Lagae LG (2005) Epilepsy and the immune system: Is there a link? Eur J Paediatr Neurol 9: 29-42.
  33. Shiihara T, Miyashita M, Yoshizumi M, et al. (2010) Peripheral lymphocyte subset and serum cytokine profiles of patients with West syndrome. Brain Dev 32: 695-702.
  34. Aronica E, Crino PB (2011) Inflammation in epilepsy: Clinical observations. Epilepsia 52: 26-32.
  35. Vezzani A, Moneta D, Richichi C, et al. (2002) Functional role of inflammatory cytokines and antiinflammatory molecules in seizures and epileptogenesis. Epilepsia 43: 30-35.
  36. Yamanaka G, Kawashima H, Oana S, et al. (2010) Increased level of serum interleukin-1 receptor antagonist subsequent to resolution of clinical symptoms in patients with West syndrome. J Neurol Sci 298: 106-109.
  37. Zarczuk R, Łukasik D, Jedrych M, et al. (2010) Immunological aspects of epilepsy. Pharmacol Rep 62: 592-607.
  38. Lehtimäki KA, Keränen T, Huhtala H, et al. (2004) Regulation of IL-6 system in cerebrospinal fluid and serum compartments by seizures: the effect of seizure type and duration. J Neuroimmunol 152: 121-125.
  39. Lundström W, Fewkes NM, Mackall CL (2012) IL-7 in human health and disease. Semin Immunol 24: 218-224.
  40. Milne CD, Paige CJ (2006) IL-7: A key regulator of B lymphopoiesis. Semin Immunol 18: 20-30.
  41. Kan AA, de Jager W, de Wit M, et al. (2012) Protein expression profiling of inflammatory mediators in human temporal lobe epilepsy reveals co-activation of multiple chemokines and cytokines. J Neuroinflammation 9: 207.
  42. Li G, Bauer S, Nowak M, et al. (2011) Cytokines and epilepsy. Seizure 20: 249-256.
  43. Moynes DM, Vanner SJ, Lomax AE (2014) Participation of interleukin 17A in neuroimmune interactions. Brain Behav Immun 41: 1-9.
  44. Hu Y, Shen F, Crellin NK, et al. (2011) The IL-17 pathway as a major therapeutic target in autoimmune diseases. Ann N Y Acad Sci 1217: 60-76.
  45. Mao LY, Ding J, Peng WF, et al. (2013) Interictal interleukin-17A levels are elevated and correlate with seizure severity of epilepsy patients. Epilepsia 54: e142-e145.
  46. Hegde M, Lowenstein DH (2014) The search for circulating epilepsy biomarkers. Biomark Med 8: 413-427.
  47. Lukasiuk K, Becker AJ (2014) Molecular biomarkers of epileptogenesis. Neurotherapeutics 11: 319-323.
  48. Dedeurwaerdere S, Friedman A, Fabene PF, et al. (2012) Finding a better drug for epilepsy: Anti-inflammatory targets. Epilepsia 53: 1113-1118.
  49. Liu ZS, Wang QW, Wang FL, et al. (2001) Serum cytokine levels are altered in patients with West syndrome. Epilepsia 23: 548-551.
  50. Tekgul H, Polat M, Tosun A, et al. (2006) Cerebrospinal fluid interleukin-6 levels in patients with west syndrome. Brain Dev 28: 19-23.
  51. Haginoya K, Noguchi R, Zhao Y, et al. (2009) Reduced levels of interleukin-1 receptor antagonist in the cerebrospinal fluid in patients with West syndrome. Epilepsy Res 85: 314-317.
  52. Uscátegui AM (2008) Inmunoterapia en epilepsia refractaria Immunotherapy in refractory epilepsy. Acta Neurológica Colombiana 24: 46-50.
  53. Marchi N, Fan Q, Ghosh C, et al. (2009) Antagonism of peripheral inflammation reduces the severity of status epilepticus. Neurobiol Dis 33: 171-181.
  54. Falip M, Salas-Puig X, Cara C (2013) Causes of CNS inflammation and potential targets for anticonvulsants. CNS Drugs 27: 611-623.
  55. Brunson KL, Avishai-Eliner S, Baram TZ (2002) ACTH treatmen of infantile spasms: mechanisms ofits effects in modulation of neuronal excitability. Int Rev Neurobiol 49: 185-197.
