ArticlePDF Available

Mitochondrial Dysfunction: A Prelude to Neuropathogenesis of SARS-CoV-2

Authors:

Abstract

The SARS-CoV-2 virus is notorious for its neuroinvasive capability, causing multiple neurological conditions. The neuropathology of SARS-CoV-2 is increasingly attributed to mitochondrial dysfunction of brain microglia cells. However, the changes in biochemical content of mitochondria that drive the progression of neuro-COVID remain poorly understood. Here we introduce a Raman microspectrometry approach that enables the molecular profiling of single cellular organelles to characterize the mitochondrial molecular makeup in the infected microglia cells. We found that microglia treated with either spike protein or heat-inactivated SARS-CoV-2 trigger a dramatic reduction in mtDNA content and an increase in phospholipid saturation levels. At the same time, no significant changes were detected in Golgi apparatus and in lipid droplets, the organelles that accommodate biogenesis and storage of lipids. We hypothesize that transformations in mitochondria are caused by increased synthesis of reactive oxygen species in these organelles. Our findings call for the development of mitochondria-targeted therapeutic approaches to limit neuropathology associated with SARS-CoV-2.
Mitochondrial Dysfunction: A Prelude to Neuropathogenesis of
SARS-CoV2
Artem Pliss, Andrey N. Kuzmin, Paras N. Prasad,*and Supriya D. Mahajan*
Cite This: ACS Chem. Neurosci. 2022, 13, 308312
Read Online
ACCESS Metrics & More Article Recommendations *
sıSupporting Information
ABSTRACT: The SARS-CoV-2 virus is notorious for its neuro-
invasive capability, causing multiple neurological conditions. The
neuropathology of SARS-CoV-2 is increasingly attributed to
mitochondrial dysfunction of brain microglia cells. However, the
changes in biochemical content of mitochondria that drive the
progression of neuro-COVID remain poorly understood. Here we
introduce a Raman microspectrometry approach that enables the
molecular proling of single cellular organelles to characterize the
mitochondrial molecular makeup in the infected microglia cells. We
found that microglia treated with either spike protein or heat-
inactivated SARS-CoV-2 trigger a dramatic reduction in mtDNA
content and an increase in phospholipid saturation levels. At the
same time, no signicant changes were detected in Golgi apparatus
and in lipid droplets, the organelles that accommodate biogenesis and storage of lipids. We hypothesize that transformations in
mitochondria are caused by increased synthesis of reactive oxygen species in these organelles. Our ndings call for the development
of mitochondria-targeted therapeutic approaches to limit neuropathology associated with SARS-CoV-2.
KEYWORDS: Microglia, mitochondria, ROS, SARS-CoV-2, neuro-COVID, Raman spectrometry
INTRODUCTION
A signicant number of COVID-19 patients develop neuro-
logical symptoms, attributed to viral encephalitis, resulting in
neuroinammation, neuronal damage, and neurocognitive
impairment. The microglia, which are the resident macro-
phages in the central nervous system, are the major players in
the brains immune response to SARS-CoV-2 infection.
Furthermore, it has been shown that functional mitochondria
are integral to initiation and maintenance of immune responses
by microglia, while neurological damage in COVID patients is
attributed to mitochondrial dysfunction. Mitochondria are the
primary site of ATP production and also regulate basic
metabolic functions and participate in homeostasis, cellular
proliferation, and apoptosis as well as in the synthesis of amino
acids, lipids, and nucleotides. In microglia these organelles also
mediate the antiviral immune response by releasing pro-
inammatory cytokines, which limit viral survival and viral
replication and trigger inammation.
14
Strikingly, SARS-CoV-
2 can evade the innate immune response of host cells via the
modulation of mitochondrial functions. The spike protein of
SARS-CoV-2 binds to the angiotensin-converting enzyme-2
(ACE-2) receptor on the human host cell
3
to enter the host,
and the transmembrane serine protease 2 (TMPRESS 2)
facilitates this attachment by priming the spike protein.
5
Notably, the ACE-2 receptor regulates mitochondrial func-
tion.
6
Reduced expression of ACE-2 is correlated with
decreased ATP synthesis and activation of NADPH oxidase
4, which contributes to the production of reactive oxygen
species (ROS).
4
Consistent with that, an invasion of SARS-
CoV-2 via the ACE-2 receptor compromises mitochondrial
regulation. Excessive ROS production exacerbates neuro-
inammation, initiating apoptosis in infected cells, which
results in neurocognitive impairments. It is known that SARS-
CoV-2 infection results in a massive inammatory response in
the brain by triggering the release of cytokines such as
interleukin (IL)-10, TNF-α, and INF-γ, which in turn further
increase mitochondrial ROS production through upregulation
of mitochondrial genes and modulation of the electron
transport chain (ETC).
7
The mitochondrial ROS stimulate
additional proinammatory cytokine production
8
in the face of
viral persistence, leading to a cytokine storm syndrome,
which underlies viral encephalopathy.
7
We recently observed
an increased oxygen consumption rate (OCR) in microglial
cells treated with SARS-CoV-2 spike protein.
9
Our data
suggested that SARS-CoV-2 induced a robust inammatory
Received: October 13, 2021
Accepted: January 18, 2022
Published: January 20, 2022
Letterpubs.acs.org/chemneuro
© 2022 American Chemical Society 308
https://doi.org/10.1021/acschemneuro.1c00675
ACS Chem. Neurosci. 2022, 13, 308312
This article is made available via the ACS COVID-19 subset for unrestricted RESEARCH re-use
and analyses in any form or by any means with acknowledgement of the original source.
These permissions are granted for the duration of the World Health Organization (WHO)
declaration of COVID-19 as a global pandemic.
Downloaded via UNIV AT BUFFALO STATE UNIV NEW YORK on February 28, 2022 at 15:23:14 (UTC).
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.
response, signicantly increasing oxidative stress and OCR, all
of which contributed to neuroinammation and associated
neuropathology of an encephalitic coronavirus infection.
In order to evade host cell immunity and facilitate virus
replication, SARS-CoV-2 viral open reading frame (ORF) 9b
localizes in mitochondria and can directly modulate
mitochondrial function, thereby contributing to COVID-19
disease progression.
4
Thus, we hypothesize that modulating mitochondrial activity
may prevent mitochondrial dysfunction following SARS-CoV-2
infection and that mitochondria-targeted pharmacological
interventions may enhance an immune response in SARS-
CoV-2 associated neuropathogenesis.
Toward verication of this hypothesis, we analyzed the
molecular composition in the mitochondria of infected cells. It
is worth noting that characterization of the mitochondria
metabolic variations by standard biochemical approaches is
extremely challenging. Traditional molecular proling ap-
proaches rely on cellular fractioning and extraction of the
analyte protein or lipid molecules from the studied organelles,
whichisacumbersomeprocedurethatisproneto
contamination. Additionally, the molecular extraction approach
inherently produces data averaging, thus masking hetero-
geneity between organelles obtained from dierent cells.
Remarkably, the capabilities of biochemical analysis have
been recently expanded with optical biosensing tools. Raman
spectrometry, one of the most valuable biosensing technolo-
gies, relies on analysis of molecular vibrational spectra and
enables the identication of diverse molecular groups in
biological samples. Because of their inherently noninvasive
properties and independence from labels, Raman-based
techniques have opened new dimensions in systemic studies
of cells and tissues.
10,11
The recently developed biomolecular
component analysis (BCA) of Raman spectra enables selective
detection and concentration measurements of the major
categories of biomolecules, including lipids, proteins, nucleic
acids, and saccharides,
12,13
in the studied samples. The high
three-dimensional resolution available in modern confocal
Raman spectrometry setups has been validated for character-
ization of microscopic subcellular structures, such as single
organelles, including the identication of abnormal biomo-
lecular signatures associated with disease progression.
1420
In this study, we employed Raman spectrometry together
with the BCA algorithm to characterize the changes in the
molecular composition of mitochondria in response to
treatment with heat-inactivated SARS-CoV-2 or the SARS-
CoV-2 spike protein. In addition, we studied key organelles
involved in lipid metabolism: Golgi apparatus (GA) and lipid
droplets (LD). The roles lipids play in viral infection include
viral endocytosis and exocytosis, viral entry into the host cell
via membrane fusion, and viral replication, and therefore, we
were interested in potential changes of the lipid signatures in
these organelles.
