Preview

Обозрение психиатрии и медицинской психологии имени В.М.Бехтерева

Расширенный поиск

Гены-кандидаты воспаления и их роль в патогенезе шизофрении: современные научные данные

https://doi.org/10.31363/2313-7053-2025-4-2-1099

Аннотация

Цель. Целью аналитического обзора стали поиск, анализ и обобщение данных об ассоциациях генов-кандидатов воспаления и их влиянии на патогенез и клинические проявления шизофрении. Материалы и методы. Проводился поиск оригинальных исследований, систематических обзоров литературы и мета-анализов в базах Google Scholar, PubMed и eLIBRARY.ru. Включались публикации за период с 1 января 2017 года по 31 декабря 2024 года без языковых ограничений, посвящённые генетическим аспектам воспаления в контексте шизофрении. Результаты. Обзор подтвердил значимую роль генетических ассоциаций воспаления в патогенезе шизофрении. Полиморфизмы генов, кодирующих провоспалительные и противовоспалительные молекулы (IL6, IL10, IL1B, IL28B, TNF-α, HLA, VEGF-A, NF-κB), ассоциированы с риском заболевания, клиническими проявлениями и изменениями структуры мозга. Также отмечена роль генетической регуляции воспалительных механизмов в формировании нарушений нейропластичности, проницаемости гематоэнцефалического барьера и окислительного стресса при шизофрении. Заключение. Наш обзор подтверждает значимую роль генетических ассоциаций воспалительных механизмов в патогенезе шизофрении. Наследственная предрасположенность к дисбалансу иммунного ответа может влиять на развитие, тяжесть и течение заболевания.

Об авторах

А. П. Отмахов
Национальный медицинский исследовательский центр им. В. А. Алмазова
Россия

Отмахов Андрей Павлович – ассистент кафедры психиатрии и психотерапии с клиникой Института медицинского образования

197341, г. Санкт-Петербург, ул. Аккуратова, д. 2



Л. Р. Бакиров
Башкирский Государственный Медицинский Университет
Россия

Бакиров Линар Рифкатович - к.м.н., доцент кафедры психиатрии, наркологии и психотерапии

450008, Республика Башкортостан, г. Уфа, ул. Ленина, д. 3



Н. Г. Незнанов
Национальный медицинский исследовательский центр психиатрии и неврологии им. В.М. Бехтерева; Первый Санкт-Петербургский государственный медицинский университет им. И.П. Павлова
Россия

Незнанов Николай Григорьевич – д.м.н., профессор, заслуженный деятель науки РФ, директор; заведующий кафедрой психиатрии и наркологии

192019, г. Санкт-Петербург, ул. Бехтерева, д. 3;

197022, г. Санкт-Петербург, ул. Льва Толстого, 6-8



А. Р. Асадуллин
Башкирский Государственный Медицинский Университет; Саратовский ГМУ им. В. И. Разумовского
Россия

Асадуллин Азат Раилевич - д.м.н., профессор кафедры психиатрии, наркологии и психотерапии; профессор кафедры психиатрии, наркологии, психотерапии и клинической психологии

450008, Республика Башкортостан, г. Уфа, ул. Ленина, д. 3;

410012, г. Саратов, ул. Большая Казачья, 112



Список литературы

1. Абдрахманова А.Е., Гильманов А.Ж., Галеева Е.Х., Ефремов И.С., Асадуллин А.Р. Нейровоспаление и воспалительные маркеры в периферической крови при тревожных и депрессивных расстройствах (обзор литературы). Психическое здоровье. 2022;17(6):55-64. https://doi.org/10.25557/2074-014X.2022.06.55-64

2. Голимбет В.Е, Клюшник Т.П. Молекулярно-генетический и иммунологический аспекты формирования психопатологических симптомов при шизофрении. Журнал неврологии и психиатрии имени С.С. Корсакова. 2022;122(10):66-71. https://doi.org/10.17116/jnevro202212210166

3. Михайлова ВА, Плакунова ВВ, Лежейко ТВ, Колесина НЮ, Голимбет ВЕ. Связь полиморфизма rs1800629 гена фактора некроза опухолей альфа с субдоменами негативных симптомов шизофрении. Медицинская генетика. 2022;21(8):20-22. https://doi.org/10.25557/2073-7998.2022.08.20-22

