Mast Cells in Atopic Diseases: More Than Just Histamine

Authors

  • Theoharis C. Theoharides, MS, MPhil, MD, PhD

DOI:

https://doi.org/10.58931/cait.2022.2s0345

Abstract

Mast cells are present in all tissues and are able to release multiple mediators in response to allergic, autoimmune, environmental, neurohormonal and pathogenic triggers. Histamine has received most of the attention in terms of pathophysiology and drug development, while tryptase remains to this date with no clear function and no known inhibitor. Mast cells can also release pro-inflammatory and pruritogenic molecules, such as IL-6 and IL-31, selectively without degranulation. One such critical molecule is platelet activating factor (PAF), which is vasoactive, can cause wheal and flare on ita own, but can also stimulate eosinophils and mast cells that are critical in the pathogenesis of chronic spontaneous urticaria (CSU) and rhinitis. Mast cell-derived cytokines and PAF have also been implicated in inflammatory processes including COVID-19. Among the second generation histamine-1 receptor antagonists, rupatadine is more effective overall, it has potent anti-PAF activity, and also inhibits activation of human mast cells and eosinophils. Rupatadine could, therefore, serve as a first-line drug for CSU and rhinitis, but may also be used, especially for patients resistant to antihistamines.

Author Biography

Theoharis C. Theoharides, MS, MPhil, MD, PhD

Dr. Theoharis C. Theoharides is Professor of Pharmacology and Internal Medicine, and Director of Molecular Immunopharmacology and Drug Discovery at Tufts University School of Medicine, Boston. He received his degrees (BA cum laude, MS, MPhil, PhD, MD) from Yale University and was awarded the Winternitz Prize in Pathology. He trained in internal medicine at New England Medical Center and received the Oliver Smith Award “recognizing excellence, compassion and service.” He received a Certificate in Global Leadership from the Tufts Fletcher School of Law and Diplomacy and a Fellowship at the Harvard Kennedy School of Government. He was awarded the Distinguished Faculty Recognition Award twice, the Excellence in Teaching ten times and the Alumni Award for Faculty Excellence. He first showed that mast cells, known for allergic reactions, are critical for neuroinflammatory conditions. He has published 470 scientific papers with over 40,000 citations and was listed in the world’s top 2% most cited scientists. He has received 37 patents and trademarks. He was inducted into the Alpha Omega Alpha National Medical Honor Society, the Rare Diseases Hall of Fame, and the World Academy of Sciences. He has been recognized with the 2018 Albert Nelson Marquis Lifetime Achievement and the Distinguished Humanitarian awards.

References

Csaba G. Mast cell, the peculiar member of the immune system: A homeostatic aspect. Acta Microbiol Immunol Hung 2015; 62(3):207-231.

Theoharides TC, Kalogeromitros D. The critical role of mast cells in allergy and inflammation. Ann N Y Acad Sci Nov;1088:78-99.

Wernersson S, Pejler G. Mast cell secretory granules: armed for battle. Nat Rev Immunol 2014; 14(7):478-494.

Castells M. Mast cell mediators in allergic inflammation and mastocytosis. Immunol Allergy Clin North Am 2006Aug;26(3):465-85.

Mukai K, Tsai M, Saito H, Galli SJ. Mast cells as sources of cytokines, chemokines, and growth factors. Immunol Rev 2018Mar;282(1):121-150.

Chen CC, Grimbaldeston MA, Tsai M, Weissman IL, Galli SJ. Identification of mast cell progenitors in adult mice. Proc Natl Acad Sci U S A 2005 Aug 9;102(32):11408-11413.

Gurish MF, Austen KF. Developmental origin and functional specialization of mast cell subsets 1. Immunity 2012 Jul 27;37(1):25-33.

Rodewald HR, Dessing M, Dvorak AM, Galli SJ. Identification of a committed precursor for the mast cell lineage. Science 1996 Feb 9; 271:818-822.

Kitamura Y, Ito A. Mast cell-committed progenitors. Proc Natl Acad Sci U S A 2005 Aug 9; 102(32):11129-11130. doi: 10.1073/pnas.0505073102.

Schmetzer O, Valentin P, Church MK, Maurer M, Siebenhaar F. Murine and human mast cell progenitors. Eur J Pharmacol 2016 May 5;778:2-10. doi: 10.1016/j.ejphar.2015.07.016. Epub 2015 Jul 8.

Theoharides TC, Valent P, Akin C. Mast Cells, Mastocytosis, and Related Disorders. N Engl J Med 2015; 373(2):163-172.

Galli SJ, Borregaard N, Wynn TA. Phenotypic and functional plasticity of cells of innate immunity: macrophages, mast cells and neutrophils. Nat Immunol 2011Oct 19;12(11):1035-44.

Theoharides TC. Skin Mast Cells: Are We Missing the Forest for the Trees? Exp Dermatol 2016 Jun;25(6):422-3.

Theoharides TC. Mast cells: the immune gate to the brain. Life Sci 1990; 46:607-617. doi: 10.1016/0024-3205(90)90129-f.

Traina G. Mast cells in the brain - Old cells, new target. J Integr Neurosci 2017; 16(s1):S69-S83.

Rozniecki JJ, Dimitriadou V, Lambracht-Hall M, Pang X, Theoharides TC. Morphological and functional demonstration of rat dura mast cell-neuron interactions in vitro and in vivo. Brain Res 1999; 849:1-15.

Polyzoidis S, Koletsa T, Panagiotidou S, Ashkan K, Theoharides TC. Mast cells in meningiomas and brain inflammation. J Neuroinflammation 2015; 12(1):170.

Theoharides TC, Konstantinidou A. Corticotropin-releasing hormone and the blood-brain-barrier. Front Biosci 2007; 12:1615-1628.

Colonna M, Butovsky O. Microglia Function in the Central Nervous System During Health and Neurodegeneration. Annu Rev Immunol 2017; 35:441-468.

Paus R, Theoharides TC, Arck PC. Neuroimmunoendocrine circuitry of the ‘brain-skin connection’. Trends Immunol 2006; 27(1):32-39.

Panula P. Histamine receptors, agonists, and antagonists in health and disease. Handb Clin Neurol 2021; 180:377-387.