Our data indicate that infection with SARS-CoV-2 causes
mitochondrial dysfunction in microglia cells, which triggers
metabolic alterations that result in a substantial increase in
glycolysis.
9
These ndings suggest that a metabolic switch to
glycolysis compensates for mitochondrial dysfunction and an
energy decit in microglia and that a consequence of this
metabolic change is an enhanced inammatory response that
contributes to neuropathology associated with COVID-19. At
the same time, the molecular content of GA and LD was not
signicantly changed, apparently because of the lack of specic
interactions between these organelles and the components of
SARS-CoV-2.
Overall, our ndings support a view that viral infection of
host cells results in higher metabolic alterations to cope with
the increased anabolic demand of the cell for viral replication.
Furthermore, SARS-CoV-2-induced manipulation of the host-
cell metabolic machineries alters transcriptional regulation of
key metabolic pathways.
METHODS
Cell Culturing and Sample Preparation. Human microglia cells
(HMC3) were obtained from ATCC (cat. no. ATCC CRL-3304) and
grown in luminescence-free 35 mm glass-bottom dishes (Fisher
Scientic Co., Hanover Park, IL). The culture medium used was
Eagles Minimum Essential Medium (EMEM) (cat. no. ATCC 30-
2003) supplemented with 5% fetal bovine serum (FBS), 100 units/
mL penicillin, and 100 μg/mL streptomycin, and the cells were grown
to 70% conuence at 37 °C in a humidied atmosphere containing
5% CO2. The mitochondria and GA were labeled with MitoTracker
Green FM and NBD C6 ceramide-BSA (Thermo Fisher Scientic),
respectively, as per the manufacturer-provided protocols. After
labeling, the cells were thoroughly washed in sterile phosphate-
buered saline (PBS).
The cells were treated with the following viral constructs: 0.5 μg/
mL recombinant spike protein from SARS-related Coronavirus 2
Wuhan-Hu-1 (BEI Resources Inc., cat. no. NR-52308, lot no.
70034410) or 5 μL/mL heat-inactivated SARS-Coronavirus 2 (HI-
SARS), isolate USA-WA1/2020, (BEI Resources Inc., cat. no. NR-
52286, lot no. 70033548, pre-inactivation titer by TCID50 assay in
Vero E6 Cells = 1.6 ×105TCID50/mL), as specied.
To target acquisition of Raman spectra to specic organelles, the
mitochondria, endoplasmic reticulum (ER), and GA were labeled
using MitoTracker Green FM, ER-Tracker Green, and NBD C6
ceramide-BSA (ThermoFisher Scientic), respectively, as per the
manufacturer-provided protocols. Then the cells were thoroughly
washed in sterile PBS, and Raman spectra were acquired in the labeled
organelles.
The Raman Microscope. The spectra were measured on a DXR2
Raman microscopy setup (Thermo Fisher Scientic, Madison, WI),
equipped with a laser source unit emitting 60 mW at 633 nm
(ROUSB-633-PLR-70-1, Ondax), a 50 μm pinhole to shape the laser
beam to a 0.7 μm×0.7 μm×1.5 μm full width at half-maximum
(fwhm), and a Plan N 100×Olympus objective lens (NA = 1.25). In
addition, the Raman microscope was equipped with a uorescence
illumination system (5-UR7005, Olympus), a green uorescence cube
(488/561EX), and a uorescence lamp (X-Cite 120 PC, Photonic
Solutions).
Acquisition of Raman Spectra. Prior to the measurements, live
cells were transferred into optically transparent Dulbeccos Modied
Eagles Medium (DMEM) (Thermo Fisher Scientic) and mounted
on the microscope stage. The spectra were acquired from the labeled
organelles in live cells as recently described.
15
Fluorescence-labeled
organelles were visualized using the 488/561EX uorescence cube.
To generate the spectra, the Raman excitation laser was overlapped
with single labeled organelles. To warrant a high-quality signal/noise
ratio, the spectra accumulation parameter was set to 6 ×20 s;
importantly, no measurable phototoxicity was observed at this
irradiation dose. During the experiments, the cells were maintained
under physiological conditions at 37 °C. We visually veried the XYZ
position of the cell before and after each measurement to ensure the
spatial precision of Raman spectra acquisition.
Biomolecular Component Analysis of Raman Spectra. The
calibration of Raman band intensities on the concentrations of
biomolecules in the sample was performed as previously
described.
15,21
Quantitative analysis of cellular spectra was performed
using BCAbox software (ACIS LLC, Bualo, NY). The description,
interface of the BCAbox software, and schematics for the spectrum
processing algorithm are shown in Figures S1 and S2. Representative
ACS Chemical Neuroscience pubs.acs.org/chemneuro Letter
https://doi.org/10.1021/acschemneuro.1c00675
ACS Chem. Neurosci. 2022, 13, 308312
309
examples of raw and preprocessed mitochondria spectra are shown in
Figure S3.
RESULTS AND DISCUSSION
In our experiments, we incubated cultured microglia cells with
SARS-CoV-2 spike protein or heat-inactivated SARS-CoV-2 to
imitate viral neuroinvasion. Untreated microglia were used as
an experimental control. Mitochondria and GA were stained
with specicuorescence probes, thus enabling acquisition of
Raman spectra in these organelles, while LD were identied by
transmitted light imaging.
The obtained Raman spectra were processed with the BCA
algorithm to quantify the concentrations of major groups of
biomolecules (Figures S6 and S7). The measurements were
performed as recently described.
18
It is worth noting that
although the Raman spectra were collected within a submicron
volume of an excitation laser focused on specic organelles, the
adjacent cytoplasm may also overlap with the laser probe and
contribute to the spectra. Nevertheless, despite this potential
contribution, there were statistically signicant dierences
between the molecular proles obtained in various organelles,
which supports the sensitivity of Raman microspectrometry to
the subcellular biochemical environment. The measured values
obtained in single mitochondria of control and treated cells are
shown in Tables S1S3.
We found that treatment with SARS-CoV-2 spike protein or
HI-SARS induced signicant alterations in the concentrations
of diverse types of biomolecules in the mitochondria. First, the
concentration of mitochondrial DNA was reduced almost 2-
fold in the infected cells, from 2.2 mg/mL in control cells to
1.2 mg/mL in the cells treated with either viral agent (Figure
1), which indicates the decrease in mitochondrial DNA copy
number. At the same time, the concentration of mtRNA was
increased from 2.25 mg/mL in the control to 2.8 mg/mL in
HI-SARS-treated cells and 4.0 mg/mL in cells treated with
the spike protein; the latter dierence was statistically
signicant. This increase in RNA is consistent with previous
reports on mitochondrial genome upregulation in cells infected
by SARS-CoV-2.
7
We also found a signicant reduction in
mitochondrial saccharides from 1.5 mg/mL in the control to
0.9 mg/mL in the HI-SARS-treated cells and 0.7 mg/mL in
the cells treated with the spike protein. The mitochondrial
saccharide fraction includes glucose and pyruvate, and its
reduction suggests a decrease of the respiratory function of
mitochondria.
Furthermore, we detected a signicant perturbation in the
saturation of phospholipids populating the mitochondrial
lipidome. The average number of unsaturated CC bonds
per phospholipid was signicantly reduced from 4.3 in the
control to 3.8 in the cells treated with the HI-SARS viral
construct and 3.7 in the cells treated with the spike protein.
At the same time, we did not record any signicant change in
the total concentration of lipids in mitochondria (Figure 1).
We thus concluded that the shift in lipidome saturation occurs
as a result of biochemical processes inside the mitochondria
and likely is not caused by tracking of the saturated
phospholipids to this organelle.
In parallel, we investigated the impact of HI-SARS on the
major organelles involved in the metabolism of lipids such as
GA. However, it appears that SARS-CoV-2 does not directly
inuence the lipid biogenesis. We found that all of the
resolvable lipidome characteristics in the control and treated
cells for these organelles were remarkably uniform. Similarly,
the composition of LD in the treated cells remained largely
unchanged. However, we found that HI-SARS induces an
increase in the number of CC bonds in the pool of
unsaturated phospholipids stored in LD (Figures S5S7).
