Helicobacter pylori Infection and Extragastric Diseases—A Focus on the Central Nervous System
Abstract
:1. Introduction
2. Helicobacter pylori Characteristics
3. Gut–Brain Axis
4. Pathophysiology of Helicobacter pylori Infection and CNS Diseases
Pathophysiology of Helicobacter pylori Infection
5. Central Nervous System Diseases and Helicobacter pylori Infection
5.1. Parkinson’s Disease
5.2. Alzheimer’s Disease
5.3. Multiple Sclerosis
5.4. Guillain–Barré Syndrome
5.5. Bickerstaff Brainstem Encephalitis
5.6. Devic Syndrome
5.7. Stroke
5.8. Migraine Headaches
6. Conclusions
Author Contributions
Funding
Conflicts of Interest
Abbreviations
AD | Alzheimer’s disease |
AIDP | Acute inflammatory demyelinating polyneuropathy |
AMAN | Acute motor axonal neuropathy |
AMSAN | Acute motor and sensory axonal neuropathy |
ANS | Autonomic nervous system |
ApoE4 | Apolioprotein E4 |
BBB | Brain-blood-barrier |
BBE | Bickerstaff brainstem encephalitis |
BMD-MCs | Bone marrow-derived mast cells |
CIS | Clinically isolated syndrome |
CMV | Cytomegalovirus |
CNS | Central nervous system |
CRF | Corticotropin releasing factor |
CSF | Cerebrospinal fluid |
EAE | Experimental autoimmune encephalitis |
EBV | Epstein–Barr virus |
EDSS | Expanded Disability Status Scale |
ENS | Enteric nervous system |
GBA | Gut–brain axis |
GBS | Guillain-Barré syndrome |
HPA | Hypothalamus–pituitary axis |
H. pylori | Helicobacter pylori |
HSP60 | Anti- H. pylori heat shock protein 60 |
HSPs | Heat shock proteins |
L-dopa | L-3,4-dihydroxyphenylalanine |
MALT-lymphoma | Marginal zone/mucosa associated lymphoid tissue lymphoma |
MAMPs | Microbial-associated molecular patterns |
MFS | Miller Fisher syndrome |
MS | Multiple sclerosis |
nAChRs | α7-Nicotinic acetylcholine receptors |
NMO | Neuromyelitis optica |
PD | Parkinson’s disease |
SCFAs | Short-chain fatty acids |
SPMS | Secondary progressive multiple sclerosis |
References
- Hooi, J.K.Y.; Lai, W.Y.; Ng, W.K.; Suen, M.M.Y.; Underwood, F.E.; Tanyingoh, D.; Malfertheiner, P.; Graham, D.Y.; Wong, V.W.S.; Wu, J.C.Y.; et al. Global Prevalence of Helicobacter pylori Infection: Systematic Review and Meta-Analysis. Gastroenterology 2017, 153, 420–429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thung, I.; Aramin, H.; Vavinskaya, V.; Gupta, S.; Park, J.Y.; Crowe, S.E.; Valasek, M.A. Review article: The global emergence of Helicobacter pylori antibiotic resistance. Aliment. Pharmacol. Ther. 2016, 43, 514–533. [Google Scholar] [CrossRef] [Green Version]
- Parsonnet, J.; Friedman, G.D.; Vandersteen, D.P.; Chang, Y.; Vogelman, J.H.; Orentreich, N.; Sibley, R.K. Helicobacter pyloriInfection and the Risk of Gastric Carcinoma. N. Engl. J. Med. 1991, 325, 1127–1131. [Google Scholar] [CrossRef] [PubMed]
- Suerbaum, S.; Michetti, P. Helicobacter pylori Infection. N. Engl. J. Med. 2002, 347, 1175–1186. [Google Scholar] [CrossRef] [Green Version]
- Kusters, J.G.; van Vliet, A.H.M.; Kuipers, E.J. Pathogenesis of Helicobacter pylori infection. Clin. Microbiol. Rev. 2006, 19, 449–490. [Google Scholar] [CrossRef] [Green Version]
- Dunn, B.E.; Cohen, H.; Blaser, M.J. Helicobacter pylori. Clin. Microbiol. Rev. 1997, 10, 720–741. [Google Scholar] [CrossRef] [PubMed]
- Kayali, S.; Manfredi, M.; Gaiani, F.; Bianchi, L.; Bizzarri, B.; Leandro, G.; Di Mario, F.; De’Angelis, G.L. Helicobacter pylori, trans-mission routes and recurrence of infection: State of the art. Acta Biomed. 2018, 89, 72–76. [Google Scholar] [CrossRef]
- Brown, L.M. Helicobacter pylori: Epidemiology and routes of transmission. Epidemiol. Rev. 2000, 22, 283–297. [Google Scholar] [CrossRef] [PubMed]
- Dimola, S.; Caruso, M.L. Helicobacter pylori in animals affecting the human habitat through the food chain. Anticancer Res. 1999, 19, 3889–3894. [Google Scholar]
- Aziz, R.K.; Khalifa, M.M.; Sharaf, R.R. Contaminated water as a source of Helicobacter pylori infection: A review. J. Adv. Res. 2015, 6, 539–547. [Google Scholar] [CrossRef] [Green Version]
- Uemura, N.; Okamoto, S.; Yamamoto, S.; Matsumura, N.; Yamaguchi, S.; Yamakido, M.; Taniyama, K.; Sasaki, N.; Schlemper, R.J. Helicobacter pyloriInfection and the Development of Gastric Cancer. N. Engl. J. Med. 2001, 345, 784–789. [Google Scholar] [CrossRef]
- Crowe, S.E. Helicobacter pylori Infection. N. Engl. J. Med. 2019, 380, 1158–1165. [Google Scholar] [CrossRef]
- Machlowska, J.; Kapusta, P.; Baj, J.; Morsink, F.H.M.; Wołkow, P.; Maciejewski, R.; Offerhaus, G.J.A.; Sitarz, R. High-Throughput Se-quencing of Gastric Cancer Patients: Unravelling Genetic Predispositions Towards an Early-Onset Subtype. Cancers 2020, 12, 1981. [Google Scholar] [CrossRef]
- Budzyński, J.; Kłopocka, M. Brain-gut axis in the pathogenesis of Helicobacter pylori infection. World J. Gastroenterol. 2014, 20, 5212–5225. [Google Scholar] [CrossRef] [PubMed]
- Kountouras, J.; Zavos, C.; Polyzos, S.A.; Deretzi, G. The gut-brain axis: Interactions between Helicobacter pylori and enteric and central nervous systems. Ann. Gastroenterol. 2015, 28, 506. [Google Scholar] [PubMed]
- Cryan, J.F.; O’Mahony, S.M. The microbiome-gut-brain axis: From bowel to behavior. Neurogastroenterol. Motil. 2011, 23, 187–192. [Google Scholar] [CrossRef]
