ArticlePDF Available

SARS-CoV-2 Infection, Inflammation, Immunonutrition, and Pathogenesis of COVID-19

Authors:
  • Institute of Integrative Omics and Applied Biotechnology (IIOAB)-India and UFMG-Brazil

Abstract

The COVID-19 pandemic, caused by the coronavirus, SARS-CoV-2, has claimed millions of lives worldwide in the past two years. Fatalities among the elderly with underlying cardiovascular disease, lung disease, and diabetes have particularly been high. A bibliometrics analysis on author's keywords was carried out, and searched for possible links between various coronavirus studies over the past 50 years, and integrated them. We found keywords like immune system, immunity, nutrition, malnutrition, micronutrients, exercise, inflammation, and hyperinflammation were highly related to each other. Based on these findings, we hypothesized that the human immune system is a multilevel super complex system, which employs multiple strategies to contain microorganism infections and restore homeostasis. It was also found that the behavior of the immune system is not able to be described by a single immunological theory. However, one main strategy is "self-destroy and rebuild", which consists of a series of inflammatory responses: 1) active self-destruction of damaged/dysfunctional somatic cells; 2) removal of debris and cells; 3) rebuilding tissues. Thus, invading microorganisms' clearance could be only a passive bystander response to this destroy-rebuild process. Microbial infections could be self-limiting and promoted as an indispensable essential nutrition for the vast number of genes existing in the microorganisms. The transient nutrition surge resulting from the degradation of the self-destroyed cell debris coupled with the existing nutrition state in the patient may play an important role in the pathogenesis of COVID-19. Finally, a few possible coping strategies to mitigate COVID-19, including vaccination, are also discussed.
Send Orders for Reprints to reprints@benthamscience.ae
Current Medicinal Chemistry, 2023, 1
0929-8673/19 $95.00+.00 © 2023 Bentham Science Publishers
ARTICLE TYPE: Review
Title: SARS-CoV-2 Infection, Inflammation, Immunonutrition, and Pathogenesis of COVID-19
Ligen Yu *a, Mohd Khanapi Abd Ghani b, Alessio Aghemo c, Debmalya Barh d, Matteo Bassetti e, Fausto Catena f, Gaetano
Gallo g, Ali Gholamrezanezhad h, Mohammad Amjad Kamal i, Amos Lal j, Kamal Kant Sahu k, Shailendra K Saxena l, Ugo
Elmore m, Farid Rahimi n, Chiara Robba o, Yuanlin Song p, Zhengyuan Xia q, Boxuan Yu r
a Talent Recruitment and Career Support (TRACS) Office, Nanyang Technological University, N2.1 B4-01, 76 Nanyang Drive, Singapore
637331
b Biomedical Computing and Engineering Technologies (BIOCORE) Applied Research Group, Faculty of Information and
Communication Technology, Universiti Teknikal Malaysia Melaka, Durian Tunggal 76100 Melaka, Malaysia
c Humanitas Research Hospital IRCCS, Rozzano, Italy
d Institute of Integrative Omics and Applied Biotechnology (IIOAB), Nonakuri, Purba Medinipur, WB-721172, India; and Departamento
de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901,
Brazil
e Infectious Diseases Clinic, Department of Health Sciences, University of Genoa and Policlinico San Martino Hospital – IRCCS, Genoa,
Italy
f Azienda Ospedaliero - Universitaria di Parma, Parma, Italy
g University of Catanzaro, Catanzaro, Italy
h Keck School of Medicine, University of Southern California (USC), Los Angeles, California, USA
i 7 Peterlee Place, Hebersham, NSW 2770, Australia
j Department of Medicine, Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA 55902
k Huntsman Cancer Centre, University of Utah, USA
l Center for Advanced Research, King George's Medical University (KGMU), Lucknow - 226003, India
m Department of Gastrointestinal Surgery, IRCCS San Raffaele Scientific Institute and San Raffaele Vita-Salute University, Milan, Italy
n Division of Biomedical Science and Biochemistry, Research School of Biology, The Australian National University, Ngunnawal and
Ngambri Country, Canberra, ACT 2600, Australia
o Anesthesia and Intensive Care, Policlinico San Martino, Largo Rosanna Benzi 15, 16100 Genova, Italy
p Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, P.R.China, 200032
q Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
r School of Computer Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798
*Address correspondence to LG Yu at the Talent Recruitment and Career Support (TRACS) Office, Nanyang Technological University, N2.1
B4-01, 76 Nanyang Drive, Singapore 637331, Singapore; Tel: ++65-6592-3248; E-mail: mlgyu@ntu.edu.sg
Abstract: The COVID-19 pandemic, caused by the coronavirus, SARS-CoV-2, has claimed millions
of lives worldwide in the past two years. Fatalities among the elderly with underlying cardiovascular
disease, lung disease, and diabetes have particularly been high. A bibliometrics analysis on author’s
keywords was carried out, and searched for possible links between various coronavirus studies over
