Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1749 2022-10-12 21:13:09 |
2 format correction Meta information modification 1749 2022-10-27 03:12:30 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Shnayder, N.A.;  Khasanova, A.K.;  Strelnik, A.I.;  Al-Zamil, M.;  Otmakhov, A.P.;  Neznanov, N.G.;  Shipulin, G.A.;  Petrova, M.M.;  Garganeeva, N.P.;  Nasyrova, R.F. Cytokine Imbalance in Treatment-Resistant Schizophrenia. Encyclopedia. Available online: https://encyclopedia.pub/entry/31465 (accessed on 26 April 2024).
Shnayder NA,  Khasanova AK,  Strelnik AI,  Al-Zamil M,  Otmakhov AP,  Neznanov NG, et al. Cytokine Imbalance in Treatment-Resistant Schizophrenia. Encyclopedia. Available at: https://encyclopedia.pub/entry/31465. Accessed April 26, 2024.
Shnayder, Natalia A., Aiperi K. Khasanova, Anna I. Strelnik, Mustafa Al-Zamil, Andrey P. Otmakhov, Nikolay G. Neznanov, German A. Shipulin, Marina M. Petrova, Natalia P. Garganeeva, Regina F. Nasyrova. "Cytokine Imbalance in Treatment-Resistant Schizophrenia" Encyclopedia, https://encyclopedia.pub/entry/31465 (accessed April 26, 2024).
Shnayder, N.A.,  Khasanova, A.K.,  Strelnik, A.I.,  Al-Zamil, M.,  Otmakhov, A.P.,  Neznanov, N.G.,  Shipulin, G.A.,  Petrova, M.M.,  Garganeeva, N.P., & Nasyrova, R.F. (2022, October 26). Cytokine Imbalance in Treatment-Resistant Schizophrenia. In Encyclopedia. https://encyclopedia.pub/entry/31465
Shnayder, Natalia A., et al. "Cytokine Imbalance in Treatment-Resistant Schizophrenia." Encyclopedia. Web. 26 October, 2022.
Cytokine Imbalance in Treatment-Resistant Schizophrenia
Edit

Treatment-resistant schizophrenia (TRS) is an important and unresolved problem in biological and clinical psychiatry. Approximately 30% of cases of schizophrenia (Sch) are TRS, which may be due to the fact that some patients with TRS may suffer from pathogenetically “non-dopamine” Sch, in the development of which neuroinflammation is supposed to play an important role. There are many factors leading to a chronic neuroinflammatory process in TRS. 

cytokines cytokine status treatment-resistant schizophrenia

1. Changes in the Functional Activity of Microglia in Treatment Resistance Schizophrenia

Microglia account for 10–20% of all cells found in the brain and are an important component of immune system of central neural system (CNS) [1]. Microglia play an important role in neuroinflammation, providing protection in the event of damage or disease to the CNS. When neuroinfection occurs, activation of microglia, synthesis and release of central pro-inflammatory cytokines, which leads to various mental and behavioral disorders [2]. There is now evidence that aging [3], neurodegeneration [4] and stress [5] lead to “sensitization” or “priming” of microglia, which subsequently causes an exaggerated immune response. Exposure of primed microglia to, for example, minor systemic inflammation leads to proliferation and increased production of pro-inflammatory cytokines [6], which, in turn, can exacerbate the immune response in the CNS and be expressed in a change in behavior [7]. One drug thought to reduce microglial activation is minocycline, a broad-spectrum tetracycline antibiotic with broad anti-inflammatory activity [8]. First and second generations of APs regulate the secretory profile of microglia in vitro. They inhibit the release of proinflammatory cytokines from activated microglia and alleviate oxidative stress [9]. However, some recent reports have shown conflicting results on the effect of some APs on the release of pro-inflammatory cytokines [10]. At the same time, not all antipsychotics (APs) have an anti-inflammatory effect, which may be due to the role of microglia in the development of TRS.