In the interpretation of our data, we point to the fact that
mitochondrial lipids are predominantly synthesized in the
endoplasmic reticulum and then transported to the mitochon-
dria through the GA. While these organelles show no
dierences in molecular composition between control and
treated cells, the mitochondria demonstrate substantial dier-
ences not only in phospholipid saturation but also in the
abundances of RNA, saccharides, and mtDNA (Figure 1). We
propose that these changes originate in virus-induced ROS
production, in part via oxidative damage to lipids and oxidation
of respiratory chain proteins, aecting metabolism and protein
import, which then induces DNA damage as reected in a
sharp decrease in the mtDNA level. Furthermore, the
mechanistic link between lipid metabolism and inammation
is well-established, wherein lipids can directly activate
inammatory pathways.
22
Thus, signicant changes in the
composition and distribution of lipids within the brain are
believed to contribute to neurocognitive decline.
23
Further-
more, SARS-CoV-2-induced oxidative stress impacts phospho-
lipid membranes, causing additional perturbations of biological
processes.
24
We propose that increased oxidative stress impacts
the uidity of phospholipid membranes, which can aect the
interactions and activity of metabolic enzymes, resulting in
membrane remodeling. The membrane fatty acid composition
is thought to be altered in response to oxidative stress by a
Figure 1. Comparative analysis of the molecular content in
nontreated mitochondria (control) and mitochondria treated with
either heat-inactivated SARS-CoV-2 or the SARS-CoV-2 spike protein
(as indicated). The top chart shows absolute concentrations of
proteins, DNA, RNA, saccharides (Gly), and lipids in live-cell
mitochondria. The bottom chart shows a decrease in the number of
CC bonds in mitochondrial phospholipids (phospholipid unsatura-
tion parameter) in both groups of treated cells. The statistically
signicant dierences are indicated by horizontal brackets and p
values.
ACS Chemical Neuroscience pubs.acs.org/chemneuro Letter
https://doi.org/10.1021/acschemneuro.1c00675
ACS Chem. Neurosci. 2022, 13, 308312
310
decrease in the number of CC bonds, which results in higher
saturation of the organellar lipidome.
24,25
The physiological
relevance of membrane remodeling remains unclear, but it may
be an adaptive response to cellular stress. These data support
our hypothesis that mitochondrial dysfunction, oxidative stress,
and inammation could lead to an increase in COVID-
associated neurological dysfunction. In addition, our data
support the premise that SARS-CoV-2 induces release of
pathogen-associated molecular patterns (PAMPS) and danger-
associated molecular patterns (DAMPS), ATP, oxidized lipids,
and heat shock proteins, all of which are associated with
apoptosis and autophagy.
26,27
Overall, our study clariestheroleofmitochondrial
dysfunction in SARS-CoV-2-induced neuropathology. Our
data suggest that mitochondrial dysfunction is among the
earliest and most prominent features of neurodegeneration. In
addition, the absence of any signicant changes in the lipidome
of GA and LD indicate a targeted impact of SARS infection on
mitochondria. Therefore, examining mitochondrial function or
mitochondrial damage markers in the microglia cells in
response to interactions with SARS-CoV-2 spike protein may
help identify pathways of viral pathogenesis, unravel
mechanisms of cellular vulnerability, and aid in the discovery
of mitochondrial biomarkers relevant to SARS-CoV-2 neuro-
inammation and progression to neuropathogenesis. Further-
more, therapeutic strategies that modulate mitochondrial
processes may be ecacious in treating patients with neuro-
COVID. Our study calls for the development of mitochondria-
targeted pharmaceutical drugs that can neutralize virus-induced
ROS production in these cellular organelles.
ASSOCIATED CONTENT
*
sıSupporting Information
The Supporting Information is available free of charge at
https://pubs.acs.org/doi/10.1021/acschemneuro.1c00675.
Figures S1S7 and Tables S1S3 (PDF)
AUTHOR INFORMATION
Corresponding Authors
Paras N. Prasad Institute for Lasers, Photonics and
Biophotonics and Department of Chemistry, University at
Bualo, The State University of New York, Bualo, New York
14260, United States; orcid.org/0000-0002-0905-7084;
Email: pnprasad@bualo.edu
Supriya D. Mahajan Department of Medicine, Division of
Allergy, Immunology, and Rheumatology, State University of
New York at Bualo, Clinical Translational Research Center,
Bualo, New York 14203, United States; Email: smahajan@
bualo.edu
Authors
Artem Pliss Institute for Lasers, Photonics and Biophotonics
and Department of Chemistry, University at Bualo, The
State University of New York, Bualo, New York 14260,
United States; orcid.org/0000-0003-4867-4074
Andrey N. Kuzmin Institute for Lasers, Photonics and
Biophotonics and Department of Chemistry, University at
Bualo, The State University of New York, Bualo, New York
14260, United States; orcid.org/0000-0001-7371-4643
Complete contact information is available at:
https://pubs.acs.org/10.1021/acschemneuro.1c00675
Author Contributions
A.P., A.N.K., P.N.P., and S.D.M. conceived the project. A.P.,
A.N.K., and S.D.M. performed the experiments. All of the
authors drafted and edited the manuscript.
Funding
Funding support by the National Institute of Drug Abuse,
National Institutes of Health (Grant 5R01DA047410-02) to
S.D.M. toward experiments in this study is duly acknowledged
Notes
The authors declare no competing nancial interest.
REFERENCES
(1) Khan, M.; Syed, G. H.; Kim, S. J.; Siddiqui, A. Mitochondrial
dynamics and viral infections: A close nexus. Bba-Mol. Cell Res. 2015,
1853, 28222833.
(2) Tiku, V.; Tan, M. W.; Dikic, I. Mitochondrial Functions in
Infection and Immunity. Trends Cell Biol. 2020,30, 263275.
(3) Cao, Z.; Wu, Y.; Faucon, E.; Sabatier, J. M. SARS-CoV-2 &
Covid-19: Key-Roles of the Renin-AngiotensinSystem/Vitamin D
Impacting Drug and Vaccine Developments. Infect Disord Drug
Targets 2020,20, 348349.
(4) Singh, K. K.; Chaubey, G.; Chen, J. Y.; Suravajhala, P. Decoding
SARS-CoV-2 hijacking of host mitochondria in COVID-19 patho-
genesis. Am. J. Physiol Cell Physiol 2020,319, C258C267.
(5) Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Krüger, N.;
Herrler, T.; Erichsen, S.; Schiergens, T. S.; Herrler, G.; Wu, N.-H.;
Nitsche, A.; Müller, M. A.; Drosten, C.; Pöhlmann, S. SARS-CoV-2
Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a
Clinically Proven Protease Inhibitor. Cell 2020,181, 271280.
(6) Shi, T. T.; Yang, F. Y.; Liu, C.; Cao, X.; Lu, J.; Zhang, X. L.;
Yuan, M. X.; Chen, C.; Yang, J. K. Angiotensin-converting enzyme 2
regulates mitochondrial function in pancreatic beta-cells. Biochem.
Biophys. Res. Commun. 2018,495, 860866.
(7) Saleh, J.; Peyssonnaux, C.; Singh, K. K.; Edeas, M. Mitochondria
and microbiota dysfunction in COVID-19 pathogenesis. Mitochond-
rion 2020,54,17.
(8) Li, X.; Fang, P.; Mai, J.; Choi, E. T.; Wang, H.; Yang, X.-f.
Targeting mitochondrial reactive oxygen species as novel therapy for
inflammatory diseases and cancers. J. Hematol. Oncol. 2013,6, No. 19.
(9) Clough, E.; Inigo, J.; Chandra, D.; Chaves, L.; Reynolds, J. L.;
Aalinkeel, R.; Schwartz, S. A.; Khmaladze, A.; Mahajan, S. D.
Mitochondrial dynamics in SARS-CoV-2 spike protein treated human
Microglia: Implications for Neuro-COVID. J. Neuroimmune Pharma-
col. 2021,16, 770784.
(10) Wu, H. W.; Volponi, J. V.; Oliver, A. E.; Parikh, A. N.;
Simmons, B. A.; Singh, S. In vivo lipidomics using single-cell Raman
spectroscopy. P Natl. Acad. Sci. USA 2011,108, 38093814.
(11) Zhang, D. M.; Xie, Y.; Mrozek, M. F.; Ortiz, C.; Davisson, V. J.;
Ben-Amotz, D. Raman detection of proteomic analytes. Anal. Chem.
2003,75, 57035709.
(12) Kuzmin, A. N.; Pliss, A.; Prasad, P. N. Ramanomics: New
Omics Disciplines Using Micro Raman Spectrometry with Bio-
molecular Component Analysis for Molecular Profiling of Biological
Structures. Biosensors (Basel) 2017,7, No. 52.