2 Current Medicinal Chemistry, 2022, Vol. 0, No. 0 Yu et al.
the past 50 years, and integrated them. We found keywords like immune system, immunity, nutrition,
malnutrition, micronutrients, exercise, inflammation, and hyperinflammation were highly related to
each other. Based on these findings, we hypothesized that the human immune system is a multilevel
super complex system, which employs multiple strategies to contain microorganism infections and
restore homeostasis. It was also found that the behavior of the immune system is not able to be
described by a single immunological theory. However, one main strategy is “self-destroy and rebuild”,
which consists of a series of inflammatory responses: 1) active self-destruction of
damaged/dysfunctional somatic cells; 2) removal of debris and cells; 3) rebuilding tissues. Thus,
invading microorganisms’ clearance could be only a passive bystander response to this destroy–rebuild
process. Microbial infections could be self-limiting and promoted as an indispensable essential
nutrition for the vast number of genes existing in the microorganisms. The transient nutrition surge
resulting from the degradation of the self-destroyed cell debris coupled with the existing nutrition state
in the patient may play an important role in the pathogenesis of COVID-19. Finally, a few possible
coping strategies to mitigate COVID-19, including vaccination, are also discussed.
CONSENT FOR PUBLICATION
Not applicable.
FUNDING
None.
CONFLICT OF INTEREST
Dr. Ligen Yu is the Section Editor of the Journal of CMC.
Acknowledgements
The authors are grateful to Dr. Melvin Hayden (MD, Professor at the University of Missouri, USA), Dr Jamie
Cunliffe (www.morphostasis.org.uk), Dr. Marcello Candelli (Catholic University of Sacred Heart of Rome,
Italy), Dr. Danilo Buonsenso (Fondazione Policlinico Universitario A, Italy), Dr. Susanna Esposito,
(University Hospital of Parma, Italy), and Dr. Sushil Kumar Middha (Maharani Lakshmi Ammanni College
For Women, India) for reading and providing insightful comments to this manuscript.
SARS-CoV-2 Infection, Inflammation, Immunonutrition, and Pathogenesis of COVID-19 Current Medicinal Chemistry, 2023, Vol. 0, No. 0 3
REFERENCES
[1] Cousins-Frankel, J. A cancer survivor had the longest documented COVID-19 infection. Here’s
what scientists learned. Science News October 19, 2021.
http://dx.doi.org/10.1126/science.acx9383; https://www.science.org/content/article/cancer-survivor-
had-longest-documented-covid-19-infection-here-s-what-scientists-learned (Accessed Dec 01, 2021)
[2] Nussenblatt, V.; Order, A.E.; Das, S.; de Wit, E.; Youn, J.H.; Banakis, S.; Mushegian, A.; Mederos,
C.; Wang, W.; Chung, M.; Pérez-Pérez, L.; Palmore, T.; Brudno, J.N.; Kochenderfer, J.N.; Ghedin,
E. Yearlong COVID-19 Infection Reveals Within-Host Evolution of SARS-CoV-2 in a Patient With
B-Cell Depletion. J. Infect. Dis., 2022, 225(7), 1118-1123.
http://dx.doi.org/10.1093/infdis/jiab622 PMID: 34940844
[3] Zhu, C.C.; Zhu, J. The effect of self-limiting on the prevention and control of the diffuse COVID-19
epidemic with delayed and temporal-spatial heterogeneous. BMC Infect. Dis., 2021, 21, 1145.
http://dx.doi.org/10.1186/s12879-021-06670-y PMID: 34753451
[4] Narain, J.P.; Dawa, N.; Bhatia, R. Health system response to COVID-19 and future pandemics. J.