2. Sensitization or Kindling in Treatment Resistance Schizophrenia

“Firing”/“sensitization” refers to the process by which the initial immune response to some stimulus (stress or infection) raises or lowers the threshold to respond to the next exposure to the same stimulus. At the same time, a weaker stimulus is required to activate the immune response or release cytokines than with the initial exposure to an unfavorable (damaging) factor. It is believed that the memory function of the acquired immune system is responsible for this process [5]. The action of factors such as systemic inflammation or stress on healthy people leads to the stimulation of the immune response. As a result, cell proliferation is activated, an increase in the production and release of pro-inflammatory cytokines is observed [11]. “Firing up”/“sensitization” supports the hypothesis that neuroinfection in early childhood may lead to increased release of cytokines when the immune system is activated later in life. These processes lead to neurotransmitter disorders. Stress induces a pro-inflammatory immune response in CNS. However, this is usually reduced after a stressful event. Psychopathological symptoms and neuroinflammation are associated with the immune response of CNS cells to stress, and neuroinflammation is involved in stress-related behavioral changes induced by cytokines and mediated by neurotransmitters. Studies have found that after exposure to chronic stress or repeated stressful events, the threshold for physiological responses of the CNS to stress decreases. As a result, less stimulus is enough to activate an immune or neurotransmitter response. In an animal study, it was shown that with age, the brain is in a sensitized state and produces more cytokines for inflammatory stimuli than the brain of young animals [6]. Repeated exposure to pro-inflammatory cytokines leads to increased neurotransmitter responses [12] as, for example, with tumor necrosis factor alpha (TNF-α) [13]. Stress causes activation and proliferation of microglia in the CNS, which may possibly mediate these cytokine effects [6]. Chronic stress is known to affect the glutamatergic system, ionotropic and metabotropic glutamate receptors and excitatory amino acid transporters [14], which may also play a role in the development of TRS, as it is associated with higher levels of glutamate in the anterior cingulate cortex [15].

3. Vulnerability-Stress-Inflammation in Treatment Resistance Schizophrenia

The risk of developing TRS increases with stressful life events or psychological stress, especially those that act at key periods in the development of the CNS. The Sch vulnerability-stress model was first proposed by Zubin and Vesna [16], who suggested that stress above the vulnerability threshold in humans contributes to the development of a psychotic episode. It is important to add inflammation to this model, forming the vulnerability-stress-inflammation model, since neuroinflammation plays an important role in the pathogenesis of TRS and can in turn be caused by stress [17]. For example, if an inflammatory response in the CNS is stimulated in a second trimester of pregnancy or offspring while the CNS is still developing, the offspring may be a risk of developing Sch. Animal studies have shown that exposure to stress at an early age leads to an increase in the level of pro-inflammatory cytokines [18], which play an important role in the development of Sch and TRS. Vulnerability-stress-inflammation-induced immune dysregulation is associated with dysregulation of many neurotransmitter systems that APs cannot therapeutically address. Thus, the development of TRS is likely associated with stress-induced inflammation [19].

4. Prenatal, Perinatal and Postnatal Infection in Treatment Resistance Schizophrenia

Existing epidemiological studies give us the idea that prenatal exposure to maternal infection is associated with an increased risk of Sch in the offspring [20]. The risk of developing Sch may be related to the direct effects of neuroinfection (e.g., disruption of structure due to cyst formation, exposure to inflammatory factors) as well as neurochemical changes such as increased dopamine levels associated with poor performance. Catechol-O-methyl transferase (COMT) and increased dopamine synthesis caused by Toxoplasma gondii infection [21]. Exposure to viruses and other infectious agents—influenza, herpes simplex virus type 2, Coronavirus Disease 2019 (COVID-19) during pregnancy and at the time of conception—is associated with a greater risk of psychotic disorders [22]. Given the changes in pro-inflammatory cytokine production in pregnant women with COVID-19, schizophrenic and psychotic disorders may potentially be a long-term risk in the offspring of pregnant women who have experienced COVID-19 [23]. Animal studies have also provided evidence for the role of pre- and perinatal infections in the later development of Sch [24]. For example, after prenatal exposure to viruses, offspring show typical symptoms of Sch, such as cognitive impairment or startle reflex abnormalities [25]. Maternal bacterial infection during pregnancy is closely associated with the development of psychosis in the offspring and varied depending on the severity of the infection and the sex of the offspring. At the same time, the effect of a multisystem bacterial infection was almost two times higher than that of a less severe localized bacterial infection [26]. Of interest are studies that have demonstrated the association of Sch development with prenatal or early childhood exposure to various viruses [27], respiratory infections [28] and infections of the genital organs or reproductive tract [29]. Because Sch develops more frequently during adolescence or adulthood, it is important to establish a possible mechanism for the association between early infection and Sch in adults. Studies in animal models show that early infection or immune activation affects several processes of neurogenesis, including dopaminergic and glutamatergic neurotransmission [30]. The study of bacterial [26] and some other infections in humans [31] are examples that highlight this connection. The risk of developing TRS is also indicated by an increased level of C-reactive protein (CRP) or cytokines in childhood [32]. In addition, neuroinfection at a later age has been shown to be associated with an increased risk of developing TRS. A large epidemiological study conducted in Denmark showed that autoimmune disorders, as well as severe infections, increase the risk of developing Sch and Sch spectrum disorders. This is especially true for patients with both risk factors for TRS [33].

