Mechanisms of SARS-CoV-2-Induced Neurological Damage: Comparison
Please note this is a comparison between Version 2 by Jason Zhu and Version 1 by Marco Velasco-Velazquez.

COVID-19 patients can exhibit a wide range of clinical manifestations affecting various organs and systems. Neurological symptoms have been reported in COVID-19 patients, both during the acute phase of the illness and in cases of long-term COVID. Moderate symptoms include ageusia, anosmia, altered mental status, and cognitive impairment, and in more severe cases can manifest as ischemic cerebrovascular disease and encephalitis.

  • COVID-19
  • SARS-CoV-2
  • neurological symptoms
  • neuroprotective compounds

1. Direct Neuronal Damage

SARS-CoV-2 can reach the CNS via the olfactory tract [84][1]. The human olfactory epithelium is a pseudostratified epithelium composed of Bowman’s glands, horizontal and globose basal cells, microvillar cells, sustentacular cells, and olfactory sensory neurons that extends a single axon towards the olfactory bulb in the brain. Sustentacular cells in the olfactory epithelium exhibit high levels of ACE2 and TMPRSS2 [75][2] and a study in hamsters showed SARS-CoV-2 active infection in those cells [85][3]. In agreement, autopsies of COVID-19 patients revealed that the olfactory sensory epithelium was severely damaged [75][2]. Despite the absence of ACE2 and TMPRSS2 in the olfactory sensory epithelium, the olfactory nerve tissue was found to be positive to SARS-CoV-2 in post-mortem examinations of COVID-19 patients [86][4]. Thus, it is possible that the replication of virions in the olfactory epithelium leads to infection in the olfactory bulb as blood vessels and pericytes in this brain region express both proteins [87,88][5][6]. These neurotrophic properties of SARS-CoV-2 explain the onset of anosmia as a prior symptom [89][7]. Interestingly, in mice, ACE2 and TMPRSS2 expression in the olfactory epithelium increases with age [75[2][8],90], suggesting a possible mechanism by which older patients are more vulnerable to the disease and neurological complications.
Furthermore, the virus can reach brain tissue by the hematogenous route, in which endothelial cells or leukocytes are infected by the virus that passes from the bloodstream to the CNS [75][2] across the blood–brain barrier (BBB) [91,92][9][10] or by transmigration of peripheral immune cells, following the “Trojan horse” mechanism [93,94][11][12]. For example, brain vascular cells and choroidal barrier cells robustly express several genes that are relevant for SARS-CoV-2 entry into the brain [95][13]. SARS-CoV-2 infects and crosses an in vitro model of the BBB comprising primary brain microvascular endothelial cells and astrocytes [96][14]. Infection of ACE2-overexpressing primary human endothelial cells by SARS-CoV-2 induces the overexpression of coagulation factors, adhesion molecules, and pro-inflammatory cytokines, as well as the formation of multinucleated syncytia and endothelial cell lysis [97][15]. Consequently, SARS-CoV-2 alters the function and integrity of the BBB, which contributes to viral encephalopathy [98,99][16][17].
Furthermore, ACE2 receptors have been found in glial cells of the brain and spinal neurons, so SARS-CoV-2 can adhere, multiply, and cause direct damage to neuronal tissue [100][18]. Neuronal infection has been associated with neurodegeneration and neurovascular remodeling [101][19], causing cerebral vascular/endothelial dysfunctions that can generate cerebral circulatory disturbances [102][20]. Helms et al., using perfusion imaging, demonstrated in patients with COVID-19 that SARS-CoV-2 neuroinvasion causes bilateral frontotemporal hypoperfusion, demonstrating cerebral circulatory impairment [103][21]. As consequences of cerebral hypoxia, COVID patients can show cerebral vasodilation, brain cell swelling, interstitial edema, obstruction of cerebral blood flow, and even headache due to ischemia and congestion [104][22].