Carthy E, Ellender T. Histamine, Neuroinflammation and Neurodevelopment: A Review. Front Neurosci 2021; 15:680214.

Mochizuki T. Histamine as an Alert Signal in the Brain. Curr Top Behav Neurosci 2022;59:413-425. doi: 10.1007/7854_2021_249.

Provensi G, Costa A, Izquierdo I, Blandina P, Passani MB. Brain histamine modulates recognition memory: possible implications in major cognitive disorders. Br J Pharmacol 2020; 177(3):539-556.

Passani MB, Benetti F, Blandina P, Furini CRG, de Carvalho MJ, Izquierdo I. Histamineregulates memory consolidation. Neurobiol Learn Mem 2017; 145:1-6.

Burgess CR. Histamine and orexin in the control of arousal, locomotion, and motivation. J Neurosci 2010; 30(8):2810-2811.

Sadek B, Saad A, Sadeq A, Jalal F, Stark H. Histamine H3 receptor as a potential target for cognitive symptoms in neuropsychiatric diseases. Behav Brain Res 2016; 312:415-430.

Schlicker E, Kathmann M. Role of the Histamine H3 Receptor in the Central Nervous System. Handb Exp Pharmacol 2017; 241:277-299.

Torrealba F, Riveros ME, Contreras M, Valdes JL. Histamine and motivation. Front Syst Neurosci 2012; 6:51.

Nomura H, Shimizume R, Ikegaya Y. Histamine: A Key Neuromodulator of Memory Consolidation and Retrieval. Curr Top Behav Neurosci 2022;59:329-353.

Rivera J, Fierro NA, Olivera A, Suzuki R. New insights on mast cell activation via the high affinity receptor for IgE. Adv Immunol 2008; 98:85-120.

Rivera J, Gilfillan AM. Molecular regulation of mast cell activation. J Allergy Clin Immunol 2006; 117(6):1214-1225.

Cheng LE, Hartmann K, Roers A, Krummel MF, Locksley RM. Perivascular mast cells dynamically probe cutaneous blood vessels to capture immunoglobulin E. Immunity 2013; 38(1):166-175.

Msallam R, Balla J, Rathore APS et al. Fetal mast cells mediate postnatal allergic responses dependent on maternal IgE. Science 2020; 370(6519):941-950.

Peters JL, Cohen S, Staudenmayer J, Hosen J, Platts-Mills TA, Wright RJ. Prenatal negative life events increases cord blood IgE: interactions with dust mite allergen and maternal atopy. Allergy 2012; 67(4):545-551.

Theoharides TC. Danger Signals and Inflammation. Clin Ther 2016; 38(5):996-999.

Redegeld FA, Yu Y, Kumari S, Charles N, Blank U. Non-IgE mediated mast cell activation. Immunol Rev 2018; 282(1):87-113.

Olivera A, Beaven MA, Metcalfe DD. Mast cells signal their importance in health and disease. J Allergy Clin Immunol 2018; 142(2):381-393.

Migalovich-Sheikhet H, Friedman S, Mankuta D, Levi-Schaffer F. Novel identified receptors on mast cells. Front Immunol 2012; 3:238.

Theoharides TC. Neuroendocrinology of mast cells: Challenges and controversies. Exp Dermatol 2017; 26(9):751-759.

Theoharides TC, Donelan JM, Papadopoulou N, Cao J, Kempuraj D, Conti P. Mast cells as targets of corticotropin-releasing factor and related peptides. Trends Pharmacol Sci 2004; 25(11):563-568.

Donelan J, Boucher W, Papadopoulou N, Lytinas M, Papaliodis D, Theoharides TC. Corticotropin-releasing hormone induces skin vascular permeability through a neurotensin-dependent process. Proc Natl Acad Sci USA 2006; 103:7759-7764.

Theoharides TC, Zhang B, Kempuraj D et al. IL-33 augments substance P-induced VEGF secretion from human mast cells and is increased in psoriatic skin. Proc Natl Acad Sci U S A 2010; 107(9):4448-4453.

Theoharides TC, Betchaku T, Douglas WW. Somatostatin-induced histamine secretion in mast cells; a comparison with known mast cell secretagogues. J Biol Chem 1990.

Theoharides TC, Douglas WW. Mast cell histamine secretion in response to somatostatin analogues and other peptides. J Biol Chem 1990.

McNeil BD, Pundir P, Meeker S et al. Identification of a mast-cell-specific receptor crucial for pseudo-allergic drug reactions. Nature 2015; 519(7542):237-241.

Sumpter TL, Ho CH, Pleet AR et al. Autocrine hemokinin-1 functions as an endogenous adjuvant for IgE-mediated mast cell inflammatory responses. J Allergy Clin Immunol 2015; 135(4):1019-1030.

Kay AB, Ying S, Ardelean E et al. Calcitonin gene-related peptide and vascular endothelial growth factor are expressed in lesional but not uninvolved skin in chronic spontaneous urticaria. Clin Exp Allergy 2014; 44(8):1053-1060.

Saluja R, Metz M, Maurer M. Role and relevance of mast cells in fungal infections. Front Immunol 2012; 3:146.

Ratnaseelan AM, Tsilioni I, Theoharides TC. Effects of Mycotoxins on Neuropsychiatric Symptoms and Immune Processes. Clin Ther 2018; 40(6):903-917.

Abraham SN, St John AL. Mast cell- orchestrated immunity to pathogens. Nat Rev Immunol 2010; 10(6):440-452.

Marshall JS, Portales-Cervantes L, Leong E. Mast Cell Responses to Viruses and Pathogen Products. Int J Mol Sci 2019; 20(17):4241.

Theoharides TC. Potential Association of Mast Cells with COVID-19. Ann Allergy Asthma Immunol 2020; 126(3):217-218.

Motta Junior JDS, Miggiolaro AFRD, Nagashima S et al. Mast Cells in Alveolar Septa of COVID-19 Patients: A Pathogenic Pathway That May Link Interstitial Edema to Immunothrombosis. Front Immunol 2020 Sep 18;11:574862.

Theoharides TC, Stewart JM. Genitourinary mast cells and survival. Transl Androl Urol 2015; 4(5):579-586.