- Mulak, A.; Bonaz, B. Brain-gut-microbiota axis in Parkinson’s disease. World J. Gastroenterol. 2015, 21, 10609. [Google Scholar] [CrossRef]
- Franceschi, F.; Gasbarrini, A.; Polyzos, S.A.; Kountouras, J. Extragastric Diseases and Helicobacter pylori. Helicobacter 2015, 20, 40–46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Honjo, K.; van Reekum, R.; Verhoeff, N.P.L.G. Alzheimer’s disease and infection: Do infectious agents contribute to progression of Alzheimer’s disease? Alzheimer’s Dement. 2009, 5, 348–360. [Google Scholar] [CrossRef]
- Kountouras, J.; Tsolaki, M.; Gavalas, E.; Boziki, M.; Zavos, C.; Karatzoglou, P.; Chatzopoulos, D.; Venizelos, I. Relationship between Helicobacter pylori infection and Alzheimer disease. Neurology 2006, 66, 938–940. [Google Scholar] [CrossRef] [PubMed]
- Gavalas, E.; Kountouras, J.; Deretzi, G.; Boziki, M.; Grigoriadis, N.; Zavos, C.; Venizelos, I. Helicobacter pylori and multiple sclerosis. J. Neuroimmunol. 2007, 188, 187–189. [Google Scholar] [CrossRef] [PubMed]
- Park, A.M.; Omura, S.; Fujita, M.; Sato, F.; Tsunoda, I. Helicobacter pylori and gut microbiota in multiple sclerosis versus Alzheimer’s disease: 10 pitfalls of microbiome studies. Clin. Exp. Neuroimmunol. 2017, 8, 215–232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dardiotis, E.; Sokratous, M.; Tsouris, Z.; Siokas, V.; Mentis, A.A.; Aloizou, A.M.; Michalopoulou, A.; Bogdanos, D.P.; Xiromerisiou, G.; Deretzi, G.; et al. Association between Helicobacter pylori infection and Guillain-Barré Syndrome: A meta-analysis. Eur. J. Clin. Investig. 2020, 50, e13218. [Google Scholar] [CrossRef] [PubMed]
- Kountouras, J.; Deretzi, G.; Zavos, C.; Tsiptsios, D.; Gavalas, E.; Vardaka, E.; Polyzos, S.A.; Klonizakis, P.; Kyriakou, P.; Pοlyzos, S.A. Helicobacter pylori infection may trigger Guillain-Barré syndrome, Fisher syndrome and Bickerstaff brainstem encephalitis. J. Neurol. Sci. 2011, 305, 167–168. [Google Scholar] [CrossRef] [PubMed]
- Schistosomes, liver flukes and Helicobacter pylori. IARC Working Group on the Evaluation of Carcinogenic Risks to Humans. Lyon, 7-14 June 1994. IARC Monogr. Eval. Carcinog. Risks Hum. 1994, 61, 1–241.
- Burucoa, C.; Axon, A. Epidemiology of Helicobacter pylori infection. Helicobacter 2017, 22, e12403. [Google Scholar] [CrossRef]
- Baj, J.; Korona-Głowniak, I.; Forma, A.; Maani, A.; Sitarz, E.; Rahnama-Hezavah, M.; Radzikowska, E.; Portincasa, P. Mechanisms of the Epithelial–Mesenchymal Transition and Tumor Microenvironment in Helicobacter pylori-Induced Gastric Cancer. Cells 2020, 9, 1055. [Google Scholar] [CrossRef] [Green Version]
- Baj, J.; Brzozowska, K.; Forma, A.; Maani, A.; Sitarz, E.; Portincasa, P. Immunological Aspects of the Tumor Microenvironment and Epithelial-Mesenchymal Transition in Gastric Carcinogenesis. Int. J. Mol. Sci. 2020, 21, 2544. [Google Scholar] [CrossRef] [Green Version]
- Kozak, J.; Forma, A.; Czeczelewski, M.; Kozyra, P.; Sitarz, E.; Radzikowska-Büchner, E.; Sitarz, M.; Baj, J. Inhibition or Reversal of the Epithelial-Mesenchymal Transition in Gastric Cancer: Pharmacological Approaches. Int. J. Mol. Sci. 2021, 22, 277. [Google Scholar] [CrossRef]
- Baj, J.; Forma, A.; Sitarz, M.; Portincasa, P.; Garruti, G.; Krasowska, D.; Maciejewski, R. Helicobacter pylori Virulence Factors-Mechanisms of Bacterial Pathogenicity in the Gastric Microenvironment. Cells 2020, 10, 27. [Google Scholar] [CrossRef]
- Chen, Z.; Maqbool, J.; Sajid, F.; Hussain, G.; Sun, T. Human gut microbiota and its association with pathogenesis and treatments of neurodegenerative diseases. Microb. Pathog. 2021, 150, 104675. [Google Scholar] [CrossRef] [PubMed]
- Collins, S.M.; Surette, M.; Bercik, P. The interplay between the intestinal microbiota and the brain. Nat. Rev. Microbiol. 2012, 10, 735–742. [Google Scholar] [CrossRef] [PubMed]
- Sharkey, K.A.; Beck, P.L.; McKay, D.M. Neuroimmunophysiology of the gut: Advances and emerging concepts focusing on the epithelium. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 765–784. [Google Scholar] [CrossRef] [PubMed]
- Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.; Sandhu, K.V.; Bastiaanssen, T.F.; Boehme, M.; Codagnone, M.G.; Cussotto, S.; Fulling, C.; Golubeva, A.V.; et al. The microbiota-gut-brain axis. Physiol. Rev. 2019, 99, 1877–2013. [Google Scholar] [CrossRef] [PubMed]
- Udit, S.; Gautron, L. Molecular anatomy of the gut-brain axis revealed with transgenic technologies: Implications in metabolic research. Front. Neurosci. 2013, 7, 134. [Google Scholar] [CrossRef] [Green Version]
- Hattori, N.; Yamashiro, Y. The Gut-Brain Axis. Ann. Nutr. Metab. 2021, 1–3. [Google Scholar] [CrossRef] [PubMed]
- Forsythe, P.; Kunze, W.A. Voices from within: Gut microbes and the CNS. Cell. Mol. Life Sci. 2013, 70, 55–69. [Google Scholar] [CrossRef]
- Mayer, E.A.; Tillisch, K.; Gupta, A. Gut/brain axis and the microbiota. J. Clin. Investig. 2015, 125, 926–938. [Google Scholar] [CrossRef]
- Delvaux, M. Alterations of sensori-motor functions of the digestive tract in the pathophysiology of irritable bowel syndrome. Best Pract. Res. Clin. Gastroenterol. 2004, 18, 747–771. [Google Scholar] [CrossRef]
- Fasano, A.; Shea-Donohue, T. Mechanisms of disease: The role of intestinal barrier function in the pathogenesis of gastrointestinal autoimmune diseases. Nat. Clin. Pract. Gastroenterol. Hepatol. 2005, 2, 416–422. [Google Scholar] [CrossRef]