2. Indirect Effects

Exacerbated inflammation participates in the damage to nervous tissue, as in other target organs. SARS-CoV-2 elicits an exacerbated and deregulated immune response of soluble immune mediators, termed a “cytokine storm” [105][23]. Multiple immune mediators, such as IL-1β, IL-6, CXCL10, TNFα, and other diverse cytokines are produced in response to SARS-CoV-2 infection and have been associated with functional alterations or tissue damage in different organs, including the brain [106][24].
In addition, elevated levels of pro-inflammatory cytokines could participate in aggravating neuropathies during critical COVID-19 illness. The overproduction of systemic inflammatory factors (cytokines, nitric oxide, and oxygen radicals) has been associated with the malfunction of peripheral nerves [107][25] as well as microvascular disorders and electrical and metabolic (channel) disturbances in muscle cells [108][26].
In addition, chronic damage to other systems can also damage the CNS through ischemia, metabolic dysfunction, and hormonal dysregulation [109][27]. Coagulopathy and endotheliopathy triggered by cytokine storms are potential mechanisms causing ischemic stroke in COVID-19 patients [110,111][28][29]. Furthermore, COVID-19 patients have elevated levels of von Willebrand factor (VWF) antigen, VWF activity, and factor VIII [112][30], leukocytosis, thrombocytopenia, increased partial thromboplastin time, and low levels of antithrombin activity [113][31]. COVID-19 patients are at an increased risk of developing venous thromboembolism and disseminated intravascular coagulation [114][32].
Cerebral venous sinus thrombosis (CVT) can be caused by the hypercoagulable state in SARS-CoV-2 infection, which may be triggered by endothelial dysfunction that predisposes vessels to thrombus formation, platelet dysfunction, hypoxia, and/or alterations of the complement system [115,116][33][34]. CVT may cause generalized neurological deficits [117][35] and there are multiple reports of its association with SARS-CoV-2 infection [118,119][36][37].
Moreover, the renin-angiotensin-aldosterone system (RAAS) can contribute to the appearance of brain damage and systemic hyperinflammatory state in COVID-19 patients [120,121][38][39]. It has been reported that during SARS-CoV-2 infection: (1) the local levels of angiotensin II (Ang II) increase, acting on angiotensin II type 1 receptors (AT1), and thus increasing arterial pressure; (2) there is endothelial dysfunction in the cerebral vessels in the CNS, which increases the risk of cerebral hemorrhage; and (3) the generation of Ang (1–7) decreases, preventing the vasodilator, neuroprotective, and antifibrotic effects of Ang (1–7)/Mas receptor signaling [122,123][40][41].

3. Oxidative Stress

An overproduction of reactive oxygen species (ROS) and the deprivation of antioxidant mechanisms are known to be crucial for viral replication and subsequent virus-associated disease, as shown by increased ROS levels and impaired antioxidant defense during SARS-CoV-2 infection [124][42]. The viral protease Mpro activates nuclear factor kappa B (NF-kB)-mediated transcription, which correlates with increased levels of intracellular ROS [125][43]. In addition, Mpro causes a significant increase in ROS production in HL-CZ cells, which, in turn, induces cellular apoptosis. Similarly, SARS-CoV-2 increases oxidative stress in nervous tissue, which contributes to neuronal cell death [126,127][44][45]. A post-mortem case study showed that 37 of 43 COVID-19 patients had astrogliosis and 34 had microglial activation in the brainstem and cerebellum [128][46]. In a preclinical trial, neuronal microgliosis in the brain has been observed to persist beyond SARS-CoV-2 clearance [129][47].

4. Protein Misfolding

Protein misfolding and aggregation have also been reported in COVID-19. Interactions between the S protein of SARS-CoV-2 and its receptor ACE2 favor the spread of cytosolic prions and tau aggregates [130][48]. The RBD domain of the S1 subunit from SARS-CoV-2 S protein (RBD SARS-CoV-2 S1) binds heparin and heparin-binding proteins, accelerating the pathological aggregation of brain proteins, including Aβ (amyloid beta), α-synuclein, tau, prion, and TDP-43 RRM [131][49]. In addition, SARS-CoV-2-infected hamsters develop microgliosis in the olfactory bulb and selective accumulation of hyperphosphorylated tau and α-synuclein in the cortex after virus clearance, indicating that proteinopathies can be generated in neurons post-infection [129][47]. Although further studies are required, this evidence suggests that protein misfolding may play a role in the neurological symptoms caused by SARS-CoV-2 infection.

5. Changes in Neurotrophins Expression

Neurotrophins are growth factors acting as regulators of neuronal survival, development, function, and plasticity [132][50]. Neurotrophins include nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3), and neurotrophin-4 (NT-4) [133][51]. In addition to their classical functions, they regulate axonal and dendritic growth and guidance, synaptic structure and connections, neurotransmitter release, and long-term potentiation, a cellular mechanism underlying memory and learning [134][52]. The circulating levels of BDNF [135][53] and NGF [136][54] are reduced in adult COVID-19 patients compared to healthy individuals. BDNF reduction is higher in patients > 60 years of age [137][55], indicating age-dependent effects. Reductions in serum BDNF correlate with the severity of the disease [137,138][55][56] and cognitive impairment after recovery [139][57]. Interestingly, adult COVID-19 patients that required supplemental oxygen had even lower BDNF serum concentrations [135][53], showing an interplay between deregulated BDNF levels and viral hypoxia. These findings support the role of neurotrophins in regulating neurological outcomes in COVID-19 patients. However, further studies are required to define the extent of their participation and the mechanisms involved, especially in the long-lasting effects of this disease.