4. Ahmad SF, Zoheir KMA, Ansari MA, Nadeem A, Bakheet SA, Al-Ayadhi LY, et al. Dysregulation of Th1, Th2, Th17, and T regulatory cell-related transcription factor signaling in children with autism. Mol Neurobiol. 2017;54:4390–4400. https://doi.org/10.1007/s12035-016-9977-0

5. Apte RS, Chen DS, Ferrara N. VEGF in signaling and disease: beyond discovery and development. Cell. 2019;176(6):1248-1264. https://doi.org/10.1016/j.cell.2019.01.021

6. Berbers RM, van der Wal MM, van Montfrans JM, Ellerbroek PM, Dalm VA, van Hagen PM, et al. Chronically activated T-cells retain their inflammatory properties in common variable immunodeficiency. J Clin Immunol. 2021;41:1621-1632. https://doi.org/10.1007/s10875-021-01084-6

7. Bishop JR, Zhang L, Lizano P. Inflammation subtypes and translating inflammation-related genetic findings in schizophrenia and related psychoses: a perspective on pathways for treatment stratification and novel therapies. Harv Rev Psychiatry. 2022;30(1):59-70. https://doi.org/10.1097/HRP.0000000000000321

8. Bleich A, Brown SL, Kahn R, van Praag HM. The role of serotonin in schizophrenia. Schizophr Bull. 1988;14(2):297-315. https://doi.org/10.1093/schbul/14.2.297

9. Boukouaci W, Lajnef M, Richard JR, Wu CL, Bouassida J, Rafik I, et al. HLA-E circulating and genetic determinants in schizophrenia and bipolar disorder. Sci Rep. 2021;11(1):20260. https://doi.org/10.1038/s41598-021-99732-9

10. Castañeda-Cabral JL, Beas-Zárate C, Rocha-Arrieta LL, Orozco-Suárez SA, Alonso-Vanegas M, Guevara-Guzmán R, et al. Increased protein expression of VEGF-A, VEGF-B, VEGF-C and their receptors in the temporal neocortex of pharmacoresistant temporal lobe epilepsy patients. J Neuroimmunol. 2019;328:68-72. https://doi.org/10.1016/j.jneuroim.2018.12.007

11. Choi KY, Choo JM, Lee YJ, Lee Y, Cho CH, Kim SH, Lee HJ. Association between the IL10 rs1800896 polymorphism and tardive dyskinesia in schizophrenia. Psychiatry Investig. 2020;17(10):1031. https://doi.org/10.30773/pi.2020.0191

12. Comer AL, Carrier M, Tremblay MÈ, Cruz-Martín A. The inflamed brain in schizophrenia: the convergence of genetic and environmental risk factors that lead to uncontrolled neuroinflammation. Front Cell Neurosci. 2020;14:274. https://doi.org/10.3389/fncel.2020.00274

13. Debnath M, Berk M, Leboyer M, Tamouza R. The MHC/HLA gene complex in major psychiatric disorders: emerging roles and implications. Curr Behav Neurosci Rep. 2018;5:179-188. https://doi.org/10.1007/s40473-018-0155-8

14. Dendrou CA, Petersen J, Rossjohn J, Fugger L. HLA variation and disease. Nat Rev Immunol. 2018;18(5):325-339. https://doi.org/10.1038/nri.2017.143

15. Dickerson F, Stallings C, Origoni A, Boronow J, Yolken R. C-reactive protein is associated with the severity of cognitive impairment but not of psychiatric symptoms in individuals with schizophrenia. Schizophr Res. 2007;93(1-3):261-265. https://doi.org/10.1016/j.schres.2007.03.022

16. Ermakov EA, Dmitrieva EM, Parshukova DA, Kazantseva DV, Vasilieva AR, Smirnova LP. Oxidative stress‐related mechanisms in schizophrenia pathogenesis and new treatment perspectives. Oxid Med Cell Longev. 2021;2021:8881770. https://doi.org/10.1155/2021/8881770

17. Fan X, Pristach C, Liu EY, Freudenreich O, Henderson DC, Goff DC. Elevated serum levels of C-reactive protein are associated with more severe psychopathology in a subgroup of patients with schizophrenia. Psychiatry Res. 2007;149(1-3):267-271. https://doi.org/10.1016/j.psychres.2006.07.011

18. Fillman SG, Weickert TW, Lenroot RK, Catts SV, Bruggemann JM, Catts VS, et al. Elevated peripheral cytokines characterize a subgroup of people with schizophrenia displaying poor verbal fluency and reduced Broca’s area volume. Mol Psychiatry. 2016;21(8):1090-1098. https://doi.org/10.1038/mp.2015.90