- Wehrwein, E.A.; Orer, H.S.; Barman, S.M. Overview of the Anatomy, Physiology, and Pharmacology of the Autonomic Nervous System. In Comprehensive Physiology; John Wiley & Sons, Inc.: Hoboken, NJ, USA, 2016; Volume 6, pp. 1239–1278. [Google Scholar] [CrossRef]
- Bonaz, B.L.; Bernstein, C.N. Brain-gut interactions in inflammatory bowel disease. Gastroenterology 2013, 144, 36–49. [Google Scholar] [CrossRef] [Green Version]
- Grenham, S.; Clarke, G.; Cryan, J.F.; Dinan, T.G. Brain-gut-microbe communication in health and disease. Front. Physiol. 2011, 2, 94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dinan, T.G.; Quigley, E.M.; Ahmed, S.M.; Scully, P.; O’Brien, S.; O’Mahony, L.; Mahony, S.O.; Shanahan, F.; Keeling, P.N. Hypothalamic-Pituitary-Gut Axis Dysregulation in Irritable Bowel Syndrome: Plasma Cytokines as a Potential Biomarker? Gastroenterology 2006, 130, 304–311. [Google Scholar] [CrossRef] [PubMed]
- Clarke, G.; Grenham, S.; Scully, P.; Fitzgerald, P.J.; Moloney, R.D.; Shanahan, F.; Dinan, T.; Cryan, J. The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol. Psychiatry 2012, 18, 666–673. [Google Scholar] [CrossRef] [Green Version]
- Sgambato, D.; Capuano, A.; Sullo, M.G.; Miranda, A.; Federico, A.; Romano, M. Gut-Brain Axis in Gastric Mucosal Damage and Protection. Curr. Neuropharmacol. 2016, 14, 959–966. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pott, J.; Hornef, M. Innate immune signalling at the intestinal epithelium in homeostasis and disease. EMBO Repo. 2012, 13, 684–698. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cani, P.D.; Everard, A.; Duparc, T. Gut microbiota, enteroendocrine functions and metabolism. Curr. Opin. Pharmacol. 2013, 13, 935–940. [Google Scholar] [CrossRef]
- Reigstad, C.S.; Salmonson, C.E.; Iii, J.F.R.; Szurszewski, J.H.; Linden, D.R.; Sonnenburg, J.L.; Farrugia, G.; Kashyap, P.C. Gut microbes promote colonic serotonin production through an effect of short-chain fatty acids on enterochromaffin cells. FASEB J. 2014, 29, 1395–1403. [Google Scholar] [CrossRef] [Green Version]
- Strandwitz, P.; Kim, K.H.; Terekhova, D.; Liu, J.K.; Sharma, A.; Levering, J.; McDonald, D.; Dietrich, D.; Ramadhar, T.R.; Lekbua, A.; et al. GABA-modulating bacteria of the human gut microbiota. Nat. Microbiol. 2018, 4, 396–403. [Google Scholar] [CrossRef]
- Rhee, S.H.; Pothoulakis, C.; Mayer, E.A. Principles and clinical implications of the brain-gut-enteric microbiota axis. Nat. Rev. Gastroenterol. Hepatol. 2009, 6, 306–314. [Google Scholar] [CrossRef] [Green Version]
- Kaelberer, M.M.; Buchanan, K.L.; Klein, M.E.; Barth, B.B.; Montoya, M.M.; Shen, X.; Bohórquez, D.V. A gut-brain neural circuit for nutrient sensory transduction. Science 2018, 361, eaat5236. [Google Scholar] [CrossRef] [Green Version]
- Berthoud, H.R.; Blackshaw, L.A.; Brookes, S.J.H.; Grundy, D. Neuroanatomy of extrinsic afferents supplying the gastrointestinal tract. Neurogastroenterol. Motil. 2004, 16 (Suppl. S1), 28–33. [Google Scholar] [CrossRef] [PubMed]
- Pavlov, V.A.; Tracey, K.J. The cholinergic anti-inflammatory pathway. Brain Behav. Immunity 2005, 19, 493–499. [Google Scholar] [CrossRef] [PubMed]
- Browning, J.S.; Houseworth, J.H. Development of new symptoms following medical and surgical treatment for duodenal ulcer. Psychosom. Med. 1953, 15, 328–336. [Google Scholar] [CrossRef]
- Whitlock, F.A. Some psychiatric consequences of gastrectomy. Br. Med. J. 1961, 1, 1560–1564. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Grimonprez, A.; Raedt, R.; Baeken, C.; Boon, P.; Vonck, K. The antidepressant mechanism of action of vagus nerve stimulation: Evidence from preclinical studies. Neurosci. Biobehav. Rev. 2015, 56, 26–34. [Google Scholar] [CrossRef] [PubMed]
- Spichak, S.; Bastiaanssen, T.F.; Berding, K.; Vlckova, K.; Clarke, G.; Dinan, T.G.; Cryan, J.F. Mining microbes for mental health: Determining the role of microbial metabolic pathways in human brain health and disease. Neurosci. Biobehav. Rev. 2021, 125, 698–761. [Google Scholar] [CrossRef]
- Anderson, G.; Rodriguez, M.; Reiter, R.J. Multiple Sclerosis: Melatonin, Orexin, and Ceramide Interact with Platelet Activation Coagulation Factors and Gut-Microbiome-Derived Butyrate in the Circadian Dysregulation of Mitochondria in Glia and Immune Cells. Int. J. Mol. Sci. 2019, 20, 5500. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Boziki, M.; Grigoriadis, N.; Papaefthymiou, A.; Doulberis, M.; Polyzos, S.A.; Gavalas, E.; Deretzi, G.; Karafoulidou, E.; Kesidou, E.; Taloumtzis, C.; et al. The trimebutine effect on Helicobacter pylori-related gastrointestinal tract and brain disorders: A hypothesis. Neurochem. Int. 2021, 144, 104938. [Google Scholar] [CrossRef] [PubMed]
- Engelhardt, B.; Liebner, S. Novel insights into the development and maintenance of the blood-brain barrier. Cell Tissue Res. 2014, 355, 687–699. [Google Scholar] [CrossRef] [Green Version]
- Bacon, C.M.; Du, M.Q.; Dogan, A. Mucosa-associated lymphoid tissue (MALT) lymphoma: A practical guide for pathologists. J Clin Pathol. 2007, 60, 361–372. [Google Scholar] [CrossRef] [Green Version]