5. Cytokine Imbalance in Treatment Resistance Schizophrenia

Based on the meta-analyses by Momtazmanesh et al. [34], it is possible to conditionally classify cytokines according to their serum levels in patients with TRS into four groups: group 1—elevated cytokines, including interleukin 6 (IL-6), tumor necrosis factor alpha (TNF-α), interleukin 1 beta (IL-1β), interleukin 12 (IL-12) and transforming growth factor beta (TGF-β); group 2—unchanged cytokines, including interleukin 2 (IL-2), interleukin 4 (IL-4) and interleukin 17 (IL-17); group 3—elevated or unchanged cytokines, including interleukin 8 (IL-8) and interferon gamma (IFN-γ); group 4—interleukin 10 (IL-10) with increased, decreased and unchanged serum levels. However, this grouping is not unambiguous and includes mainly pro-inflammatory cytokines. In addition, the authors did not provide an analysis of the relationship between the levels of pro-inflammatory and anti-inflammatory cytokines in patients with TRS.
Higher serum levels of pro-inflammatory cytokines are characteristic of both patients with the first episode of Sch and patients with relapse and TRS, compared with the control group [35]. IL-1β, IL-6 and TGF-β were elevated at the first psychotic episode, and Sch flare normalized after AP treatment. Conversely, the levels of IL-12, IFN-γ, TNF-α and soluble interleukin 2 receptor (sIL-2R) remained elevated during exacerbations and during AP therapy [36]. A study of interleukins in the cerebrospinal fluid (CSF) showed that the levels of interleukin 6 (IL-6) and IL-8 were increased in Sch, but not significantly increased in affective disorders [37]. A meta-analysis of CSF cytokines showed higher levels of pro-inflammatory cytokines and lower levels of anti-inflammatory cytokines in patients with Sch and TRS [38].
It is known that dopaminergic dysfunction is a significant feature in the pathophysiology of TRS [39]. Interactions between cytokines and neurotransmitters in certain areas of the brain, and also during brain development, are important in the pathophysiology of TRS. Apparently, the pro-inflammatory cytokine IL-1β, which induces the transformation of rat mesencephalic progenitor cells into a dopaminergic phenotype [40], and IL-6, which reduces the survival of serotonergic neurons in the fetal brain, seem to play an important role in influencing neurotransmitter systems in TRS. [41]. Studies have found abnormalities in the cytokine system in patients with TRS [42][43]. There is evidence that the levels of IL-2 and IL-6 were elevated in patients with TRS, which is probably associated with the activation of the inflammatory response system (IRS). Moreover, serum IL-2 or IL-6 and cortisol are positively correlated with Sch, supporting the hypothesis that hypercortisolemia may also be caused by pro-inflammatory cytokines [44][45].
The delicate balance between pro-inflammatory and anti-inflammatory cytokines determines the net effect of a neuroinflammatory response in patients with TRS. Perturbations in this equilibrium can drive the patient defense immune response towards chronic neuroinflammation (pro-inflammatory) or towards healing (anti-inflammatory). Thus, a cytokine imbalance may be beneficial to the patient with TRS by initiating the neuroinflammatory response. However, overproduction or underproduction of pro-inflammatory or anti-inflammatory endogenous mediators (cytokines) may actually be deleterious to the patient with “non-dopamine” TRS. In addition, chronic neuroinflammation, supported by an imbalance between pro-inflammatory and anti-inflammatory cytokines, and persistent dopaminergic neurotransmission disorder can be considered as an overlap syndrome in patients with “dopamine” TRS.