Pang X, Cotreau-Bibbo MM, Sant GR, Theoharides TC. Bladder mast cell expression of high affinity estrogen receptors in patients with interstitial cystitis. Br J Urol 1995; 75:154-161.

Csaba G, Kovacs P. Hormones in the nucleus of mast cells: confocal microscopic immunocytochemical observations. Horm Metab Res 2009; 41(8):621-625.

Kempuraj D, Papadopoulou NG, Lytinas M et al. Corticotropin-releasing hormoneand its structurally related urocortin are synthesized and secreted by human mast cells. Endocrinology 2004; 145:43-48.

Grounds MD, Radley HG, Gebski BL, Bogoyevitch MA, Shavlakadze T. Implications of cross-talk between tumour necrosis factor and insulin-like growth factor-1 signalling in skeletal muscle. Clin Exp Pharmacol Physiol 2008; 35(7):846-851.

Baumann A, Gonnenwein S, Bischoff SC et al. The circadian clock is functional in eosinophils and mast cells. Immunology 2013; 140(4):465-474.

Wang X, Reece SP, Van Scott MR, Brown JM. A circadian clock in murine bone marrow-derived mast cells modulates IgE-dependent activation in vitro. Brain Behav Immun 2011; 25(1):127-134.

Christ P, Sowa AS, Froy O, Lorentz A. The Circadian Clock Drives Mast Cell Functions in Allergic Reactions. Front Immunol 2018; 9:1526.

Pham L, Baiocchi L, Kennedy L et al. The interplay between mast cells, pineal gland, and circadian rhythm: Links between histamine, melatonin, and inflammatory mediators. J Pineal Res 2021; 70(2):e12699.

Csaba G. Regulation of mast cell formation. Studia Biolog Acad Scient Hung 1972; 11:7-148.

Uvnas B. Histamine storage and release. Fed Proc 1974 Oct;33(10):2172-6.

Borriello F, Iannone R, Marone G. Histamine Release from Mast Cells and Basophils. Handb Exp Pharmacol 2017; 241:121-139.

Cochrane DE, Carraway RE, Feldberg RS, Boucher W, Gelfand JM. Stimulated rat mast cells generate histamine-releasing peptide from albumin. Peptides 1993; 14:117-123.

Boyce JA. Mast cells and eicosanoid mediators: a system of reciprocal paracrine and autocrine regulation. Immunol Rev 2007; 217:168-185.

Varvara G, Tettamanti L, Gallenga CE et al. Stimulated mast cells release inflammatory cytokines: potential suppression and therapeutical aspects. J Biol Regul Homeost Agents 2018;32(6):1355-1360.

Alevizos M, Karagkouni A, Vasiadi M et al. Rupatadine inhibits inflammatory mediator release from human LAD2 cultured mast cells stimulated by PAF . Ann Allergy Asthma Immunol 2013; 111(6):524-527.

Demopoulos C, Antonopoulou S, Theoharides TC. COVID-19, microthromboses, inflammation, and platelet activating factor. Biofactors 2020; 46(6):927-933.

Theoharides TC, Antonopoulou S, Demopoulos CA. Coronavirus 2019, Microthromboses, and Platelet Activating Factor. Clin Ther 2020; 42(10):1850-1852.

Ashraf MA, Nookala V. Biochemistry of Platelet Activating Factor. 2021.

Demopoulos CA, Pinckard RN, Hanahan DJ. Platelet-activating factor. Evidence for 1-O-alkyl-2-acetyl-sn-glyceryl-3-phosphorylcholine as the active component (a new class of lipid chemical mediators). J Biol Chem 1979; 254(19):9355-9358.

Demopoulos CA, Andrikopoulos NK, Antonopoulou S. A simple and precise method for the routine determination of platelet-activating factor in blood and urine. Lipids 1994; 29(4):305-309.

Theoharides TC, Bondy PK, Tsakalos ND, Askenase PW. Differential release of serotonin and histamine from mast cells. Nature 1982; 297:229-231.

Kandere-Grzybowska K, Letourneau R, Kempuraj D et al. IL-1 induces vesicular secretion of IL-6 without degranulation from human mast cells. J Immunol 2003; 171(9):4830-4836.

Cao J, Papadopoulou N, Kempuraj D et al. Human mast cells express corticotropin-releasing hormone (CRH) receptors and CRH leads to selective secretion of vascular endothelial growth factor. J Immunol 2005; 174(12):7665-7675.

Theoharides TC, Kempuraj D, Tagen M, Conti P, Kalogeromitros D. Differential release of mast cell mediators and the pathogenesis of inflammation. Immunol Rev 2007; 217:65-78.

Enoksson M, Lyberg K, Moller-Westerberg C, Fallon PG, Nilsson G, Lunderius-Andersson C. Mast cells as sensors of cell injury through IL-33 recognition. J Immunol 2011; 186(4):2523-2528.

Saluja R, Khan M, Church MK, Maurer M. The role of IL-33 and mast cells in allergy and inflammation. Clin Transl Allergy 2015; 5:33.

Theoharides TC, Leeman SE. Effect of IL-33 on de novo synthesized mediators from human mast cells. J Allergy Clin Immunol 2019; 143:451.

Cristinziano L, Poto R, Criscuolo G et al. IL-33 and Superantigenic Activation of Human Lung Mast Cells Induce the Release of Angiogenic and Lymphangiogenic Factors. Cells 2021; 10(1).

Petra AI, Tsilioni I, Taracanova A, Katsarou-Katsari A, Theoharides TC. Interleukin 33 and interleukin 4 regulate interleukin 31 gene expression and secretion from human laboratory of allergic diseases 2 mast cells stimulated by substance P and/or immunoglobulin E. Allergy Asthma Proc 2018;39(2):153-160.

Taracanova A, Alevizos M, Karagkouni A et al. SP and IL-33 together markedly enhance TNF synthesis and secretion from human mast cells mediated by the interaction of their receptors. Proc Natl Acad Sci U S A 2017; 114(20):E4002-E4009.