- Mayer, E.A.; Tillisch, K.; Bradesi, S. Review article: Modulation of the brain-gut axis as a therapeutic approach in gastrointestinal disease. Aliment. Pharmacol. Ther. 2006, 24, 919–933. [Google Scholar] [CrossRef]
- Sticlaru, L.; Stăniceanu, F.; Cioplea, M.; Nichita, L.; Bastian, A.; Micu, G.; Popp, C. Dangerous Liaison: Helicobacter pylori, Ganglionitis, and Myenteric Gastric Neurons: A Histopathological Study. Anal. Cell. Pathol. (Amst.) 2019, 2019, 3085181. [Google Scholar] [CrossRef] [Green Version]
- Gravina, A.G.; Zagari, R.M.; De Musis, C.; Romano, L.; Loguercio, C.; Romano, M. Helicobacter pylori and extragastric diseases: A review. World J. Gastroenterol. 2018, 24, 3204–3221. [Google Scholar] [CrossRef]
- Carabotti, M.; Scirocco, A.; Maselli, M.A.; Severi, C. The gut-brain axis: Interactions between enteric microbiota, central and enteric nervous systems. Ann. Gastroenterol. 2015, 28, 203–209. [Google Scholar] [PubMed]
- Forsythe, P.; Bienenstock, J.; Kunze, W.A. Vagal pathways for microbiome-brain-gut axis communication. Adv. Exp. Med. Biol. 2014, 817, 115–133. [Google Scholar] [CrossRef]
- Deretzi, G.; Kountouras, J.; Polyzos, S.A.; Zavos, C.; Giartza-Taxidou, E.; Gavalas, E.; Tsiptsios, I. Gastrointestinal immune system and brain dialogue implicated in neuroinflammatory and neurodegenerative diseases. Curr. Mol. Med. 2011, 11, 696–707. [Google Scholar] [CrossRef] [PubMed]
- Deretzi, G.; Kountouras, J.; Grigoriadis, N.; Zavos, C.; Chatzigeorgiou, S.; Koutlas, E.; Tsiptsios, I. From the “little brain” gastrointestinal infection to the “big brain” neuroinflammation: A proposed fast axonal transport pathway involved in multiple sclerosis. Med. Hypotheses 2009, 73, 781–787. [Google Scholar] [CrossRef] [PubMed]
- Doulberis, M.; Kotronis, G.; Thomann, R.; Polyzos, S.A.; Boziki, M.; Gialamprinou, D.; Deretzi, G.; Katsinelos, P.; Kountouras, J. Review: Impact of Helicobacter pylori on Alzheimer’s disease: What do we know so far? Helicobacter 2018, 23, e12451. [Google Scholar] [CrossRef]
- McClain, M.S.; Cover, T.L. Expression of Helicobacter pylori vacuolating toxin in Escherichia coli. Infect Immun. 2003, 71, 2266–2271. [Google Scholar] [CrossRef] [Green Version]
- Gorlé, N.; Bauwens, E.; Haesebrouck, F.; Smet, A.; Vandenbroucke, R.E. Helicobacter and the Potential Role in Neurological Disorders: There Is More Than Helicobacter pylori. Front. Immunol. 2021, 11, 584165. [Google Scholar] [CrossRef] [PubMed]
- Yağci, M.; Yamaç, K.; Acar, K.; Cingi, E.; Kitapçi, M.; Haznedar, R. Gastric emptying in patients with vitamin B(12) deficiency. Eur. J. Nucl. Med. Mol. Imaging 2002, 29, 1125–1127. [Google Scholar] [CrossRef]
- Tsay, F.W.; Hsu, P.I. H. pylori infection and extra-gastroduodenal diseases. J. Biomed. Sci. 2018, 25, 65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Miller, A.; Korem, M.; Almog, R.; Galboiz, Y. Vitamin B12, demyelination, remyelination and repair in multiple sclerosis. J. Neurol. Sci. 2005, 233, 93–97. [Google Scholar] [CrossRef]
- Yahn, G.B.; Abato, J.E.; Jadavji, N.M. Role of vitamin B12 deficiency in ischemic stroke risk and outcome. Neural Regen. Res. 2021, 16, 470–474. [Google Scholar] [CrossRef]
- Altschuler, E. Gastric Helicobacter pylori infection as a cause of idiopathic Parkinson disease and non-arteric anterior optic ischemic neuropathy. Med. Hypotheses 1996, 47, 413–414. [Google Scholar] [CrossRef]
- Charlett, A.; Dobbs, R.J.; Dobbs, S.M.; Weller, C.; Brady, P.; Peterson, D.W. Parkinsonism: Siblings share Helicobacter pylori seropositivity and facets of syndrome. Acta Neurol. Scand. 1999, 99, 26–35. [Google Scholar] [CrossRef] [PubMed]
- Bjarnason, I.T.; Charlett, A.; Dobbs, R.J.; Dobbs, S.M.; Ibrahim, M.A.; Kerwin, R.W.; Mahler, R.F.; Oxlade, N.L.; Peterson, D.W.; Plant, J.M.; et al. Role of chronic infection and inflammation in the gastrointestinal tract in the etiology and pathogenesis of idiopathic parkinsonism. Part 2: Response of facets of clinical idiopathic parkinsonism to Helicobacter pylori eradication. A randomized, double-blind, placebo-controlled efficacy study. Helicobacter 2005, 10, 276–287. [Google Scholar] [CrossRef] [PubMed]
- Shen, X.; Yang, H.; Wu, Y.; Zhang, D.; Jiang, H. Meta-analysis: Association of Helicobacter pylori infection with Parkinson’s diseases. Helicobacter 2017, 22, e12398. [Google Scholar] [CrossRef]
- Huang, H.K.; Wang, J.H.; Lei, W.Y.; Chen, C.L.; Chang, C.Y.; Liou, L.S. Helicobacter pylori infection is associated with an increased risk of Parkinson’s disease: A population-based retrospective cohort study. Parkinsonism Relat. Disord. 2018, 47, 26–31. [Google Scholar] [CrossRef] [PubMed]
- Gravina, A.G.; Priadko, K.; Ciamarra, P.; Granata, L.; Facchiano, A.; Miranda, A.; Dallio, M.; Federico, A.; Romano, M. Extra-Gastric Manifestations of Helicobacter pylori Infection. J. Clin. Med. 2020, 9, 3887. [Google Scholar] [CrossRef]
- Béraud, D.; Maguire-Zeiss, K.A. Misfolded α-synuclein and Toll-like receptors: Therapeutic targets for Parkinson’s disease. Parkinsonism Relat. Disord. 2012, 18 (Suppl. S1), S17–S20. [Google Scholar] [CrossRef] [Green Version]
- Mridula, K.R.; Borgohain, R.; Chandrasekhar Reddy, V.; Bandaru, V.; Suryaprabha, T. Association of Helicobacter pylori with Parkinson’s Disease. J. Clin. Neurol. (Seoul, Korea) 2017, 13, 181–186. [Google Scholar] [CrossRef] [Green Version]