References

  1. Borst, K.; Dumas, A.A.; Prinz, M. Microglia: Immune and non-immune functions. Immunity 2021, 54, 2194–2208.
  2. Lasselin, J. Back to the future of psychoneuroimmunology: Studying inflammation-induced sickness behavior. Brain Behav. Immun. Health 2021, 18, 100379.
  3. Martínez-Cué, C.; Rueda, N. Cellular Senescence in Neurodegenerative Diseases. Front. Cell. Neurosci. 2020, 14, 16.
  4. Lecours, C.; Bordeleau, M.; Cantin, L.; Parent, M.; Paolo, T.D.; Tremblay, M.È. Microglial implication in Parkinson’s disease: Loss of beneficial physiological roles or gain of inflammatory functions? Front. Cell. Neurosci. 2018, 12, 282.
  5. Fonken, L.K.; Frank, M.G.; Gaudet, A.D.; Maier, S.F. Stress and aging act through common mechanisms to elicit neuroinflammatory priming. Brain Behav. Immun. 2018, 73, 133–148.
  6. Woodburn, S.C.; Bollinger, J.L.; Wohleb, E.S. The semantics of microglia activation: Neuroinflammation, homeostasis, and stress. J. Neuroinflamm. 2021, 18, 258.
  7. Dantzer, R. Neuroimmune interactions: From the brain to the immune system and vice versa. Physiol. Rev. 2018, 98, 477–504.
  8. Mondelli, V.; Vernon, A.C.; Turkheimer, F.; Dazzan, P.; Pariante, C.M. Brain microglia in psychiatric disorders. Lancet Psychiatry 2017, 4, 563–572.
  9. Amato, D.; Beasley, C.L.; Hahn, M.K.; Vernon, A.C. Neuroadaptations to antipsychotic drugs: Insights from pre-clinical and human post-mortem studies. Neurosci. Biobehav. Rev. 2017, 76, 317–335.
  10. Stapel, B.; Sieve, I.; Falk, C.S.; Bleich, S.; Hilfiker-Kleiner, D.; Kahl, K.G. Second generation atypical antipsychotics olanzapine and aripiprazole reduce expression and secretion of inflammatory cytokines in human immune cells. J. Psychiatr. Res. 2018, 105, 95–102.
  11. Frank, M.G.; Fonken, L.K.; Annis, J.L.; Watkins, L.R.; Maier, S.F. Stress disinhibits microglia via down-regulation of CD200R: A mechanism of neuroinflammatory priming. Brain Behav. Immun. 2018, 69, 62–73.
  12. Jeon, S.W.; Kim, Y.K. The role of neuroinflammation and neurovascular dysfunction in major depressive disorder. J. Inflamm. Res. 2018, 11, 179.
  13. Schwarz, M.J. Cytokines, neurophysiology, neuropsychology, and psychiatric symptoms. Dialogues Clin. Neurosci. 2022, 5, 139–153.
  14. Réus, G.Z.; Abelaira, H.M.; Coutellier, L.D.; Manosso, L.M.; Pavlovic, Z.M. Role of Glutamatergic Neurotransmission in the Pathophysiology of Stress-Related Disorders and Chronic Stress Response. In Glutamate and Neuropsychiatric Disorders; Springer International Publishing: Berlin/Heidelberg, Germany, 2022; pp. 65–112.
  15. Kumar, V.; Manchegowda, S.; Jacob, A.; Rao, N.P. Glutamate metabolites in treatment resistant schizophrenia: A meta-analysis and systematic review of 1H-MRS studies. Psychiatry Res. Neuroimaging 2020, 300, 111080.
  16. Zubin, J.; Spring, B. Vulnerability-a new view of schizophrenia. J. Abnorm. Psychol. 1977, 86, 103–126.
  17. Müller, N. Inflammation in Schizophrenia: Pathogenetic Aspects and Therapeutic Considerations. Schizophr. Bull. 2018, 44, 973–982.
  18. Lumertz, F.S.; Kestering-Ferreira, E.; Orso, R.; Creutzberg, K.C.; Tractenberg, S.G.; Stocchero, B.A.; Viola, T.W.; Grassi-Oliveira, R. Effects of early life stress on brain cytokines: A systematic review and meta-analysis of rodent studies. Neurosci. Biobehav. Rev. 2022, 139, 104746.