Health Manag., 2020, 22(2), 138-145.
http://dx.doi.org/10.1177/0972063420935538
[5] Pieniawska-Śmiech, K.; Kuraszewicz, A.; Sado, J.; Śmiech, K.; Lewandowicz-Uszyńska, A.
Assessment of COVID-19 Incidence and the Ability to Synthesise Anti-SARS-CoV-2 Antibodies of
Paediatric Patients with Primary Immunodeficiency. J. Clin. Med., 2021, 10(21), 5111.
http://dx.doi.org/10.3390/jcm10215111 PMID: 34768630
[6] Bansal, N.; Ovchinsky, N.; Foca, M.; Lamour, J.M.; Kogan-Liberman, D.; Hsu, D.T.; Beddows, K.;
Abraham, L.; Coburn, M.; Cunningham, R.; Nguyen, T.; Hayde, N. COVID-19 infection in pediatric
solid organ transplant patients. Pediatr Transplant., 2021, 11, 14156.
http://dx.doi.org/10.1111/petr.14156 PMID: 34633125
[7] Levin, B.R.; Baquero, F.; Ankomah, P.; McCall, I.C. Phagocytes, Antibiotics, and Self-Limiting
Bacterial Infections . Trends Microbiol., 2017, 25(11), 878-892.
http://dx.doi.org/10.1016/j.tim.2017.07.005 PMID: 28843668
[8] Levin, B.R.; Antia, R. Why we don't get sick: The within-host population dynamics of bacterial
infections. Science, 2001, 292, 1112-1115.
http://dx.doi.org/10.1126/science.1058879 PMID: 11352067
[9] Choi, B.; Choudhary, M.C.; Regan, J.; Sparks, J.A.; Padera, R.F.; Qiu, X.; Solomon, I.H.; Kuo,
H.H.; Boucau, J.; Bowman, K.; Adhikari, U.D.; Winkler, M.L.; Mueller, A.A.; Hsu, T.Y.;
4 Current Medicinal Chemistry, 2022, Vol. 0, No. 0 Yu et al.
Desjardins, M.; Baden, L.R.; Chan, B.T.; Walker, B.D.; Lichterfeld, M.; Brigl, M.; Kwon, D.S.;
Kanjilal, S.; Richardson, E.T.; Jonsson, A.H.; Alter, G.; Barczak, A.K.; Hanage, W.P.; Yu, X.G.;
Gaiha, G.D.; Seaman, M.S.; Cernadas, M.; Li J.Z. Persistence and Evolution of SARS-CoV-2 in an
Immunocompromised Host. N. Engl. J. Med., 2020, 383(23), 2291-2293.
http://dx.doi.org/10.1056/NEJMc2031364 PMID: 33176080
[10] Wu, Z.; McGoogan, J.M. Characteristics of and Important Lessons From the Coronavirus Disease
2019 (COVID-19) Outbreak in China: Summary of a Report of 72 314 Cases From the Chinese
Center for Disease Control and Prevention. JAMA, 2020, 323(13), 1239-1242.
http://dx.doi.org/10.1001/jama.2020.2648 PMID: 32091533
[11] Merad, M.; Martin, J.C. Pathological inflammation in patients with COVID-19: a key role for
monocytes and macrophages. Nat. Rev. Immunol., 2020, 20, 355–362.
http://dx.doi.org/10.1038/s41577-020-0331-4 PMID: 32376901
[12] Cao, X. COVID-19: immunopathology and its implications for therapy. Nat. Rev. Immunol., 2020,
20, 269–270.
