Chronological brain lesions after SARS-CoV-2 infection in hACE2-transgenic mice

Enric Vidal, Carlos López-Figueroa, Jordi Rodon, Mónica Pérez, Marco Brustolin, Guillermo Cantero, Víctor Guallar, Nuria Izquierdo-Useros, Jorge Carrillo, Julià Blanco, Bonaventura Clotet, Júlia Vergara-Alert, Joaquim Segalés, Enric Vidal, Carlos López-Figueroa, Jordi Rodon, Mónica Pérez, Marco Brustolin, Guillermo Cantero, Víctor Guallar, Nuria Izquierdo-Useros, Jorge Carrillo, Julià Blanco, Bonaventura Clotet, Júlia Vergara-Alert, Joaquim Segalés

Abstract

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes respiratory disease, but it can also affect other organs including the central nervous system. Several animal models have been developed to address different key questions related to Coronavirus Disease 2019 (COVID-19). Wild-type mice are minimally susceptible to certain SARS-CoV-2 lineages (beta and gamma variants), whereas hACE2-transgenic mice succumb to SARS-CoV-2 and develop a fatal neurological disease. In this article, we aimed to chronologically characterize SARS-CoV-2 neuroinvasion and neuropathology. Necropsies were performed at different time points, and the brain and olfactory mucosa were processed for histopathological analysis. SARS-CoV-2 virological assays including immunohistochemistry were performed along with a panel of antibodies to assess neuroinflammation. At 6 to 7 days post inoculation (dpi), brain lesions were characterized by nonsuppurative meningoencephalitis and diffuse astrogliosis and microgliosis. Vasculitis and thrombosis were also present and associated with occasional microhemorrhages and spongiosis. Moreover, there was vacuolar degeneration of virus-infected neurons. At 2 dpi, SARS-CoV-2 immunolabeling was only found in the olfactory mucosa, but at 4 dpi intraneuronal virus immunolabeling had already reached most of the brain areas. Maximal distribution of the virus was observed throughout the brain at 6 to 7 dpi except for the cerebellum, which was mostly spared. Our results suggest an early entry of the virus through the olfactory mucosa and a rapid interneuronal spread of the virus leading to acute encephalitis and neuronal damage in this mouse model.

Keywords: Coronavirus Disease 2019; SARS-CoV-2; animal model; brain; hACE2-transgenic mice; meningoencephalitis; neuropathology.

Conflict of interest statement

Declaration of Conflicting Interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Figures