  19. Howes, O.D.; Thase, M.E.; Pillinger, T. Treatment resistance in psychiatry: State of the art and new directions. Mol. Psychiatry 2022, 27, 58–72.
  20. Cheslack-Postava, K.; Brown, A.S. Prenatal infection and schizophrenia: A decade of further progress. Schizophr. Res. 2022, 247, 7–15.
  21. Rovira, P.; Gutiérrez, B.; Sorlózano-Puerto, A.; Gutiérrez-Fernández, J.; Molina, E.; Rivera, M.; Martínez-Leal, R.; Ibanez-Casas, I.; Martín-Laguna, M.V.; Rosa, A.; et al. Toxoplasma gondii Seropositivity Interacts with Catechol-O-methyltransferase Val105/158Met Variation Increasing the Risk of Schizophrenia. Genes 2022, 13, 1088.
  22. Stilo, S.A.; Murray, R.M. Non-genetic factors in schizophrenia. Curr. Psychiatry Rep. 2019, 21, 100.
  23. Cavalcante, M.B.; Cavalcante, C.T.D.M.B.; Sarno, M.; Barini, R.; Kwak-Kim, J. Maternal immune responses and obstetrical outcomes of pregnant women with COVID-19 and possible health risks of offspring. J. Reprod. Immunol. 2021, 143, 103250.
  24. Minakova, E.; Warner, B.B. Maternal immune activation, central nervous system development and behavioral phenotypes. Birth Defects Res. 2018, 110, 1539–1550.
  25. Benros, M.E.; Mortensen, P.B. Role of Infection, Autoimmunity, Atopic Disorders, and the Immune System in Schizophrenia: Evidence from Epidemiological and Genetic Studies. Curr. Top. Behav. Neurosci. 2020, 44, 141–159.
  26. Lee, Y.H.; Cherkerzian, S.; Seidman, L.J.; Papandonatos, G.D.; Savitz, D.A.; Tsuang, M.T.; Goldstein, J.M.; Buka, S.L. Maternal Bacterial Infection During Pregnancy and Offspring Risk of Psychotic Disorders: Variation by Severity of Infection and Offspring Sex. Am. J. Psychiatry 2020, 177, 66–75.
  27. Vasistha, N.A.; Pardo-Navarro, M.; Gasthaus, J.; Weijers, D.; Müller, M.K.; García-González, D.; Malwade, S.; Korshunova, I.; Pfisterer, U.; von Engelhardt, J.; et al. Maternal inflammation has a profound effect on cortical interneuron development in a stage and subtype-specific manner. Mol. Psychiatry 2020, 25, 2313–2329.
  28. Lydholm, C.N.; Köhler-Forsberg, O.; Nordentoft, M.; Yolken, R.H.; Mortensen, P.B.; Petersen, L.; Benros, M.E. Parental Infections Before, During, and After Pregnancy as Risk Factors for Mental Disorders in Childhood and Adolescence: A Nationwide Danish Study. Biol. Psychiatry 2019, 85, 317–325.
  29. Chaves Filho, A.; Mottin, M.; Lós, D.B.; Andrade, C.H.; Macedo, D.S. The tetrapartite synapse in neuropsychiatric disorders: Matrix metalloproteinases (MMPs) as promising targets for treatment and rational drug design. Biochimie 2022, 201, 79–99.
  30. Meehan, C.; Harms, L.; Frost, J.D.; Barreto, R.; Todd, J.; Schall, U.; Shannon Weickert, C.; Zavitsanou, K.; Michie, P.T.; Hodgson, D.M. Effects of immune activation during early or late gestation on schizophrenia-related behaviour in adult rat offspring. Brain Behav. Immun. 2017, 63, 8–20.
  31. Müller, N. Inflammation and Immunity in Schizophrenia. Immuno-Psychiatry 2021, 2, 227–240.
