A Potential Role of the Spike Protein in Neurodegenerative Diseases: A Narrative Review

Stephanie Seneff, Anthony M Kyriakopoulos, Greg Nigh, Peter A McCullough, Stephanie Seneff, Anthony M Kyriakopoulos, Greg Nigh, Peter A McCullough

Abstract

Human prion protein and prion-like protein misfolding are widely recognized as playing a causal role in many neurodegenerative diseases. Based on in vitro and in vivo experimental evidence relating to prion and prion-like disease, we extrapolate from the compelling evidence that the spike glycoprotein of SARS-CoV-2 contains extended amino acid sequences characteristic of a prion-like protein to infer its potential to cause neurodegenerative disease. We propose that vaccine-induced spike protein synthesis can facilitate the accumulation of toxic prion-like fibrils in neurons. We outline various pathways through which these proteins could be expected to distribute throughout the body. We review both cellular pathologies and the expression of disease that could become more frequent in those who have undergone mRNA vaccination. Specifically, we describe the spike protein's contributions, via its prion-like properties, to neuroinflammation and neurodegenerative diseases; to clotting disorders within the vasculature; to further disease risk due to suppressed prion protein regulation in the context of widely prevalent insulin resistance; and to other health complications. We explain why these prion-like characteristics are more relevant to vaccine-related mRNA-induced spike proteins than natural infection with SARS-CoV-2. We note with an optimism an apparent loss of prion-like properties among the current Omicron variants. We acknowledge that the chain of pathological events described throughout this paper is only hypothetical and not yet verified. We also acknowledge that the evidence we usher in, while grounded in the research literature, is currently largely circumstantial, not direct. Finally, we describe the implications of our findings for the general public, and we briefly discuss public health recommendations we feel need urgent consideration. An earlier version of this article was previously posted to the Authorea preprint server on August 16, 2022.

Keywords: amyloidosis; cd16+ monocytes; diabetes; exosomes; g quadruplexes; mrna vaccines; neurodegeneration; prion disease; sars-cov-2; spike protein.

Conflict of interest statement

The authors have declared that no competing interests exist.

Copyright © 2023, Seneff et al.

Figures

Figure 1. Schematic of pathways and consequences…
Figure 1. Schematic of pathways and consequences of spike protein binding to the TLR4 receptor in neurons and stimulating the NF-kB signaling response, leading to upregulation of miR-146a and subsequent sequelae.
Source: [55,56,58,70,72,73].
Figure 2. Schematic of the sequelae to…
Figure 2. Schematic of the sequelae to mRNA injection into the deltoid muscle, ultimately leading to neurodegeneration in the brain.
APCs: Antigen-presenting cells; LNPs: Lipid nanoparticles; CJD: Creutzfeldt-Jakob disease; ALS: Amyotrophic lateral sclerosis; AD: Alzheimer’s disease; PD: Parkinson’s disease.