http://dx.doi.org/10.1038/s41577-020-0308-3 PMID: 32273594
[13] van Eijk, L.E.; Binkhorst, M.; Bourgonje, A.R.; Offringa, A.K.; Mulder, D.J.; Bos, E.M.; Kolundzic,
N.; Abdulle, A.E.; van der Voort, P.H.; Olde Rikkert, M.G.; van der Hoeven, J.G.; den Dunnen,
W.F.; Hillebrands, J.L.; van Goor, H. COVID-19: immunopathology, pathophysiological
mechanisms, and treatment options. J. Pathol., 2021, 254(4), 307-331.
http://dx.doi.org/10.1002/path.5642 PMID: 33586189
[14] Fox, S.E.; Akmatbekov, A.; Harbert, J.L.; Li, G.; Brown, J.Q.; Vander Heide, R.S. Pulmonary and
cardiac pathology in African American patients with COVID-19: an autopsy series from New
Orleans. Lancet Respir. Med., 2020, 8, 681-686.
http://dx.doi.org/10.1016/S2213-2600(20)30243-5 PMID: 32473124
[15] Prasad, A.; Prasad, M. Single Virus Targeting Multiple Organs: What We Know and Where We Are
Heading? Front. Med., 2020, 7, 370.
http://dx.doi.org/10.3389/fmed.2020.00370 PMID: 32850890
[16] Raza, A.; Estepa, A.; Chan, V.; Jafar, M.S. Acute Renal Failure in Critically Ill COVID19 Patients
with a Focus on the Role of Renal Replacement Therapy: A Review of What We Know So Far.
Cureus, 2020, 12, 8429.
http://dx.doi.org/10.7759/cureus.8429 PMID: 32642345
SARS-CoV-2 Infection, Inflammation, Immunonutrition, and Pathogenesis of COVID-19 Current Medicinal Chemistry, 2023, Vol. 0, No. 0 5
[17] Su, H.; Yang, M.; Wan, C.; Yi, L.X.; Tang, F.; Zhu, H.Y.; Yi, F.; Yang, H.C.; Fogo, A.B.; Nie, X.;
Zhang, C. Renal histopathological analysis of 26 postmortem findings of patients with COVID-19 in
China. Kidney Int., 2020, 98, 219–227.
http://dx.doi.org/10.1016/j.kint.2020.04.003 PMID: 32327202
[18] Ritter, A.; Kreis, N.N.; Louwen, F.; Yuan, J.P. Obesity and COVID-19: Molecular Mechanisms
Linking Both Pandemics. Int. J. Mol. Sci., 2020, 21, 5793.
http://dx.doi.org/10.3390/ijms21165793 PMID: 32806722
[19] Mauvais-Jarvis, F. Aging, Male Sex, Obesity, and Metabolic Inflammation Create the Perfect Storm
for COVID-19. Diabetes, 2020, 69, 1857-1863.
http://dx.doi.org/10.2337/dbi19-0023 PMID: 32669390
[20] Petrakis, D.; Margina, D.; Tsarouhas, K.; Tekos, F.; Stan, M.; Nikitovic, D.; Kouretas, D.;
Spandidos, D.A.; Tsatsakis, A. Obesity-a risk factor for increased COVID-19 prevalence, severity
and lethality. Mol. Med. Rep., 2020, 22, 9-19.
http://dx.doi.org/10.3892/mmr.2020.11127 PMID: 32377709
[21] Flaherty, G.T.; Hession, P.; Liew, C.H.; Lim, B.C.W.; Leong, T.K.; Lim, V.; Sulaiman, L.H.