Taracanova A, Tsilioni I, Conti P, Norwitz ER, Leeman SE, Theoharides TC. Substance P and IL-33 administered together stimulate a marked secretion of IL-1beta from human mast cells, inhibited by methoxyluteolin. Proc Natl Acad Sci U S A 2018; 115(40):E9381-E9390.

Bawazeer MA, Theoharides TC. IL-33 stimulates human mast cell release of CCL5 and CCL2 via MAPK and NF-kappaB, inhibited by methoxyluteolin. Eur J Pharmacol 2019; 865:172760.

Brown MA. Studies of Mast Cells: Adventures in Serendipity. Front Immunol 2018;9:520.

Conti P, Caraffa A, Tete G et al. Mast cells activated by SARS-CoV-2 release histamine which increases IL-1 levels causing cytokine storm and inflammatory reaction in COVID-19. J Biol Regul Homeost Agents 2020;34(5):1629-1632.

Gagari E, Tsai M, Lantz CS, Fox LG, Galli SJ. Differential release of mast cell interleukin-6 via c-kit. Blood 1997; 89:2654-2663.

Theoharides TC, Boucher W, Spear K. Serum interleukin-6 reflects disease severity and osteoporosis in mastocytosis patients. Int Arch Allergy Immunol 2002; 128:344-350.

Brockow K, Akin C, Huber M, Metcalfe DD. IL-6 levels predict disease variant and extent of organ involvement in patients with mastocytosis. Clin Immunol 2005;115(2):216-223.

Mayado A, Teodosio C, Garcia-Montero AC et al. Increased IL6 plasma levels in indolent systemic mastocytosis patients are associated with high risk of disease progression. Leukemia 2015; 30(1):124-130.

Herold T, Jurinovic V, Arnreich C et al. Elevated levels of IL-6 and CRP predict the need for mechanical ventilation in COVID-19. J Allergy Clin Immunol 2020; 146(1):128-136.

Copaescu A, Smibert O, Gibson A, Phillips EJ, Trubiano JA. The role of IL-6 and other mediators in the cytokine storm associated with SARS-CoV-2 infection. J Allergy Clin Immunol 2020; 146(3):518-534.

Kaur D, Gomez E, Doe C et al. IL-33 drives airway hyper-responsiveness through IL-13-mediated mast cell: airway smooth muscle crosstalk. Allergy 2015; 70(5):556-567.

Tobio A, Bandara G, Morris DA et al. Oncogenic D816V-KIT signaling in mast cells causes persistent IL-6 production. Haematologica 2020; 105(1):124-135.

Theoharides TC. Mast cell: a neuroimmunoendocrine master player. Int J Tissue React 1996; 18(1):1-21.

Esposito P, Chandler N, Kandere-Grzybowska K et al. Corticotropin-releasing hormone (CRH) and brain mast cells regulate blood-brain-barrier permeability induced by acute stress. J Pharmacol Exp Ther 2002;303:1061-1066.

Fiorentino M, Sapone A, Senger S et al. Blood-brain barrier and intestinal epithelial barrier alterations in autism spectrum disorders. Mol Autism 2016; 7:49.

Rozniecki JJ, Sahagian GG, Kempuraj D et al. Brain metastases of mouse mammary adenocarcinoma is increased by acute stress. Brain Res 2010; 1366:204-210.

Theoharides TC, Rozniecki JJ, Sahagian G et al. Impact of stress and mast cells on brain metastases. J Neuroimmunol 2008;205(1-2):1-7.

Abbott NJ. Inflammatory mediators and modulation of blood-brain barrier permeability. Cell Mol Neurobiol 2000; 20:131-147.

Pan W, Stone KP, Hsuchou H, Manda VK, Zhang Y, Kastin AJ. Cytokine signaling modulates blood-brain barrier function. Curr Pharm Des 2011; 17(33):3729-3740.

Theoharides TC. Effect of psychological stress on mast cells. Annals Allergy, Asthma, Immunology 125[4], 388-392. 8-10-2020.

Kandere-Grzybowska K, Gheorghe D, Priller J et al. Stress-induced dura vascular permeability does not develop in mast cell-deficient and neurokinin-1 receptor knockout mice 1. Brain Res 2003;980(2):213-220.

Matsumoto I, Inoue Y, Shimada T, Aikawa T. Brain mast cells act as an immune gate to the hypothalamic-pituitary-adrenal axis in dogs. J Exp Med 2001; 194:71-78.

Bugajski AJ, Chlap Z, Gadek-Michalska A, Borycz J, Bugajski J. Degranulation and decrease in histamine levels of thalamic mast cells coincides with corticosterone secretion induced by compound 48/80. Inflamm Res 1995; 44 Suppl 1:S50-S51.

Kalogeromitros D, Syrigou EI, Makris M et al. Nasal provocation of patients with allergic rhinitis and the hypothalamic-pituitary-adrenal axis. Annals Allergy, Asthma, Immunology 2007; 98:269-273.

Scaccianoce S, Lombardo K, Nicolai R, Affricano D, Angelucci L. Studies on the involvement of histamine in the hypothalamic- pituitary-adrenal axis activation induced by nerve growth factor. Life Sci 2000; 67:3143-3152.

Mastorakos G, Chrousos GP, Weber JS. Recombinant interleukin-6 activates the hypothalamic-pituitary-adrenal axis in humans. J Clin Endocrinol Metab 1993; 77:1690-1694.

Theoharides TC, Tsilioni I, Bawazeer M. Mast Cells, Neuroinflammation and Pain in Fibromyalgia Syndrome. Front Cell Neurosci 2019; 13:353.

Xu H, Shi X, Li X et al. Neurotransmitter and neuropeptide regulation of mast cell function: a systematic review. J Neuroinflammation 2020; 17(1):356.

Okada T, Hirayama Y, Kishi S, Miyayasu K, Hiroi J, Fujii T. Functional neurokinin NK-1 receptor expression in rat peritoneal mast cells. Inflamm Res 1999; 48:274-279.

Asadi S, Alysandratos KD, Angelidou A et al. Substance P (SP) induces expression of functional corticotropin-releasing hormone receptor-1 (CRHR-1) in human mast cells. J Invest Dermatol 2012; 132(2):324-329.