References

    1. Role of prion protein aggregation in neurotoxicity. Corsaro A, Thellung S, Villa V, Nizzari M, Florio T. Int J Mol Sci. 2012;13:8648–8669.
    1. Prion-like C-terminal domain of TDP-43 and α-synuclein interact synergistically to generate neurotoxic hybrid fibrils. Dhakal S, Wyant CE, George HE, Morgan SE, Rangachari V. J Mol Biol. 2021;433:166953.
    1. Potent prion-like behaviors of pathogenic α-synuclein and evaluation of inactivation methods. Tarutani A, Arai T, Murayama S, Hisanaga SI, Hasegawa M. Acta Neuropathol Commun. 2018;6:29.
    1. Amyloidogenesis of Tau protein. Nizynski B, Dzwolak W, Nieznanski K. Protein Sci. 2017;26:2126–2150.
    1. Amyloid fibril structure of α-synuclein determined by cryo-electron microscopy. Li Y, Zhao C, Luo F, et al. Cell Res. 2018;28:897–903.
    1. More than just two peas in a pod: common amyloidogenic properties of tau and alpha-synuclein in neurodegenerative diseases. Lee VM, Giasson BI, Trojanowski JQ. Trends Neurosci. 2004;27:129–134.
    1. TDP-43 prions. Nonaka T, Hasegawa M. Cold Spring Harb Perspect Med. 2018;8
    1. Prion-like transmission of protein aggregates in neurodegenerative diseases. Brundin P, Melki R, Kopito R. Nat Rev Mol Cell Biol. 2010;11:301–307.
    1. Intercellular transmission of a synthetic bacterial cytotoxic prion-like protein in mammalian cells. Revilla-García A, Fernández C, Moreno-Del Álamo M, de Los Ríos V, Vorberg IM, Giraldo R. mBio. 2020;11
    1. Seeding brain protein aggregation by SARS-CoV-2 as a possible long-term complication of COVID-19 infection. Tavassoly O, Safavi F, Tavassoly I. ACS Chem Neurosci. 2020;11:3704–3706.
    1. SARS-CoV-2 spike protein interactions with amyloidogenic proteins: potential clues to neurodegeneration. Idrees D, Kumar V. Biochem Biophys Res Commun. 2021;554:94–98.
    1. Heparin-assisted amyloidogenesis uncovered through molecular dynamics simulations. Khurshid B, Rehman AU, Luo R, Khan A, Wadood A, Anwar J. ACS Omega. 2022;7:15132–15144.
    1. The amplification of CNS damage in Alzheimer's disease due to SARS-CoV2 infection. Nuovo GJ, Suster D, Sawant D, Mishra A, Michaille JJ, Tili E. Ann Diagn Pathol. 2022;61:152057.
    1. Modifications in an emergency: the role of N1-methylpseudouridine in COVID-19 vaccines. Nance KD, Meier JL. ACS Cent Sci. 2021;7:748–756.
    1. Worse than the disease? Reviewing some possible unintended consequences of the mRNA vaccines against COVID-19. Seneff S, Nigh G. Int J Vaccine Theory Pract Res. 2021;2:38–79.
    1. Persistence of SARS CoV-2 S1 protein in CD16+ monocytes in post-acute sequelae of COVID-19 (PASC) up to 15 months post-infection. Patterson BK, Francisco EB, Yogendra R, et al. Front Immunol. 2021;12:746021.
    1. Innate immune suppression by SARS-CoV-2 mRNA vaccinations: the role of G-quadruplexes, exosomes, and microRNAs. Seneff S, Nigh G, Kyriakopoulos AM, McCullough PA. Food Chem Toxicol. 2022;164:113008.
    1. Vaccine mRNA can be detected in blood at 15 days post-vaccination. Fertig TE, Chitoiu L, Marta DS, et al. Biomedicines. 2022;10
    1. Prion-like domains in eukaryotic viruses. Tetz G, Tetz V. Sci Rep. 2018;8:8931.
    1. Prion-like domains in spike protein of SARS-CoV-2 differ across its variants and enable changes in affinity to ACE2. Tetz G, Tetz V. Microorganisms. 2022;10
    1. The possible role of prion-like viral protein domains on the emergence of novel viruses as SARS-CoV-2. Shahzad S, Willcox M. J Mol Evol. 2022;90:227–230.
    1. Prion-like proteins and their computational identification in proteomes. Batlle C, Iglesias V, Navarro S, Ventura S. Expert Rev Proteomics. 2017;14:335–350.
    1. A yeast prion provides a mechanism for genetic variation and phenotypic diversity. True HL, Lindquist SL. Nature. 2000;407:477–483.