19. Gallego JA, Blanco EA, Husain-Krautter S, Fagen EM, Moreno-Merino P, del Ojo-Jiménez JA, et al. Cytokines in cerebrospinal fluid of patients with schizophrenia spectrum disorders: New data and an updated meta-analysis. Schizophr Res. 2018;202:64-71. https://doi.org/10.1016/j.schres.2018.07.019

20. Gandal MJ, Haney JR, Parikshak NN, Leppa V, Ramaswami G, Hartl C, et al. Shared molecular neuropathology across major psychiatric disorders parallels polygenic overlap. Science. 2018;359(6376):693-697. https://doi.org/10.1126/science.aad646

21. Gnanasekaran A, Kelchen MN, Brogden NK, Smith RM. Vascular endothelial growth factor (VEGF) expression and neuroinflammation is increased in the frontopolar cortex of individuals with autism spectrum disorder. 2019. https://doi.org/10.1101/627083

22. Golimbet V, Lezheiko T, Mikhailova V, Korovaitseva G, Kolesina N, Plakunova V, et al. A study of the association between polymorphisms in the genes for interleukins IL-6 and IL-10 and negative symptoms subdomains in schizophrenia. Indian J Psychiatry. 2022;64(5):484-488. https://doi.org/10.4103/indianjpsychiatry.indianjpsychiatry_212_22

23. Halstead S, Siskind D, Amft M, Wagner E, Yakimov V, Liu Z, et al. Alteration patterns of peripheral concentrations of cytokines and associated inflammatory proteins in acute and chronic stages of schizophrenia: a systematic review and network meta-analysis. Lancet Psychiatry. 2023;10(4):260-271. https://doi.org/10.1016/S2215-0366(23)00025-1

24. Haque R, Kim Y, Park K, Jang H, Kim SY, Lee H, et al. Altered distributions in circulating follicular helper and follicular regulatory T cells accountable for imbalanced cytokine production in multiple sclerosis. Clin Exp Immunol. 2021;205(1):75-88. https://doi.org/10.1111/cei.13596

25. Hayden MS, Ghosh S. NF-κB in immunobiology. Cell Res. 2011;21(2):223-244. https://doi.org/10.1038/cr.2011.13

26. Hudson ZD, Miller BJ. Meta-analysis of cytokine and chemokine genes in schizophrenia. Clin Schizophr Relat Psychoses. 2018;12(3):121-129B. https://doi.org/10.3371/csrp.humi.070516

27. Jacomb I, Stanton C, Vasudevan R, Powell H, O'Donnell M, Lenroot R, et al. C-reactive protein: higher during acute psychotic episodes and related to cortical thickness in schizophrenia and healthy controls. Front Immunol. 2018;9:2230. https://doi.org/10.3389/fimmu.2018.02230

28. Javitt DC. Glutamatergic theories of schizophrenia. Isr J Psychiatry. 2010;47(1):4.

29. Jayaraman A, Htike TT, James R, Picon C, Reynolds R. TNF-mediated neuroinflammation is linked to neuronal necroptosis in Alzheimer's disease hippocampus. Acta Neuropathol Commun. 2021;9:1-21. https://doi.org/10.1186/s40478-021-01264-w

30. Jin M, Günther R, Akgün K, Hermann A, Ziemssen T. Peripheral proinflammatory Th1/Th17 immune cell shift is linked to disease severity in amyotrophic lateral sclerosis. Sci Rep. 2020;10(1):5941. https://doi.org/10.1038/s41598-020-62756-8

31. Kadasah S, Arfin M, Rizvi S, Al-Asmari M, Al-Asmari A. Tumor necrosis factor-α and-β genetic polymorphisms as a risk factor in Saudi patients with schizophrenia. Neuropsychiatr Dis Treat. 2017;1081-1088. https://doi.org/10.2147/NDT.S131144

32. Kang N, Shin W, Jung S, Bang M, Lee SH. The effect of TNF-alpha polymorphism on white matter structures and memory function in patients with schizophrenia: A pilot study. Psychiatry Investig. 2022;19(12):1027. https://doi.org/10.30773/pi.2021.0326