- Fasano, A.; Bove, F.; Gabrielli, M.; Petracca, M.; Zocco, M.A.; Ragazzoni, E.; Barbaro, F.; Piano, C.; Fortuna, S.; Tortora, A.; et al. The role of small intestinal bacterial overgrowth in Parkinson’s disease. Mov. Disord. 2013, 28, 1241–1249. [Google Scholar] [CrossRef] [PubMed]
- Huang, W.S.; Yang, T.Y.; Shen, W.C.; Lin, C.L.; Lin, M.C.; Kao, C.H. Association between Helicobacter pylori infection and dementia. J. Clin. Neurosci. 2014, 21, 1355–1358. [Google Scholar] [CrossRef] [PubMed]
- Roubaud Baudron, C.; Letenneur, L.; Langlais, A.; Buissonnière, A.; Mégraud, F.; Dartigues, J.F.; Salles, N.; Personnes Agées QUID Study. Does Helicobacter pylori infection increase incidence of dementia? The Personnes Agées QUID Study. J. Am. Geriatr. Soc. 2013, 61, 74–78. [Google Scholar] [CrossRef]
- Beydoun, M.A.; Beydoun, H.A.; Shroff, M.R.; Kitner-Triolo, M.H.; Zonderman, A.B. Helicobacter pylori seropositivity and cognitive performance among US adults: Evidence from a large national survey. Psychosom. Med. 2013, 75, 486–496. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kountouras, J.; Boziki, M.; Gavalas, E.; Zavos, C.; Deretzi, G.; Grigoriadis, N.; Tsolaki, M.; Chatzopoulos, D.; Katsinelos, P.; Tzilves, D.; et al. Increased cerebrospinal fluid Helicobacter pylori antibody in Alzheimer’s disease. Int. J. Neurosci. 2009, 119, 765–777. [Google Scholar] [CrossRef]
- Kountouras, J.; Boziki, M.; Gavalas, E.; Zavos, C.; Deretzi, G.; Chatzigeorgiou, S.; Katsinelos, P.; Grigoriadis, N.; Giartza-Taxidou, E.; Venizelos, I. Five-year survival after Helicobacter pylori eradication in Alzheimer disease patients. Cogn. Behav. Neurol. 2010, 23, 199–204. [Google Scholar] [CrossRef]
- Kountouras, J.; Boziki, M.; Gavalas, E.; Zavos, C.; Grigoriadis, N.; Deretzi, G.; Tzilves, D.; Katsinelos, P.; Tsolaki, M.; Chatzopoulos, D.; et al. Eradication of Helicobacter pylori may be beneficial in the management of Alzheimer’s disease. J. Neurol. 2009, 256, 758–767. [Google Scholar] [CrossRef]
- Goni, E.; Franceschi, F. Helicobacter pylori and extragastric diseases. Helicobacter 2016, 21 (Suppl. S1), 45–48. [Google Scholar] [CrossRef]
- Chang, Y.P.; Chiu, G.F.; Kuo, F.C.; Lai, C.L.; Yang, Y.H.; Hu, H.M.; Chang, P.Y.; Chen, C.Y.; Wu, D.C.; Yu, F.J. Eradication of Helicobacter pylori Is Associated with the Progression of Dementia: A PopulationBased Study. Gastroenterol. Res. Pract. 2013, 2013, 175729. [Google Scholar] [CrossRef] [Green Version]
- Shiota, S.; Murakami, K.; Yoshiiwa, A.; Yamamoto, K.; Ohno, S.; Kuroda, A.; Mizukami, K.; Hanada, K.; Okimoto, T.; Kodama, M.; et al. The relationship between Helicobacter pylori infection and Alzheimer’s disease in Japan. J. Neurol. 2011, 258, 1460–1463. [Google Scholar] [CrossRef] [Green Version]
- Santos, C.Y.; Snyder, P.J.; Wu, W.C.; Zhang, M.; Echeverria, A.; Alber, J. Pathophysiologic relationship between Alzheimer’s disease, cerebrovascular disease, and cardiovascular risk: A review and synthesis. Alzheimers Dement. (Amst.) 2017, 7, 69–87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kira, J.-I.; Isobe, N. Helicobacter pylori infection and demyelinating disease of the central nervous system. J. Neuroimmunol. 2019, 329, 14–19. [Google Scholar] [CrossRef] [PubMed]
- Ebers, G.C. Environmental factors and multiple sclerosis. Lancet Neurol. 2008, 7, 268–277. [Google Scholar] [CrossRef]
- Sand, I.K.; Zhu, Y.; Ntranos, A.; Clemente, J.C.; Cekanaviciute, E.; Brandstadter, R.; Crabtree-Hartman, E.; Singh, S.; Bencosme, Y.; Debelius, J.; et al. Disease-modifying therapies alter gut microbial composition in MS. Neurol.-Neuroimmunol. Neuroinflammation 2018, 6, e517. [Google Scholar] [CrossRef] [Green Version]
- Cossu, D.; Yokoyama, K.; Hattori, N. Bacteria–host interactions in multiple sclerosis. Front. Microbiol. 2018, 9, 2966. [Google Scholar] [CrossRef]
- Li, W.; Minohara, M.; Su, J.J.; Matsuoka, T.; Osoegawa, M.; Ishizu, T.; Kira, J. Helicobacter pylori infection is a potential protective factor against conventional multiple sclerosis in the Japanese population. J. Neuroimmunol. 2007, 184, 227–231. [Google Scholar] [CrossRef]
- Cook, K.W.; Crooks, J.; Hussain, K.; O’Brien, K.; Braitch, M.; Kareem, H.; Constantinescu, C.S.; Robinson, K.; Gran, B. Helicobacter pylori infection reduces disease severity in an experimental model of multiple sclerosis. Front. Microbiol. 2015, 6, 52. [Google Scholar] [CrossRef] [Green Version]
- Smyk, D.S.; Koutsoumpas, A.L.; Mytilinaiou, M.G.; Rigopoulou, E.I.; Sakkas, L.I.; Bogdanos, D.P. Helicobacter pylori and autoimmune disease: Cause or bystander. World J. Gastroenterol. 2014, 20, 613–629. [Google Scholar] [CrossRef] [PubMed]
- Ram, M.; Barzilai, O.; Shapira, Y.; Anaya, J.-M.; Tincani, A.; Stojanovich, L.; Bombardieri, S.; Bizzaro, N.; Kivity, S.; Levin, N.A.; et al. Helicobacter pylori serology in autoimmune diseases – fact or fiction? Clin. Chem. Lab. Med. 2013, 51, 1075–1082. [Google Scholar] [CrossRef] [PubMed]
- Relationship between Helicobacter pylori Infection and Multiple Sclerosis—ScienceOpen n.d. Available online: https://www.scienceopen.com/document?vid=4c548e26-5a70-4735-8991-80c42e3ea4ff (accessed on 23 April 2021).