(13) Kuzmin, A. N.; Pliss, A.; Rzhevskii, A.; Lita, A.; Larion, M.
BCAbox Algorithm Expands Capabilities of Raman Microscope for
Single Organelles Assessment. Biosensors (Basel) 2018,8, 106.
(14) Lita, A.; Pliss, A.; Kuzmin, A.; Yamasaki, T.; Zhang, L. M.;
Dowdy, T.; Burks, C.; de Val, N.; Celiku, O.; Ruiz-Rodado, V.; Nicoli,
E. R.; Kruhlak, M.; Andresson, T.; Das, S.; Yang, C. Z.; Schmitt, R.;
Herold-Mende, C.; Gilbert, M. R.; Prasad, P. N.; Larion, M. IDH1
mutations induce organelle defects via dysregulated phospholipids.
Nat. Commun. 2021,12, No. 614.
(15) Lita, A.; Kuzmin, A. N.; Pliss, A.; Baev, A.; Rzhevskii, A.;
Gilbert, M. R.; Larion, M.; Prasad, P. N. Toward Single-Organelle
Lipidomics in Live Cells. Anal. Chem. 2019,91, 1138011387.
ACS Chemical Neuroscience pubs.acs.org/chemneuro Letter
https://doi.org/10.1021/acschemneuro.1c00675
ACS Chem. Neurosci. 2022, 13, 308312
311
(16) Yadav, N.; Pliss, A.; Kuzmin, A.; Rapali, P.; Sun, L.; Prasad, P.;
Chandra, D. Transformations of the macromolecular landscape at
mitochondria during DNA-damage-induced apoptotic cell death. Cell
Death Dis. 2014,5, No. e1453.
(17) OMalley, J.; Kumar, R.; Kuzmin, A. N.; Pliss, A.; Yadav, N.;
Balachandar, S.; Wang, J. M.; Attwood, K.; Prasad, P. N.; Chandra, D.
Lipid quantification by Raman microspectroscopy as a potential
biomarker in prostate cancer. Cancer Lett. 2017,397,5260.
(18) Pliss, A.; Kuzmin, A. N.; Lita, A.; Kumar, R.; Celiku, O.; Atilla-
Gokcumen, G. E.; Gokcumen, O.; Chandra, D.; Larion, M.; Prasad, P.
N. A Single-Organelle Optical Omics Platform for Cell Science and
Biomarker Discovery. Anal. Chem. 2021,93, 82818290.
(19) Kuzmin, A. N.; Levchenko, S. M.; Pliss, A.; Qu, J.; Prasad, P. N.
Molecular profiling of single organelles for quantitative analysis of
cellular heterogeneity. Sci. Rep 2017,7, 6512.
(20) Levchenko, S. M.; Kuzmin, A. N.; Pliss, A.; Qu, J.; Prasad, P. N.
Macromolecular Profiling of Organelles in Normal Diploid and
Cancer Cells. Anal. Chem. 2017,89, 1098510990.
(21) Pliss, A.; Kuzmin, A. N.; Kachynski, A. V.; Prasad, P. N.
Nonlinear Optical Imaging and Raman Microspectrometry of the Cell
Nucleus throughout the Cell Cycle. Biophys. J. 2010,99, 34833491.
(22) Batista-Gonzalez, A.; Vidal, R.; Criollo, A.; Carreno, L. J. New
Insights on the Role of Lipid Metabolism in the Metabolic
Reprogramming of Macrophages. Front. Immunol 2020,10, No. 2993.
(23) Panza, F.; DIntrono, A.; Colacicco, A. M.; Capurso, C.;
Pichichero, G.; Capurso, S. A.; Capurso, A.; Solfrizzi, V. Lipid
metabolism in cognitive decline and dementia. Brain Res. Rev. 2006,
51, 275292.
(24) Fernandes, I. G.; de Brito, C. A.; dos Reis, V. M. S.; Sato, M.
N.; Pereira, N. Z. SARS-CoV-2 and Other Respiratory Viruses: What
Does Oxidative Stress Have to Do with It? Oxid Med. Cell. Longevity
2020,2020, No. 8844280.
(25) Rodrigo, R.; Fernandez-Gajardo, R.; Gutierrez, R.; Matamala, J.
M.; Carrasco, R.; Miranda-Merchak, A.; Feuerhake, W. Oxidative
Stress and Pathophysiology of Ischemic Stroke: Novel Therapeutic
Opportunities. Cns Neurol Disord-Dr 2013,12, 698714.
(26) Tang, D. L.; Comish, P.; Kang, R. The hallmarks of COVID-19
disease. PLoS Pathog. 2020,16, No. e1008536.
(27) Kumar, P.; Sobhanan, J.; Takano, Y.; Biju, V. Molecular
recognition in the infection, replication, and transmission of COVID-
19-causing SARS-CoV-2: an emerging interface of infectious disease,
biological chemistry, and nanoscience. NPG Asia Mater. 2021,13, 14.
ACS Chemical Neuroscience pubs.acs.org/chemneuro Letter
https://doi.org/10.1021/acschemneuro.1c00675
ACS Chem. Neurosci. 2022, 13, 308312
312
... Conversely, the downregulated intersecting genes were mainly enriched in pathways related to mitochondrial function, ion transport, neural development, and behavior ( Figure 2D). The manipulation of mitochondria via SARS-CoV-2 could induce mitochondria dysfunction and then increase mitochondria-derived double-membrane vehicles in which the virus can hide and replicate [79]. Mitochondria have been reported to be involved in the process of inflammation in both innate and adaptive immunity [80]. ...
... Mitochondrial dysfunction induced by manipulations of host mitochondria using SRAS-CoV-2 viral open reading frames (ORFs) has also been reported [115]. A recent study reported that mitochondrial dysfunction, as the earliest feature of neurodegeneration, is a prelude to SARS-CoV-2-induced neuropathogenesis [79]. In our study, the common down-regulated DEGs shared in AD and COVID-19 patients are enriched in mitochondrion organization and the mitochondrial respiratory chain complex assembly. ...
... Similarly, among the top 50 down-regulated AD DEGs, 54% of them (27 genes) are downexpressed upon SARS-CoV-2 infection. NDUFA (NDUFA9, NDUFAB1), ATF (ATP5F1A, ATP5PB, ATP5MC3, ATP6V1D, ATP6V1E1), PSMA (PSMA1, PSMA5), and MRPS (MRPS16, MRPS23) families are enriched in these 27 genes.The most enriched GO term for these 27 genes is the mitochondrial respiratory chain complex assembly, which highlights the critical role of mitochondria in AD and COVID-19 progression[79]. When the common 12 up-expressed and 27 down-expressed DEGs are combined, these genes are enriched in a new GO term of antigen receptor-mediated signaling pathway, which again supports the observation that inflammatory responses play an essential role in both AD progression and SARS-CoV-2 infection. ...
Article
Full-text available
SARS-CoV-2 caused the COVID-19 pandemic. COVID-19 may elevate the risk of cognitive impairment and even cause dementia in infected individuals; it may accelerate cognitive decline in elderly patients with dementia, possibly in Alzheimer’s disease (AD) patients. However, the mechanisms underlying the interplay between AD and COVID-19 are still unclear. To investigate the underlying mechanisms and associations between AD progression and SARS-CoV-2 infection, we conducted a series of bioinformatics research into SARS-CoV-2-infected cells, COVID-19 patients, AD patients, and SARS-CoV-2-infected AD patients. We identified the common differentially expressed genes (DEGs) in COVID-19 patients, AD patients, and SARS-CoV-2-infected cells, and these DEGs are enriched in certain pathways, such as immune responses and cytokine storms. We constructed the gene interaction network with the signaling transduction module in the center and identified IRF7, STAT1, STAT2, and OAS1 as the hub genes. We also checked the correlations between several key transcription factors and the SARS-CoV-2 and COVID-19 pathway-related genes. We observed that ACE2 expression is positively correlated with IRF7 expression in AD and coronavirus infections, and interestingly, IRF7 is significantly upregulated in response to different RNA virus infections. Further snRNA-seq analysis indicates that NRGN neurons or endothelial cells may be responsible for the increase in ACE2 and IRF7 expression after SARS-CoV-2 infection. The positive correlation between ACE2 and IRF7 expressions is confirmed in the hippocampal formation (HF) of SARS-CoV-2-infected AD patients. Our findings could contribute to the investigation of the molecular mechanisms underlying the interplay between AD and COVID-19 and to the development of effective therapeutic strategies for AD patients with COVID-19.
