Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 3108 word(s) 3108 2021-12-17 08:40:17 |
2 update definition and layout + 4 word(s) 3112 2021-12-27 03:18:28 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Ahmad, L. MicroRNAs in Migraine. Encyclopedia. Available online: https://encyclopedia.pub/entry/17550 (accessed on 27 July 2024).
Ahmad L. MicroRNAs in Migraine. Encyclopedia. Available at: https://encyclopedia.pub/entry/17550. Accessed July 27, 2024.
Ahmad, Lara. "MicroRNAs in Migraine" Encyclopedia, https://encyclopedia.pub/entry/17550 (accessed July 27, 2024).
Ahmad, L. (2021, December 24). MicroRNAs in Migraine. In Encyclopedia. https://encyclopedia.pub/entry/17550
Ahmad, Lara. "MicroRNAs in Migraine." Encyclopedia. Web. 24 December, 2021.
MicroRNAs in Migraine
Edit

Preliminary but convergent findings suggest a role for microRNAs (miRNAs) in the generation and maintenance of chronic pain and migraine. Initial observations showed that serum levels of miR-382-5p and miR-34a-5p expression were increased in serum during the migraine attack, with miR-382-5p increasing in the interictal phase as well. By contrast, miR-30a-5p levels were lower in migraine patients compared to healthy controls. Of note, antimigraine treatments proved to be capable of influencing the expression of these miRNAs. Altogether, these observations suggest that miRNAs may represent migraine biomarkers, but several points are yet to be elucidated. A major concern is that these miRNAs are altered in a broad spectrum of painful and non-painful conditions, and thus it is not possible to consider them as truly “migraine-specific” biomarkers.  These miRNAs may represent useful tools to uncover and define different phenotypes across the migraine spectrum with different treatment susceptibilities and clinical features, although further studies are needed to confirm the hypothesis. In this narrative entry , an update and a critical analysis of available data on miRNAs and migraines was provided,  in order to propose possible interpretations. The main objective is to stimulate research in an area that holds promise when it comes to providing reliable biomarkers for theoretical and practical scientific advances.

miRNA biomarker pain headache CGRP

1. Current evidence on miRNA expression in migraine.

Table 1. Current evidence on miRNA expression in migraine.
miRNA Specimen Population Summary of Study Results Reference
miR-382-5p Serum 8 migraine patients without any medication
12 migraine patients with normal medication habits
8 healthy controls
4.1-fold increase expression during migraine attack.
Higher expression during the interictal period in migraine patients vs. healthy subjects.
Andersen et al. (2016) [1]
Peripheral blood mononuclear cells 27 patients with EM
28 patients with CM–MOH
Higher levels in CM–MOH vs. EM.
Positive correlation with CGRP levels.
Greco et al. (2020) [2]
miR-34a-5p Serum 8 migraine patients without any medication
12 migraine patients with normal medication habits
8 healthy controls
9-fold increase expression during migraine attack. Andersen et al. (2016) [1]
Peripheral blood mononuclear cells 27 patients with EM and 28 with CM–MOH Higher levels in CM–MOH vs. EM.
Positive correlation with CGRP levels.
Greco et al. (2020) [2]
miR-30a Serum Patients with migraine with or without aura (sample size not defined) Significant lower expression in patients with migraine vs. healthy controls. Zhai et al. (2018) [3]
Other miRNAs Peripheral blood mononuclear cells 15 female patients with migraine without aura
13 healthy controls
Reduced miR-181a, let-7b and miR-22 levels in EM vs. HC.
Increased miR-27b levels in EM vs. HC.
Tafuri et al. (2015) [4]
Serum 8 migraine patients without any medication
12 migraine patients with normal medication habits
8 healthy controls
Increased miR-29c-5p and miR-26b-3p expression in EM patients. Andersen et al. (2016) [1]
Serum 30 patients with EM
30 healthy controls
Increased expression of miR-155, miR-126, and let-7 in EM vs. HC. Chen et al. (2018) [5]
Legend: EM: episodic migraine. CM: chronic migraine. MOH: medication overuse headache. HC: healthy controls. CGRP: calcitonin gene-related peptide.
Table 2. Treatment driven modifications of miRNAs expression in migraine.
Treatment Posology and Duration Population miRNAs Modifications and Timing Reference
Detoxification 7-day standardized detoxification protocol in hospitalized patients: abrupt withdrawal of overused drugs associated to intravenous therapy twice daily with isotonic 0.9% NaCl saline 500 mL + cyanocobalamin 2500 mcg + folic acid 0.70 mg + nicotinamide 12 mg + ascorbic acid 150 mg + sodic glutathione 600 mg + delorazepam 0.5 mg 28 patients with CM–MOH Significant reduction of miR-382-5p and miR-34a-5p two months after detoxification in CM–MOH. Greco et al. (2020) [2]
Erenumab 70 mg One administration s.c. every 28 days for a total of three administrations 7 patients with CM
33 patients with CM–MOH
Reduction of miR-382-5p and miR-34a-5p in the overall study population after three months of treatment with erenumab.
No differences between 30% Responders and NON-responders after three months of erenumab treatment.
De Icco et al. (2020) [6]
NSAIDs and long-term magnesium Chronic treatment with magnesium (400 mg/day for three months) + Abortive treatment with acetaminophen (15 mg/kg) or ibuprofen (10 mg/kg) 24 children and adolescents affected by migraine without aura divided into two groups (treated, and untreated) and 12 healthy controls Decreased expression of miR-34a-5p in migraine patients treated with NSAIDs and long-term magnesium vs. untreated migraine patients.