- Long, Y.; Gao, C.; Qiu, W.; Hu, X.; Shu, Y.; Peng, F.; Lu, Z. Helicobacter pyloriInfection in Neuromyelitis Optica and Multiple Sclerosis. Neuroimmunomodulation 2013, 20, 107–112. [Google Scholar] [CrossRef]
- Deretzi, G.; Gavalas, E.; Boziki, M.; Tsiptsios, D.; Polyzos, S.A.; Venizelos, I.; Zavos, C.; Koutlas, E.; Tsiptsios, I.; Katsinelos, P.; et al. Impact ofHelicobacter pylorion multiple sclerosis-related clinically isolated syndrome. Acta Neurol. Scand. 2015, 133, 268–275. [Google Scholar] [CrossRef] [PubMed]
- Gerges, S.E.; Alosh, T.K.; Khalil, S.H.; el Din, M.M.W. Relevance of Helicobacter pylori infection in Egyptian multiple sclerosis patients. Egypt. J. Neurol. Psychiatry Neurosurg. 2018, 54, 41. [Google Scholar] [CrossRef] [PubMed]
- Fourie, K.R.; Wilson, H.L. Understanding groel and dnak stress response proteins as antigens for bacterial diseases. Vaccines 2020, 8, 773. [Google Scholar] [CrossRef] [PubMed]
- Loshaj-Shala, A.; Regazzoni, L.; Daci, A.; Orioli, M.; Brezovska, K.; Panovska, A.P.; Beretta, G.; Suturkova, L. Guillain Barré syndrome (GBS): New insights in the molecular mimicry between C. jejuni and human peripheral nerve (HPN) proteins. J. Neuroimmunol. 2015, 289, 168–176. [Google Scholar] [CrossRef]
- Elfaitouri, A.; Herrmann, B.; Bölin-Wiener, A.; Wang, Y.; Gottfries, C.-G.; Zachrisson, O.; Pipkorn, R.; Rönnblom, L.; Blomberg, J. Epitopes of Microbial and Human Heat Shock Protein 60 and Their Recognition in Myalgic Encephalomyelitis. PLoS ONE 2013, 8, e81155. [Google Scholar] [CrossRef] [PubMed]
- Chiricosta, L.; Gugliandolo, A.; Bramanti, P.; Mazzon, E. Could the heat shock proteins 70 family members exacerbate the immune response in multiple sclerosis? An in silico study. Genes 2020, 11, 615. [Google Scholar] [CrossRef]
- Yao, G.; Wang, P.; Luo, X.D.; Yu, T.M.; Harris, R.A.; Zhang, X.M. Meta-analysis of association between Helicobacter pylori infection and multiple sclerosis. Neurosci. Lett. 2016, 620, 1–7. [Google Scholar] [CrossRef]
- Pedrini, M.J.F.; Seewann, A.; Bennett, K.A.; Wood, A.J.T.; James, I.; Burton, J.; Marshall, B.J.; Carroll, W.M.; Kermode, A.G. Helicobacter pyloriinfection as a protective factor against multiple sclerosis risk in females. J. Neurol. Neurosurg. Psychiatry 2015, 86, 603–607. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mohebi, N.; Mamarabadi, M.; Moghaddasi, M. Relation of Helicobacter pylori infection and multiple sclerosis in Iranian patients. Neurol. Int. 2013, 5, 31–33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kiani, S.; Vakilian, A.; Kamiab, Z.; Shamsizadeh, A. Correlation of dietary intake and Helicobacter pylori infection with multiple sclerosis, a case-control study in Rafsanjan, Iran, 2017–2018. Qatar Med. J. 2021, 2020, 45. [Google Scholar] [CrossRef]
- Ranjbar, R.; Karampoor, S.; Jalilian, F.A. The protective effect of Helicobacter pylori infection on the susceptibility of multiple sclerosis. J. Neuroimmunol. 2019, 337, 577069. [Google Scholar] [CrossRef]
- Robinson, K.; Stephens, J.; Constantinescu, C.S.; Gran, B. Helicobacter pylori, experimental autoimmune encephalomyelitis, and multiple sclerosis. In Neuro-Immuno-Gastroenterology; Springer International Publishing: Cham, Switzerland, 2016; pp. 97–122. [Google Scholar] [CrossRef]
- Arnold, I.C.; Hitzler, I.; Müller, A. The immunomodulatory properties of Helicobacter pylori confer protection against allergic and chronic inflammatory disorders. Front. Cell. Infect. Microbiol. 2012, 2, 10. [Google Scholar] [CrossRef] [Green Version]
- Cook, K.; Letley, D.P.; Ingram, R.J.M.; Staples, E.; Skjoldmose, H.; Atherton, J.C.; Robinson, K. CCL20/CCR6-mediated migration of regulatory T cells to theHelicobacter pylori-infected human gastric mucosa. Gut 2014, 63, 1550–1559. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Comerford, I.; Bunting, M.; Fenix, K.; Haylock-Jacobs, S.; Litchfield, W.; Harata-Lee, Y.; Turvey, M.; Brazzatti, J.; Gregor, C.; Nguyen, P.; et al. An immune paradox: How can the same chemokine axis regulate both immune tolerance and activation? BioEssays 2010, 32, 1067–1076. [Google Scholar] [CrossRef]
- Elhofy, A.; DePaolo, R.W.; Lira, S.A.; Lukacs, N.W.; Karpus, W.J. Mice deficient for CCR6 fail to control chronic experimental autoimmune encephalomyelitis. J. Neuroimmunol. 2009, 213, 91–99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liston, A.; Kohler, R.E.; Townley, S.; Haylock-Jacobs, S.; Comerford, I.; Caon, A.C.; Webster, J.; Harrison, J.M.; Swann, J.; Clark-Lewis, I.; et al. Inhibition of CCR6 Function Reduces the Severity of Experimental Autoimmune Encephalomyelitis via Effects on the Priming Phase of the Immune Response. J. Immunol. 2009, 182, 3121–3130. [Google Scholar] [CrossRef] [Green Version]