... Furthermore, viral infections have also been shown to trigger mitochondrial dysfunction in affected cells due to impaired mitophagy (Thangaraj et al., 2018;Foo et al., 2022;Pliss et al., 2022). Mitophagy is a selective form of autophagy in which damaged mitochondria are degraded and recycled for mitochondrial biogenesis. ...
... Next, the role of UA in attenuating mitochondrial damage and oxidative stress, which have been shown to occur in viral infections, was investigated (Thangaraj et al., 2018;Foo et al., 2022;Pliss et al., 2022). In contrast to other studies showing that UA induces mitophagy by decreasing mitochondrial accumulation and thus the expression of mitochondrial proteins such as COX4, here UA instead led to a very slight increase in COX4 expression. ...
Article
Full-text available
Neuroinflammation can be triggered by various stimuli, including viral infections. Viruses can directly invade the brain and infect neuronal cells or indirectly trigger a “cytokine storm” in the periphery that eventually leads to microglial activation in the brain. While this initial activation of microglial cells is important for viral clearance, chronic activation leads to excessive inflammation and oxidative stress, which can be neurotoxic. Remarkebly, recent studies have shown that certain viruses such as influenza A virus, coronavirus, herpes virus and Epstein–Barr virus may be involved in the development of neurodegenerative diseases such as Parkinson’s disease, Alzheimer’s disease, and multiple sclerosis. Therefore, it is important to find therapeutic strategies against chronic neuroinflammation triggered by viral infections. Here, we investigated the effects of urolithin A (UA) on microglial activation in vitro induced by a viral mimetic, poly I:C, in a triple co-culture system of neurons, astrocytes and microglial cells. Immunocytochemistry was used to perform a comprehensive single-cell analysis of the morphological changes of microglia as an indicator of their reactive state. Treatment with UA significantly prevented the poly I:C-induced reactive state of microglia, which was characterized by increased expression of the microglial activation markers CD68 and IBA-1. UA restored the poly I:C-induced morphology by restoring microglial ramification. In addition, UA was able to reduce the release of the pro-inflammatory mediators CCL2, TNF-α, and IL-1β and showed a trend toward attenuation of cellular ROS production in poly I:C-treated cultures. Overall, this study suggests that UA as a component of a healthy diet may help prevent virus-induced neuroinflammation and may have therapeutic potential for future studies to prevent or treat neurodegenerative diseases by targeting the associated neuroinflammatory processes.
... Microglia are the immune cells of the CNS and mediate homeostasis, during viral neurotropic infections. In a recent study, we observed that microglia treated with either SARS-CoV2 spike protein or heat-inactivated SARS-CoV2 trigger a dramatic reduction in mtDNA content and an increase in phospholipid saturation levels [17]. We also showed that SARS-CoV2 spike protein increases the levels of pro-inflammatory cytokines and ROS production, increased cell apoptosis and oxygen consumption rate (OCR) in microglial cells indicative of increased oxidative stress. ...
... We have previously shown that METH independently cause significant changes in lipoproteins [4]. We also showed microglia treated with either spike protein SARS-CoV2 trigger a dramatic reduction in mtDNA content and an increase in phospholipid saturation levels [17]. We speculate that METH and SARS-CoV2 disrupt redox homeostasis induces oxidative stress and lipid peroxidation which results in cell apoptosis due to the accumulation of lipid peroxides. ...
Article
Full-text available
Acute and chronic use of Methamphetamine (METH) has critical immunological implications and METH users are more vulnerable to SARS-CoV2 infection. Inflammasomes are activated in response to SARS-CoV2 infection. METH also activates NLRP3 inflammasome in microglia and promotes neuro cognitive deficits. The goal of the study was to examine the involvement of NLRP3 inflammasome in METH and/or SARS-CoV2 induced neuro-oxidative stress in microglial cells. Our results suggests that METH +/− SARS-CoV2 initiated a neuro immune-inflammatory response and mitochondrial oxidative stress via NLRP3 inflammasome activation induced increased Caspase −1 and increased lipid peroxidation. Our data suggests that SARS-CoV2 infection in METH abusing subjects may result in long-term neurological deficits resulting from microglial dysfunction and apoptosis attributed to NLRP3 inflammasome activation.
... On the other hand, aberrations of cerebral endothelium respiration are related to mitochondrial damage induced by S1-and Trimer proteins (Kim et al., 2021). When microglial cells were treated with S-protein or inactivated SARS-CoV-2 virions, specific alterations in mitochondrial biogenesis occurred through increased ROS levels, leading to a decrease in mitogenome copies and increased phospholipid saturation (Pliss et al., 2022). Those harmful responses within the central nervous system can be further exacerbated among patients suffering from additional diseases, for example, neurodegenerative ones (Denaro et al., 2022). ...
Article
Full-text available
The rapid development of the COVID-19 pandemic resulted in a closer analysis of cell functioning during beta-coronavirus infection. This review will describe evidence for COVID-19 as a syndrome with a strong, albeit still underestimated, mitochondrial component. Due to the sensitivity of host mitochondria to coronavirus infection, SARS-CoV-2 affects mitochondrial signaling, modulates the immune response, modifies cellular energy metabolism, induces apoptosis and ageing, worsening COVID-19 symptoms which can sometimes be fatal. Various aberrations across human systems and tissues and their relationships with mitochondria were reported. In this review, particular attention is given to characterization of multiple alterations in gene expression pattern and mitochondrial metabolism in COVID-19; the complexity of interactions between SARS-CoV-2 and mitochondrial proteins is presented. The participation of mitogenome fragments in cell signaling and the occurrence of SARS-CoV-2 subgenomic RNA within membranous compartments, including mitochondria is widely discussed. As SARS-CoV-2 severely affects the quality system of mitochondria, the cellular background for aberrations in mitochondrial dynamics in COVID-19 is additionally characterized. Finally, perspectives on the mitigation of COVID-19 symptoms by affecting mitochondrial biogenesis by numerous compounds and therapeutic treatments are briefly outlined.
... SAR-COV2 could induce m-Dys, activate mitochondrial-dependent intrinsic apoptotic pathways, and cause microglial and neuronal apoptosis leading to neuropathological symptoms in COVID-19 and PASC patients 169,170 . In the current pandemic, about 40% of COVID-19 patients demonstrated neurological symptoms, lingering neuro-inflammation, where neuronal damage in PASC patients has emerged as a novel syndrome, the 'Neuro-COVID' 169,171 . ...