    1. Emergence and evolution of yeast prion and prion-like proteins. An L, Fitzpatrick D, Harrison PM. BMC Evol Biol. 2016;16:24.
    1. Giant viruses of amoebae as potential human pathogens. Colson P, La Scola B, Raoult D. Intervirology. 2013;56:376–385.
    1. A pan-coronavirus fusion inhibitor targeting the HR1 domain of human coronavirus spike. Xia S, Yan L, Xu W, et al. Sci Adv. 2019;5:0.
    1. Fusion mechanism of 2019-nCoV and fusion inhibitors targeting HR1 domain in spike protein. Xia S, Zhu Y, Liu M, et al. Cell Mol Immunol. 2020;17:765–767.
    1. Structure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptor. Lan J, Ge J, Yu J, et al. Nature. 2020;581:215–220.
    1. Amyloidogenesis of SARS-CoV-2 spike protein. Nyström S, Hammarström P. J Am Chem Soc. 2022;144:8945–8950.
    1. Neutrophil elastase and proteinase 3 cleavage sites are adjacent to the polybasic sequence within the proteolytic sensitive activation loop of the SARS-CoV-2 spike protein. Mustafa Z, Zhanapiya A, Kalbacher H, Burster T. ACS Omega. 2021;6:7181–7185.
    1. Amyloid fibrils trigger the release of neutrophil extracellular traps (NETs), causing fibril fragmentation by NET-associated elastase. Azevedo EP, Guimarães-Costa AB, Torezani GS, et al. J Biol Chem. 2012;287:37206–37218.
    1. Dementia with Lewy bodies: an update and outlook. Outeiro TF, Koss DJ, Erskine D, et al. Mol Neurodegener. 2019;14:5.
    1. SARS-CoV-2 proteins interact with alpha synuclein and induce Lewy body-like pathology in vitro. Wu Z, Zhang X, Huang Z, Ma K. Int J Mol Sci. 2022;23
    1. COVID-19 infection enhances susceptibility to oxidative stress-induced parkinsonism. Smeyne RJ, Eells JB, Chatterjee D, et al. Mov Disord. 2022;37:1394–1404.
    1. Initiation and propagation of α-synuclein aggregation in the nervous system. Hijaz BA, Volpicelli-Daley LA. Mol Neurodegener. 2020;15:19.
    1. Creutzfeldt-Jakob disease in a man with COVID-19: SARS-CoV-2-accelerated neurodegeneration? Young MJ, O'Hare M, Matiello M, Schmahmann JD. Brain Behav Immun. 2020;89:601–603.
    1. SARS-CoV-2, long COVID, prion disease and neurodegeneration. Zhao Y, Jaber VR, Lukiw WJ. Front Neurosci. 2022;16:1002770.
    1. Towards the emergence of a new form of the neurodegenerative Creutzfeldt-Jakob disease: twenty-six cases of CJD declared a few days after a COVID-19 “vaccine” Jab. Perez JC, Moret-Chalmin C, RIP Montagnier L. Zenodo. 2022;14
    1. In silico characterization of human prion-like proteins: beyond neurological diseases. Iglesias V, Paladin L, Juan-Blanco T, Pallarès I, Aloy P, Tosatto SC, Ventura S. Front Physiol. 2019;10:314.
    1. Mitogen activated protein kinase (MAPK) activation, p53, and autophagy inhibition characterize the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein induced neurotoxicity. Kyriakopoulos AM, Nigh G, McCullough PA, Seneff S. Cureus. 2022;14:0.
    1. Pathogenesis of mouse scrapie: dynamics of agent replication in spleen, spinal cord and brain after infection by different routes. Kimberlin RH, Walker CA. J Comp Pathol. 1979;89:551–562.
    1. The amyloid precursor protein: beyond amyloid. Zheng H, Koo EH. Mol Neurodegener. 2006;1:5.
    1. Expression and function of APP and its metabolites outside the central nervous system. Puig KL, Combs CK. Exp Gerontol. 2013;48:608–611.
    1. Preclinical and clinical demonstration of immunogenicity by mRNA vaccines against H10N8 and H7N9 influenza viruses. Bahl K, Senn JJ, Yuzhakov O, et al. Mol Ther. 2017;25:1316–1327.
    1. Nonclinical safety assessment of repeated administration and biodistribution of a novel rabies self-amplifying mRNA vaccine in rats. Stokes A, Pion J, Binazon O, et al. Regul Toxicol Pharmacol. 2020;113:104648.
    1. The germinal centre B cell response to SARS-CoV-2. Laidlaw BJ, Ellebedy AH. Nat Rev Immunol. 2022;22:7–18.