- Willison, H.J.; Jacobs, B.C.; van Doorn, P.A. Guillain-Barré syndrome. Lancet 2016, 388, 717–727. [Google Scholar] [CrossRef] [Green Version]
- Leonhard, S.E.; Mandarakas, M.R.; Gondim, F.A.A.; Bateman, K.; Ferreira, M.L.B.; Cornblath, D.R.; Van Doorn, P.A.; Dourado, M.E.; Hughes, R.A.C.; Islam, B.; et al. Diagnosis and management of Guillain–Barré syndrome in ten steps. Nat. Rev. Neurol. 2019, 15, 671–683. [Google Scholar] [CrossRef] [PubMed]
- Cao-Lormeau, V.-M.; Blake, A.; Mons, S.; Lastère, S.; Roche, C.; Vanhomwegen, J.; Dub, T.; Baudouin, L.; Teissier, A.; Larre, P.; et al. Guillain-Barré Syndrome outbreak associated with Zika virus infection in French Polynesia: A case-control study. Lancet 2016, 387, 1531–1539. [Google Scholar] [CrossRef] [Green Version]
- van den Berg, B.; van der Eijk, A.A.; Pas, S.D.; Hunter, J.G.; Madden, R.G.; Tio-Gillen, A.P.; Dalton, H.R.; Jacobs, B.C. Guillain-Barré syndrome associated with preceding hepatitis E virus infection. Neurology 2014, 82, 491–497. [Google Scholar] [CrossRef] [PubMed]
- Jacobs, B.C.; Rothbarth, P.H.; van der Meché, F.; Herbrink, P.; Schmitz, P.I.; de Klerk, M.A.; van Doorn, P.A. The spectrum of antecedent infections in Guillain-Barré syndrome. Neurology 1998, 51, 1110–1115. [Google Scholar] [CrossRef] [PubMed]
- Dimachkie, M.M.; Barohn, R.J. Guillain-Barré syndrome and variants. Neurol. Clin. 2013, 31, 491–510. [Google Scholar] [CrossRef] [Green Version]
- Rocha Cabrero, F.; Morrison, E.H. Miller Fisher Syndrome. In StatPearls [Internet]; StatPearls Publishing: Treasure Island, FL, USA, 28 April 2021. Available online: https://www.ncbi.nlm.nih.gov/books/NBK507717/ (accessed on 23 April 2021).
- Steer, A.C.; Starr, M.; Kornberg, A.J. Bickerstaff brainstem encephalitis associated with Mycoplasma pneumoniae infection. J. Child Neurol. 2006, 21, 533–534. [Google Scholar] [CrossRef]
- Ho, T.W.; Mishu, B.; Li, C.Y.; Gao, C.Y.; Cornblath, D.R.; Griffin, J.W.; Asbury, A.K.; Blaser, M.J.; McKhann, G.M. Guillain-Barré syndrome in northern China. Relationship to Campylobacter jejuni infection and anti-glycolipid antibodies. Brain 1995, 118 Pt 3, 597–605. [Google Scholar] [CrossRef]
- Hafer-Macko, C.E.; Sheikh, K.A.; Li, C.Y.; Ho, T.W.; Cornblath, D.R.; McKhann, G.M.; Asbury, A.K.; Griffin, J.W. Immune attack on the Schwann cell surface in acute inflammatory demyelinating polyneuropathy. Ann. Neurol. 1996, 39, 625–635. [Google Scholar] [CrossRef] [PubMed]
- Kountouras, J.; Deretzi, G.; Zavos, C.; Karatzoglou, P.; Touloumis, L.; Nicolaides, T.; Chatzopoulos, D.; Venizelos, I. Association between Helicobacter pylori infection and acute inflammatory demyelinating polyradiculoneuropathy. Eur. J. Neurol. 2005, 12, 139–143. [Google Scholar] [CrossRef] [PubMed]
- Moran, A.P.; Prendergast, M.M. Molecular mimicry in Campylobacter jejuni and Helicobacter pylori lipopolysaccharides: Contribution of gastrointestinal infections to autoimmunity. J. Autoimmun. 2001, 16, 241–256. [Google Scholar] [CrossRef]
- Chiba, S.; Sugiyama, T.; Yonekura, K.; Tanaka, S.; Matsumoto, H.; Fujii, N.; Ebisu, S.; Sekiguchi, K. An antibody to VacA of Helicobacter pylori in cerebrospinal fluid from patients with Guillain-Barre syndrome. J. Neurol. Neurosurg. Psychiatry 2002, 73, 76–78. [Google Scholar] [CrossRef] [Green Version]
- Álvarez-Arellano, L. Helicobacter pylori and neurological diseases: Married by the laws of inflammation. World J. Gastrointest. Pathophysiol. 2014, 5, 400. [Google Scholar] [CrossRef]
- Bickerstaff, E.R. Brain-stem Encephalitis. BMJ 1957, 1, 1384–1390. [Google Scholar] [CrossRef] [Green Version]
- Chiba, A.; Kusunoki, S.; Shimizu, T.; Kanazawa, I. Serum IgG antibody to ganglioside GQ1b is a possible marker of Miller Fisher syndrome. Ann. Neurol. 1992, 31, 677–679. [Google Scholar] [CrossRef] [PubMed]
- Yuki, N.; Sato, S.; Tsuji, S.; Hozumi, I.; Miyatake, T. An immunologic abnormality common to Bickerstaff’s brain stem encephalitis and Fisher’s syndrome. J. Neurol. Sci. 1993, 118, 83–87. [Google Scholar] [CrossRef]
- Odaka, M.; Yuki, N.; Hirata, K. Anti-GQ1b IgG antibody syndrome: Clinical and immunological range. J. Neurol. Neurosurg. Psychiatry 2001, 70, 50–55. [Google Scholar] [CrossRef] [Green Version]
- Tatsumoto, M.; Koga, M.; Gilbert, M.; Odaka, M.; Hirata, K.; Kuwabara, S.; Yuki, N. Spectrum of neurological diseases associated with antibodies to minor gangliosides GM1b and GalNAc-GD1a. J. Neuroimmunol. 2006, 177, 201–208. [Google Scholar] [CrossRef] [PubMed]
- Ito, M.; Kuwabara, S.; Odaka, M.; Misawa, S.; Koga, M.; Hirata, K.; Yuki, N. Bickerstaff’s brainstem encephalitis and Fisher syndrome form a continuous spectrum. J. Neurol. 2008, 255, 674–682. [Google Scholar] [CrossRef] [PubMed]