Alysandratos KD, Asadi S, Angelidou A et al. Neurotensin and CRH interactions augment human mast cell activation. PloS One 2012;7(11):e48934.

Xu H, Bin NR, Sugita S. Diverse exocytic pathways for mast cell mediators. Biochem Soc Trans 2018; 46(2):235-247.

Gilfillan AM, Tkaczyk C. Integrated signalling pathways for mast-cell activation. Nat Rev Immunol 2006; 6(3):218-230.

Gaudenzio N, Sibilano R, Marichal T et al. Different activation signals induce distinct mast cell degranulation strategies. J Clin Invest 2016;126(10):3981-3998.

Sibilano R, Frossi B, Pucillo CE. Mast cell activation: a complex interplay of positive and negative signaling pathways. Eur J Immunol 2014; 44(9):2558-2566.

Crivellato E, Nico B, Gallo VP, Ribatti D. Cell secretion mediated by granule-associated vesicle transport: a glimpse at evolution. Anat Rec (Hoboken ) 2010; 293(7):1115-1124.

Ogulur I, Pat Y, Ardicli O et al. Advances and highlights in biomarkers of allergic diseases. Allergy 2021.

Siebenhaar F, Redegeld FA, Bischoff SC, Gibbs BF, Maurer M. Mast Cells as Drivers of Disease and Therapeutic Targets. Trends Immunol 2018; 39(2):151-162.

Gallenga CE, Pandolfi F, Caraffa A et al. Interleukin-1 family cytokines and mast cells: activation and inhibition. J Biol Regul Homeost Agents 2019; 33(1):1-6.

Marone G, Galli SJ, Kitamura Y. Probing the roles of mast cells and basophils in natural and acquired immunity, physiology and disease. Trends Immunol 2002; 23:425-427.

Galli SJ, Grimbaldeston M, Tsai M. Immunomodulatory mast cells: negative, as well as positive, regulators of immunity. Nat Rev Immunol 2008; 8(6):478-486.

Hakim-Rad K, Metz M, Maurer M. Mast cells: makers and breakers of allergic inflammation. Curr Opin Allergy Clin Immunol 2009; 9(5):427-430.

Theoharides TC, Alysandratos KD, Angelidou A et al. Mast cells and inflammation. Biochim Biophys Acta 2012; 1822(1):21-33.

He L, Yi W, Huang X, Long H, Lu Q. Chronic Urticaria: Advances in Understanding of the Disease and Clinical Management. Clin Rev Allergy Immunol 2021.

Saini S, Shams M, Bernstein JA, Maurer M. Urticaria and Angioedema Across the Ages. J Allergy Clin Immunol Pract 2020;8(6):1866-1874.

Goncalo M, Gimenez-Arnau A, Al-Ahmad M et al. The global burden of chronic urticaria for the patient and society. Br J Dermatol 2021;184(2):226-236.

Church MK, Kolkhir P, Metz M, Maurer M. The role and relevance of mast cells in urticaria.Immunol Rev 2018; 282(1):232-247.

Altrichter S, Frischbutter S, Fok JS et al. The role of eosinophils in chronic spontaneous urticaria. J Allergy Clin Immunol 2020;145(6):1510-1516.

Kolkhir P, Andre F, Church MK, Maurer M, Metz M. Potential blood biomarkers in chronic spontaneous urticaria. Clin Exp Allergy 2017;47(1):19-36.

Deza G, Ricketti PA, Gimenez-Arnau AM, Casale TB. Emerging Biomarkers and Therapeutic Pipelines for Chronic Spontaneous Urticaria. J Allergy Clin Immunol Pract 2018;6(4):1108-1117.

Wong MM, Keith PK. Presence of positive skin prick tests to inhalant allergens and markers of T2 inflammation in subjects with chronic spontaneous urticaria (CSU): a systematic literature review. Allergy Asthma Clin Immunol 2020; 16:72.

Cao TBT, Cha HY, Yang EM, Ye YM. Elevated MRGPRX2 Levels Related to Disease Severity in Patients With Chronic Spontaneous Urticaria. Allergy Asthma Immunol Res 2021;13(3):498-506.

Kolkhir P, Elieh-Ali-Komi D, Metz M, Siebenhaar F, Maurer M. Understanding human mast cells: lesson from therapies for allergic and non-allergic diseases. Nat Rev Immunol 2021.

Vadas P, Gold M, Perelman B et al. Platelet-activating factor, PAF acetylhydrolase, and severe anaphylaxis. N Engl J Med 2008;358(1):28-35.

Gill P, Jindal NL, Jagdis A, Vadas P. Platelets in the immune response: Revisiting platelet-activating factor in anaphylaxis. J Allergy Clin Immunol 2015; 135(6):1424-1432.

Vadas P, Perelman B, Liss G. Platelet-activating factor, histamine, and tryptase levels in human anaphylaxis. J Allergy Clin Immunol 2013; 131(1):144-149.

Arias K, Baig M, Colangelo M et al. Concurrent blockade of platelet-activating factor and histamine prevents life-threatening peanut-induced anaphylactic reactions. J Allergy Clin Immunol 2009;124(2):307-14, 314.

Palgan K, Bartuzi Z. Platelet activating factor in allergies. Int J Immunopathol Pharmacol 2015; 28(4):584-589.

Furukawa M, Ogura M, Tsutsumi T, Tsuji H, Yamashita T. Presence of platelet-activating factor in nasal polyps and eosinophils. Acta Otolaryngol 2002; 122(8):872-876.

Munoz-Cano RM, Casas-Saucedo R, Valero SA, Bobolea I, Ribo P, Mullol J. Platelet-Activating Factor (PAF) in Allergic Rhinitis: Clinical and Therapeutic Implications. J Clin Med 2019; 8(9).

Rihoux JP, Fadel R, Juhlin L. Platelet-activating factor-induced immediate and late cutaneous reactions. Int Arch Allergy Appl Immunol 1991; 94(1-4):299-300.

Ulambayar B, Yang EM, Cha HY, Shin YS, Park HS, Ye YM. Increased platelet activating factor levels in chronic spontaneous urticaria predicts refractoriness to antihistamine treatment: an observational study. Clin Transl Allergy 2019; 9:33.

Tedeschi A, Palumbo G, Milazzo N, Miadonna A. Nasal neutrophilia and eosinophilia induced by challenge with platelet activating factor. J Allergy Clin Immunol 1994; 93(2):526-533.

Kato M, Kita H, Tachibana A, Hayashi Y, Tsuchida Y, Kimura H. Dual signaling and effector pathways mediate human eosinophil activation by platelet-activating factor. Int Arch Allergy Immunol 2004; 134 Suppl 1:37-43.

Zoratti EM, Sedgwick JB, Vrtis RR, Busse WW. The effect of platelet-activating factor on the generation of superoxide anion in human eosinophils and neutrophils. J Allergy Clin Immunol 1991; 88(5):749-758.

Austin CE, Foreman JC. The effect of platelet-activating factor on the responsiveness of the human nasal airway. Br J Pharmacol 1993; 110(1):113-118.

Gutierrez S, Palacios I, Egido J et al. IL-1 beta and IL-6 stimulate the production of platelet-activating factor (PAF) by cultured rabbit synovial cells. Clin Exp Immunol 1995;99(3):364-368.

Biffl WL, Moore EE, Moore FA, Barnett CC, Jr., Silliman CC, Peterson VM. Interleukin-6 stimulates neutrophil production of platelet-activating factor. J Leukoc Biol 1996;59(4):569-574.

Thivierge M, Rola-Pleszczynski M. Platelet-activating factor enhances interleukin-6 production by alveolar macrophages. J Allergy Clin Immunol 1992; 90(5):796-802.

Keglowich L, Baraket M, Tamm M, Borger P. Hypoxia exerts dualistic effects on inflammatory and proliferative responses of healthy and asthmatic primary human bronchial smooth muscle cells. PLoS ONE 2014;9(2):e89875.

Hamel-Cote G, Lapointe F, Veronneau S, Mayhue M, Rola-Pleszczynski M, Stankova J. Regulation of platelet-activating factor-mediated interleukin-6 promoter activation by the 48 kDa but not the 45 kDa isoform ofprotein tyrosine phosphatase non-receptor type2. Cell Biosci 2019; 9:51.

Krause K, Gimenez-Arnau A, Martinez-Escala E et al. Platelet-activating factor (PAF) induces wheal and flare skin reactions independent of mast cell degranulation. Allergy 2013; 68(2):256-258.

Saini SS, Kaplan AP. Chronic Spontaneous Urticaria: The Devil’s Itch. J Allergy Clin Immunol Pract 2018; 6(4):1097-1106.

Kim HJ. Pruritus in autoimmune connective tissue diseases. Ann Transl Med 2021; 9(5):441.

Gibbs BF, Patsinakidis N, Raap U. Role of the Pruritic Cytokine IL-31 in Autoimmune Skin Diseases. Front Immunol 2019; 10:1383.

Datsi A, Steinhoff M, Ahmad F, Alam M, Buddenkotte J. Interleukin-31: The “itchy” cytokine in inflammation and therapy. Allergy 2021; 76(10):2982-2997.

Murdaca G, Greco M, Tonacci A et al. IL-33/IL-31 Axis in Immune-Mediated and Allergic Diseases. Int J Mol Sci 2019; 20(23).

Chaowattanapanit S, Choonhakarn C, Salao K et al. Increased serum IL-31 levels in chronic spontaneous urticaria and psoriasis with pruritic symptoms. Heliyon 2020; 6(12):e05621.

Golpanian RS, Kim HS, Yosipovitch G. Effects of Stress on Itch. Clin Ther 2020;42(5):745-756.

Huang Y, Xiao Y, Jing D et al. Association of Chronic Spontaneous Urticaria With Anxiety and Depression in Adolescents: A Mediation Analysis. Front Psychiatry 2021; 12:655802.

Ayasse MT, Buddenkotte J, Alam M, Steinhoff M. Role of neuroimmune circuits and pruritus in psoriasis. Exp Dermatol 2020; 29(4):414-426.

Nakashima C, Ishida Y, Kitoh A, Otsuka A, Kabashima K. Interaction of peripheral nerves and mast cells, eosinophils, and basophils in the development of pruritus. Exp Dermatol 2019;28(12):1405-1411.

Nakasaki T, Masuyama K, Fukui H et al. Effects of PAF on histamine H1 receptor mRNA expression in rat trigeminal ganglia. Prostaglandins Other Lipid Mediat 1999;58(1):29-41.

Ackermann M, Verleden SE, Kuehnel M et al. Pulmonary Vascular Endothelialitis, Thrombosis, and Angiogenesis in Covid-19. N Engl J Med 2020; 383(2):120-128.

Calabretta E, Moraleda JM, Iacobelli M et al. COVID-19-induced endotheliitis: emerging evidence and possible therapeutic strategies. Br J Haematol 2021; 193(1):43-51.

Wang J, Doran J. The Many Faces of Cytokine Release Syndrome-Related Coagulopathy. Clin Hematol Int 2021; 3(1):3-12.

Klein M, Dao V, Khan F. A Review of Platelet-Activating Factor As a Potential Contributor to Morbidity and Mortality Associated with Severe COVID-19. Clin Appl Thromb Hemost 2021; 27:10760296211051764.

Theoharides TC. COVID-19, pulmonary mast cells, cytokine storms, and beneficial actions of luteolin. Biofactors 2020; 46(3):306-308.

Theoharides TC, Conti P. COVID-19 and Multisystem Inflammatory Syndrome, or is it Mast Cell Activation Syndrome? J Biol Regul Homeost Agents 2020; 34(5):1633.-1636.

Afrin LB, Weinstock LB, Molderings GJ. Covid-19 hyperinflammation and post-Covid-19 illness may be rooted in mast cell activation syndrome. Int J Infect Dis 2020; 100:327-332.

Hafezi B, Chan L, Knapp JP et al. Cytokine Storm Syndrome in SARS-CoV-2 Infections: A Functional Role of Mast Cells. Cells 2021; 10(7).

Lam HY, Tergaonkar V, Kumar AP, Ahn KS. Mast cells: Therapeutic targets for COVID-19 and beyond. IUBMB Life 2021; 73(11):1278-1292.

Tan J, Anderson DE, Rathore APS et al. Signatures of mast cell activation are associated with severe COVID-19. medRxiv 2021.

Gebremeskel S, Schanin J, Coyle KM et al. Mast Cell and Eosinophil Activation Are Associated With COVID-19 and TLR-Mediated Viral Inflammation: Implications for an Anti-Siglec-8 Antibody. Front Immunol 2021;12:650331.

Rodriguez L, Pekkarinen PT, Lakshmikanth T et al. Systems-Level Immunomonitoring from Acute to Recovery Phase of Severe COVID-19. Cell Rep Med 2020; 1(5):100078.

Algaadi SA. Urticaria and COVID-19: A review. Dermatol Ther 2020; 33(6):e14290.

Moreno-Perez O, Merino E, Leon-Ramirez JM et al. Post-acute COVID-19 Syndrome. Incidence and risk factors: a Mediterranean cohort study. J Infect 2021; 82(3):378-383.

Nalbandian A, Sehgal K, Gupta A et al. Post-acute COVID-19 syndrome. Nat Med 2021; 27(4):601-615.

Montagne A, Nation DA, Sagare AP et al. APOE4 leads to blood-brain barrier dysfunction predicting cognitive decline. Nature 2020; 581(7806):71-76.

Sudre CH, Murray B, Varsavsky T et al. Attributes and predictors of long COVID. Nat Med 2021; 27(4):626-631.

Dennis A, Wamil M, Alberts J et al. Multiorgan impairment in low-risk individuals with post-COVID-19 syndrome: a prospective, community-based study. BMJ Open 2021; 11(3):e048391.

Baig AM. Chronic COVID Syndrome: Need for an appropriate medical terminology for Long-COVID and COVID Long-Haulers. J Med Virol 2020.

Higgins V, Sohaei D, Diamandis EP, Prassas I. COVID-19: from an acute to chronic disease? Potential long-term health consequences. Crit Rev Clin Lab Sci 2020;1-23.

Huang C, Huang L, Wang Y et al. 6-month consequences of COVID-19 in patients discharged from hospital: a cohort study. Lancet 2021; 397(10270):220-232.

de Erausquin GA, Snyder H, Carrillo M, Hosseini AA, Brugha TS, Seshadri S. The chronic neuropsychiatric sequelae of COVID-19: The need for a prospective study of viral impact on brain functioning. Alzheimers Dement 2021;17(6):1056-1065.

Baig AM. Deleterious Outcomes in Long-Hauler COVID-19: The Effects of SARS-CoV-2 on the CNS in Chronic COVID Syndrome. ACS Chem Neurosci 2020; 11(24):4017-4020.

Theoharides TC, Cholevas C, Polyzoidis K, Politis A. Long-COVID syndrome-associated brain fog and chemofog: Luteolin to the rescue. Biofactors 2021; 47(2):232-241.

Graham EL, Clark JR, Orban ZS et al. Persistent neurologic symptoms and cognitive dysfunction in non-hospitalized Covid-19 “long haulers”. Ann Clin Transl Neurol 2021;8(5):1073-1085.

Stefano GB, Buttiker P, Weissenberger S, Martin A, Ptacek R, Kream RM. Editorial: The Pathogenesis of Long-Term Neuropsychiatric COVID-19 and the Role of Microglia, Mitochondria, and Persistent Neuroinflammation: A Hypothesis. Med Sci Monit 2021; 27:e933015.

Bell ML, Catalfamo CJ, Farland LV et al. Post-acute sequelae of COVID-19 in a non-hospitalized cohort: Results from the Arizona CoVHORT. PLoS ONE 2021; 16(8):e0254347.

Hugon J, Msika EF, Queneau M, Farid K, Paquet C. Long COVID: cognitive complaints (brain fog) and dysfunction of the cingulate cortex. J Neurol 2021; Jun 18:1-3.

sadi-Pooya AA, Akbari A, Emami A et al. Long COVID syndrome-associated brain fog. J Med Virol 2021.

Zhang X, Dong H, Li N et al. Activated brain mast cells contribute to postoperative cognitive dysfunction by evoking microglia activation and neuronal apoptosis 1. J Neuroinflammation 2016;13(1):127.

Moura DS, Sultan S, Georgin-Lavialle S et al. Evidence for cognitive impairment in mastocytosis: prevalence, features and correlations to depression. PLoS ONE 2012; 7(6):e39468.

Afrin LB, Pohlau D, Raithel M et al. Mast cell activation disease: An underappreciated cause of neurologic and psychiatric symptoms and diseases. Brain Behav Immun 2015; 50:314-321.

Akin C, Valent P, Metcalfe DD. Mast cell activation syndrome: Proposed diagnostic criteria. J Allergy Clin Immunol 2010;126(6):1099-104.

Theoharides TC, Tsilioni I, Ren H. Recent advances in our understanding of mast cell activation - or should it be mast cell mediator disorders? Expert Rev Clin Immunol 2019;15(6):639-656.

Motta Junior JDS, Miggiolaro AFRD, Nagashima S et al. Mast Cells in Alveolar Septa of COVID-19 Patients: A PathogenicPathway That May Link Interstitial Edema to Immunothrombosis. Front Immunol 2020;11:574862.

Theoharides TC, Alysandratos KD, Angelidou A et al. Mast cells and inflammation. Biochim Biophys Acta 2012; 1822(1):21-33.

Finn DF, Walsh JJ. Twenty-first century mast cell stabilizers 1. Br J Pharmacol 2013 Sep; 170(1):23-37.

Caslin HL, Kiwanuka KN, Haque TT et al. Controlling Mast Cell Activation and Homeostasis: Work Influenced by Bill Paul That Continues Today. Front Immunol 2018; 9:868.

Heinrich MC, Griffith DJ, Druker BJ, Wait CL, Ott KA, Zigler AJ. Inhibition of c-kit receptor tyrosine kinase activity by STI 571, a selective tyrosine kinase inhibitor 1. Blood 2000; 96(3):925-932.

Gotlib J, Kluin-Nelemans HC, George TI et al. Efficacy and Safety of Midostaurin in Advanced Systemic Mastocytosis. N Engl J Med 2016; 374(26):2530-2541.