33. Kappelmann N, Khandaker GM, Dal H, Stochl J, Kosidou K, Jones PB, et al. Systemic inflammation and intelligence in early adulthood and subsequent risk of schizophrenia and other non-affective psychoses: A longitudinal cohort and co-relative study. Psychol Med. 2019;49(2):295-302. https://doi.org/10.1017/S0033291718000831

34. Kaur H, Ghorai SM. Role of cytokines as immunomodulators. In: Immunomodulators and Human Health. Singapore: Springer Nature Singapore; 2022. p. 371-414. https://doi.org/10.1007/978-981-16-6379-6_13

35. Kerschensteiner M, Meinl E, Hohlfeld R. Neuro-immune crosstalk in CNS diseases. Neuroscience. 2009;158(3):1122-1132. https://doi.org/10.1016/j.neuroscience.2008.09.009

36. Kirkpatrick B, Miller BJ. Inflammation and schizophrenia. Schizophr Bull. 2013;39(6):1174-1179. https://doi.org/10.1093/schbul/sbt141

37. Koistinaho J, Koskuvi M, Pörsti E, Wu YC, Trontti K, McQuade A, et al. Genetic contribution to microglial activation in schizophrenia. 2022. https://doi.org/10.21203/rs.3.rs-1980131/v1

38. Korovaitseva GI, Gabaeva MV, Oleichik IV, Golimbet VE. The effect of INDEL polymorphism of the human leukocyte antigen G (HLA-G) and the season of birth on the risk of schizophrenia and its clinical features. Russ J Genet. 2021;57(2):221-226. https://doi.org/10.1134/S102279542102006X

39. Kustrimovic N, Comi C, Magistrelli L, E Rasini, Legnaro M, Bombelli R, et al. Parkinson’s disease patients have a complex phenotypic and functional Th1 bias: cross-sectional studies of CD4+ Th1/Th2/T17 and Treg in drug-naive and drug-treated patients. J Neuroinflammation. 2018;15:1-17. https://doi.org/10.1186/s12974-018-1248-8

40. Lee BH, Hong JP, Hwang JA, Ham BJ, Na KS, Kim WJ, et al. Alterations in plasma vascular endothelial growth factor levels in patients with schizophrenia before and after treatment. Psychiatry Res. 2015;228(1):95-99. https://doi.org/10.1016/j.psychres.2015.04.020

41. Leza JC, García-Bueno B, Bioque M, Arango C, Parellada M, Do K, et al. Inflammation in schizophrenia: a question of balance. Neurosci Biobehav Rev. 2015;55:612-626. https://doi.org/10.1016/j.neubiorev.2015.05.014

42. Li W, Luo Y, Xu H, Ma Q, Yao Q. Imbalance between T helper 1 and regulatory T cells plays a detrimental role in experimental Parkinson’s disease in mice. J Int Med Res. 2021;49(4):0300060521998471. https://doi.org/10.1177/030006052199847

43. Lingappan K. NF-κB in oxidative stress. Curr Opin Toxicol. 2018;7:81-86. https://doi.org/10.1016/j.cotox.2017.11.002

44. Liu B, Shao Y, Fu R. Current research status of HLA in immune‐related diseases. Immun Inflamm Dis. 2021;9(2):340-350. https://doi.org/10.1002/iid3.416

45. Liu T, Zhang L, Joo D, Sun SC. NF-κB signaling in inflammation. Signal Transduct Target Ther. 2017;2(1):1-9. https://doi.org/10.1038/sigtrans.2017.23

46. Lizano P, Lutz O, Ling G, Padmanabhan J, Tandon N, Sweeney J, et al. VEGFA gene variation influences hallucinations and frontotemporal morphology in psychotic disorders: a B-SNIP study. Transl Psychiatry. 2018;8(1):215. https://doi.org/10.1038/s41398-018-0271-y

47. Long J, Huang G, Liang W, Liang B, Chen Q, Xie J, et al. The prevalence of schizophrenia in mainland China: evidence from epidemiological surveys. Acta Psychiatr Scand. 2014;130(4):244-256. https://doi.org/10.1111/acps.12296

48. Lv H, Guo M, Guo C, He K. The interrelationships between cytokines and schizophrenia: a systematic review. Int J Mol Sci. 2024;25(15):8477. https://doi.org/10.3390/ijms25158477

49. Matern BM, Olieslagers TI, Voorter CE, Groeneweg M, Tilanus MGJ. Insights into the polymorphism in HLA‐DRA and its evolutionary relationship with HLA haplotypes. HLA. 2020;95(2):117-127. https://doi.org/10.1111/tan.13730