Article
Full-text available
SARS‐CoV‐2, the etiological agent of COVID-19, is devoid of any metabolic capacity; therefore, it is criticalfor the viral pathogen to hijack host cellular metabolic machinery for its replication and propagation. Thissingle-stranded RNA virus with a 29.9 kb genome encodes 14 open reading frames (ORFs) and initiates aplethora of virus–host protein–protein interactions in the human body. These extensive viral proteininteractions with host-specific cellular targets could trigger severe human metabolic reprogramming/dysregulation (HMRD), a rewiring of sugar-, amino acid-, lipid-, and nucleotide-metabolism(s), as well asaltered or impaired bioenergetics, immune dysfunction, and redox imbalance in the body. In the infectiousprocess, the viral pathogen hijacks two major human receptors, angiotensin-converting enzyme (ACE)-2and/or neuropilin (NRP)-1, for initial adhesion to cell surface; then utilizes two major host proteases,TMPRSS2 and/or furin, to gain cellular entry; and finally employs an endosomal enzyme, cathepsin L (CTSL)for fusogenic release of its viral genome. The virus-induced HMRD results in 5 possible infectiousoutcomes: asymptomatic, mild, moderate, severe to fatal episodes; while the symptomatic acuteCOVID-19 condition could manifest into 3 clinical phases: (i) hypoxia and hypoxemia (Warburg effect), (ii)hyperferritinemia (‘cytokine storm’), and (iii) thrombocytosis (coagulopathy). The mean incubation period forCOVID-19 onset was estimated to be 5.1 days, and most cases develop symptoms after 14 days. The meanviral clearance times were 24, 30, and 39 days for acute, severe, and ICU-admitted COVID-19 patients,respectively. However, about 25–70% of virus-free COVID-19 survivors continue to sustain virus-inducedHMRD and exhibit a wide range of symptoms that are persistent, exacerbated, or new ‘onset’ clinicalincidents, collectively termed as post-acute sequelae of COVID-19 (PASC) or long COVID. PASC patientsexperience several debilitating clinical condition(s) with >200 different and overlapping symptoms that maylast for weeks to months. Chronic PASC is a cumulative outcome of at least 10 different HMRD-relatedpathophysiological mechanisms involving both virus-derived virulence factors and a multitude of innatehost responses. Based on HMRD and virus-free clinical impairments of different human organs/systems,PASC patients can be categorized into 4 different clusters or sub-phenotypes: sub-phenotype-1 (33.8%)with cardiac and renal manifestations; sub-phenotype-2 (32.8%) with respiratory, sleep and anxietydisorders; sub-phenotype-3 (23.4%) with skeleto-muscular and nervous disorders; and sub-phenotype-4(10.1%) with digestive and pulmonary dysfunctions. This narrative review elucidates the effects of viralhijack on host cellular machinery during SARS-CoV-2 infection, ensuing detrimental effect(s) of virus-induced HMRD on human metabolism, consequential symptomatic clinical implications, and damage tomultiple organ systems; as well as chronic pathophysiological sequelae in virus-free PASC patients. Wehave also provided a few evidence-based, human randomized controlled trial (RCT)-tested, precisionnutrients to reset HMRD for health recovery of PASC patients. (
... Pathway analysis of DEGs in SARS-CoV-2 infected OM-ALI cells also revealed alterations of genes involved in oxidative phosphorylation and mitochondrial function. Others have reported mitochondrial dysfunction in infected brain cells, which has been attributed to the neuropathogenesis of SARS-CoV-2 infection [81]. Furthermore, mitochondrial dysfunction is a hallmark of many neurodegenerative diseases, including AD, and alterations in mitochondrially located genes and mitochondrial function in OM cells of individuals with AD have been previously demonstrated [30]. ...
Article
Full-text available
Background The neurological effects of the coronavirus disease of 2019 (COVID-19) raise concerns about potential long-term consequences, such as an increased risk of Alzheimer's disease (AD). Neuroinflammation and other AD-associated pathologies are also suggested to increase the risk of serious SARS-CoV-2 infection. Anosmia is a common neurological symptom reported in COVID-19 and in early AD. The olfactory mucosa (OM) is important for the perception of smell and a proposed site of viral entry to the brain. However, little is known about SARS-CoV-2 infection at the OM of individuals with AD. Methods To address this gap, we established a 3D in vitro model of the OM from primary cells derived from cognitively healthy and AD individuals. We cultured the cells at the air–liquid interface (ALI) to study SARS-CoV-2 infection under controlled experimental conditions. Primary OM cells in ALI expressed angiotensin-converting enzyme 2 (ACE-2), neuropilin-1 (NRP-1), and several other known SARS-CoV-2 receptor and were highly vulnerable to infection. Infection was determined by secreted viral RNA content and confirmed with SARS-CoV-2 nucleocapsid protein (NP) in the infected cells by immunocytochemistry. Differential responses of healthy and AD individuals-derived OM cells to SARS-CoV-2 were determined by RNA sequencing. Results Results indicate that cells derived from cognitively healthy donors and individuals with AD do not differ in susceptibility to infection with the wild-type SARS-CoV-2 virus. However, transcriptomic signatures in cells from individuals with AD are highly distinct. Specifically, the cells from AD patients that were infected with the virus showed increased levels of oxidative stress, desensitized inflammation and immune responses, and alterations to genes associated with olfaction. These results imply that individuals with AD may be at a greater risk of experiencing severe outcomes from the infection, potentially driven by pre-existing neuroinflammation. Conclusions The study sheds light on the interplay between AD pathology and SARS-CoV-2 infection. Altered transcriptomic signatures in AD cells may contribute to unique symptoms and a more severe disease course, with a notable involvement of neuroinflammation. Furthermore, the research emphasizes the need for targeted interventions to enhance outcomes for AD patients with viral infection. The study is crucial to better comprehend the relationship between AD, COVID-19, and anosmia. It highlights the importance of ongoing research to develop more effective treatments for those at high risk of severe SARS-CoV-2 infection. Graphical Abstract
... There is much speculation about how exactly mitochondria help the coronavirus replicate and evade the immune response, but there is little concrete data [104][105][106]. On the other hand, there are quite a lot of studies showing that mitochondria are disrupted during COVID-19 in platelets [107], leukocytes [108], microglia [109], and cardiomyocytes [110]. In most cases, this is discussed in the context of the long-term consequences of the disease. ...
Article
Full-text available
The hematological effects of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are important in COVID-19 pathophysiology. However, the interactions of SARS-CoV-2 with platelets and red blood cells are still poorly understood. There are conflicting data regarding the mechanisms and significance of these interactions. The aim of this review is to put together available data and discuss hypotheses, the known and suspected effects of the virus on these blood cells, their pathophysiological and diagnostic significance, and the potential role of platelets and red blood cells in the virus’s transport, propagation, and clearance by the immune system. We pay particular attention to the mutual activation of platelets, the immune system, the endothelium, and blood coagulation and how this changes with the evolution of SARS-CoV-2. There is now convincing evidence that platelets, along with platelet and erythroid precursors (but not mature erythrocytes), are frequently infected by SARS-CoV-2 and functionally changed. The mechanisms of infection of these cells and their role are not yet entirely clear. Still, the changes in platelets and red blood cells in COVID-19 are significantly associated with disease severity and are likely to have prognostic and pathophysiological significance in the development of thrombotic and pulmonary complications.
... Mitochondria, crucial for host innate immunity, are exploited by many viruses, including SARS-CoV-2, to avert detection and ensure proper replication in host cells. For example, SARS-CoV-2 antigens (nonstructural protein 4 and 9 (NSP4, NSP9) and open reading frame 9C (ORF9C) disrupt the organelle, generating mitochondrial reactive oxygen species (mROS), which promote the development of pTau [9,[26][27][28]. In this regard, assays of phosphorylated Tau at threonine 217 (pTau-217) and threonine 181 (pTau-181), developed by Lilly and Simoa ®® , are being utilized as tauopathy blood markers, respectively. ...
Article
Full-text available
Long COVID, also called post-acute sequelae of SARS-CoV-2, is characterized by a multitude of lingering symptoms, including impaired cognition, that can last for many months. This symptom, often called "brain fog", affects the life quality of numerous individuals, increasing medical complications as well as healthcare expenditures. The etiopathogenesis of SARS-CoV-2-induced cognitive deficit is unclear, but the most likely cause is chronic inflammation maintained by a viral remnant thriving in select body reservoirs. These viral sanctuaries are likely comprised of fused, senescent cells, including microglia and astrocytes, that the pathogen can convert into neurotoxic phenotypes. Moreover, as the enteric nervous system contains neurons and glia, the virus likely lingers in the gastrointestinal tract as well, accounting for the intestinal symptoms of long COVID. Fusogens are proteins that can overcome the repulsive forces between cell membranes, allowing the virus to coalesce with host cells and enter the cytoplasm. In the intracellular compartment, the pathogen hijacks the actin cytoskeleton, fusing host cells with each other and engendering pathological syncytia. Cell-cell fusion enables the virus to infect the healthy neighboring cells. We surmise that syncytia formation drives cognitive impairment by facilitating the "seeding" of hyperphosphorylated Tau, documented in COVID-19. In our previous work, we hypothesized that the SARS-CoV-2 virus induces premature endothelial senescence, increasing the permeability of the intestinal and blood-brain barrier. This enables the migration of gastrointestinal tract microbes and/or their components into the host circulation, eventually reaching the brain where they may induce cognitive dysfunction. For example, translocated lipopolysaccharides or microbial DNA can induce Tau hyperphosphorylation, likely accounting for memory problems. In this perspective article, we examine the pathogenetic mechanisms and potential biomarkers of long COVID, including microbial cell-free DNA, interleukin 22, and phosphorylated Tau, as well as the beneficial effect of transcutaneous vagal nerve stimulation.