References

  1. Li, Y.C.; Bai, W.Z.; Hashikawa, T. The neuroinvasive potential of SARS-CoV2 may play a role in the respiratory failure of COVID-19 patients. J. Med. Virol. 2020, 92, 552–555.
  2. Bilinska, K.; Jakubowska, P.; Von Bartheld, C.S.; Butowt, R. Expression of the SARS-CoV-2 entry proteins, ACE2 and TMPRSS2, in cells of the olfactory epithelium: Identification of cell types and trends with age. ACS Chem. Neurosci. 2020, 11, 1555–1562.
  3. Bryche, B.; St Albin, A.; Murri, S.; Lacôte, S.; Pulido, C.; Ar Gouilh, M.; Lesellier, S.; Servat, A.; Wasniewski, M.; Picard-Meyer, E.; et al. Massive transient damage of the olfactory epithelium associated with infection of sustentacular cells by SARS-CoV-2 in golden Syrian hamsters. Brain Behav. Immun. 2020, 89, 579–586.
  4. Fabbri, V.P.; Foschini, M.P.; Lazzarotto, T.; Gabrielli, L.; Cenacchi, G.; Gallo, C.; Aspide, R.; Frascaroli, G.; Cortelli, P.; Riefolo, M.; et al. Brain ischemic injury in COVID-19-infected patients: A series of 10 post-mortem cases. Brain Pathol. 2021, 31, 205–210.
  5. Brann, D.H.; Tsukahara, T.; Weinreb, C.; Lipovsek, M.; Van den Berge, K.; Gong, B.; Chance, R.; Macaulay, I.C.; Chou, H.J.; Fletcher, R.B.; et al. Non-neuronal expression of SARS-CoV-2 entry genes in the olfactory system suggests mechanisms underlying COVID-19-associated anosmia. Sci. Adv. 2020, 6, eabc5801.
  6. Lechien, J.R.; Radulesco, T.; Calvo-Henriquez, C.; Chiesa-Estomba, C.M.; Hans, S.; Barillari, M.R.; Cammaroto, G.; Descamps, G.; Hsieh, J.; Vaira, L.; et al. ACE2 & TMPRSS2 Expressions in Head & Neck Tissues: A Systematic Review. Head Neck Pathol. 2021, 15, 225–235.
  7. Al-olama, M.; Rashid, A.; Garozzo, D. COVID-19-associated meningoencephalitis complicated with intracranial hemorrhage: A case report. Acta. Neurochir. 2020, 162, 1495–1499.
  8. Brechbühl, J.; Lopes, A.C.; Wood, D.; Bouteiller, S.; de Vallière, A.; Verdumo, C.; Broillet, M.C. Age-dependent appearance of SARS-CoV-2 entry sites in mouse chemosensory systems reflects COVID-19 anosmia-ageusia symptoms. Commun. Biol. 2021, 4, 880.
  9. Proust, A.; Queval, C.J.; Harvey, R.; Adams, L.; Bennett, M.; Wilkinson, R. Differential effects of SARS-CoV-2 variants on central nerv-ous system cells and blood–brain barrier functions. J. Neuroinflam. 2023, 20, 1–17.
  10. Reynolds, J.L.; Mahajan, S.D. SARS-CoV2 alters blood brain barrier integrity contributing to neuro-inflammation. J. Neuroimmune Pharmacol. 2021, 16, 4–6.
  11. Dahm, T.; Rudolph, H.; Schwerk, C.; Schroten, H.; Tenenbaum, T. Neuroinvasion and inflammation in viral central nervous system infections. Mediat. Inflamm. 2016, 2016, 8562805.
  12. Gu, J.; Gong, E.; Zhang, B.; Zheng, J.; Gao, Z.; Zhong, Y.; Zou, W.; Zhan, J.; Wang, S.; Xie, Z.; et al. Multiple organ infection and the pathogenesis of SARS. J. Exp. Med. 2005, 202, 415–424.
  13. Yang, A.C.; Kern, F.; Losada, P.M.; Agam, M.R.; Maat, C.A.; Schmartz, G.P.; Fehlmann, T.; Stein, J.A.; Schaum, N.; Lee, D.P.; et al. Dysregulation of brain and choroid plexus cell types in severe COVID-19. Nature 2021, 595, 565–571.