    1. Regulation of exosome production and cargo sorting. Wei H, Chen Q, Lin L, et al. Int J Biol Sci. 2021;17:163–177.
    1. Linkage between endosomal escape of LNP-mRNA and loading into EVs for transport to other cells. Maugeri M, Nawaz M, Papadimitriou A, et al. Nat Commun. 2019;10:4333.
    1. Transneuronal propagation of pathologic α-synuclein from the gut to the brain models Parkinson's disease. Kim S, Kwon SH, Kam TI, et al. Neuron. 2019;103:627–641.
    1. Centers for Disease Control and Prevention. About the Vaccine Adverse Event Reporting System (VAERS)
    1. Prions and exosomes: from PrPc trafficking to PrPsc propagation. Porto-Carreiro I, Février B, Paquet S, Vilette D, Raposo G. Blood Cells Mol Dis. 2005;35:143–148.
    1. Exosomes taken up by neurons hijack the endosomal pathway to spread to interconnected neurons. Polanco JC, Li C, Durisic N, Sullivan R, Götz J. Acta Neuropathol Commun. 2018;6:10.
    1. Depletion of microglia and inhibition of exosome synthesis halt tau propagation. Asai H, Ikezu S, Tsunoda S, et al. Nat Neurosci. 2015;18:1584–1593.
    1. Alzheimer's disease: Tau pathology and dysfunction of endocytosis. Ando K, Houben S, Homa M, et al. Front Mol Neurosci. 2020;13:583755.
    1. Extracellular vesicles isolated from the brains of rTg4510 mice seed Tau protein aggregation in a threshold-dependent manner. Polanco JC, Scicluna BJ, Hill AF, Götz J. J Biol Chem. 2016;291:12445–12466.
    1. The role of RNA structure at 5' untranslated region in microRNA-mediated gene regulation. Gu W, Xu Y, Xie X, Wang T, Ko JH, Zhou T. RNA. 2014;20:1369–1375.
    1. MicroRNAs: small RNAs with a big role in gene regulation. He L, Hannon GJ. Nat Rev Genet. 2004;5:522–531.
    1. Exosome-delivered microRNAs modulate the inflammatory response to endotoxin. Alexander M, Hu R, Runtsch MC, et al. Nat Commun. 2015;6:7321.
    1. Role of miRNAs as biomarkers of COVID-19: a scoping review of the status and future directions for research in this field. Visacri MB, Nicoletti AS, Pincinato EC, et al. Biomark Med. 2021;15:1785–1795.
    1. Transfer of extracellular vesicle-microRNA controls germinal center reaction and antibody production. Fernández-Messina L, Rodríguez-Galán A, de Yébenes VG, et al. EMBO Rep. 2020;21:0.
    1. MicroRNA-146a-5p, neurotropic viral infection and prion disease (PrD) Pogue AI, Lukiw WJ. Int J Mol Sci. 2021;22
    1. Upregulation of micro RNA-146a (miRNA-146a), a marker for inflammatory neurodegeneration, in sporadic Creutzfeldt-Jakob disease (sCJD) and Gerstmann-Straussler-Scheinker (GSS) syndrome. Lukiw WJ, Dua P, Pogue AI, Eicken C, Hill JM. J Toxicol Environ Health A. 2011;74:1460–1468.
    1. MicroRNA-146a suppresses ROCK1 allowing hyperphosphorylation of tau in Alzheimer's disease. Wang G, Huang Y, Wang LL, et al. Sci Rep. 2016;6:26697.
    1. ROCK1 functions as a suppressor of inflammatory cell migration by regulating PTEN phosphorylation and stability. Vemula S, Shi J, Hanneman P, Wei L, Kapur R. Blood. 2010;115:1785–1796.
    1. A monoclonal antibody against staphylococcal enterotoxin B superantigen inhibits SARS-CoV-2 entry in vitro. Cheng MH, Porritt RA, Rivas MN, et al. Structure. 2021;29:951–962.
    1. SARS-CoV-2 spike antagonizes innate antiviral immunity by targeting interferon regulatory factor 3. Freitas RS, Crum TF, Parvatiyar K. Front Cell Infect Microbiol. 2021;11:789462.
    1. SARS-CoV-2 spike protein suppresses ACE2 and type I interferon expression in primary cells from macaque lung bronchoalveolar lavage. Sui Y, Li J, Venzon DJ, Berzofsky JA. Front Immunol. 2021;12:658428.
    1. MicroRNA-146A contributes to abnormal activation of the type I interferon pathway in human lupus by targeting the key signaling proteins. Tang Y, Luo X, Cui H, et al. Arthritis Rheum. 2009;60:1065–1075.