  56. Brunson KL, Eghbal-Ahmadi M, Baram TZ (2001) How do the many etiologies of West syndrome lead to excitability and seizures? The corticotropin releasing hormone excess hypothesis. Brain Dev 23: 533-538.
  57. Verhelst H, Boon P, Buyse G, et al. (2005) Steroids in intractable childhood epilepsy: Clinical experience and review of the literature. Seizure 14: 412-421.
  58. Lin HC, Young C, Wang PJ, et al. (2006) ACTH therapy for Taiwanese children with West syndrome -efficacy and impact on long-term prognosis. Brain Dev 28: 196-201.
  59. Go CY, Mackay MT, Weiss SK, et al. (2012) Evidence-based guideline update: Medical treatment of infantile spasms. Report of the Guideline Development Subcommittee of the American Academy of Neurology and the Practice Committee of the Child Neurology Society. Neurology 12: 1974-1980.
  60. Brunson KL, Khan N, Eghbal-Ahmadi M, et al. (2001) Corticotropin (ACTH) acts directly on amygdala neurons to down-regulate corticotropin-releasing hormone gene expression. Ann Neurol 49: 304-312.
  61. Wang J, Wang J, Zhang Y, et al. (2012) Proteomic analysis of adrenocorticotropic hormone treatment of an infantile spasm model induced by N-methyl-D-aspartic acid and prenatal stress. PLoS One 7: e45347.
  62. Cohen-Sadan S, Kramer U, Ben-Zeev B, et al. (2009) Multicenter long-term follow-up of children with idiopathic West syndrome: ACTH versus vigabatrin. Eur J Neurol 16: 482-487.
  63. Hancock E, Osborne J, Edwards S (2013) Treatment of infantile spasms. Cochrane Database of Sysematic Review 5.
  64. Lux AL, Edwards SW, Hancock E, et al. (2005) The United Kingdom Infantile Spasms Study (UKISS) comparing hormone treatment with vigabatrin on developmental and epilepsy outcomes to age 14 months: A multicentre randomised trial. Lancet Neurol 4: 712-717.
  65. Oguni H, Yanagaki S, Hayashi K, et al. (2006) Extremely low-dose ACTH step-up protocol for West syndrome: Maximum therapeutic effect with minimal side effects. Brain Dev 28: 8-13.
  66. Guevara-Campos J, González de Guevara L (2007) Síndrome de Landau-Kleffner : Análisis de 10 casos en Venezuela. Rev Neurol 44: 652-656.
  67. Okumura A, Tsuji T, Kato T, et al. (2006) ACTH therapy for generalized seizures other than spasms. Seizure 15: 469-475.
  68. Kalra V, Sharma S, Arya R (2009) ACTH Therapy in refractory generalized epilepsy. Indian J Pediatr 76: 91-93.
  69. Medina Malo CH, Guío LV, Uscátegui AM RJ (2008) Experiencia con el uso de hormona adrenocorticotropica (ACTH) en pacientes con encefalopatía epiléptica Neuropediatria Liga Central Contra la Epilepsia. Acta Neurol Colomb 24: 78.
  70. Lehtimäki KA, Liimatainen S, Peltola J, et al. (2011) The serum level of interleukin-6 in patients with intellectual disability and refractory epilepsy. Epilepsy Res 95: 184-187.
  71. Kalra V, Gulati S, Pandey RM, et al. (2002) West syndrome and other infantile epileptic encephalopathies--Indian hospital experience. Brain Dev 24: 130-139.

Abstract


Epileptic encephalopathies make up a group of electroclinical syndromes that are characterized by to have an established presenting age, clinical neurological deterioration secondary to persistent epileptic activity and an irregular response to classical pharmacological treatments. Its etiology is variable, and its precipitating factors are controversial. Since the 90's, the role of inflammation in epilepsy has been described, through to the findings like the presence of inflammatory cells and molecules in cerebrospinal fluid and surgical specimen from affected patients. Among the molecules that actively participate in this process we can find interleukins, which may be pro-inflammatory or anti-inflammatory and that may alter the permeability of the blood-brain barrier, modify the liberation of neurotransmitters and induce permanent neuroexcitation. The persistence of the neuronal excitability promotes the recurrence of seizure and refractariety to the conventional treatments. This is why we propose that the effectiveness of the immunomodulation therapies in patients with epileptic encephalopathies might be due to control the subjacent inflammatory state.