Article
Full-text available
Mitochondria have multiple functions such as supplying energy, regulating the redox status, and producing proteins encoded by an independent genome. They are closely related to the physiology and pathology of many organs and tissues, among which the brain is particularly prominent. The brain demands 20% of the resting metabolic rate and holds highly active mitochondrial activities. Considerable research shows that mitochondria are closely related to brain function, while mitochondrial defects induce or exacerbate pathology in the brain. In this review, we provide comprehensive research advances of mitochondrial biology involved in brain functions, as well as the mitochondria-dependent cellular events in brain physiology and pathology. Furthermore, various perspectives are explored to better identify the mitochondrial roles in neurological diseases and the neurophenotypes of mitochondrial diseases. Finally, mitochondrial therapies are discussed. Mitochondrial-targeting therapeutics are showing great potentials in the treatment of brain diseases.
Article
Full-text available
SARS‐CoV‐2, the etiological agent of COVID-19, is devoid of any metabolic capacity; therefore, it is critical for the viral pathogen to hijack host cellular metabolic machinery for its replication and propagation. This single-stranded RNA virus with a 29.9 kb genome encodes 14 open reading frames (ORFs) and initiates a plethora of virus–host protein–protein interactions in the human body. These extensive viral protein interactions with host-specific cellular targets could trigger severe human metabolic reprogramming/dysregulation (HMRD), a rewiring of sugar-, amino acid-, lipid-, and nucleotide-metabolism(s), as well as altered or impaired bioenergetics, immune dysfunction, and redox imbalance in the body. In the infectious process, the viral pathogen hijacks two major human receptors, angiotensin-converting enzyme (ACE)-2 and/or neuropilin (NRP)-1, for initial adhesion to cell surface; then utilizes two major host proteases, TMPRSS2 and/or furin, to gain cellular entry; and finally employs an endosomal enzyme, cathepsin L (CTSL) for fusogenic release of its viral genome. The virus-induced HMRD results in 5 possible infectious outcomes: asymptomatic, mild, moderate, severe to fatal episodes; while the symptomatic acute COVID-19 condition could manifest into 3 clinical phases: (i) hypoxia and hypoxemia (Warburg effect), (ii) hyperferritinemia (‘cytokine storm’), and (iii) thrombocytosis (coagulopathy). The mean incubation period for COVID-19 onset was estimated to be 5.1 days, and most cases develop symptoms after 14 days. The mean viral clearance times were 24, 30, and 39 days for acute, severe, and ICU-admitted COVID-19 patients, respectively. However, about 25–70% of virus-free COVID-19 survivors continue to sustain virus-induced HMRD and exhibit a wide range of symptoms that are persistent, exacerbated, or new ‘onset’ clinical incidents, collectively termed as post-acute sequelae of COVID-19 (PASC) or long COVID. PASC patients experience several debilitating clinical condition(s) with >200 different and overlapping symptoms that may last for weeks to months. Chronic PASC is a cumulative outcome of at least 10 different HMRD-related pathophysiological mechanisms involving both virus-derived virulence factors and a multitude of innate host responses. Based on HMRD and virus-free clinical impairments of different human organs/systems, PASC patients can be categorized into 4 different clusters or sub-phenotypes: sub-phenotype-1 (33.8%) with cardiac and renal manifestations; sub-phenotype-2 (32.8%) with respiratory, sleep and anxiety disorders; sub-phenotype-3 (23.4%) with skeleto-muscular and nervous disorders; and sub-phenotype-4 (10.1%) with digestive and pulmonary dysfunctions. This narrative review elucidates the effects of viral hijack on host cellular machinery during SARS-CoV-2 infection, ensuing detrimental effect(s) of virus-induced HMRD on human metabolism, consequential symptomatic clinical implications, and damage to multiple organ systems; as well as chronic pathophysiological sequelae in virus-free PASC patients. We have also provided a few evidence-based, human randomized controlled trial (RCT)-tested, precision nutrients to reset HMRD for health recovery of PASC patients.
Article
Full-text available
Emerging clinical data from the current COVID-19 pandemic suggests that ~ 40% of COVID-19 patients develop neurological symptoms attributed to viral encephalitis while in COVID long haulers chronic neuro-inflammation and neuronal damage result in a syndrome described as Neuro-COVID. We hypothesize that SAR-COV2 induces mitochondrial dysfunction and activation of the mitochondrial-dependent intrinsic apoptotic pathway, resulting in microglial and neuronal apoptosis. The goal of our study was to determine the effect of SARS-COV2 on mitochondrial biogenesis and to monitor cell apoptosis in human microglia non-invasively in real time using Raman spectroscopy, providing a unique spatio-temporal information on mitochondrial function in live cells. We treated human microglia with SARS-COV2 spike protein and examined the levels of cytokines and reactive oxygen species (ROS) production, determined the effect of SARS-COV2 on mitochondrial biogenesis and examined the changes in molecular composition of phospholipids. Our results show that SARS- COV2 spike protein increases the levels of pro-inflammatory cytokines and ROS production, increases apoptosis and increases the oxygen consumption rate (OCR) in microglial cells. Increases in OCR are indicative of increased ROS production and oxidative stress suggesting that SARS-COV2 induced cell death. Raman spectroscopy yielded significant differences in phospholipids such as Phosphatidylinositol (PI), phosphatidylserine (PS), phosphatidylethanolamine (PE) and phosphatidylcholine (PC), which account for ~ 80% of mitochondrial membrane lipids between SARS-COV2 treated and untreated microglial cells. These data provide important mechanistic insights into SARS-COV2 induced mitochondrial dysfunction which underlies neuropathology associated with Neuro-COVID. Graphical Abstract
Article
Full-text available
A coronavirus (CoV) commonly known as SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) and causing COVID-19 (coronavirus disease of 2019) has become a pandemic following an outbreak in Wuhan. Although mutations in the SARS-CoV-2 spike glycoprotein (SGP) are obvious from comparative genome studies, the novel infectious nature of the virus, its new varients detected in the UK, and outside and recovery–death ratios of COVID-19 inspired us to review the mechanisms of the infection, replication, release, and transmission of progeny virions and the immune response in the host cell. In addition to the specificity of SARS-CoV-2 binding to angiotensin-converting enzyme 2 receptor and transmembrane protease serine 2, the varied symptoms and severity of the infection by the original and mutated forms of the virus suggest the significance of correlating the host innate and adaptive immunity with the binding of the virus to the mannose receptor via lipopolysaccharides (LPSs), toll-like receptors via LPS/proteins/RNA, and sialic acid (Sia) via hemagglutinin, or sugar-acid segments of glycans. HA-to-Sia binding is considered based on the innate Sia N-acetylneuraminic acid and the acquired Sia N-glycolylneuraminic acid in the epithelial cells and the sialidase/neuraminidase- or esterase-hydrolyzed release and transmission of CoVs. Furthermore, the cytokine storms common to aged humans infected with SARS-CoV-2 and aged macaques infected with SARS-CoV encourage us to articulate the mechanism by which the nuclear capsid protein and RNAs bypass the pattern recognition-induced secretion of interferons (IFNs), which stimulate IFN genes through the Janus-activated kinase-signal transducer and activator of a transcription pathway, leading to the secretion of antiviral proteins such as myxovirus resistance protein A/B. By considering the complexities of the structure, and the infectious nature of the virus and the structures and functions of the molecules involved in CoV infection, replication, and immune response, a new interface among virology, immunology, chemistry, imaging technology, drug delivery, and nanoscience is proposed and will be developed. This interface can be an essential platform for researchers, technologists, and physicians to collaborate and develop vaccines and medicines against COVID-19 and other pandemics in the future. Scientists in Japan have reviewed how nanoscience is helping us understand infection with SARS-CoV-2, the virus responsible for Covid-19, and the immune response it produces. The coronavirus pandemic has driven international scientific collaboration to identify treatments and develop a vaccine, not only between virologists and immunologists but also with researchers from a broad range of other disciplines including chemists, physicists and materials scientists. Vasudevanpillai Biju from Hokkaido University, Sapporo, and colleagues have reviewed the ongoing research at the interface of infectious diseases, biological chemistry and nanoscience aimed at answering key questions on how the virus functions. The authors summarize the use of nanomaterials in imaging techniques, vaccine development and drug delivery, while investigating problems associated with the toxicity of nanomaterials. Understanding these molecular interactions will help to fight this and future pandemics. Despite the hopeful signs of progress of COVID-19 vaccine development and vaccination, the highly infectious nature and mutations of SARS-CoV-2 are warnings of an infighting annual revival of the virus. This article clarifies the complexities of COVID-19 by referring to the molecular-level mechanisms of the infection, immune response, replication, and transmission of SARS-CoV-2, which are essential during the development of an effective vaccine or a drug to fight the pandemic. Furthermore, this article underscores the significance of an interface among chemistry, nanoscience, cell biology, immunology, and virology to resolve the challenges of COVID-19.