    1. SARS-CoV-2 S1 protein persistence in SARS-CoV-2 negative post-vaccination individuals with long COVID/ PASC-like symptoms. Patterson BK, Francisco EB, Yogendra R, et al. Research Square. 2022;12:1–14.
    1. Potential mechanisms for human genome integration of genetic code from SARS-CoV-2 mRNA vaccination. Kyriakopoulos AM, McCullough PA, Nigh G, et al. J Neurol Disord. 2022;10:519.
    1. Intracellular reverse transcription of Pfizer BioNTech COVID-19 mRNA vaccine BNT162b2 in vitro in human liver cell line. Aldén M, Olofsson Falla F, Yang D, Barghouth M, Luan C, Rasmussen M, De Marinis Y. Curr Issues Mol Biol. 2022;44:1115–1126.
    1. Retroviral Gag protein-RNA interactions: implications for specific genomic RNA packaging and virion assembly. Olson ED, Musier-Forsyth K. Semin Cell Dev Biol. 2019;86:129–139.
    1. Prion protein PrP nucleic acid binding and mobilization implicates retroelements as the replicative component of transmissible spongiform encephalopathy. Lathe R, Darlix JL. Arch Virol. 2020;165:535–556.
    1. LINE-1 retrotransposition in the nervous system. Thomas CA, Paquola AC, Muotri AR. Annu Rev Cell Dev Biol. 2012;28:555–573.
    1. Aberrantly high levels of somatic LINE-1 expression and retrotransposition in human neurological disorders. Terry DM, Devine SE. Front Genet. 2019;10:1244.
    1. SARS-CoV-2 spike protein S1 induces fibrin(ogen) resistant to fibrinolysis: implications for microclot formation in COVID-19. Grobbelaar LM, Venter C, Vlok M, et al. Biosci Rep. 2021;41
    1. A central role for amyloid fibrin microclots in long COVID/PASC: origins and therapeutic implications. Kell DB, Laubscher GJ, Pretorius E. Biochem J. 2022;479:537–559.
    1. Cellular prion protein is released on exosomes from activated platelets. Robertson C, Booth SA, Beniac DR, Coulthart MB, Booth TF, McNicol A. Blood. 2006;107:3907–3911.
    1. Alzheimer's disease peptide beta-amyloid interacts with fibrinogen and induces its oligomerization. Ahn HJ, Zamolodchikov D, Cortes-Canteli M, Norris EH, Glickman JF, Strickland S. Proc Natl Acad Sci USA. 2010;107:21812–21817.
    1. CD16 regulates TRIF-dependent TLR4 response in human monocytes and their subsets. Shalova IN, Kajiji T, Lim JY, et al. J Immunol. 2012;188:3584–3593.
    1. Effect of TLR4/MyD88 signaling pathway on expression of IL-1β and TNF-α in synovial fibroblasts from temporomandibular joint exposed to lipopolysaccharide. Lin X, Kong J, Wu Q, Yang Y, Ji P. Mediators Inflamm. 2015;2015:329405.
    1. Regulation of toll-like receptor (TLR)2 and TLR4 on CD14dimCD16+ monocytes in response to sepsis-related antigens. Skinner NA, MacIsaac CM, Hamilton JA, Visvanathan K. Clin Exp Immunol. 2005;141:270–278.
    1. SARS-CoV-2 spike protein interacts with and activates TLR41. Zhao Y, Kuang M, Li J, et al. Cell Res. 2021;31:818–820.
    1. SARS-CoV-2 spike S1 subunit induces neuroinflammatory, microglial and behavioral sickness responses: evidence of PAMP-like properties. Frank MG, Nguyen KH, Ball JB, et al. Brain Behav Immun. 2022;100:267–277.
    1. The role of Toll-like receptors and neuroinflammation in Parkinson's disease. Heidari A, Yazdanpanah N, Rezaei N. J Neuroinflammation. 2022;19:135.
    1. Neuroinflammation and protein pathology in Parkinson's disease dementia. Kouli A, Camacho M, Allinson K, Williams-Gray CH. Acta Neuropathol Commun. 2020;8:211.
    1. Toll-like receptor expression in the blood and brain of patients and a mouse model of Parkinson's disease. Drouin-Ouellet J, St-Amour I, Saint-Pierre M, Lamontagne-Proulx J, Kriz J, Barker RA, Cicchetti F. Int J Neuropsychopharmacol. 2014;18
    1. Parallels in sepsis and COVID-19 conditions: implications for managing severe COVID-19. Olwal CO, Nganyewo NN, Tapela K, Djomkam Zune AL, Owoicho O, Bediako Y, Duodu S. Front Immunol. 2021;12:602848.
    1. The novel subset of CD14+/CD16+ blood monocytes is expanded in sepsis patients. Fingerle G, Pforte A, Passlick B, et al. Blood. 1993;82:3170–3176.
    1. The CD14+ CD16+ blood monocytes: their role in infection and inflammation. Ziegler-Heitbrock L. J Leukoc Biol. 2007;81:584–592.
    1. Unique monocyte subset in patients with AIDS dementia. Pulliam L, Gascon R, Stubblebine M, McGuire D, McGrath MS. Lancet. 1997;349:692–695.
    1. Monocyte based correlates of immune activation and viremia in HIV-infected long-term non-progressors. Prabhu VM, Singh AK, Padwal V, Nagar V, Patil P, Patel V. Front Immunol. 2019;10:2849.
    1. Circulating CD14+ CD16+ monocytes are expanded in sarcoidosis patients. Okamoto H, Mizuno K, Horio T. J Dermatol. 2003;30:503–509.
    1. Elevated blood levels of inflammatory monocytes (CD14+ CD16+ ) in patients with complex regional pain syndrome. Ritz BW, Alexander GM, Nogusa S, Perreault MJ, Peterlin BL, Grothusen JR, Schwartzman RJ. Clin Exp Immunol. 2011;164:108–117.
    1. Monocytes in sarcoidosis are potent tumour necrosis factor producers and predict disease outcome. Lepzien R, Liu S, Czarnewski P, et al. Eur Respir J. 2021;58
    1. Distinct immune regulatory receptor profiles linked to altered monocyte subsets in sarcoidosis. Fraser SD, Crooks MG, Kaye PM, Hart SP. ERJ Open Res. 2021;7
    1. The role of shed PrP(c) in the neuropathogenesis of HIV infection. Megra BW, Eugenin EA, Berman JW. J Immunol. 2017;199:224–232.
    1. Structural similarity between HIV-1 gp41 and SARS-CoV S2 proteins suggests an analogous membrane fusion mechanism. Wu Zhang X, Leng Yap Y. Theochem. 2004;677:73–76.
    1. Prions and protein-folding diseases. Norrby E. J Intern Med. 2011;270:1–14.
    1. Deadly conformations--protein misfolding in prion disease. Horwich AL, Weissman JS. Cell. 1997;89:499–510.
    1. Role of cellular prion protein in splenic CD4(+) T cell differentiation in cerebral ischaemic/reperfusion. Zhang B, Yin X, Lang Y, Han X, Shao J, Bai R, Cui L. Ann Clin Transl Neurol. 2021;8:2040–2051.
    1. Role of prion protein in premature senescence of human fibroblasts. Boilan E, Winant V, Dumortier E, et al. Mech Ageing Dev. 2018;170:106–113.
    1. SARS-CoV-2 causes senescence in human cells and exacerbates the senescence-associated secretory phenotype through TLR-3. Tripathi U, Nchioua R, Prata LG, et al. Aging (Albany NY) 2021;13:21838–21854.
    1. Do messenger RNA vaccines induce pathological syncytia? Sfera A, Thomas KG, Sfera DO, et al. Int J Pathol Clin Res. 2022;8:137.
    1. Flagging fusion: phosphatidylserine signaling in cell-cell fusion. Whitlock JM, Chernomordik LV. J Biol Chem. 2021;296:100411.
    1. An aged immune system drives senescence and ageing of solid organs. Yousefzadeh MJ, Flores RR, Zhu Y, et al. Nature. 2021;594:100–105.
    1. Macromolecular crowding converts the human recombinant PrPC to the soluble neurotoxic beta-oligomers. Huang L, Jin R, Li J, et al. FASEB J. 2010;24:3536–3543.
    1. Baseline characteristics and outcomes of 1591 patients infected with SARS-CoV-2 admitted to ICUs of the Lombardy Region, Italy. Grasselli G, Zangrillo A, Zanella A, et al. JAMA. 2020;323:1574–1581.
    1. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Zhou F, Yu T, Du R, et al. Lancet. 2020;395:1054–1062.
    1. Coronavirus infection (SARS-CoV-2) in obesity and diabetes comorbidities: is heat shock response determinant for the disease complications? Krause M, Gerchman F, Friedman R. Diabetol Metab Syndr. 2020;12:63.
    1. Chaperoning to the metabolic party: the emerging therapeutic role of heat-shock proteins in obesity and type 2 diabetes. Henstridge DC, Whitham M, Febbraio MA. Mol Metab. 2014;3:781–793.
    1. Heat shock proteins and diabetes. Zilaee M, Shirali S. Can J Diabetes. 2016;40:594–602.