Figure 1.
Figure 1.
Progression of weight loss after inoculation of hACE2-transgenic mice with either 103 or 104 SARS-CoV-2 TCID50 per animal. Significant differences between groups are shown as asterisks above each time point. Red asterisks show differences between the 104 TCID50-inoculated group and noninfected animals. Blue asterisks show differences between 103 TCID50-inoculated group and noninfected animals. Green asterisks show differences between 104 and 103 TCID50-inoculated groups. ns, P > .05; *, P ≤ .05; **, P ≤ .01; ***, P ≤ .001; ****, P ≤ .0001. The bars show the mean ± standard deviation. Figure 2. Survival curves of hACE2-transgenic mice inoculated with either 103 or 104 TCID50 per animal of SARS-CoV-2 and the negative controls.
Figures 3–5.
Figures 3–5.
Virological studies. No significant differences in viral loads in brain were observed between the 2 dose groups at 4 and 7 days postinoculation (dpi). In the high-dose group, viral genomic DNA was found in the brain as early as 2 dpi. Figure 3. Viral load in the brain of SARS-CoV-2-inoculated hACE2-transgenic mice measured by means of viral genomic RNA. Figure 4. Viral load in the brain of SARS-CoV-2-inoculated hACE2-transgenic mice measured by means of viral subgenomic RNA. Figure 5. Viral load in the brain of SARS-CoV-2-inoculated hACE2-transgenic mice measured by means of infectious virus (in cell culture). Each dot represents a single animal. ns, nonsignificant differences. The bars show the mean ± standard deviation.
Figures 6–11.
Figures 6–11.
SARS-CoV-2 infection, brain, hACE2-transgenic mice, at 7 days postinoculation, 104 TCID50 dose, hematoxylin and eosin. Figure 6. Occipital cortex. Mononuclear cell infiltrate in the leptomeninges with a thrombus in a blood vessel (arrow). Figure 7. Mesencephalon. A perivascular cuff of mononuclear cells, and a thrombus within a blood vessel. Figure 8. Occipital cortex, gray to white matter transition. There is vasculitis (arrow) and small hemorrhages (bottom left). Figure 9. Thalamus. Intravascular thrombus (arrow) in a small blood capillary. Figure 10. Hippocampal formation. Vacuolar degeneration of neurons in the dorsal end of the subiculum. Figure 11. Medulla oblongata. Myelin sheath vacuolation, limited to the root of the facial nerve.
Figure 12.
Figure 12.
SARS-CoV2 immunolabeling in the nervous system of hACE2-transgenic mice. Mice were inoculated with 103 (blue; euthanized at 4 and 7 dpi) or 104 (pink; euthanized at 2, 4, 6, and 7 dpi) TCID50 of SARS-CoV-2. Different areas of the brain and nasal cavity were semiquantitatively scored from 0 (no immunolabeling) to 3 (maximum immunolabeling) and the mean scores in each group are shown. Levels from 0 to 3 are shown by concentric circles, with the outside circle being 3. At 2 dpi, virus was only found in the olfactory mucosa, but at 4 dpi the virus had reached most brain areas (for both doses of virus). Virus throughout the brain was maximal at 6 and 7 dpi, with the exception of the cerebellum which is almost spared. At 7 dpi, the virus is no longer observed in the olfactory mucosa. See individual scores in Supplemental File S2. Each axis of the star-graph represents a studied region (Om, olfactory mucosa; Cc, cerebellar cortex; Mob, medulla oblongata; P, pons; M, mesencephalon; HT, hypothalamus; T, thalamus; S, striatum; Oc, occipital cortex; Tc, temporal cortex; Pc, parietal cortex; Fc, frontal cortex; H, hippocampus; Pfc, piriform cortex; Ob, olfactory bulb).
Figures 13–18.
Figures 13–18.