Decreased expression in healthy controls vs. untreated migraine patients.
Comparable expression between migraine patients treated with NSAIDs and long-term magnesium and healthy controls.
Gallelli et al. (2019) [7]
Biphasic ketogenic diet Phase 1: 3 weeks with <30 g of carbohydrates
Phase 2: 3 weeks with <120 g of carbohydrates
6 female obese patients with migraine Reduction of has-miR-590-5p and has-miR-660-3p expression after a 6-week ketogenic diet Cannataro et al. (2020) [8]
Legend: CM: chronic migraine. MOH: medication overuse headache. CGRP: calcitonin gene-related peptide. NSAIDs: nonsteroidal anti-inflammatory drugs.

2. miR-382-5p and miR-34a-5p in Migraine

There is mounting evidence that miR-382-5p and miR-34a-5p are dysregulated in migraine (Table 1 and Table 2).
Andersen et al. first detected the aberrant serum expression of 32 miRNAs in migraine patients in 2016 and identified a nine-fold increase in miR-34a-5p expression with a four-fold increase in miR-382-5p expression during the migraine attack when compared to the interictal phase [1]. Intriguingly, they also detected a significant higher miR-382-5p expression in migraine patients during the interictal period when compared to healthy subjects, and thus proposed this miRNA as a possible actor in migraine pathophysiology and a specific biomarker of the disease [1]. A main limitation of the study was that the migraine population was not well characterized, thus it was not possible to correlate the results obtained with the migraine phenotype (episodic or chronic) and with headache features.
A previously suggested hypothesis was that miR-382-5p serum expression was explained by increased blood–brain barrier permeability, which was suggested to occur during the migraine attack in a pre-clinical model [9], but this hypothesis has never been confirmed in humans and needs further confirmation.
Previous studies revealed that both miR-382-5p and miR-34a-5p negatively modulate target genes linked to GABAergic facilitating signaling, a neurotransmitter involved in trigeminal pain analgesia [1]. This may represent a protective compensatory mechanism in migraine self-limitation; indeed, higher GABA levels have been detected in the cerebrospinal fluid and in the saliva during migraine attacks [10][11].
miR-382-5p is also involved in Interleukin (IL)-10 anti-inflammatory signaling, where it acts via a negative modulation of the interleukin 10 receptor alpha subunit (IL-10RA), an endogenous inhibitor of pro-inflammatory IL-1β signaling [1].
The seminal work of Gallelli et al. paved the way for the identification of miRNAs as potential peripheral biological markers of drug response. They demonstrated lower miR-34a-5p and miR-375-5p expression in serum and saliva of children and adolescents treated with magnesium and acute nonsteroidal anti-inflammatory drugs (NSAIDs) compared to untreated subjects with migraine without aura [7]. It is noteworthy that in the first cohort of patients, the expression of these specific miRNAs was comparable to a group of healthy controls [7].
A recent study specifically analyzed miR-382-5p and miR-34a-5p expression in subjects with EM or CM and MOH (CM–MOH) [2]. The authors described a significantly higher interictal expression of miR-34a-5p and miR-382-5p in the plasma of CM–MOH subjects [2]. This analysis detected for the first time the association between specific miRNAs and the migraine phenotype as well as disease severity [2]. Notably, in this study CGRP plasma levels positively correlated with miR-382-5p and miR-34a-5p expression [2].
Although analyzed as a secondary endpoint, miR-382-5p and miR-34a-5p expression was reduced two months after detoxification from acute medications (in-hospital detoxification program), regardless of the clinical outcome [2]. This observation suggests the possibility of a direct role for acute medications in the expression of both miRNAs.
miR-382-5p and miR-34a-5p were also negatively modulated by erenumab, a monoclonal antibody directed against the CGRP receptor, in a population of difficult-to-treat CM patients with and without MOH [6]. The expression of each miRNA was reduced after three monthly s.c. administrations of erenumab, irrespective of the clinical outcome, which suggests a direct role for CGRP in miRNA regulation [6]. By contrast, central sensitization improved only in migraine patients who responded to erenumab treatment with an at least 30% reduction in monthly migraine days [6].

3. miR-382-5p and miR-34a-5p in Pain and Putative Mechanisms

miR-34a expression was reported to be altered in several human diseases besides migraine, including osteoarthritis [12][13], Alzheimer’s disease [14], cancer [15][16] and leukemia [17]. The connection between miR-34a and these pathologies resides in its strong contribution to the regulation of cell cycles and apoptosis, with miR-34a being a major transcriptional target of p53 [18][19]. The involvement of miR-34a in several painful and non-painful conditions clearly suggests that, when considered alone, it cannot represent a migraine-specific biomarker. Nonetheless, it may still play a role as part of a multi-biomarker panel signature of migraine [20].