  14. Zhang, B.Y.; Chen, M.; Chen, X.C.; Cao, K.; You, Y.; Qian, Y.J.; Yu, W.K. Berberine reduces circulating inflammatory mediators in patients with severe COVID-19. Br. J. Surg. 2021, 108, E9–E11.
  15. Conde, J.N.; Schutt, W.R.; Gorbunova, E.E.; Mackow, E.R. Recombinant ACE2 expression is required for SARS-CoV-2 to infect primary human endothelial cells and induce inflammatory and procoagulative responses. mBio 2020, 11, e03185-20.
  16. Erickson, M.A.; Rhea, E.M.; Knopp, R.C.; Banks, W.A. Interactions of SARS-CoV-2 with the blood–brain barrier. Int. J. Mol. Sci. 2021, 22, 2681.
  17. Buzhdygan, T.P.; DeOre, B.J.; Baldwin-Leclair, A.; Bullock, T.A.; McGary, H.M.; Khan, J.A.; Razmpour, R.; Hale, J.F.; Galie, P.A.; Potula, R.; et al. The SARS-CoV-2 spike protein alters barrier function in 2D static and 3D microfluidic in-vitro models of the human blood–brain barrier. Neurobiol. Dis. 2020, 146, 105131.
  18. Ahmad, I.; Rathore, F.A. Neurological manifestations and complications of COVID-19: A literature review. J. Clin. Neurosci. 2020, 77, 8–12.
  19. Song, E.; Zhang, C.; Israelow, B.; Lu-Culligan, A.; Prado, A.V.; Skriabine, S.; Lu, P.; Weizman, O.E.; Liu, F.; Dai, Y.; et al. Neuroinvasion of SARS-CoV-2 in human and mouse brain. J. Exp. Med. 2021, 218.
  20. Ghasemi, M.; Umeton, R.P.; Keyhanian, K.; Mohit, B.; Rahimian, N.; Eshaghhosseiny, N.; Davoudi, V. SARS-CoV-2 and acute cerebrovascular events: An overview. J. Clin. Med. 2021, 10, e20202135.
  21. Helms, J.; Kremer, S.; Merdji, H.; Clere-Jehl, R.; Schenck, M.; Kummerlen, C.; Collange, O.; Boulay, C.; Fafi-Kremer, S.; Ohana, M.; et al. Neurologic Features in Severe SARS-CoV-2 Infection. N. Engl. J. Med. 2020, 382, 2268–2270.
  22. Guo, Y.R.; Cao, Q.D.; Hong, Z.S.; Tan, Y.Y.; Chen, S.D.; Jin, H.J.; Tan, K.S.; Wang, D.Y.; Yan, Y. The origin, transmission and clinical therapies on coronavirus disease 2019 (COVID-19) outbreak—An update on the status. Mil. Med. Res. 2020, 7, 1–10.
  23. Huang, C.; Wang, Y.; Li, X.; Ren, L.; Zhao, J.; Hu, Y.; Zhang, L.; Fan, G.; Xu, J.; Gu, X.; et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020, 395, 497–506.
  24. Conti, P.; Ronconi, G.; Caraffa, A.; Gallenga, C.E.; Ross, R.; Frydas, I.; Kritas, S.K. Induction of pro-inflammatory cytokines (IL-1 and IL-6) and lung inflammation by Coronavirus-19 (COVI-19 or SARS-CoV-2): Anti-inflammatory strategies. J. Biol. Regul. Homeost. Agents 2020, 34, 327–331.
  25. Hasan, L.K.; Deadwiler, B.; Haratian, A.; Bolia, I.K.; Weber, A.E.; Petrigliano, F.A. Effects of COVID-19 on the musculoskeletal system: Clinician’s guide. Orthop. Res. Rev. 2021, 13, 141–150.
  26. Disser, N.P.; De Micheli, A.J.; Schonk, M.M.; Konnaris, M.A.; Piacentini, A.N.; Edon, D.L.; Toresdahl, B.G.; Rodeo, S.A.; Casey, E.K.; Mendias, C.L. Musculoskeletal Consequences of COVID-19. J. Bone Jt. Surg. Am. 2020, 102, 1197–1204.
  27. Iadecola, C.; Anrather, J.; Kamel, H. Effects of COVID-19 on the nervous system. Cell 2020, 183, 16–27.e11.
  28. Vogrig, A.; Gigli, G.L.; Bnà, C.; Morassi, M. Stroke in patients with COVID-19: Clinical and neuroimaging characteristics. Neurosci. Lett. 2021, 743, 135564.