- Kountouras, J.; Deretzi, G.; Grigoriadis, N.; Zavos, C.; Boziki, M.; Gavalas, E.; Katsinelos, P.; Tzilves, D.; Giouleme, O.; Lazaraki, G. Guillain-Barré syndrome. Lancet Neurol. 2008, 7, 1080–1081. [Google Scholar] [CrossRef]
- Wingerchuk, D.M.; Lennon, V.A.; Lucchinetti, C.F.; Pittock, S.J.; Weinshenker, B.G. The spectrum of neuromyelitis optica. Lancet Neurol. 2007, 6, 805–815. [Google Scholar] [CrossRef]
- Wingerchuk, D.M.; Hogancamp, W.F.; O’Brien, P.C.; Weinshenker, B.G. The clinical course of neuromyelitis optica (Devic’s syndrome). Neurology 1999, 53, 1107–1114. [Google Scholar] [CrossRef]
- Sellner, J.; Hemmer, B.; Mühlau, M. The clinical spectrum and immunobiology of parainfectious neuromyelitis optica (Devic) syndromes. J. Autoimmun. 2010, 34, 371–379. [Google Scholar] [CrossRef]
- Jarius, S.; Wildemann, B. AQP4 antibodies in neuromyelitis optica: Diagnostic and pathogenetic relevance. Nat. Rev. Neurol. 2010, 6, 383–392. [Google Scholar] [CrossRef]
- Li, W.; Minohara, M.; Piao, H.; Matsushita, T.; Masaki, K.; Matsuoka, T.; Isobe, N.; Su, J.J.; Ohyagi, Y.; Kira, J.-I. Association of anti-Helicobacter pylori neutrophil-activating protein antibody response with anti-aquaporin-4 autoimmunity in Japanese patients with multiple sclerosis and neuromyelitis optica. Mult. Scler. J. 2009, 15, 1411–1421. [Google Scholar] [CrossRef] [PubMed]
- Kira, J.I. Neuromyelitis optica and opticospinal multiple sclerosis: Mechanisms and pathogenesis. Pathophysiology 2011, 18, 69–79. [Google Scholar] [CrossRef]
- Wang, Z.W.; Li, Y.; Huang, L.Y.; Guan, Q.K.; Xu, D.W.; Zhou, W.K.; Zhang, X.Z. Helicobacter pylori infection contributes to high risk of ischemic stroke: Evidence from a meta-analysis. J. Neurol. 2012, 259, 2527–2537. [Google Scholar] [CrossRef]
- Chen, Y.; Segers, S.; Blaser, M.J. Association between Helicobacter pylori and mortality in the NHANES III study. Gut 2013, 62, 1262–1269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Franceschi, F.; Tortora, A.; Gasbarrini, G.; Gasbarrini, A. Helicobacter pylori and extragastric diseases. Helicobacter 2014, 19 (Suppl. S1), 52–58. [Google Scholar] [CrossRef]
- Tunca, A.; Türkay, C.; Tekin, O.; Kargili, A.; Erbayrak, M. Is Helicobacter pylori infection a risk factor for migraine? A case-control study. Acta Neurol. Belg. 2004, 104, 161–164. [Google Scholar] [PubMed]
- Hosseinzadeh, M.; Khosravi, A.; Saki, K.; Ranjbar, R. Evaluation of Helicobacter pylori infection in patients with common migraine headache. Arch. Med. Sci. 2011, 7, 844–849. [Google Scholar] [CrossRef] [Green Version]
Virulence Factors | |
---|---|
Urease | Flagellum |
Cytotoxin-associated gene A | Vacuolating cytotoxin A |
Catalase | Superoxidase dismutase |
Lewis antigens | Arginase |
Phospholipases | Lipopolysaccharide |
Blood group antigen-binding adhesin | Sialic acid-binding adhesin |
Outer inflammatory protein A | Duodenal ulcer promoting gene A |
Adherence-associated lipoprotein A and B | LacdiNAc-specific adhesin |
Helicobacter pylori outer membrane protein Q | Helicobacter pylori outer membrane protein Z |
Induced by contact with epithelium gene A | Cholesteryl α-glucosyltransferase |
γ-glutamyl-transpeptidase | Neutrophil-activating protein |
High temperature requirement A | Heat shock proteins |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Baj, J.; Forma, A.; Flieger, W.; Morawska, I.; Michalski, A.; Buszewicz, G.; Sitarz, E.; Portincasa, P.; Garruti, G.; Flieger, M.; et al. Helicobacter pylori Infection and Extragastric Diseases—A Focus on the Central Nervous System. Cells 2021, 10, 2191. https://doi.org/10.3390/cells10092191
Baj J, Forma A, Flieger W, Morawska I, Michalski A, Buszewicz G, Sitarz E, Portincasa P, Garruti G, Flieger M, et al. Helicobacter pylori Infection and Extragastric Diseases—A Focus on the Central Nervous System. Cells. 2021; 10(9):2191. https://doi.org/10.3390/cells10092191
Chicago/Turabian StyleBaj, Jacek, Alicja Forma, Wojciech Flieger, Izabela Morawska, Adam Michalski, Grzegorz Buszewicz, Elżbieta Sitarz, Piero Portincasa, Gabriella Garruti, Michał Flieger, and et al. 2021. "Helicobacter pylori Infection and Extragastric Diseases—A Focus on the Central Nervous System" Cells 10, no. 9: 2191. https://doi.org/10.3390/cells10092191
APA StyleBaj, J., Forma, A., Flieger, W., Morawska, I., Michalski, A., Buszewicz, G., Sitarz, E., Portincasa, P., Garruti, G., Flieger, M., & Teresiński, G. (2021). Helicobacter pylori Infection and Extragastric Diseases—A Focus on the Central Nervous System. Cells, 10(9), 2191. https://doi.org/10.3390/cells10092191