  32. Metcalf, S.A.; Jones, P.B.; Nordstrom, T.; Timonen, M.; Mäki, P.; Miettunen, J.; Jääskeläinen, E.; Järvelin, M.R.; Stochl, J.; Murray, G.K.; et al. Serum C-reactive protein in adolescence and risk of schizophrenia in adulthood: A prospective birth cohort study. Brain Behav. Immun. 2017, 59, 253–259.
  33. Orlovska-Waast, S.; Benros, M.E. Autoimmune Diseases and Infections as Risk Factors for Mental Disorders. In Immuno-Psychiatry; Springer: Berlin/Heidelberg, Germany, 2021; pp. 3–16.
  34. Momtazmanesh, S.; Zare-Shahabadi, A.; Rezaei, N. Cytokine alterations in schizophrenia: An updated review. Front. Psychiatry 2019, 10, 892.
  35. Goldsmith, D.R.; Rapaport, M.H.; Miller, B.J. A meta-analysis of blood cytokine network alterations in psychiatric patients: Comparisons between schizophrenia, bipolar disorder and depression. Mol. Psychiatry 2016, 21, 1696–1709.
  36. Miller, B.J.; Buckley, P.; Seabolt, W.; Mellor, A.; Kirkpatrick, B. Meta-analysis of cytokine alterations in schizophrenia: Clinical status and antipsychotic effects. Biol. Psychiatry 2011, 70, 663–671.
  37. Orlovska-Waast, S.; Köhler-Forsberg, O.; Brix, S.W.; Nordentoft, M.; Kondziella, D.; Krogh, J.; Benros, M.E. Cerebrospinal fluid markers of inflammation and infections in schizophrenia and affective disorders: A systematic review and meta-analysis. Mol. Psychiatry 2019, 24, 869–887.
  38. Wang, A.K.; Miller, B.J. Meta-analysis of Cerebrospinal Fluid Cytokine and Tryptophan Catabolite Alterations in Psychiatric Patients: Comparisons Between Schizophrenia, Bipolar Disorder, and Depression. Schizophr. Bull. 2018, 44, 75–83.
  39. Brandon, A.; Cui, X.; Luan, W.; Ali, A.A.; Pertile, R.A.N.; Alexander, S.A.; Eyles, D.W. Prenatal hypoxia alters the early ontogeny of dopamine neurons. Transl. Psychiatry 2022, 12, 238.
  40. Wang, M.; Ling, K.H.; Tan, J.J.; Lu, C.B. Development and differentiation of midbrain dopaminergic neuron: From bench to bedside. Cells 2020, 9, 1489.
  41. Jarskog, L.F.; Xiao, H.; Wilkie, M.B.; Lauder, J.M.; Gilmore, J.H. Cytokine regulation of embryonic rat dopamine and serotonin neuronal survival in vitro. Int. J. Dev. Neurosci. Off. J. Int. Soc. Dev. Neurosci. 1997, 15, 711–716.
  42. Romeo, B.; Brunet-Lecomte, M.; Martelli, C.; Benyamina, A. Kinetics of cytokine levels during antipsychotic treatment in schizophrenia: A meta-analysis. Int. J. Neuropsychopharmacol. 2018, 21, 828–836.
  43. Scheiber, C.; Schulz, T.; Schneider, J.M.; Bechter, K.; Schneider, E.M. Old and New Biomarkers for Infection, Inflammation, and Autoimmunity in Treatment-Resistant Affective and Schizophrenic Spectrum Disorders. Pharmaceuticals 2022, 15, 299.
  44. Dziurkowska, E.; Wesolowski, M. Cortisol as a Biomarker of Mental Disorder Severity. J. Clin. Med. 2021, 10, 5204.
  45. Zhang, X.Y.; Zhou, D.F.; Cao, L.Y.; Wu, G.Y.; Shen, Y.C. Cortisol and cytokines in chronic and treatment-resistant patients with schizophrenia: Association with psychopathology and response to antipsychotics. Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol. 2005, 30, 1532–1538.
More
Information
Subjects: Neurosciences
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , , , , ,
View Times: 413
Entry Collection: Neurodegeneration
Revisions: 2 times (View History)
Update Date: 27 Oct 2022
1000/1000