50. Melamud MM, Bobrik DV, Brit PI, Efremov IS, Buneva VN, Nevinsky GA, et al. Biochemical, hematological, inflammatory, and gut permeability biomarkers in patients with alcohol withdrawal syndrome with and without delirium tremens. J Clin Med. 2024;13(10):2776. https://doi.org/10.3390/jcm13102776

51. Melincovici CS, Boşca AB, Şuşman S, Mărginean M, Mihu C, Istrate M, et al. Vascular endothelial growth factor (VEGF)-key factor in normal and pathological angiogenesis. Rom J Morphol Embryol. 2018;59(2):455-467. PMID: 30173249.

52. Mitchell JP, Carmody RJ. NF-κB and the transcriptional control of inflammation. Int Rev Cell Mol Biol. 2018;335:41-84. https://doi.org/10.1016/bs.ircmb.2017.07.007

53. Mohammadi A, Rashidi E, Amooeian VG. Brain, blood, cerebrospinal fluid, and serum biomarkers in schizophrenia. Psychiatry Res. 2018;265:25-38. https://doi.org/10.1016/j.psychres.2018.04.036

54. Mongan D, Ramesar M, Föcking M, Cannon M, Cotter D, et al. Role of inflammation in the pathogenesis of schizophrenia: A review of the evidence, proposed mechanisms and implications for treatment. Early Interv Psychiatry. 2020;14(4):385-397. https://doi.org/10.1111/eip.12859

55. Munkvad I, Fog R, Randrup A. Dopamine and schizophrenia. Clin Physiol. 1981;1:102-107. https://doi.org/10.1111/j.1475-097X.1981.tb00948.x

56. Murphy CE, Lawther AJ, Webster MJ, Asai M, Kondo Y, Matsumoto M, et al. Nuclear factor kappa B activation appears weaker in schizophrenia patients with high brain cytokines than in non-schizophrenic controls with high brain cytokines. J Neuroinflammation. 2020;17:1-13. https://doi.org/10.1186/s12974-020-01890-6

57. Murphy CE, Walker AK, O’Donnell M, Galletly C, Lloyd AR, Liu D, et al. Peripheral NF-κB dysregulation in people with schizophrenia drives inflammation: putative anti-inflammatory functions of NF-κB kinases. Transl Psychiatry. 2022;12(1):21. https://doi.org/10.1038/s41398-021-01764-2

58. Müller N. Inflammation in schizophrenia: pathogenetic aspects and therapeutic considerations. Schizophr Bull. 2018;44(5):973-982. https://doi.org/10.1093/schbul/sby024

59. Müller N, Weidinger E, Leitner B, Schwarz MJ. The role of inflammation in schizophrenia. Front Neurosci. 2015;9:372. https://doi.org/10.3389/fnins.2015.00372

60. Oberstein TJ, Taha L, Spitzer P, Hellstern J, Herrmann M, Kornhuber J, et al. Imbalance of circulating Th17 and regulatory T cells in Alzheimer’s disease: a case control study. Front Immunol. 2018;9:1213. https://doi.org/10.3389/fimmu.2018.01213

61. Pantelis C, Yücel M, Wood SJ, McGorry PD, Velakoulis D. Early and late neurodevelopmental disturbances in schizophrenia and their functional consequences. Aust N Z J Psychiatry. 2003;37(4):399-406. https://doi.org/10.1046/j.1440-1614.2003.01193.x

62. Peach CJ, Mignone VW, Arruda MA, Alcobia DC, Hill SJ, Kilpatrick LE et al. Molecular pharmacology of VEGF-A isoforms: binding and signalling at VEGFR2. Int J Mol Sci. 2018;19(4):1264. https://doi.org/10.3390/ijms19041264

63. Pillai A, Howell KR, Ahmed AO, Weinberg D, Allen KM, Bruggemann J et al. Association of serum VEGF levels with prefrontal cortex volume in schizophrenia. Mol Psychiatry. 2016;21(5):686-692. https://doi.org/10.1038/mp.2015.96

64. Pong S, Karmacharya R, Sofman M, Bishop JR, Lizano P. The role of brain microvascular endothelial cell and blood-brain barrier dysfunction in schizophrenia. Complex Psychiatry. 2020;6(1-2):30-46. https://doi.org/10.1159/000511552