Regarding pain-related disorders, miR-34a plays a role in the modulation of the inflammatory response [21][22]. Upregulation of mir-34a has been reported in osteoarthritic tissues specimens and in patients with low back pain related to intervertebral disc degeneration [23][24][25]. miR-34a expression was enhanced by IL-1β release [12], although miR-34a itself can up-regulate the IL-1β/cyclooxygenase 2 (COX-2)/Prostaglandin E2 (PEG2) inflammation pathway [26] by targeting the deacetylase silencing information regulator 1 (SIRT1) [26][27][28]. miR-34a also inhibits SIRT1 by increasing the acetylation of the nuclear factor kappa B (NF-κB) P65 subunit, ultimately enhancing the transcription of inflammatory mediators, including IL-1β, IL-6 and tumor necrosis factor-α [27][29]. In an animal model of inflammatory pain, Chen et al. reported an increase in miR-34a expression in the spinal cord and showed that its inhibition by a miR-34a antagonist exerted analgesic effects by increasing spinal SIRT1 expression [28]. The inhibition of SIRT1 was reported to enhance the release of CGRP in rat trigeminal ganglia in vitro, probably as a consequence of IL-1β/COX-2/PGE2 pathway activation [26]. Other genes coding for proteins that are involved in chronic pain pathways have been validated as targets of miR-34a, such as sodium voltage-gated channel beta subunit 2 (Scn2b) and vesicle-associated membrane protein 2 (VAMP2) [30]. This latter belongs to the SNARE (soluble N-ethyl-maleimide-sensitive factor attachment protein receptor) protein complex that regulates presynaptic neurotransmitter release as well as neurotransmitter receptor trafficking and synaptic plasticity [31], whose variants have been reported to be linked with migraine susceptibility [32]. It must be noted that miR-34a was found to be down-regulated in rats’ dorsal root ganglions 12 days after the experimental induction of neuropathic pain, with the concomitant up-regulation of VAMP2 [30]. Since the increase of synaptic proteins is related to higher glutamate release [33], this may ultimately lead to an enhancement of nociception. Nonetheless, miR-34a was predicted to target the genes coding for specific GABA receptors subunits (GABBR2, Gamma-aminobutyric acid type B receptor subunit 2; GABRA3, Gamma-aminobutyric acid receptor subunit alpha-3) [1]. In this context, an up-regulation of this microRNA, as reported in serum of migraine patients, could contribute to the reduction of the GABA inhibitory signal [1]. Although some controversies exist, it is reasonable to assert that the role of miR-34a may vary across different pain disorders according to the time of evaluation and, more importantly, depending on specimen collection. However, together these observations strongly point towards the contribution of miR-34a to the regulation of pain-related neurotransmission.
Like miR-34a, miR-382-5p was predicted to target the GABRA5 (Gamma-aminobutyric acid receptor subunit alpha-5) GABA receptor subunit and also IL-10RA [1]. Thus, the observed up-regulation in the serum levels of miR-382-5p in migraine patients may be explained by reduced GABAergic and anti-inflammatory signaling [1][34][35]. The increased miR-382-5p serum levels in migraine patients may be also related to high protein levels of transforming growth factor β (TGF-β). In agreement with this hypothesis, increased TGF-β levels were found in the plasma and cerebrospinal fluid of patients with EM [36][37]. TGF-β is able to increase the expression of miR-382-5p [38][39][40] by inhibiting transcription factors’ repressors [41]. TGF-β modulated miR-382-5p in vitro (human CD34+ stem cells), resulting in the increased accumulation of reactive oxygen species (ROS). ROS accumulation was explained by miR-382-5p-induced inhibition of superoxide dismutase 2 (SOD2), a direct target of this mRNA [39][40]. This latter issue appears to be relevant in the context of migraine pain, since oxidative stress and mitochondrial dysfunction was suggested as actors in migraine pathophysiology [42][43][44]. miR-382-5p regulation is also influenced by the long non-coding RNA Ftx (lncRNA Ftx), which exerts its action as a competing endogenous RNA (ceRNA), competing with miR-382-5p at targets level [45]. An increase in miR-382-5p expression together with a down-regulation of lncRNA Ftx and neuregulin-1 (Nrg-1) was reported in an animal model of neuropathic pain [45]. The lack of inhibition exerted by lncRNA Ftx on miR-382, is supposed to increase the inhibition of Nrg-1, thus facilitating pain and inflammation [45]. The involvement of Nrg-1 in pain was also reported in an animal model of inflammatory pain. Its reduction has been found in the dorsal root ganglia and spinal cord of mice, contributing to hypersensitivity [46]. Notably, increasing Nrg-1 expression facilitates analgesia and tissue repair by modulating glial cells proliferation, central nervous system injury repair and the reduction of the release of pro-inflammatory cytokine [45][47][48][49]. Increased miR-382-5p expression in pain conditions may also be related to nuclear factor kappa B (NF-κB) signaling, implicated in migraine pain [50][51][52]. NF-κB acts as a transcription factor for many miRNAs [3], including miR-382 [4]. Indeed, the inhibition of NF-κB significantly suppressed miR-382 in vitro [4].
These observations suggest that the activation of the inflammatory pathways may lead to an up-regulation of miR-34a and miR-382 which in turn modulate the release of inflammatory and pain mediators (Figure 1).