COVID-19 in adult patients with pre-existing chronic cardiac, respiratory and metabolic disease: a
critical literature review with clinical recommendations. Trop. Dis. Travel Med. Vaccines, 2020, 6,
16.
http://dx.doi.org/10.1186/s40794-020-00118-y PMID: 32868984
[22] Yoshikawa, N.; Yoshikawa, T.; Hill, T.; Huang, C.; Watts, D.M.; Makino, S.; Milligan, G.; Chan,
T.; Peters, C.J. & Tseng, C.T. Differential virological and immunological outcome of severe acute
respiratory syndrome coronavirus infection in susceptible and resistant transgenic mice expressing
human angiotensin-converting enzyme 2. J. virol., 2009, 83(11), 5451–5465.
http://dx.doi.org/10.1128/JVI.02272-08 PMID: 19297479
[23] Tseng, C.T.; Perrone, L.A.; Zhu, H.; Makino, S.; Peters, C.J. Severe acute respiratory syndrome and
the innate immune responses: modulation of effector cell function without productive infection. J.
Immunol., 2005, 174(12), 7977-85.
http://dx.doi.org/10.4049/jimmunol.174.12.7977 PMID: 15944304
[24] Chivukula, R.R.; Maley, J.H.; Dudzinski, D.M.; Hibbert, K.; Hardin, C.C. Evidence-Based
Management of the Critically Ill Adult With SARS-CoV-2 Infection. J. Intensive Care Med., 2021,
36(1), 18-41.
http://dx.doi.org/10.1177/0885066620969132 PMID: 33111601
6 Current Medicinal Chemistry, 2022, Vol. 0, No. 0 Yu et al.
[25] Wilson, J.G.; Simpson, L.J.; Ferreira, A.M.; Rustagi, A.; Roque, J.; Asuni, A.; Ranganath, T.; Grant,
P.M.; Subramanian, A.; Rosenberg-Hasson, Y.; Maecker, H.T.; Holmes, S.P.; Levitt, J.E.; Blish,
C.A.; Rogers, A.J. Cytokine profile in plasma of severe COVID-19 does not differ from ARDS and
sepsis. JCI Insight, 2020, 5(17), 140289.
http://dx.doi.org/10.1172/jci.insight.140289 PMID: 32706339
[26] El Zowalaty, M.E.; Järhult, J.D. From SARS to COVID-19: A previously unknown SARS- related
coronavirus (SARS-CoV-2) of pandemic potential infecting humans - Call for a One Health
approach. One Health, 2020, 9, 100124. http://dx.doi.org/10.1016/j.onehlt.2020.100124. PMID:
32195311; PMCID: PMC7075990.
[27] Jorwal, P.; Bharadwaj, S.; Jorwal, P. One health approach and COVID-19: A perspective. J. Family
Med. Prim. Care, 2020, 9(12), 5888-5891. http://dx.doi.org/10.4103/jfmpc.jfmpc_1058_20
PMID: 33681013
[28] Li, Q.; Bergquist, R.; Grant, L.; Song, J.X.; Feng, X.Y.; Zhou, X.N. Consideration of COVID-19
beyond the human-centred approach of prevention and control: the ONE-HEALTH perspective.
Emerg Microbes Infect. 2022, 11(1), 2520-2528. http://dx.doi.org/10.1080/22221751.2022.2125343
PMID: 36102336
[29] van Eck, N.J.; Waltman, L. Software survey: VOSviewer, a computer program for bibliometric
mapping. Scientometrics, 2010, 84(2), 523-538.
http://dx.doi.org/10.1007/s11192-009-0146-3 PMID: 20585380
[30] Gupta, A.; Gupta, R.; Singh, R.L. Microbes and Environment. In: Principles and Applications of
Environmental Biotechnology for a Sustainable Future. Applied Environmental Science and
Engineering for a Sustainable Future. Singh, R. Ed., Springer, Singapore, 2017, 43-84.
https://doi.org/10.1007/978-981-10-1866-4_3
[31] Millen, D.D.; De Beni Arrigoni, M. and Lauritano Pacheco, R.D. Rumenology. Cham, Switzerland:
Springer International Publishing, 2016. ISBN 978-3-319-30531-8 ISBN 978-3-319-30533-2
(eBook).
http://dx.doi.org/10.1007/978-3-319-30533-2
[32] Cammack, K.M.; Austin, K.J.; Lamberson, W.R.; Conant, G.C.; Cunningham, H.C. Ruminant
Nutrition Symposium: Tiny but mighty: the role of the rumen microbes in livestock production. J.