  29. Altable, M.; de la Serna, J.M. Cerebrovascular disease in COVID-19: Is there a higher risk of stroke? Brain Behav. Immun. Health 2020, 6, 100092.
  30. Leonard-Lorant, I.; Delabranche, X.; Severac, F.; Helms, J.; Pauzet, C.; Collange, O.; Schneider, F.; Labani, A.; Bilbault, P.; Moliere, S.; et al. Acute pulmonary embolism in patients with COVID-19 at CT angiography and relationship to d-dimer levels. Radiology 2020, 296, E189–E191.
  31. Zhang, Y.; Xiao, M.; Zhang, S.; Xia, P.; Cao, W.; Jiang, W.; Chen, H.; Ding, X.; Zhao, H.; Zhang, H.; et al. Coagulopathy and Antiphospholipid Antibodies in Patients with COVID-19. N. Engl. J. Med. 2020, 382, e38.
  32. Pavoni, V.; Gianesello, L.; Pazzi, M.; Stera, C.; Meconi, T.; Frigieri, F.C. Evaluation of coagulation function by rotation thromboelastometry in critically ill patients with severe COVID-19 pneumonia. J. Thromb. Thrombolysis 2020, 50, 281–286.
  33. Cavalcanti, D.D.; Raz, E.; Shapiro, M.; Dehkharghani, S.; Yaghi, S.; Lillemoe, K.; Nossek, E.; Torres, J.; Jain, R.; Riina, H.A.; et al. Cerebral venous thrombosis associated with COVID-19. AJNR Am. J. Neuroradiol. 2020, 41, 1370–1376.
  34. Ghosh, R.; Roy, D.; Mandal, A.; Pal, S.K.; Chandra Swaika, B.; Naga, D.; Pandit, A.; Ray, B.K.; Benito-León, J. Cerebral venous thrombosis in COVID-19. Diabetes Metab. Syndr. 2021, 15, 1039–1045.
  35. Capecchi, M.; Abbattista, M.; Martinelli, I. Cerebral venous sinus thrombosis. J. Thromb. Haemost. 2018, 16, 1918–1931.
  36. Christopher, H.; Tom, N.; Martin, P.; Christian, S.; Salah, E. Cerebral venous sinus thrombosis as a presentation of COVID-19. Eur. J. Case. Rep. Intern. Med. 2020, 7, 001691.
  37. Essajee, F.; Solomons, R.; Goussard, P.; Van Toorn, R. Child with tuberculous meningitis and COVID-19 coinfection complicated by extensive cerebral sinus venous thrombosis. BMJ Case Rep. 2020, 13, e238597.
  38. Zanza, C.; Tassi, M.F.; Romenskaya, T.; Piccolella, F.; Abenavoli, L.; Franceschi, F.; Piccioni, A.; Ojetti, V.; Saviano, A.; Canonico, B.; et al. Lock, stock and barrel: Role of renin-angiotensin-aldosterone system in coronavirus disease 2019. Cells 2021, 10, 1752.
  39. Panariello, F.; Cellini, L.; Speciani, M.; De Ronchi, D.; Atti, A.R. How does SARS-CoV-2 affect the central nervous system? A working hypothesis. Front. Psychiatry 2020, 11, 582345.
  40. Christian, W.; Julian, K.-B. Epidemiology, prognosis and prevention of non-traumatic intracerebral hemorrhage. Curr. Pharm. Des. 2017, 15, 2193–2219.
  41. Benger, M.; Williams, O.; Siddiqui, J.; Sztriha, L. Intracerebral haemorrhage and COVID-19: Clinical characteristics from a case series. Brain Behav. Immun. 2020, 88, 940–944.
  42. Delgado-Roche, L.; Mesta, F. Oxidative stress as key player in severe acute respiratory syndrome coronavirus (SARS-CoV) infection. Arch. Med. Res. 2020, 51, 384–387.
  43. Lin, C.W.; Lin, K.H.; Hsieh, T.H.; Shiu, S.Y.; Li, J.Y. Severe acute respiratory syndrome coronavirus 3C-like protease-induced apoptosis. FEMS Microbiol. Immunol. 2006, 46, 375–380.