Article
Full-text available
Infiltrating gliomas are devastating and incurable tumors. Amongst all gliomas, those harboring a mutation in isocitrate dehydrogenase 1 mutation (IDH1mut) acquire a different tumor biology and clinical manifestation from those that are IDH1WT. Understanding the unique metabolic profile reprogrammed by IDH1 mutation has the potential to identify new molecular targets for glioma therapy. Herein, we uncover increased monounsaturated fatty acids (MUFA) and their phospholipids in endoplasmic reticulum (ER), generated by IDH1 mutation, that are responsible for Golgi and ER dilation. We demonstrate a direct link between the IDH1 mutation and this organelle morphology via D-2HG-induced stearyl-CoA desaturase (SCD) overexpression, the rate-limiting enzyme in MUFA biosynthesis. Inhibition of IDH1 mutation or SCD silencing restores ER and Golgi morphology, while D-2HG and oleic acid induces morphological defects in these organelles. Moreover, addition of oleic acid, which tilts the balance towards elevated levels of MUFA, produces IDH1mut-specific cellular apoptosis. Collectively, these results suggest that IDH1mut-induced SCD overexpression can rearrange the distribution of lipids in the organelles of glioma cells, providing new insight into the link between lipid metabolism and organelle morphology in these cells, with potential and unique therapeutic implications. The understanding of altered lipid metabolism by isocitrate dehydrogenase 1 (IDH1) mutations in gliomas at a compartment-specific level is limited. Here, the authors use Raman spectroscopy to monitor organelle-specific metabolic changes and report that IDH1 mutations induce phospholipid imbalances which lead to ER and Golgi dilation.
Article
Full-text available
The phenomenon of oxidative stress, characterized as an imbalance in the production of reactive oxygen species and antioxidant responses, is a well-known inflammatory mechanism and constitutes an important cellular process. The relationship of viral infections, reactive species production, oxidative stress, and the antiviral response is relevant. Therefore, the aim of this review is to report studies showing how reactive oxygen species may positively or negatively affect the pathophysiology of viral infection. We focus on known respiratory viral infections, especially severe acute respiratory syndrome coronaviruses (SARS-CoVs), in an attempt to provide important information on the challenges posed by the current COVID-19 pandemic. Because antiviral therapies for severe acute respiratory syndrome coronaviruses (e.g., SARS-CoV-2) are rare, knowledge about relevant antioxidant compounds and oxidative pathways may be important for understanding viral pathogenesis and identifying possible therapeutic targets.
Article
Full-text available
Due to ongoing pandemic around the world, the mechanisms underlying the SARS-CoV-2 induced COVID-19 are subject to intense investigation. Based on available data for the SARS-CoV-1 virus, we suggest how CoV-2 localization of RNA transcripts in mitochondria hijacks the host cell's mitochondrial function to viral advantage. Besides viral RNA transcripts, RNA also localizes to mitochondria. SARS-CoV-2 may manipulate mitochondrial function indirectly, first by ACE2 regulation of mitochondrial function, and once it enters the host cell, ORFS such as ORF-9b can directly manipulate mitochondrial function to evade host cell immunity and facilitate virus replication and COVID-19 disease. Manipulations of host mitochondria by viral ORFs can release mitochondrial DNA (mtDNA) in the cytoplasm and activate mtDNA induced inflammasome and suppress innate and adaptive immunity. We argue that a decline in ACE2 function in aged individuals, coupled with the age-associated decline in mitochondrial functions resulting in chronic metabolic disorders like diabetes or cancer, may make the host more vulnerable to infection and health complications to mortality. These observations suggest that distinct localization of viral RNA and proteins in mitochondria must play essential roles in SARS-CoV-2 pathogenesis. Understanding the mechanisms underlying virus communication with host mitochondria may provide critical insights into COVID-19 pathologies. An investigation into the SARS-CoV-2 hijacking of mitochondria should lead to novel approaches to prevent and treat COVID-19.
Article
Full-text available
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is a novel coronavirus that has caused a worldwide pandemic of the human respiratory illness COVID-19, resulting in a severe threat to public health and safety. Analysis of the genetic tree suggests that SARS-CoV-2 belongs to the same Betacoronavirus group as severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV). Although the route for viral transmission remains a mystery, SARS-CoV-2 may have originated in an animal reservoir, likely that of bat. The clinical features of COVID-19, such as fever, cough, shortness of breath, and fatigue, are similar to those of many acute respiratory infections. There is currently no specific treatment for COVID-19, but antiviral therapy combined with supportive care is the main strategy. Here, we summarize recent progress in understanding the epidemiological, virological, and clinical characteristics of COVID-19 and discuss potential targets with existing drugs for the treatment of this emerging zoonotic disease.
Article
Research in fundamental cell biology and pathology could be revolutionized by developing the capacity for quantitative molecular analysis of subcellular structures. To that end, we introduce the Ramanomics platform, based on confocal Raman microspectrometry coupled to a biomolecular component analysis algorithm, which together enable us to molecularly profile single organelles in a live-cell environment. This emerging omics approach categorizes the entire molecular makeup of a sample into about a dozen of general classes and subclasses of biomolecules and quantifies their amounts in submicrometer volumes. A major contribution of our study is an attempt to bridge Raman spectrometry with big-data analysis in order to identify complex patterns of biomolecules in a single cellular organelle and leverage discovery of disease biomarkers. Our data reveal significant variations in organellar composition between different cell lines. We also demonstrate the merits of Ramanomics for identifying diseased cells by using prostate cancer as an example. We report large-scale molecular transformations in the mitochondria, Golgi apparatus, and endoplasmic reticulum that accompany the development of prostate cancer. Based on these findings, we propose that Ramanomics datasets in distinct organelles constitute signatures of cellular metabolism in healthy and diseased states.
Article
The COVID-19 pandemic caused by the coronavirus (SARS-CoV-2) has taken the world by surprise into a major crisis of overwhelming morbidity and mortality. This highly infectious disease is associated with respiratory failure unusual in other coronavirus infections. Mounting evidence link the accelerated progression of the disease in COVID-19 patients to the hyper-inflammatory state termed as the “cytokine storm” involving major systemic perturbations. These include iron dysregulation manifested as hyperferritinemia associated with disease severity. Iron dysregulation induces reactive oxygen species (ROS) production and promotes oxidative stress. The mitochondria are the hub of cellular oxidative homeostasis. In addition, the mitochondria may circulate “cell-free” in non-nucleated platelets, in extracellular vesicles and mitochondrial DNA is found in the extracellular space. The heightened inflammatory/oxidative state may lead to mitochondrial dysfunction leading to platelet damage and apoptosis. The interaction of dysfunctional platelets with coagulation cascades aggravates clotting events and thrombus formation. Furthermore, mitochondrial oxidative stress may contribute to microbiota dysbiosis, altering coagulation pathways and fueling the inflammatory/oxidative response leading to the vicious cycle of events. Here, we discuss various cellular and systemic incidents caused by SARS-CoV-2 that may critically impact intra and extracellular mitochondrial function, and contribute to the progression and severity of the disease. It is crucial to understand how these key modulators impact COVID-19 pathogenesis in the quest to identify novel therapeutic targets that may reduce fatal outcomes of the disease.
Article
The recent emergence of the novel, pathogenic SARS-coronavirus 2 (SARS-CoV-2) in China and its rapid national and international spread pose a global health emergency. Cell entry of coronaviruses depends on binding of the viral spike (S) proteins to cellular receptors and on S protein priming by host cell proteases. Unravelling which cellular factors are used by SARS-CoV-2 for entry might provide insights into viral transmission and reveal therapeutic targets. Here, we demonstrate that SARS-CoV-2 uses the SARS-CoV receptor ACE2 for entry and the serine protease TMPRSS2 for S protein priming. A TMPRSS2 inhibitor approved for clinical use blocked entry and might constitute a treatment option. Finally, we show that the sera from convalescent SARS patients cross-neutralized SARS-2-S-driven entry. Our results reveal important commonalities between SARS-CoV-2 and SARS-CoV infection and identify a potential target for antiviral intervention.