Anim. Sci., 2018, 96(2), 752-770.
http://dx.doi.org/10.1093/jas/skx053 PMID: 29385535
SARS-CoV-2 Infection, Inflammation, Immunonutrition, and Pathogenesis of COVID-19 Current Medicinal Chemistry, 2023, Vol. 0, No. 0 7
[33] Storm, E.; Orskov, E.R.; Smart, R. The Nutritive-Value of Rumen Microorganisms in Ruminants .2.
The Apparent Digestibility and Net Utilization of Microbial-N for Growing Lambs. Br. J. Nutr.,
1983, 50, 471-478.
http://dx.doi.org/10.1079/BJN19830115 PMID: 6615775
[34] Hackmann, T.J.; Firkins, J.L. Maximizing efficiency of rumen microbial protein production. Front.
Microbiol., 2015, 6, 465.
http://dx.doi.org/10.3389/fmicb.2015.00465 PMID: 26029197
[35] Gilbert, S.F.; Sapp, J; Tauber, A.I. A Symbiotic View of Life: We Have Never Been Individuals. Q.
Rev. Biol., 2012, 87(4): 325-341.
http://dx.doi.org/10.1086/668166 PMID: 23397797
[36] Alexander, K.L.; Targan, S.R.; Elson, C.O. Microbiota activation and regulation of innate and
adaptive immunity. Immunol. Rev., 2014, 260, 206-220.
http://dx.doi.org/10.1111/imr.12180 PMID: 24942691
[37] Georgountzou, A.; Papadopoulos, N.G. Postnatal innate immune Development: From Birth to
Adulthood. Front. Immunol., 2017, 8, 957.
http://dx.doi.org/10.3389/fimmu.2017.00957 PMID: 28848557
[38] Kloc, M.; Ghobrial, R.M.; Kuchar, E.; Lewicki, S.; Kubiak, J.Z. Development of child immunity in
the context of COVID-19 pandemic. Clin. Immunol., 2020, 217, 108510.
http://dx.doi.org/10.1016/j.clim.2020.108510 PMID: 32544611
[39] Backhed, F.; Ley, R.E.; Sonnenburg, J.L.; Peterson, D.A.; Gordon, J.I. Host-bacterial mutualism in
the human intestine. Science, 2005, 307, 1915-1920.
http://dx.doi.org/10.1126/science.1104816 PMID: 15790844
[40] McFall-Ngai, M.; Hadfield, M.G.; Bosch, T.C.G.; Carey, H.V.; Domazet-Lošo, T.; Douglas, A.E.;
Dubilier, N.; Eberl, G.; Fukami, T.; Gilbert, S.F.; Hentschel, U.; King, N.; Kjelleberg, S.; Knoll,
A.H.; Kremer, N.; Mazmanian, S.K.; Metcalf, J.L.; Nealson, K.; Pierce, N.E.; Rawls, J.F.; Reid, A.;
Ruby, E.G.; Rumpho, M.; Sanders, J.G.; Tautz, D.; Wernegreen, J.J. Animals in a bacterial world, a
new imperative for the life sciences. Proc. Natl. Acad. Sci. USA, 2013, 110, 3229-3236.
http://dx.doi.org/10.1073/pnas.1218525110 PMID: 23391737
[41] Cho, I.; Blaser, M.J. The human microbiome: at the interface of health and disease. Nat. Rev. Genet.,
2012, 13(4), 260-270.
http://dx.doi.org/10.1038/nrg3182 PMID: 22411464
[42] Lahiri, S.; Kim, H.J.; Garcia-Perez, I.; Reza, M.M.; Martin, K.A.; Kundu, P.; Cox, L.M.; Selkrig, J.;
Posma, J.M.; Zhang, H.; Padmanabhan, P.; Moret, C.; Gulyás, B.; Blaser, M.J.; Auwerx, J.; Holmes,
8 Current Medicinal Chemistry, 2022, Vol. 0, No. 0 Yu et al.
E.; Nicholson, J.; