MERTK in the rat trigeminal system: a potential novel target for cluster headache?

Headache Classification Committee of the International Headache Society (IHS) (2018) The International classification of Headache disorders. 3rd edition. Cephalalgia 38(1):1–211

Article  Google Scholar 

Fischera M, Marziniak M, Gralow I, Evers S (2008) The incidence and prevalence of cluster headache: a meta-analysis of population-based studies. Cephalalgia 28(6):614–618

Article  CAS  PubMed  Google Scholar 

Carmine Belin A, Ran C, Edvinsson L (2020) Calcitonin gene-related peptide (CGRP) and cluster headache. Brain Sci 10(1):30

Article  PubMed  PubMed Central  Google Scholar 

Hoffmann J, May A (2018) Diagnosis, pathophysiology, and management of cluster headache. Lancet Neurol 17(1):75–83

Article  PubMed  Google Scholar 

Nesbitt AD, Goadsby PJ (2012) Cluster headache. BMJ. 344

Belin AC, Barloese MC (2023) The genetics and chronobiology of cluster headache. Cephalalgia 43(10):03331024231208126

Article  Google Scholar 

Edvinsson J, Viganò A, Alekseeva A, Alieva E, Arruda R, De Luca C et al (2020) The fifth cranial nerve in headaches. J Headache Pain 21:1–17

Article  Google Scholar 

Liu Y, Broman J, Zhang M, Edvinsson L (2009) Brainstem and thalamic projections from a craniovascular sensory nervous centre in the rostral cervical spinal dorsal horn of rats. Cephalalgia 29(9):935–948

Article  CAS  PubMed  Google Scholar 

Russo AF, Hay DL (2023) CGRP physiology, pharmacology, and therapeutic targets: migraine and beyond. Physiol Rev 103(2):1565–1644

Article  CAS  PubMed  Google Scholar 

Messlinger K, Balcziak LK, Russo AF (2020) Cross-talk signaling in the trigeminal ganglion: role of neuropeptides and other mediators. J Neural Transm 127:431–444

Article  PubMed  Google Scholar 

O’Connor E, Fourier C, Ran C, Sivakumar P, Liesecke F, Southgate L et al (2021) Genome-wide association study identifies risk loci for cluster headache. Ann Neurol 90(2):193–202

Article  PubMed  Google Scholar 

Harder AVE, Winsvold BS, Noordam R, Vijfhuizen LS, Børte S, Kogelman LJA et al (2021) Genetic Susceptibility Loci in Genomewide Association Study of Cluster Headache. Ann Neurol 90(2):203–216

Article  CAS  PubMed  PubMed Central  Google Scholar 

Chen SP, Hsu CL, Wang YF, Yang FC, Chen TH, Huang JH et al (2022) Genome-wide analyses identify novel risk loci for cluster headache in Han Chinese residing in Taiwan. J Headache Pain 23(1):147

Article  PubMed  PubMed Central  Google Scholar 

Winsvold BS, Harder AVE, Ran C, Chalmer MA, Dalmasso MC, Ferkingstad E et al (2023) Cluster headache genomewide association study and meta-analysis identifies eight loci and implicates smoking as causal risk factor. Ann Neurol 94(4):713–726

Article  CAS  PubMed  PubMed Central  Google Scholar 

Duncan JL, LaVail MM, Yasumura D, Matthes MT, Yang H, Trautmann N et al (2003) An RCS-like retinal dystrophy phenotype in mer knockout mice. Investig Ophthalmol Vis Sci 44(2):826–838

Article  Google Scholar 

Fourgeaud L, Través PG, Tufail Y, Leal-Bailey H, Lew ED, Burrola PG et al (2016) TAM receptors regulate multiple features of microglial physiology. Nature 532(7598):240–244

Article  CAS  PubMed  PubMed Central  Google Scholar 

Akalu YT, Mercau ME, Ansems M, Hughes LD, Nevin J, Alberto EJ et al (2022) Tissue-specific modifier alleles determine Mertk loss-of-function traits. Elife 11:e80530

Article  CAS  PubMed  PubMed Central  Google Scholar 

Ma GZ, Stankovich J, Australia, Consortium NZMSG, Kilpatrick TJ, Binder MD et al (2011) Polymorphisms in the receptor tyrosine kinase MERTK gene are associated with multiple sclerosis susceptibility. PLoS ONE 6(2):e16964

Article  CAS  PubMed  PubMed Central  Google Scholar 

Shen K, Reichelt M, Kyauk RV, Ngu H, Shen Y-AA, Foreman O et al (2021) Multiple sclerosis risk gene Mertk is required for microglial activation and subsequent remyelination. Cell Rep. 34(10)

Audo I, Mohand-Said S, Boulanger‐Scemama E, Zanlonghi X, Condroyer C, Démontant V et al (2018) MERTK mutation update in inherited retinal diseases. Hum Mutat 39(7):887–913

Article  CAS  PubMed  Google Scholar 

Patin E, Kutalik Z, Guergnon J, Bibert S, Nalpas B, Jouanguy E et al (2012) Genome-wide association study identifies variants associated with progression of liver fibrosis from HCV infection. Gastroenterology 143(5):1244–1252 e12

Article  CAS  PubMed  Google Scholar 

Graham DK, Dawson TL, Mullaney DL, Snodgrass HR, Earp HS (1994) Cloning and mRNA expression analysis of a novel human protooncogene, c-mer. Cell Growth Differentiation: Mol Biology J Am Association Cancer Res 5(6):647–657

CAS  Google Scholar 

Graham DK, Salzberg DB, Kurtzberg J, Sather S, Matsushima GK, Keating AK et al (2006) Ectopic expression of the proto-oncogene mer in pediatric T-cell acute lymphoblastic leukemia. Clin Cancer Res 12(9):2662–2669

Article  CAS  PubMed  Google Scholar 

Cummings CT, DeRyckere D, Earp HS, Graham DK (2013) Molecular pathways: MERTK signaling in cancer. Clin Cancer Res 19(19):5275–5280

Article  CAS  PubMed  PubMed Central  Google Scholar 

Dransfield I, Zagórska A, Lew E, Michail K, Lemke G (2015) Mer receptor tyrosine kinase mediates both tethering and phagocytosis of apoptotic cells. Cell Death Dis 6(2):e1646–e

Article  CAS  PubMed  PubMed Central  Google Scholar 

Thorp E, Cui D, Schrijvers DM, Kuriakose G, Tabas I (2008) Mertk receptor mutation reduces efferocytosis efficiency and promotes apoptotic cell accumulation and plaque necrosis in atherosclerotic lesions of apoe–/– mice. Arterioscler Thromb Vasc Biol 28(8):1421–1428

Article  CAS  PubMed  PubMed Central  Google Scholar 

Scott RS, McMahon EJ, Pop SM, Reap EA, Caricchio R, Cohen PL et al (2001) Phagocytosis and clearance of apoptotic cells is mediated by MER. Nature 411(6834):207–211

Article  CAS  PubMed  Google Scholar 

Karlsson A, Christenson K, Matlak M, Björstad Å, Brown KL, Telemo E et al (2009) Galectin-3 functions as an opsonin and enhances the macrophage clearance of apoptotic neutrophils. Glycobiology 19(1):16–20

Article  CAS  PubMed  Google Scholar 

López E, del Pozo V, Miguel T, Sastre B, Seoane C, Civantos E et al (2006) Inhibition of chronic airway inflammation and remodeling by galectin-3 gene therapy in a murine model. J Immunol 176(3):1943–1950

Article  PubMed  Google Scholar 

Jeon SB, Yoon HJ, Chang CY, Koh HS, Jeon SH, Park EJ (2010) Galectin-3 exerts cytokine-like regulatory actions through the JAK-STAT pathway. J Immunol 185(11):7037–7046

Article  CAS  PubMed  Google Scholar 

Shin T (2013) The pleiotropic effects of galectin-3 in neuroinflammation: a review. Acta Histochem 115(5):407–411

Article  CAS  PubMed  Google Scholar 

Yucel Y, Tanriverdi H, Arıkanoglu A, Varol S, Kaplan I, Akil E et al (2014) Increased fibrinogen, D-dimer and galectin-3 levels in patients with migraine. Neurol Sci 35:545–549

Article  PubMed  Google Scholar 

Hedef A, Baylan FA, Dülgeroğlu Y, Berktaş DT, Tolun Fİ (2023) Serum levels of Neopterin, Galectin-3, Tissue Necrosis Factor Alpha, and Interleukin-10 during the interictal and attack periods of patients with migraine. Turkish J Neurology/Turk Noroloji Dergisi. 29(1)

Gürger M, Atescelik M, Yılmaz M, Yildiz M, Kalayci H, Kobat MA et al (2018) Can we define migraine patients with blood high-sensitivity C-reactive protein and galectin-3 levels in the emergency department? Archives Med Sci 14(2):307–312

Article  Google Scholar 

RCore T (2016) R: a language and environment for statistical computing. R Foundation for Statistical Computing, Vienna, Austria

Google Scholar 

Petersen AS, Lund N, Meßlinger K, Christensen SL, Barloese M, Jørgensen NR et al (2024) Reduced plasma calcitonin gene-related peptide level identified in cluster headache: a prospective and controlled study. Cephalalgia 44(3):3331024231223970

Article  PubMed  Google Scholar 

Hautakangas H, Winsvold BS, Ruotsalainen SE, Bjornsdottir G, Harder AVE, Kogelman LJA et al (2022) Genome-wide analysis of 102,084 migraine cases identifies 123 risk loci and subtype-specific risk alleles. Nat Genet 54(2):152–160

Article  CAS  PubMed  PubMed Central  Google Scholar 

Pannese E, Pannese E (2018) Biology and pathology of perineuronal satellite cells in sensory ganglia. Springer

Suadicani SO, Cherkas PS, Zuckerman J, Smith DN, Spray DC, Hanani M (2010) Bidirectional calcium signaling between satellite glial cells and neurons in cultured mouse trigeminal ganglia. Neuron Glia Biol 6(1):43–51

Article  PubMed  Google Scholar 

Hanani M, Spray DC (2020) Emerging importance of satellite glia in nervous system function and dysfunction. Nat Rev Neurosci 21(9):485–498

Article  CAS  PubMed  PubMed Central  Google Scholar 

Zhang X, Chen Y, Wang C, Huang L-Y (2007) Neuronal somatic ATP release triggers neuron–satellite glial cell communication in dorsal root ganglia. Proceedings of the National Academy of Sciences. 104(23):9864-9

Messlinger K, Russo AF (2019) Current understanding of trigeminal ganglion structure and function in headache. Cephalalgia 39(13):1661–1674

留言 (0)

沒有登入
gif