  44. Forcados, G.E.; Muhammad, A.; Oladipo, O.O.; Makama, S.; Meseko, C.A. Metabolic implications of oxidative stress and inflammatory process in SARS-CoV-2 pathogenesis: Therapeutic potential of natural antioxidants. Front. Cell Infect. Microbiol. 2021, 11, 654813.
  45. Mingoti, M.E.D.; Bertollo, A.G.; Simões, J.L.B.; Francisco, G.R.; Bagatini, M.D.; Ignácio, Z.M. COVID-19, oxidative stress, and neuroinflammation in the depression route. J. Mol. Neurosci. 2022, 72, 1166–1181.
  46. Matschke, J.; Lütgehetmann, M.; Hagel, C.; Sperhake, J.P.; Schröder, A.S.; Edler, C.; Mushumba, H.; Fitzek, A.; Allweiss, L.; Dandri, M.; et al. Neuropathology of patients with COVID-19 in Germany: A post-mortem case series. Lancet Neurol. 2020, 19, 919–929.
  47. Kaufer, C.; Schreiber, C.S.; Hartke, A.-S.; Denden, I.; Stanelle-Bertram, S.; Beck, S.; Kouassi, N.M.; Beythien, G.; Becker, K.; Schreiner, T.; et al. Microgliosis and neuronal proteinopathy in brain persist beyond viral clearance in SARS-CoV-2 hamster model. EBioMedicine 2022, 79, 103999.
  48. Liu, S.; Hossinger, A.; Heumüller, S.E.; Hornberger, A.; Buravlova, O.; Konstantoulea, K.; Müller, S.A.; Paulsen, L.; Rousseau, F.; Schymkowitz, J.; et al. Highly efficient intercellular spreading of protein misfolding mediated by viral ligand-receptor interactions. Nat. Commun. 2021, 12, 5739.
  49. Idrees, D.; Kumar, V. SARS-CoV-2 spike protein interactions with amyloidogenic proteins: Potential clues to neurodegeneration. Biochem. Biophys. Res. Commun. 2021, 554, 94–98.
  50. Dechant, G.; Neumann, H. Neurotrophins. Adv. Exp. Med. 2002, 513, 303–334.
  51. Hallböök, F. Evolution of the vertebrate neurotrophin and Trk receptor gene families. Curr. Opin. Neurobiol. 1999, 9, 616–621.
  52. Chao, M.V. Neurotrophins and their receptors: A convergence point for many signalling pathways. Nat. Rev. Neurosci. 2003, 4, 299–309.
  53. Asgarzadeh, A.; Fouladi, N.; Asghariazar, V.; Sarabi, S.F.; Khiavi, H.A.; Mahmoudi, M.; Safarzadeh, E. Serum brain-derived neurotrophic factor (BDNF) in COVID-19 patients and its association with the COVID-19 manifestations. J. Mol. Neurosci. 2022, 72, 1820–1830.
  54. Biamonte, F.; Re, A.; Balzamino, B.O.; Ciasca, G.; Santucci, D.; Napodano, C.; Nocca, G.; Fiorita, A.; Marino, M.; Basile, U.; et al. Circulating and salivary NGF and BDNF levels in SARS-CoV-2 infection: Potential predictor biomarkers of COVID-19 disease-preliminary data. J. Pers. Med. 2022, 12, 1877.
  55. Minuzzi, L.G.; Seelaender, M.; Silva, B.S.A.; Cunha, E.D.B.B.; Deus, M.C.; Vasconcellos, F.T.F.; Marqueze, L.F.B.; Gadotti, A.C.; Baena, C.P.; Pereira, T.; et al. COVID-19 outcome relates with circulating bdnf, according to patient adiposity and age. Front. Nutr. 2021, 10, 784429.
  56. Azoulay, D.; Shehadeh, M.; Chepa, S.; Shaoul, E.; Baroum, M.; Horowitz, N.A.; Kaykov, E. Recovery from SARS-CoV-2 infection is associated with serum BDNF restoration. J. Infect. 2020, 81, e79–e81.
  57. Demir, B.; Beyazyüz, E.; Beyazyüz, M.; Çelikkol, A.; Albayrak, Y. Long-lasting cognitive effects of COVID-19: Is there a role of BDNF? Eur. Arch. Psychiatry. Clin. Neurosci. 2023, 273, 1339–1347.
More
Video Production Service