Figure 1. Schematic representation of putative mechanisms through which miR-34a, miR-382 and miR-30a interfere with pain. Legend: in pain conditions, miR-34a and miR-382 were upregulated, while miR30a expression was downregulated. Other molecules are also implicated in the inhibition or enhancement of miRNA expression in pain disorders. The physiological activity of miRNAs is the downregulation of target mRNAs, thus the final effects depend on the role of the mRNAs involved. High levels of miR-34a and miR-382 promote the inhibition of pathways that reduce pain (e.g., GABA signaling). These miRNAs may also downregulate up-stream gene regulators, which in physiological conditions would have blocked pain pathways (e.g., SIRT1). The downregulation of miR30a in pain conditions leads to a reduction of its inhibitory effects. This would lead to an increase in pain-related pathways, since miR-30a targets genes that facilitate pain (like Calca). miRNA = microRNA; mRNA = messenger RNA; BDNF = brain-derived neurotrophic factor; Becn1 = Beclin 1 gene; Calca = Calcitonin related polypeptide alpha gene; CGRP = calcitonin gene-related peptide; COX-2 = cyclooxygenase 2; EP300 = E1A Binding Protein P300; GABBR2 = Gamma-aminobutyric acid type B receptor subunit 2; GABRA3 = Gamma-aminobutyric acid receptor subunit alpha-3; GABRA5 = Gamma-aminobutyric acid receptor subunit alpha-5; IL-10RA = interleukin 10 receptor alpha subunit; IL-1β = Interleukin 1β; IL-6 = Interleukin 6; lncRNA Ftx = long non-coding RNA Ftx; NF-κB = nuclear factor kappa B; Nrg-1 = neuregulin-1; PGE2 = Prostaglandin E2; PI3K = phosphatidyl Inositol 3-kinase; ROS = reactive oxygen species; Scn2b = voltage-gated channel beta subunit 2; SIRT1 = silencing information regulator 1; SNARE = soluble N-ethyl-maleimide-sensitive factor attachment protein receptor; SOD2 = superoxide dismutase 2; STAT3 = Signal transducer and activator of transcription 3; TET1 = ten-eleven translocation methylcytosine dioxygenase 1; TGF-β =Transforming growth factor β; TNF-α = tumor necrosis factor-α; VAMP2 = vesicle-associated membrane protein 2.

4. Role of miR-30a in Migraine and Pain

Like many other miRNAs, miR-30a can target several different genes [53][54][55][56], of which many are connected with pathways related with pain transmission/modulation. Zhai and Zhu reported lower serum miR-30a levels in migraine patients compared to healthy subjects [3] (Table 1 and Table 2). Interestingly, the authors also showed that miR-30a overexpression can degrade calcitonin/alpha-CGRP (Calca) gene in vitro, thus reducing CGRP levels [3].
TGF-β is known to downregulate miR-30a [57][58]. Given that high protein levels of TGF-β were found in migraine patients [34][35], it is worth speculating that, in contrast to what happens with miR-382, the increased TGF-β levels might negatively regulate miR-30a expression. This would lead to increased Calca expression and, consequently, to augmented CGRP levels. This pathway appears to be relevant for migraine, as it was hypothesized that Calca degradation may represent a protective mechanism in migraine progression [3].
In line with the above-mentioned observation, low levels of miR-30a were also found in animal models of neuropathic pain [59][60]. The involvement of miR-30a in pain modulation resides in its ability to regulate different pathways contributing to pain. For instance, miR-30a directly binds phosphatidyl Inositol 3-kinase (PI3K) [61] and Becn1 [62], the gene coding for Beclin 1, a core subunit of the PI3K complex. Up-regulation of miR-30a inhibits the PI3K/Akt/mTOR signaling pathway, thus resulting in a reduced central sensitization and pain relief [63][64][65]. In addition, miR-30a targets ten-eleven translocation methylcytosine dioxygenase 1 (TET1) [66], which is involved in the DNA demethylation of some genes linked to pain hypersensitivity, like brain-derived neurotrophic factor (BDNF) [67][68]. Accordingly, miR-30a levels were also found to be reduced in rats with spinal cord injuries, thus confirming the hypothesis that miR-30a may have a protective role when it comes to neuropathic pain [60].
Taken together, these findings support the role of miR-30a in pain modulation and suggest that a down-regulation of miR-30a in pain conditions may lead to an up-regulation of different pro-nociceptive effectors possibly involved in migraine pain. The overexpression of miR-30a may be a useful pathway to counteract pain in general and possibly also in migraine pain (Figure 1).

Conclusion

 The initial evidence prompts a possible role for miRNA profiling in migraine research. Clinical data on miR-382-5p, miR-34a-5p and miR-30a seem promising, but their scientific strength is limited by the lack of consistency across studies due to: (i) inhomogeneity or poor definition of migraine cohorts (EM, CM with and without MOH) and related comorbidities, (ii) small sample sizes and (iii) different types of specimens (e.g., blood, saliva, CSF) collected for miRNAs analysis. Further studies are therefore needed to confirm and expand on the role of miRNAs in migraine pathophysiology and management. In the future, it will be important to tackle the different components of migraine in their dynamic manifestations. Indeed, migraine is a complex disorder characterized by recurring attacks, each defined by a sequence of phases: a prodromal phase, a pain phase either preceded or not by aura symptoms and a postdrome phase. The prodromal and postdrome phases have attracted the attention of the scientific community as they may represent the true timeframe for the study of the mechanisms involved in the origin and cessation of the migraine attack. The availability of reliable and validated human migraine models will facilitate the study of miRNA expression in the different phases of the migraine cycle, prompting the possibility to set experimentally controlled conditions.

References

  1. Andersen, H.H.; Duroux, M.; Gazerani, P. Serum MicroRNA Signatures in Migraineurs During Attacks and in Pain-Free Periods. Mol. Neurobiol. 2016, 53, 1494–1500.