    1. Suppressed anti-inflammatory heat shock response in high-risk COVID-19 patients: lessons from basic research (inclusive bats), light on conceivable therapies. Heck TG, Ludwig MS, Frizzo MN, Rasia-Filho AA, Homem de Bittencourt PI. Clin Sci (Lond) 2020;134:1991–2017.
    1. Heat shock factor 1 represses transcription of the IL-1beta gene through physical interaction with the nuclear factor of interleukin 6. Xie Y, Chen C, Stevenson MA, Auron PE, Calderwood SK. J Biol Chem. 2002;277:11802–11810.
    1. Potential pathophysiological mechanisms underlying multiple organ dysfunction in cytokine release syndrome. Chen P, Tang Y, He W, Yang R, Lan Z, Chen R, Zhang P. Mediators Inflamm. 2022;2022:7137900.
    1. Heat shock proteins and exercise: a primer. Noble EG, Milne KJ, Melling CW. Appl Physiol Nutr Metab. 2008;33:1050–1065.
    1. Inducible heat shock protein 70 and its role in preconditioning and exercise. Madden LA, Sandström ME, Lovell RJ, McNaughton L. Amino Acids. 2008;34:511–516.
    1. Initiation of the immune response by extracellular Hsp72: chaperokine activity of Hsp72. Asea A. Curr Immunol Rev. 2006;2:209–215.
    1. Extracellular HSP70 binding to surface receptors present on antigen presenting cells and endothelial/epithelial cells. Thériault JR, Mambula SS, Sawamura T, Stevenson MA, Calderwood SK. FEBS Lett. 2005;579:1951–1960.
    1. The role of heat shock proteins in antigen cross presentation. Murshid A, Gong J, Calderwood SK. Front Immunol. 2012;3:63.
    1. Role of glycogen synthase kinase-3 in insulin resistance and type 2 diabetes. Henriksen EJ, Dokken BB. Curr Drug Targets. 2006;7:1435–1441.
    1. Glycogen synthase kinase 3beta negatively regulates both DNA-binding and transcriptional activities of heat shock factor 1. Xavier IJ, Mercier PA, McLoughlin CM, Ali A, Woodgett JR, Ovsenek N. J Biol Chem. 2000;275:29147–29152.
    1. HSF1 is required for extra-embryonic development, postnatal growth and protection during inflammatory responses in mice. Xiao X, Zuo X, Davis AA, McMillan DR, Curry BB, Richardson JA, Benjamin IJ. EMBO J. 1999;18:5943–5952.
    1. Human Hsp70 disaggregase reverses Parkinson's-linked α-synuclein amyloid fibrils. Gao X, Carroni M, Nussbaum-Krammer C, et al. Mol Cell. 2015;59:781–793.
    1. Molecular dissection of amyloid disaggregation by human HSP70. Wentink AS, Nillegoda NB, Feufel J, et al. Nature. 2020;587:483–488.
    1. Prion disease is accelerated in mice lacking stress-induced heat shock protein 70 (HSP70) Mays CE, Armijo E, Morales R, et al. J Biol Chem. 2019;294:13619–13628.
    1. Cell death and pathological findings of the spleen in COVID-19 patients. Ping H, Zhang K, Wang Y, et al. Pathol Res Pract. 2021;227:153610.
    1. Apoptotic cell-derived exosomes: messages from dying cells. Kakarla R, Hur J, Kim YJ, Kim J, Chwae YJ. Exp Mol Med. 2020;52:1–6.
    1. Stimulating the release of exosomes increases the intercellular transfer of prions. Guo BB, Bellingham SA, Hill AF. J Biol Chem. 2016;291:5128–5137.
    1. RNA G-quadruplexes and their potential regulatory roles in translation. Song J, Perreault JP, Topisirovic I, Richard S. Translation (Austin) 2016;4:0.
    1. G-quadruplexes within prion mRNA: the missing link in prion disease? Olsthoorn RC. Nucleic Acids Res. 2014;42:9327–9333.
    1. Differences in vaccine and SARS-CoV-2 replication derived mRNA: implications for cell biology and future disease. McKernan K, Kyriakopoulos AM, McCullough PA. Preprint. 2021
    1. Risk of infection, hospitalisation, and death up to 9 months after a second dose of COVID-19 vaccine: a retrospective, total population cohort study in Sweden. Nordström P, Ballin M, Nordström A. Lancet. 2022;399:814–823.
    1. Adverse effects of COVID-19 vaccines and measures to prevent them. Yamamoto K. Virol J. 2022;19:100.

Source: PubMed

3
Subskrybuj