References

  1. Berg AT, Berkovic SF, Brodie MJ, et al. (2010) Revised terminology and concepts for organization of seizures and epilepsies: Report of the ILAE Commission on Classification and Terminology, 2005-2009. Epilepsia 51: 676-685.
  2. West WJ (1841) On a peculiar form of infantile convulsions. The Lancet 1: 724-725.
  3. Vezzani A, Granata T (2005) Brain inflammation in epilepsy: Experimental and clinical evidence. Epilepsia 46: 1724-1743.
  4. Yu N, Liu H, Di Q (2013) Modulation of immunity and the inflammatory response: A new target for treating drug-resistant epilepsy. Curr Neuropharmacol 11: 114-127.
  5. Vezzani A, Aronica E, Mazarati A, et al. (2013) Epilepsy and brain inflammation. Exp Neurol 244: 11-21.
  6. Lucas SM, Rothwell NJ, Gibson RM (2006) The role of inflammation in CNS injury and disease. Br J Pharmacol 147: S232-S240.
  7. Xanthos DN, Sandkühler J (2014) Neurogenic neuroinflammation: Inflammatory CNS reactions in response to neuronal activity. Nat Rev Neurosci 15: 43-53.
  8. Devinsky O, Vezzani A, Najjar S, et al. (2013) Glia and epilepsy: Excitability and inflammation. Trends Neurosci 36: 174-184.
  9. Choi J, Koh S (2008) Role of brain inflammation in epileptogenesis. Yonsei Med J 49: 1-18.
  10. Gibbons MB, Smeal RM, Takahashi DK, et al. (2013) Contributions of astrocytes to epileptogenesis following status epilepticus: Opportunities for preventive therapy? Neurochem Int 63: 660-669.
  11. Xu D, Miller SD, Koh S (2013) Immune mechanisms in epileptogenesis. Front Cell Neurosci 7: 1-8.
  12. Fabene PF, Bramanti P, Constantin G (2010) The emerging role for chemokines in epilepsy. J Neuroimmunol 224: 22-27.
  13. Verrotti, Alberto, Latini, et al. (2007) The role of inflammation in epilepsy. Current Pediatric Reviews 3: 312-316.
  14. Marchi N, Granata T, Janigro D (2014) Inflammatory pathways of seizure disorders. Trends Neurosci 37: 55-65.
  15. Allan S, Rothwell NJ (2003) Inflammation in central nervous system injury. Philos Trans R Soc Lond B Biol Sci 358: 1669-1677.
  16. Vezzani A, Friedman A, Dingledine RJ (2013) The role of inflammation in epileptogenesis. Neuropharmacology 69: 16-24.
  17. Specchio N, Fusco L, Claps D, et al. (2010) Epileptic encephalopathy in children possibly related to immune-mediated pathogenesis. Brain Dev 32: 51-56.
  18. Vezzani A (2014) Epilepsy and inflammation in the brain: Overview and pathophysiology. Epilepsy Curr 14: 3-7.
  19. Vezzani A, French J, Bartfai T, et al. (2011) The role of inflammation in epilepsy. Nat Rev Neurol 7: 31-40.
  20. Youngah Y, In Kyung S, In Goo L (2013) The role of cytokines in seizures: Interleukin (IL)-1β, IL-1Ra, IL-8, and IL-10. Korean J Pediatr 56: 271-274.
  21. Vezzani A, Balosso S, Ravizza T (2008) The role of cytokines in the pathophysiology of epilepsy. Brain Behav Immun 22: 797-803.
  22. Modzikowska-Albrecht J, Steinborn B, Zarowski M (2007) Cytokines, epilepsy and antiepileptic drugs--is there a mutual influence ? Pharmacol Rep 59: 129-138.
  23. Frost D, Hrachovy RA (2005) Pathogenesis of infantile spasms: A model based on developmental desynchronization. J Clin Neurophysiol 22: 25-36.
  24. Friedman A, Dingledine R (2011) Molecular cascades that mediate the influence of inflammation on epilepsy. Epilepsia 52: 33-39.
  25. Rijkers K, Majoie HJ, Hoogland G, et al. (2009) The role of interleukin-1 in seizures and epilepsy: A critical review. Exp Neurol 216: 258-271.
  26. Viviani B, Gardoni F, Marinovich M (2007) Cytokines and neuronal ion channels in health and disease. Int Rev Neurobiol 82: 247-263.