  2. Greco, R.; De Icco, R.; Demartini, C.; Zanaboni, A.M.; Tumelero, E.; Sances, G.; Allena, M.; Tassorelli, C. Plasma levels of CGRP and expression of specific microRNAs in blood cells of episodic and chronic migraine subjects: Towards the identification of a panel of peripheral biomarkers of migraine? J. Headache Pain 2020, 21, 122.
  3. Zhai, Y.; Zhu, Y.Y. MiR-30a relieves migraine by degrading CALCA. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 2022–2028.
  4. Tafuri, E.; Santovito, D.; De Nardis, V.; Marcantonio, P.; Paganelli, C.; Affaitati, G.; Bucci, M.; Mezzetti, A.; Giamberardino, M.A.; Cipollone, F. MicroRNA profiling in migraine without aura: Pilot study. Ann. Med. 2015, 47, 468–473.
  5. Cheng, C.Y.; Chen, S.P.; Liao, Y.C.; Fuh, J.L.; Wang, Y.F.; Wang, S.J. Elevated circulating endothelial-specific microRNAs in migraine patients: A pilot study. Cephalalgia 2018, 38, 1585–1591.
  6. De Icco, R.; Fiamingo, G.; Greco, R.; Bottiroli, S.; Demartini, C.; Zanaboni, A.M.; Allena, M.; Guaschino, E.; Martinelli, D.; Putortì, A.; et al. Neurophysiological and biomolecular effects of erenumab in chronic migraine: An open label study. Cephalalgia 2020, 40, 1336–1345.
  7. Gallelli, L.; Cione, E.; Peltrone, F.; Siviglia, S.; Verano, A.; Chirchiglia, D.; Zampogna, S.; Guidetti, V.; Sammartino, L.; Montana, A.; et al. Hsa-miR-34a-5p and hsa-miR-375 as Biomarkers for Monitoring the Effects of Drug Treatment for Migraine Pain in Children and Adolescents: A Pilot Study. J. Clin. Med. 2019, 8, 928.
  8. Cannataro, R.; Caroleo, M.C.; Siniscalchi, A.; Gallelli, L.; De Sarro, G.; Cione, E. Ketogenic Diet Modifies the Expression of MicroRNAs Linked to Migraine. Acta Sci. Nutr. Health 2020, 4, 34–41.
  9. Yamanaka, G.; Suzuki, S.; Morishita, N.; Takeshita, M.; Kanou, K.; Takamatsu, T.; Suzuki, S.; Morichi, S.; Watanabe, Y.; Ishida, Y.; et al. Role of neuroinflammation and blood-brain barrier permutability on migraine. Int. J. Mol. Sci. 2021, 22, 8929.
  10. Welch, K.M.A.; Chabi, E.V.A.; Bartosh, K.; Achar, V.S.; Meyer, J.S. Cerebrospinal Fluid Y Aminobutyric Acid Levels in Migraine. BMJ 1975, 516–517.
  11. Marukawa, H.; Shimomura, T.; Takahashi, K. Salivary substance P, 5-hydroxytryptamine, and γ-aminobutyric acid levels in migraine and tension-type headache. Headache 1996, 36, 100–104.
  12. Abouheif, M.M.; Nakasa, T.; Shibuya, H.; Niimoto, T.; Kongcharoensombat, W.; Ochi, M. Silencing microRNA-34a inhibits chondrocyte apoptosis in a rat osteoarthritis model in vitro. Rheumatology 2010, 49, 2054–2060.
  13. Endisha, H.; Datta, P.; Sharma, A.; Nakamura, S.; Rossomacha, E.; Younan, C.; Ali, S.A.; Tavallaee, G.; Lively, S.; Potla, P.; et al. MicroRNA-34a-5p Promotes Joint Destruction During Osteoarthritis. Arthritis Rheumatol. 2021, 73, 426–439.
  14. Cosín-Tomás, M.; Antonell, A.; Lladó, A.; Alcolea, D.; Fortea, J.; Ezquerra, M.; Lleó, A.; Martí, M.J.; Pallàs, M.; Sanchez-Valle, R.; et al. Plasma miR-34a-5p and miR-545-3p as Early Biomarkers of Alzheimer’s Disease: Potential and Limitations. Mol. Neurobiol. 2017, 54, 5550–5562.
  15. Tazawa, H.; Tsuchiya, N.; Izumiya, M.; Nakagama, H. Tumor-suppressive miR-34a induces senescence-like growth arrest through modulation of the E2F pathway in human colon cancer cells. Proc. Natl. Acad. Sci. USA 2007, 104, 15472–15477.
  16. Zarone, M.R.; Misso, G.; Grimaldi, A.; Zappavigna, S.; Russo, M.; Amler, E.; Di Martino, M.T.; Amodio, N.; Tagliaferri, P.; Tassone, P.; et al. Evidence of novel miR-34a-based therapeutic approaches for multiple myeloma treatment. Sci. Rep. 2017, 7, 17949.
  17. Zanette, D.L.; Rivadavia, F.; Molfetta, G.A.; Barbuzano, F.G.; Proto-Siqueira, R.; Falcão, R.P.; Zago, M.A.; Silva, W.A. miRNA expression profiles in chronic lymphocytic and acute lymphocytic leukemia. Braz. J. Med. Biol. Res. 2007, 40, 1435–1440.
  18. Raver-Shapira, N.; Marciano, E.; Meiri, E.; Spector, Y.; Rosenfeld, N.; Moskovits, N.; Bentwich, Z.; Oren, M. Transcriptional Activation of miR-34a Contributes to p53-Mediated Apoptosis. Mol. Cell 2007, 26, 731–743.
  19. Chang, T.C.; Wentzel, E.A.; Kent, O.A.; Ramachandran, K.; Mullendore, M.; Lee, K.H.; Feldmann, G.; Yamakuchi, M.; Ferlito, M.; Lowenstein, C.J.; et al. Transactivation of miR-34a by p53 Broadly Influences Gene Expression and Promotes Apoptosis. Mol. Cell 2007, 26, 745–752.
  20. Ashina, M.; Terwindt, G.M.; Al-Karagholi, M.A.M.; de Boer, I.; Lee, M.J.; Hay, D.L.; Schulte, L.H.; Hadjikhani, N.; Sinclair, A.J.; Ashina, H.; et al. Migraine: Disease characterisation, biomarkers, and precision medicine. Lancet 2021, 397, 1496–1504.
  21. Jiang, P.; Liu, R.; Zheng, Y.; Liu, X.; Chang, L.; Xiong, S.; Chu, Y. MiR-34a inhibits lipopolysaccharide-induced inflammatory response through targeting Notch1 in murine macrophages. Exp. Cell Res. 2012, 318, 1175–1184.
  22. Mathé, E.; Nguyen, G.H.; Funamizu, N.; He, P.; Moake, M.; Croce, C.M.; Hussain, S.P. Inflammation regulates microRNA expression in cooperation with p53 and nitric oxide. Int. J. Cancer 2012, 131, 760–765.
  23. Tian, F.; Wang, J.; Zhang, Z.; Yang, J. LncRNA SNHG7/miR-34a-5p/SYVN1 axis plays a vital role in proliferation, apoptosis and autophagy in osteoarthritis. Biol. Res. 2020, 53, 9.
  24. Yan, S.; Wang, M.; Zhao, J.; Zhang, H.; Zhou, C.; Jin, L.; Zhang, Y.; Qiu, X.; Ma, B.; Fan, Q. MicroRNA-34a affects chondrocyte apoptosis and proliferation by targeting the SIRT1/p53 signaling pathway during the pathogenesis of osteoarthritis. Int. J. Mol. Med. 2016, 38, 201–209.
  25. Brinjikji, W.; Diehn, F.E.; Jarvik, J.G.; Carr, C.M.; Kallmes, D.F.; Murad, M.H.; Luetmer, P.H. MRI findings of disc degeneration are more prevalent in adults with low back pain than in asymptomatic controls: A systematic review and meta-analysis. Am. J. Neuroradiol. 2015, 36, 2394–2399.
  26. Zhang, H.; Zhang, X.; Zong, D.; Ji, X.; Jiang, H.; Zhang, F.; He, S. miR-34a-5p up-regulates the IL-1β/COX2/PGE2 inflammation pathway and induces the release of CGRP via inhibition of SIRT1 in rat trigeminal ganglion neurons. FEBS Open Bio 2021, 11, 300–311.
  27. Zhang, H.; He, S.D.; Zong, D.D.; Zhang, X.M.; Luo, J.; Zheng, J.K. Effects of electroacupuncture on miR-34a-5p/SIRT1 signaling in the trigeminal ganglion of rats with migraine. Zhen Ci Yan Jiu 2020, 45, 868–874.
  28. Chen, S.; Gu, Y.; Dai, Q.; He, Y.; Wang, J. Spinal miR-34a regulates inflammatory pain by targeting SIRT1 in complete Freund’s adjuvant mice. Biochem. Biophys. Res. Commun. 2019, 516, 1196–1203.
  29. Yang, H.; Zhang, W.; Pan, H.; Feldser, H.G.; Lainez, E.; Miller, C.; Leung, S.; Zhong, Z.; Zhao, H.; Sweitzer, S.; et al. SIRT1 Activators Suppress Inflammatory Responses through Promotion of p65 Deacetylation and Inhibition of NF-κB Activity. PLoS ONE 2012, 7, e46364.
  30. Brandenburger, T.; Johannsen, L.; Prassek, V.; Kuebart, A.; Raile, J.; Wohlfromm, S.; Köhrer, K.; Huhn, R.; Hollmann, M.W.; Hermanns, H. MiR-34a is differentially expressed in dorsal root ganglia in a rat model of chronic neuropathic pain. Neurosci. Lett. 2019, 708, 134365.
  31. Madrigal, M.P.; Portalés, A.; SanJuan, M.P.; Jurado, S. Postsynaptic SNARE Proteins: Role in Synaptic Transmission and Plasticity. Neuroscience 2019, 420, 12–21.
  32. Quintas, M.; Neto, J.L.; Sequeiros, J.; Sousa, A.; Pereira-Monteiro, J.; Lemos, C.; Alonso, I. Going Deep into Synaptic Vesicle Machinery Genes and Migraine Susceptibility—A Case-Control Association Study. Headache 2020, 60, 2152–2165.
  33. Hung, K.L.; Wang, S.J.; Wang, Y.C.; Chiang, T.R.; Wang, C.C. Upregulation of presynaptic proteins and protein kinases associated with enhanced glutamate release from axonal terminals (synaptosomes) of the medial prefrontal cortex in rats with neuropathic pain. Pain 2014, 155, 377–387.
  34. Ishizaki, K.; Takeshima, T.; Fukuhara, Y.; Araki, H.; Nakaso, K.; Kusumi, M.; Nakashima, K. Increased plasma transforming growth factor-β1 in migraine. Headache 2005, 45, 1224–1228.
  35. Bø, S.H.; Davidsen, E.M.; Gulbrandsen, P.; Dietrichs, E.; Bovim, G.; Stovner, L.J.; White, L.R. Cerebrospinal fluid cytokine levels in migraine, tension-type headache and cervicogenic headache. Cephalalgia 2009, 29, 365–372.
  36. Lin, C.A.; Duan, K.Y.; Wang, X.W.; Zhang, Z.S. Study on the role of Hsa-miR-382-5p in epidural fibrosis. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 3663–3668.
  37. Rossi, C.; Zini, R.; Rontauroli, S.; Ruberti, S.; Prudente, Z.; Barbieri, G.; Bianchi, E.; Salati, S.; Genovese, E.; Bartalucci, N.; et al. Role of TGF-β1/miR-382-5p/SOD2 axis in the induction of oxidative stress in CD34+ cells from primary myelofibrosis. Mol. Oncol. 2018, 12, 2102–2123.
  38. Kriegel, A.J.; Fang, Y.; Liu, Y.; Tian, Z.; Mladinov, D.; Matus, I.R.; Ding, X.; Greene, A.S.; Liang, M. MicroRNA-target pairs in human renal epithelial cells treated with transforming growth factor β1: A novel role of miR-382. Nucleic Acids Res. 2010, 38, 8338–8347.
  39. Kato, M.; Zhang, J.; Wang, M.; Lanting, L.; Yuan, H.; Rossi, J.J.; Natarajan, R. MicroRNA-192 in diabetic kidney glomeruli and its function in TGF-β-induced collagen expression via inhibition of E-box repressors. Proc. Natl. Acad. Sci. USA 2007, 104, 3432–3437.
  40. Yorns, W.R.; Hardison, H.H. Mitochondrial dysfunction in migraine. Semin. Pediatr. Neurol. 2013, 20, 188–193.
  41. Borkum, J.M. Migraine Triggers and Oxidative Stress: A Narrative Review and Synthesis. Headache 2016, 56, 12–35.
  42. Li, R.; Liu, Y.; Chen, N.; Zhang, Y.; Song, G.; Zhang, Z. Valproate attenuates nitroglycerin-induced trigeminovascular activation by preserving mitochondrial function in a Rat model of migraine. Med. Sci. Monit. 2016, 22, 3229–3237.
  43. Xiang, W.; Jiang, L.; Zhou, Y.; Li, Z.; Zhao, Q.; Wu, T.; Cao, Y.; Zhou, J. The lncRNA Ftx/miR-382-5p/Nrg1 axis improves the inflammation response of microglia and spinal cord injury repair. Neurochem. Int. 2021, 143, 104929.
  44. Wan, C.; Xu, Y.; Cen, B.; Xia, Y.; Yao, L.; Zheng, Y.; Zhao, J.; He, S.; Chen, Y. Neuregulin1-ErbB4 Signaling in Spinal Cord Participates in Electroacupuncture Analgesia in Inflammatory Pain. Front. Neurosci. 2021, 15, 636348.
  45. Kataria, H.; Alizadeh, A.; Karimi-Abdolrezaee, S. Neuregulin-1/ErbB network: An emerging modulator of nervous system injury and repair. Prog. Neurobiol. 2019, 180, 101643.
  46. Wang, G.; Dai, D.; Chen, X.; Yuan, L.; Zhang, A.; Lu, Y.; Zhang, P. Upregulation of neuregulin-1 reverses signs of neuropathic pain in rats. Int. J. Clin. Exp. Pathol. 2014, 7, 5916–5921.
  47. Alizadeh, A.; Dyck, S.M.; Kataria, H.; Shahriary, G.M.; Nguyen, D.H.; Santhosh, K.T.; Karimi-Abdolrezaee, S. Neuregulin-1 positively modulates glial response and improves neurological recovery following traumatic spinal cord injury. Glia 2017, 65, 1152–1175.
  48. Reuter, U.; Chiarugi, A.; Bolay, H.; Moskowitz, M.A. Nuclear factor-κB as a molecular target for migraine therapy. Ann. Neurol. 2002, 51, 507–516.
  49. Greco, R.; Tassorelli, C.; Cappelletti, D.; Sandrini, G.; Nappi, G. Activation of the Transcription Factor NF-κB in the nucleus trigeminalis caudalis in an animal model of migraine. Neurotoxicology 2005, 26, 795–800.
  50. Sarchielli, P.; Floridi, A.; Mancini, M.L.; Rossi, C.; Coppola, F.; Baldi, A.; Pini, L.A.; Calabresi, P. NF-κB activity and iNOS expression in monocytes from internal jugular blood of migraine without aura patients during attacks. Cephalalgia 2006, 26, 1071–1079.
  51. Novák, J.; Olejníčková, V. microRNA: Basic Science; Springer International Publishing: Cham, Switzerland, 2015; Volume 887, pp. 79–100.