65. Rampino A, Annese T, Torretta S, Tamma R, Falcone RM, Ribatti D. Involvement of vascular endothelial growth factor in schizophrenia. Neurosci Lett. 2021;760:136093. https://doi.org/10.1016/j.neulet.2021.136093

66. Requena-Ocaña N, Flores-Lopez M, Papaseit E, García-Marchena N, Ruiz JJ, Ortega-Pinazo J, et al. Vascular endothelial growth factor as a potential biomarker of neuroinflammation and frontal cognitive impairment in patients with alcohol use disorder. Biomedicines. 2022;10(5):947. https://doi.org/10.3390/biomedicines10050947

67. Rizzo FR, Musella A, De Vito F, Fresegna D, Bullitta S, Vanni V et al. Tumor necrosis factor and interleukin‐1β modulate synaptic plasticity during neuroinflammation. Neural Plast. 2018;2018:8430123. https://doi.org/10.1155/2018/8430123

68. Robinson J, Guethlein LA, Cereb N, Yang SY, Norman PJ, Marsh SG et al. Distinguishing functional polymorphism from random variation in the sequences of >10,000 HLA-A,-B and -C alleles. PLoS Genet. 2017;13(6):e1006862. https://doi.org/10.1371/journal.pgen.1006862

69. Saha S, Chant D, Welham J, McGrath J. A systematic review of the prevalence of schizophrenia. PLoS Med. 2005;2(5):e141. https://doi.org/10.1371/journal.pmed.0020141

70. Sahbaz C, Zibandey N, Kurtulmus A, Duran Y, Gokalp M, Kırpınar I et al. Reduced regulatory T cells with increased proinflammatory response in patients with schizophrenia. Psychopharmacology. 2020;237:1861-1871. https://doi.org/10.1007/s00213-020-05504-0

71. Saoud H, Aflouk Y, Afia AB, Gaha L, Jrad BBH. Association of VEGF-A and KDR polymorphisms with the development of schizophrenia. Hum Immunol. 2022;83(6):528-53. https://doi.org/10.1016/j.humimm.2022.04.003

72. Shivakumar V, Debnath M, Venugopal D, Rajasekaran A, Kalmady SV, Subbanna M et al. Influence of correlation between HLA-G polymorphism and Interleukin-6 (IL6) gene expression on the risk of schizophrenia. Cytokine. 2018;107:59-64. https://doi.org/10.1016/j.cyto.2017.11.016

73. Shivakumar V, Sreeraj VS, Subbanna M, Kalmady SV, Amaresha AC, Narayanaswamy JC et al. Differential impact of interleukin-6 promoter gene polymorphism on hippocampal volume in antipsychotic-naïve schizophrenia patients. Indian J Psychiatry. 2020;62(1):36-42. https://doi.org/10.4103/psychiatry.IndianJPsychiatry_486_19

74. Solleiro-Villavicencio H, Rivas-Arancibia S. Effect of chronic oxidative stress on neuroinflammatory response mediated by CD4+ T cells in neurodegenerative diseases. Front Cell Neurosci. 2018;12:114. https://doi.org/10.3389/fncel.2018.00114

75. Stein L, Wise CD. Possible etiology of schizophrenia: Progressive damage to the noradrenergic reward system by 6-hydroxydopamine. Science. 1971;171(3975):1032-1036. https://doi.org/10.1126/science.171.3975.103

76. Suchanek-Raif R, Raif P, Kowalczyk M, Paul-Samojedny M, Kucia K, Merk W et al. Promoter polymorphisms of TNF-α gene as a risk factor for schizophrenia. Arch Med Res. 2018;49(4):248-254. https://doi.org/10.1016/j.arcmed.2018.09.007

77. Swain SA, Sarangi P, Rattan R, Sahu PK, Lamare AA. A study of nuclear factor-kappa B1 gene polymorphism types in schizophrenia patients and their correlation with disease severity. Cureus. 2022;14(4):e24401. https://doi.org/10.7759/cureus.24401

78. Tamouza R, Krishnamoorthy R, Leboyer M. Understanding the genetic contribution of the human leukocyte antigen system to common major psychiatric disorders in a world pandemic context. Brain Behav Immun. 2021;91:731-739. https://doi.org/10.1016/j.bbi.2020.09.033

79. Trovão N, Prata J, VonDoellinger O, Santos S, Barbosa M, Coelho R et al. Peripheral biomarkers for first-episode psychosis—opportunities from the neuroinflammatory hypothesis of schizophrenia. Psychiatry Investig. 2019;16(3):177. https://doi.org/10.30773/pi.2018.12.19.1