  27. Silveira G, de Oliveira ACP, Teixeira AL (2012) Insights into inflammation and epilepsy from the basic and clinical sciences. J Clin Neurosci 19: 1071-1075.
  28. Von Wedel Parlow M, Wölte P, Galla HJ (2009) Regulation of major efflux transporters under inflammatory conditions at the blood-brain barrier in vitro. J Neurochem 111: 111-118.
  29. Barrier B, Hartz AM, Miller DS (2007) Tumor Necrosis Factor Alpha and Endothelin-1 Increase P-Glycoprotein Expression and Transport Activity at the. Mol Pharmacol 71: 667-675.
  30. Galic M, Riazi K, Pittman QJ (2012) Cytokines and brain excitability. Front Neuroendocrinol 33: 116-125.
  31. Yirmiya R, Goshen I (2011) Immune modulation of learning, memory, neural plasticity and neurogenesis. Brain Behav Immun 25: 181-213.
  32. Billiau AD, Wouters CH, Lagae LG (2005) Epilepsy and the immune system: Is there a link? Eur J Paediatr Neurol 9: 29-42.
  33. Shiihara T, Miyashita M, Yoshizumi M, et al. (2010) Peripheral lymphocyte subset and serum cytokine profiles of patients with West syndrome. Brain Dev 32: 695-702.
  34. Aronica E, Crino PB (2011) Inflammation in epilepsy: Clinical observations. Epilepsia 52: 26-32.
  35. Vezzani A, Moneta D, Richichi C, et al. (2002) Functional role of inflammatory cytokines and antiinflammatory molecules in seizures and epileptogenesis. Epilepsia 43: 30-35.
  36. Yamanaka G, Kawashima H, Oana S, et al. (2010) Increased level of serum interleukin-1 receptor antagonist subsequent to resolution of clinical symptoms in patients with West syndrome. J Neurol Sci 298: 106-109.
  37. Zarczuk R, Łukasik D, Jedrych M, et al. (2010) Immunological aspects of epilepsy. Pharmacol Rep 62: 592-607.
  38. Lehtimäki KA, Keränen T, Huhtala H, et al. (2004) Regulation of IL-6 system in cerebrospinal fluid and serum compartments by seizures: the effect of seizure type and duration. J Neuroimmunol 152: 121-125.
  39. Lundström W, Fewkes NM, Mackall CL (2012) IL-7 in human health and disease. Semin Immunol 24: 218-224.
  40. Milne CD, Paige CJ (2006) IL-7: A key regulator of B lymphopoiesis. Semin Immunol 18: 20-30.
  41. Kan AA, de Jager W, de Wit M, et al. (2012) Protein expression profiling of inflammatory mediators in human temporal lobe epilepsy reveals co-activation of multiple chemokines and cytokines. J Neuroinflammation 9: 207.
  42. Li G, Bauer S, Nowak M, et al. (2011) Cytokines and epilepsy. Seizure 20: 249-256.
  43. Moynes DM, Vanner SJ, Lomax AE (2014) Participation of interleukin 17A in neuroimmune interactions. Brain Behav Immun 41: 1-9.
  44. Hu Y, Shen F, Crellin NK, et al. (2011) The IL-17 pathway as a major therapeutic target in autoimmune diseases. Ann N Y Acad Sci 1217: 60-76.
  45. Mao LY, Ding J, Peng WF, et al. (2013) Interictal interleukin-17A levels are elevated and correlate with seizure severity of epilepsy patients. Epilepsia 54: e142-e145.
  46. Hegde M, Lowenstein DH (2014) The search for circulating epilepsy biomarkers. Biomark Med 8: 413-427.
  47. Lukasiuk K, Becker AJ (2014) Molecular biomarkers of epileptogenesis. Neurotherapeutics 11: 319-323.
  48. Dedeurwaerdere S, Friedman A, Fabene PF, et al. (2012) Finding a better drug for epilepsy: Anti-inflammatory targets. Epilepsia 53: 1113-1118.
  49. Liu ZS, Wang QW, Wang FL, et al. (2001) Serum cytokine levels are altered in patients with West syndrome. Epilepsia 23: 548-551.
  50. Tekgul H, Polat M, Tosun A, et al. (2006) Cerebrospinal fluid interleukin-6 levels in patients with west syndrome. Brain Dev 28: 19-23.