SARS-CoV-2 infection, olfactory mucosa (Fig. 13), and brain (Figs. 14–18), hACE2-transgenic mice, immunohistochemistry for SARS-CoV-2. Figure 13. Small clusters of immunolabeled cells in the olfactory epithelium (2 dpi, 104 TCID50 dose). Figure 14. Widespread intracytoplasmic neuronal immunolabeling in all layers of the occipital cortex (7 dpi, 104 TCID50 dose). Inset: immunolabeled cytoplasmic inclusion bodies in a neuron (arrowheads). Figure 15. The early distribution of SARS-CoV-2 antigen immunolabeling with patchy involvement of thalamus, CA1, CA2, CA3, and subiculum of the hippocampus (the lateral portion of CA1 and the dentate gyrus are mostly spared) and the parietal cortex (4 dpi, 104 TCID50 dose). Figure 16. The cerebellar cortex is mostly spared, and only the perikaryon and dendrites of a few piriform (Purkinje) cells are immunolabeled (7 dpi, 104 TCID50 dose). Figure 17. Parietal cortex. All degenerating neurons with cytoplasmic vacuoles are immunolabeled (7 dpi, 104 TCID50 dose). Figure 18. Mesencephalon. There is immunolabeling of neurons but not of the endothelium in a vessel that has a cuff of mononuclear cells (the same blood vessel depicted in Fig. 7; 7 dpi, 104 TCID50 dose).
Figures 19–20.
Figures 19–20.
Quantification of the neuroinflammatory response in brains of hACE2-transgenic SARS-CoV-2-infected mice (103 TCID50 dose) at 4 dpi and 7 dpi, compared to mock-inoculated negative controls. Different areas of the brain were semiquantitatively scored from 0 (no immunolabeling) to 4 (maximum immunolabeling) and the mean scores in each group are shown. Levels from 1 to 4 are shown by concentric circles, with the outside circle being 4. Each axis of the star-graph represents a brain region; the value given is the average score for all the animals in that group in that particular area (Pfc, piriform cortex; Cc, cerebellar cortex; Mob, medulla oblongata; P, pons; M, mesencephalon; HT, hypothalamus; T, thalamus; S, striatum; Oc, occipital cortex; Tc, temporal cortex; Fc, frontal cortex; Pc, parietal cortex; H, hippocampus). Figure 19. Scoring of GFAP immunolabeling. Astrogliosis (hypertrophy and hyperplasia of astrocytes) is most evident in the brainstem (thalamus, hypothalamus, mesencephalon, pons, and medulla oblongata), piriform cortex, and occipital cortex, and is evident at 4 dpi (green squares) but more intense at 7 dpi (yellow triangles). The hippocampus, parietal cortex, striatum, and cerebellar cortex show small or no differences compared to uninoculated negative controls (gray circles). Figure 20. Scoring of the IBA1 immunolabeling. Widespread microgliosis is most severe at 7 dpi (yellow triangles), but was similar at 4 dpi (green squares) as in the negative controls (gray circles). Microglial spares only the cerebellar cortex. See individual scores in Supplemental Table S2.
Figures 21–23.
Figures 21–23.
SARS-CoV-2 infection (103 TCID50 dose), brain (mesencephalon), hACE2-transgenic mice. (a) Hematoxylin and eosin (HE), (b) immunohistochemistry (IHC) for GFAP, and (c) IHC for IBA1. Figure 21. Negative control mouse (mock-inoculated). Figure 22. SARS-CoV-2-infected at 4 dpi. (a) The increase in cellularity is not striking in HE-stained sections. (b) Moderate increase in number and hypertrophy of astroglial cells. (c) Moderate increase in number and activation of microglial cells. Figure 23. SARS-CoV-2-infected at 7 dpi. (a) Markedly increased cellularity. (b) Marked increase in number and size of astrocytes near vessels and in neuropil. (c) Marked increase in number and size of microglial cells.
Figure 24.
Figure 24.
SARS-CoV-2 infection (104 TCID50 dose, 7 dpi), brain (mesencephalon), hACE2-transgenic mice. Immunohistochemistry. (a) Several CD3-positive cells (T cells) are in most perivascular cuffs (black arrows) and occasional T cells infiltrate the neuropil (white arrows). (b) Comparatively, there are low numbers of CD20-positive cells near vessels and in neuropil (B cells, arrows). (c) Numerous IBA1-positive cells (macrophages) are observed in perivascular cuffs (arrows). IBA1-positive activated microglial cells are present in the neuropil.

References

    1. Bao L, Deng W, Huang B, et al. The pathogenicity of SARS-CoV-2 in hACE2 transgenic mice. Nature. 2020;583(7818):830–833.
    1. Bilinska K, von Bartheld CS, Butowt R. Expression of the ACE2 virus entry protein in the nervus terminalis reveals the potential for an alternative route to brain infection in COVID-19. Front Cell Neurosci. 2021;15:674123.
    1. Bodro M, Compta Y, Sánchez-Valle R. Presentations and mechanisms of CNS disorders related to COVID-19. Neurol Neuroimmunol Neuroinflam. 2021; 8(1):e923.
    1. Brustolin M, Rodon J, Rodríguez de la Concepción ML, et al. Protection against reinfection with D614- or G614-SARS-CoV-2 isolates in golden Syrian hamster. Emerg Microbes Infect. 2021;10(1):797–809.
    1. Bulfamante G, Bocci T, Falleni M, et al. Brainstem neuropathology in two cases of COVID-19: SARS-CoV-2 trafficking between brain and lung. J Neurol. 2021;68(12):4486–4491.
    1. Butowt R, von Bartheld CS. Connecting the dots: trafficking of neurotrophins, lectins and diverse pathogens by binding to the neurotrophin receptor p75NTR. Eur J Neurosci. 2003;17(4):673–680.
    1. Cantuti-Castelvetri L, Ojha R, Pedro LD, et al. Neuropilin-1 facilitates SARS-CoV-2 cell entry and infectivity. Science. 2020;370(6518):856–860.
    1. Carossino M, Montanaro P, O’Connell A, et al. Fatal neuroinvasion of SARS-CoV-2 in K18-hACE2 mice is partially dependent on hACE2 expression. bioRxiv. Preprint. doi:10.1101/2021.01.13.425144
    1. Chakravarty D, Das Sarma J. Murine-β-coronavirus-induced neuropathogenesis sheds light on CNS pathobiology of SARS-CoV2. J NeuroVirol. 2021;27(2):197–216.
    1. Chow YH, O’Brodovich H, Plumb J, et al. Development of an epithelium-specific expression cassette with human DNA regulatory elements for transgene expression in lung airways. Proc Natl Acad Sci U S A. 1997;94(26):14695–14700.
    1. Corman VM, Landt O, Kaiser M, et al. Detection of 2019 novel coronavirus (2019-nCoV) by real-time RT-PCR. Euro Surveill. 2020;25(3):2000045.
    1. DosSantos MF, Devalle S, Aran V, et al. Neuromechanisms of SARS-CoV-2: a review. Front Neuroanat. 2020;14:37.
    1. Dubé M, Le Coupanec A, Wong AHM, et al. Axonal transport enables neuron-to-neuron propagation of human coronavirus OC43. J Virol. 2018;92(17):e00404–e00418.
    1. Fodoulian L, Tuberosa J, Rossier D, et al. SARS-CoV-2 receptors and entry genes are expressed in the human olfactory neuroepithelium and brain. iScience. 2020;23(12):101839.
    1. Golden JW, Cline CR, Zeng X, et al. Human angiotensin-converting enzyme 2 transgenic mice infected with SARS-CoV-2 develop severe and fatal respiratory disease. JCI Insight. 2020;5(19):e142032.
    1. Hamming I, Timens W, Bulthuis MLC, et al. Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. J Pathol. 2004;203(2):631–637.
    1. Johnson KD, Harris C, Cain JK, et al. Pulmonary and extra-pulmonary clinical manifestations of COVID-19. Front Med (Lausanne). 2020;7:526.
    1. Karuppan MKM, Devadoss D, Nair M, et al. SARS-CoV-2 infection in the central and peripheral nervous system-associated morbidities and their potential mechanism. Mol Neurobiol. 2021;58(6):2465–2480.
    1. Korber B, Fischer WM, Gnanakaran S, et al. Tracking changes in SARS-CoV-2 spike: evidence that D614G increases infectivity of the COVID-19 virus. Cell. 2020;182(4):812–827.e19.
    1. Kumari P, Rothan HA, Natekar JP, et al. Neuroinvasion and encephalitis following intranasal inoculation of SARS-CoV-2 in K18-hACE2 mice. Viruses. 2021;13(1):132.
    1. Le Coupanec A, Desforges M, Kaufer B, et al. Potential differences in cleavage of the S protein and type-1 interferon together control human coronavirus infection, propagation, and neuropathology within the central nervous system. J Virol. 2021;95(10):e00140-21.
    1. Li YC, Bai WZ, 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(6):552–555.
    1. Li Z, Liu T, Yang N, et al. Neurological manifestations of patients with COVID-19: potential routes of SARS-CoV-2 neuroinvasion from the periphery to the brain. Front Med. 2020;14(5):533–541.