80. Upthegrove R, Khandaker GM. Cytokines, oxidative stress and cellular markers of inflammation in schizophrenia. Neuroinflammation Schizophr. 2020;49-66. https://doi.org/10.1007/7854_2018_88

81. Volk DW, Moroco AE, Roman KM, Edelson JR, Lewis DA et al. The role of the nuclear factor-κB transcriptional complex in cortical immune activation in schizophrenia. Biol Psychiatry. 2019;85(1):25-34. https://doi.org/10.1016/j.biopsych.2018.06.015

82. Wang M, Claesson MH. Classification of human leukocyte antigen (HLA) supertypes. Immunoinformatics. 2014;309-317. https://doi.org/10.1007/978-1-4939-1115-8_17

83. Wiszniak S, Schwarz Q. Exploring the intracrine functions of VEGF-A. Biomolecules. 2021;11(1):128. https://doi.org/10.3390/biom11010128

84. Wong-Guerra M, Calfio C, Maccioni RB, Rojo LE. Revisiting the neuroinflammation hypothesis in Alzheimer’s disease: A focus on the druggability of current targets. Front Pharmacol. 2023;14:1161850. https://doi.org/10.3389/fphar.2023.1161850

85. Wu EQ, Shi L, Birnbaum H, Hudson T, Kessler R et al. Annual prevalence of diagnosed schizophrenia in the USA: a claims data analysis approach. Psychol Med. 2006;36(11):1535-1540. https://doi.org/10.1017/S0033291706008191

86. Xavier RM, Vorderstrasse A. Genetic basis of positive and negative symptom domains in schizophrenia. Biol Res Nurs. 2017;19(5):559-575. https://doi.org/10.1177/1099800417715907

87. Xu J, Li J, Xiao K, Zou S, Yan P, Xie X et al. Dynamic changes in human HLA‐DRA gene expression and Th cell subsets in sepsis: Indications of immunosuppression and associated outcomes. Scand J Immunol. 2020;91(1):e12813. https://doi.org/10.1111/sji.12813

88. Yang QQ, Zhou JW. Neuroinflammation in the central nervous system: Symphony of glial cells. Glia. 2019;67(6):1017-1035. https://doi.org/10.1002/glia.23571

89. Yu S, Qu Y, Du Z, Ou M, Lu R, Yuan J et al. The expression of immune related genes and potential regulatory mechanisms in schizophrenia. Schizophr Res. 2024;267:507-518. https://doi.org/10.1016/j.schres.2023.11.007

90. Yu YQ, Wang H. Imbalance of Th1 and Th2 cytokines and stem cell therapy in pathological pain. CNS Neurol Disord Drug Targets. 2024;23(1):88-101. https://doi.org/10.2174/1871527322666221226145828

91. Yuan J, Amin P, Ofengeim D. Necroptosis and RIPK1-mediated neuroinflammation in CNS diseases. Nat Rev Neurosci. 2019;20(1):19-33. https://doi.org/10.1038/s41583-018-0093-1

92. Zhang Y, Fang X, Fan W, Tang W, Cai J, Song L et al. Interaction between BDNF and TNF-α genes in schizophrenia. Psychoneuroendocrinology. 2018;89:1-6. https://doi.org/10.1016/j.psyneuen.2017.12.024


Рецензия

Для цитирования:


Отмахов А.П., Бакиров Л.Р., Незнанов Н.Г., Асадуллин А.Р. Гены-кандидаты воспаления и их роль в патогенезе шизофрении: современные научные данные. Обозрение психиатрии и медицинской психологии имени В.М.Бехтерева. https://doi.org/10.31363/2313-7053-2025-4-2-1099

For citation:


Otmakhov A.P., Bakirov L.R., Neznanov N.G., Asadullin A.R. Inflammation-related genes and their role in the pathogenesis of schizophrenia: current scientific evidence. V.M. BEKHTEREV REVIEW OF PSYCHIATRY AND MEDICAL PSYCHOLOGY. (In Russ.) https://doi.org/10.31363/2313-7053-2025-4-2-1099

Просмотров: 10


Creative Commons License
Контент доступен под лицензией Creative Commons Attribution-NonCommercial-ShareAlike 4.0.


ISSN 2313-7053 (Print)
ISSN 2713-055X (Online)