  51. Haginoya K, Noguchi R, Zhao Y, et al. (2009) Reduced levels of interleukin-1 receptor antagonist in the cerebrospinal fluid in patients with West syndrome. Epilepsy Res 85: 314-317.
  52. Uscátegui AM (2008) Inmunoterapia en epilepsia refractaria Immunotherapy in refractory epilepsy. Acta Neurológica Colombiana 24: 46-50.
  53. Marchi N, Fan Q, Ghosh C, et al. (2009) Antagonism of peripheral inflammation reduces the severity of status epilepticus. Neurobiol Dis 33: 171-181.
  54. Falip M, Salas-Puig X, Cara C (2013) Causes of CNS inflammation and potential targets for anticonvulsants. CNS Drugs 27: 611-623.
  55. Brunson KL, Avishai-Eliner S, Baram TZ (2002) ACTH treatmen of infantile spasms: mechanisms ofits effects in modulation of neuronal excitability. Int Rev Neurobiol 49: 185-197.
  56. Brunson KL, Eghbal-Ahmadi M, Baram TZ (2001) How do the many etiologies of West syndrome lead to excitability and seizures? The corticotropin releasing hormone excess hypothesis. Brain Dev 23: 533-538.
  57. Verhelst H, Boon P, Buyse G, et al. (2005) Steroids in intractable childhood epilepsy: Clinical experience and review of the literature. Seizure 14: 412-421.
  58. Lin HC, Young C, Wang PJ, et al. (2006) ACTH therapy for Taiwanese children with West syndrome -efficacy and impact on long-term prognosis. Brain Dev 28: 196-201.
  59. Go CY, Mackay MT, Weiss SK, et al. (2012) Evidence-based guideline update: Medical treatment of infantile spasms. Report of the Guideline Development Subcommittee of the American Academy of Neurology and the Practice Committee of the Child Neurology Society. Neurology 12: 1974-1980.
  60. Brunson KL, Khan N, Eghbal-Ahmadi M, et al. (2001) Corticotropin (ACTH) acts directly on amygdala neurons to down-regulate corticotropin-releasing hormone gene expression. Ann Neurol 49: 304-312.
  61. Wang J, Wang J, Zhang Y, et al. (2012) Proteomic analysis of adrenocorticotropic hormone treatment of an infantile spasm model induced by N-methyl-D-aspartic acid and prenatal stress. PLoS One 7: e45347.
  62. Cohen-Sadan S, Kramer U, Ben-Zeev B, et al. (2009) Multicenter long-term follow-up of children with idiopathic West syndrome: ACTH versus vigabatrin. Eur J Neurol 16: 482-487.
  63. Hancock E, Osborne J, Edwards S (2013) Treatment of infantile spasms. Cochrane Database of Sysematic Review 5.
  64. Lux AL, Edwards SW, Hancock E, et al. (2005) The United Kingdom Infantile Spasms Study (UKISS) comparing hormone treatment with vigabatrin on developmental and epilepsy outcomes to age 14 months: A multicentre randomised trial. Lancet Neurol 4: 712-717.
  65. Oguni H, Yanagaki S, Hayashi K, et al. (2006) Extremely low-dose ACTH step-up protocol for West syndrome: Maximum therapeutic effect with minimal side effects. Brain Dev 28: 8-13.
  66. Guevara-Campos J, González de Guevara L (2007) Síndrome de Landau-Kleffner : Análisis de 10 casos en Venezuela. Rev Neurol 44: 652-656.
  67. Okumura A, Tsuji T, Kato T, et al. (2006) ACTH therapy for generalized seizures other than spasms. Seizure 15: 469-475.
  68. Kalra V, Sharma S, Arya R (2009) ACTH Therapy in refractory generalized epilepsy. Indian J Pediatr 76: 91-93.
  69. Medina Malo CH, Guío LV, Uscátegui AM RJ (2008) Experiencia con el uso de hormona adrenocorticotropica (ACTH) en pacientes con encefalopatía epiléptica Neuropediatria Liga Central Contra la Epilepsia. Acta Neurol Colomb 24: 78.
  70. Lehtimäki KA, Liimatainen S, Peltola J, et al. (2011) The serum level of interleukin-6 in patients with intellectual disability and refractory epilepsy. Epilepsy Res 95: 184-187.
  71. Kalra V, Gulati S, Pandey RM, et al. (2002) West syndrome and other infantile epileptic encephalopathies--Indian hospital experience. Brain Dev 24: 130-139.