Theoharides TC, Sieghart W, Greengard P, Douglas WW. Antiallergic drug cromolyn may inhibit histamine secretion by regulating phosphorylation of a mast cell protein. Science 1980; 207(4426):80-82.

Oka T, Kalesnikoff J, Starkl P, Tsai M, Galli SJ. Evidence questioning cromolyn’s effectiveness and selectivity as a ‘mast cell stabilizer’ in mice. Lab Invest 2012; 92(10):1472-1482.

Thangam EB, Jemima EA, Singh H et al. The Role of Histamine and Histamine Receptors in Mast Cell-Mediated Allergy and Inflammation: The Hunt for New Therapeutic Targets. Front Immunol 2018; 9:1873.

Duguay BA, Lu L, Arizmendi N, Unsworth LD, Kulka M. The Possible Uses and Challenges of Nanomaterials in Mast Cell Research. J Immunol 2020; 204(8):2021-2032.

Duan S, Arlian BM, Nycholat CM et al. Nanoparticles Displaying Allergen and Siglec-8 Ligands Suppress IgE-FcepsilonRI-Mediated Anaphylaxis and Desensitize Mast Cells to Subsequent Antigen Challenge. J Immunol 2021; 206(10):2290-2300.

Podder I, Jaiswal S, Das A. Dietary strategies for chronic spontaneous urticaria: an evidence-based review. Int J Dermatol 2023 Feb;62(2):143-153. doi: 10.1111/ijd.15988

Li Y, Cao Z, Guo J, Li Q, Su J. Effectsof Serum Vitamin D Levels and Vitamin D Supplementation on Urticaria: A Systematic Review and Meta-Analysis. Int J Environ Res Public Health 2021; 18(9).

Theoharides TC. Vitamin D and Atopy. Clin Ther 2017 May; 39(5):880-883. doi: 10.1016/j.clinthera.2017.03.002.

Zhao JW, Ping JD, Wang YF et al. Vitamin D suppress the production of vascular endothelial growth factor in mast cell by inhibiting PI3K/Akt/p38 MAPK/HIF-1alpha pathway in chronic spontaneous urticaria. Clin Immunol 2020; 215:108444.

Shi Y, Zhou S, Zheng Q et al. Systematic reviews of pharmacological and nonpharmacological treatments for patients with chronic urticaria: An umbrella systematic review. Medicine (Baltimore) 2019;98(20):e15711.

Yanai K, Yoshikawa T, Church MK. Efficacy and Safety of Non-brain Penetrating H1-Antihistamines for the Treatment of Allergic Diseases. Curr Top Behav Neurosci 2021.

Iriarte SP, Armisen M, Usero-Barcena T et al. Efficacy and Safety of Up-dosing Antihistamines in Chronic Spontaneous Urticaria: A Systematic Review of the Literature. J Investig Allergol Clin Immunol 2021;31(4):282-291.

Sarti L, Barni S, Giovannini M, Liccioli G, Novembre E, Mori F. Efficacy and tolerability of the updosing of second-generation non-sedating H1 antihistamines in children with chronic spontaneous urticaria. Pediatr Allergy Immunol 2021; 32(1):153-160.

Gonzalez-Nunez V, Bachert C, Mullol J. Rupatadine: global safety evaluation in allergic rhinitis and urticaria. Expert Opin Drug Saf 2016; 15(10):1439-1448.

Merlos M, Giral M, Balsa D et al. Rupatadine, a new potent, orally active dual antagonist of histamine and platelet-activating factor (PAF). J Pharmacol Exp Ther 1997; 280(1):114-121.

Church MK. Efficacy and tolerability of rupatadine at four times the recommended dose against histamine- and platelet-activating factor-induced flare responses and ex vivo platelet aggregation in healthy males. Br J Dermatol 2010; 163(6):1330-1332.

Sanchez-Borges M, Ansotegui I, Jimenez JM, Rojo MI, Serrano C, Yanez A. Comparative efficacy of non-sedating antihistamine updosing in patients with chronic urticaria. World Allergy Organ J 2014; 7(1):33.

Phinyo P, Koompawichit P, Nochaiwong S, Tovanabutra N, Chiewchanvit S, Chuamanochan M. Comparative Efficacy and Acceptability of Licensed Dose Second-Generation Antihistamines in Chronic Spontaneous Urticaria: A Network Meta-Analysis. J Allergy Clin Immunol Pract 2021;9(2):956-970.

Vasiadi M, Kalogeromitros D, Kempuraj D et al. Rupatadine inhibits proinflammatory mediator secretion from human mast cells triggered by different stimuli. Int Arch Allergy Immunol 2010;151(1):38-45.

Munoz-Cano R, insua-Enrich E, Torres- Atencio I et al. Effects of Rupatadine on Platelet- Activating Factor-Induced Human Mast Cell Degranulation Compared With Desloratadine and Levocetirizine (The MASPAF Study). J Investig Allergol Clin Immunol 2017; 27(3):161-168.

Agache I, Rocha C, Pereira A et al. Efficacy and safety of treatment with omalizumab for chronic spontaneous urticaria: A systematic review for the EAACI Biologicals Guidelines. Allergy 2021;76(1):59-70.

Amin K. The role of mast cells in allergic inflammation. Respir Med 2012; 106(1):9-14.

Bachelet I, Levi-Schaffer F, Mekori YA. Mast cells: not only in allergy. Immunol Allergy Clin North Am 2006; 26(3):407-425.

Theoharides TC, Cochrane DE. Critical role of mast cells in inflammatory diseases and the effect of acute stress. J Neuroimmunol 2004; 146(1-2):1-12.

Galli SJ, Tsai M, Piliponsky AM. The development of allergic inflammation. Nature 2008; 454(7203):445-454.

Mast cells in atopic diseases: more than just histamine

Published

2022-03-01

How to Cite

1.
Theoharides TC. Mast Cells in Atopic Diseases: More Than Just Histamine. Can Allergy Immunol Today [Internet]. 2022 Mar. 1 [cited 2024 Sep. 16];2(s03):3–15. Available from: https://canadianallergyandimmunologytoday.com/article/view/1-s03-theoharides

Issue

Section

Articles