  52. Wang, X.; Xue, N.; Zhao, S.; Shi, Y.; Ding, X.; Fang, Y. Upregulation of miR-382 contributes to renal fibrosis secondary to aristolochic acid-induced kidney injury via PTEN signaling pathway. Cell Death Dis. 2020, 11, 620.
  53. Zhu, Q.; Li, H.; Li, Y.; Jiang, L. MicroRNA-30a functions as tumor suppressor and inhibits the proliferation and invasion of prostate cancer cells by down-regulation of SIX1. Hum. Cell 2017, 30, 290–299.
  54. Shepard, A.; Hoxha, S.; Troutman, S.; Harbaugh, D.; Kareta, M.S.; Kissil, J.L. Transcriptional regulation of miR-30a by YAP impacts PTPN13 and KLF9 levels and Schwann cell proliferation. J. Biol. Chem. 2021, 297, 100962.
  55. Li, J.; Xie, Y.; Li, L.; Li, X.; Shen, L.; Gong, J.; Zhang, R. MicroRNA-30a Modulates Type I Interferon Responses to Facilitate Coxsackievirus B3 Replication Via Targeting Tripartite Motif Protein 25. Front. Immunol. 2021, 11, 603437.
  56. Li, Y.; Zhang, J.; Liu, Y.; Zhang, B.; Zhong, F.; Wang, S.; Fang, Z. MiR-30a-5p confers cisplatin resistance by regulating IGF1R expression in melanoma cells. BMC Cancer 2018, 18, 404.
  57. Shi, S.; Yu, L.; Zhang, T.; Qi, H.; Xavier, S.; Ju, W.; Bottinger, E. Smad2-Dependent Downregulation of miR-30 Is Required for TGF-β-Induced Apoptosis in Podocytes. PLoS ONE 2013, 8, e75572.
  58. Volkmann, I.; Kumarswamy, R.; Pfaff, N.; Fiedler, J.; Dangwal, S.; Holzmann, A.; Batkai, S.; Geffers, R.; Lother, A.; Hein, L.; et al. MicroRNA-mediated epigenetic silencing of sirtuin1 contributes to impaired angiogenic responses. Circ. Res. 2013, 113, 997–1003.
  59. Chen, H.; Wang, Y.; Xu, Y.; Wang, G.N. Overexpression of miR-30a attenuates neuropathic pain by targeting SOCS1 in rats with chronic constriction injury. Int. J. Clin. Exp. Pathol. 2016, 9, 1258–1266.
  60. Tan, M.; Shen, L.; Hou, Y. Epigenetic modification of BDNF mediates neuropathic pain via miR-30a-3p/EP300 axis in CCI rats. Biosci. Rep. 2020, 40, BSR20194442.
  61. Zhong, M.; Bian, Z.; Wu, Z. MiR-30a suppresses cell migration and invasion through downregulation of PIK3CD in colorectal carcinoma. Cell. Physiol. Biochem. 2013, 31, 209–218.
  62. Zhang, L.; Cheng, R.; Huang, Y. MiR-30a inhibits BECN1-mediated autophagy in diabetic cataract. Oncotarget 2017, 8, 77360–77368.
  63. Chen, S.-P.; Zhou, Y.-Q.; Liu, D.-Q.; Zhang, W.; Manyande, A.; Guan, X.-H.; Tian, Y.; Ye, D.-W.; Omar, D.M. PI3K/Akt Pathway: A Potential Therapeutic Target for Chronic Pain. Curr. Pharm. Des. 2017, 23, 1860–1868.
  64. Liu, W.; Lv, Y.; Ren, F. PI3K/Akt Pathway is Required for Spinal Central Sensitization in Neuropathic Pain. Cell. Mol. Neurobiol. 2018, 38, 747–755.
  65. Guo, J.-R.; Wang, H.; Jin, X.-J.; Jia, D.-L.; Zhou, X.; Tao, Q. Effect and mechanism of inhibition of PI3K/Akt/mTOR signal pathway on chronic neuropathic pain and spinal microglia in a rat model of chronic constriction injury. Oncotarget 2017, 8, 52923–52934.
  66. Zhang, S.; Liu, H.; Liu, Y.; Zhang, J.; Li, H.; Liu, W.; Cao, G.; Xv, P.; Zhang, J.; Lv, C.; et al. miR-30a as potential therapeutics by targeting tet1 through regulation of Drp-1 promoter hydroxymethylation in idiopathic pulmonary fibrosis. Int. J. Mol. Sci. 2017, 18, 633.
  67. Hsieh, M.C.; Lai, C.Y.; Ho, Y.C.; Wang, H.H.; Cheng, J.K.; Chau, Y.P.; Peng, H.Y. Tet1-dependent epigenetic modification of BDNF expression in dorsal horn neurons mediates neuropathic pain in rats. Sci. Rep. 2016, 6, 37411.
  68. Pan, Z.; Xue, Z.Y.; Li, G.F.; Sun, M.L.; Zhang, M.; Hao, L.Y.; Tang, Q.Q.; Zhu, L.J.; Cao, J.L. DNA Hydroxymethylation by Ten-eleven Translocation Methylcytosine Dioxygenase 1 and 3 Regulates Nociceptive Sensitization in a Chronic Inflammatory Pain Model. Anesthesiology 2017, 127, 147–163.
More
Information
Subjects: Neurosciences
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 354
Revisions: 2 times (View History)
Update Date: 29 Mar 2022
1000/1000
Video Production Service