    1. Lu R, Zhao X, Li J, et al. Genomic characterisation and epidemiology of 2019 novel coronavirus: implications for virus origins and receptor binding. Lancet. 2020;395(10224):565–574.
    1. Mao L, Jin H, Wang M, et al. Neurologic manifestations of hospitalized patients with coronavirus disease 2019 in Wuhan, China. JAMA Neurol. 2020;77(6):683–690.
    1. Matschke J, Lütgehetmann M, Hagel C, et al. Neuropathology of patients with COVID-19 in Germany: a post-mortem case series. Lancet Neurol. 2020;19(11):919–929.
    1. McCray PB, Jr, Pewe L, Wohlford-Lenane C, et al. Lethal infection of K18-hACE2 mice infected with severe acute respiratory syndrome coronavirus. J Virol. 2007;81(2):813–821.
    1. Meinhardt J, Radke J, Dittmayer C, et al. Olfactory transmucosal SARS-CoV-2 invasion as a port of central nervous system entry in individuals with COVID-19. Nat Neurosci. 2021;24(2):168–175.
    1. Messlinger K, Neuhuber W, May A. Activation of the trigeminal system as a likely target of SARS-CoV-2 may contribute to anosmia in COVID-19. Cephalalgia. Published online August 18, 2021. doi:10.1177/03331024211036665
    1. Muñoz-Fontela C, Dowling WE, Funnell SGP, et al. Animal models for COVID-19. Nature. 2020;586(7830):509–515.
    1. Netland J, Meyerholz DK, Moore S, et al. Severe acute respiratory syndrome coronavirus infection causes neuronal death in the absence of encephalitis in mice transgenic for human ACE2. J Virol. 2008;82(15):7264–7275.
    1. Ng JH, Sun A, Je HS, et al. Unravelling pathophysiology of neurological and psychiatric complications of COVID-19 using brain organoids. Neuroscientist. Published online May 26, 2021. doi:10.1177/10738584211015136
    1. Oladunni FS, Park JG, Pino PA, et al. Lethality of SARS-CoV-2 infection in K18 human angiotensin-converting enzyme 2 transgenic mice. Nat Commun. 2020;11(1):6122.
    1. Otte MS, Bork ML, Zimmermann PH, et al. Patients with COVID-19-associated olfactory impairment also show impaired trigeminal function. Auris Nasus Larynx. Published online July 26, 2021. doi:10.1016/j.anl.2021.07.012
    1. Perez-Zsolt D, Muñoz-Basagoiti J, Rodon J, et al. Siglec-1 on dendritic cells mediates SARS-CoV-2 trans-infection of target cells while on macrophages triggers proinflammatory responses. bioRxiv. Preprint. doi:10.1101/2021.05.11.443572
    1. Plante JA, Liu Y, Liu J, et al. Spike mutation D614G alters SARS-CoV-2 fitness. Nature. 2021;592(7852):116–121.
    1. Ramani A, Müller L, Ostermann PN, et al. SARS-CoV-2 targets neurons of 3D human brain organoids. EMBO J. 2020;39(20):e106230.
    1. Rockx B, Kuiken T, Herfst S, et al. Comparative pathogenesis of COVID-19, MERS, and SARS in a nonhuman primate model. Science. 2020;368(6494):1012–1015.
    1. Seehusen F, Clark JJ, Sharma P, et al. Viral neuroinvasion and neurotropism without neuronal damage in the hACE2 mouse model of COVID-19. bioRxiv. Preprint. doi:10.1101/2021.04.16.440173
    1. Song E, Bartley CM, Chow RD, et al. Divergent and self-reactive immune responses in the CNS of COVID-19 patients with neurological symptoms. Cell Rep Med. 2021;2(5):100288.
    1. Song E, Zhang C, Israelow B, et al. Neuroinvasion of SARS-CoV-2 in human and mouse brain. J Exp Med. 2021;218(3):e20202135.
    1. Sriwastava S, Tandon M, Podury S, et al. COVID-19 and neuroinflammation: a literature review of relevant neuroimaging and CSF markers in central nervous system inflammatory disorders from SARS-COV2. J Neurol. 2021;268(12):4448–4478.
    1. Tang D, Comish P, Kang R. The hallmarks of COVID-19 disease. PLoS Pathogens. 2020;16(5):e1008536.
    1. Vilensky JA. The neglected cranial nerve: nervus terminalis (cranial nerve N). Clin Anat. 2014;27(1):46–53.
    1. Wölfel R, Corman VM, Guggemos W, et al. Virological assessment of hospitalized patients with COVID-2019. Nature. 2020;581(7809):465–469.
    1. Zhu N, Zhang D, Wang W, et al. A novel coronavirus from patients with pneumonia in China, 2019. N Engl J Med. 2020;382(8):727–733.

Source: PubMed

3
Abonnieren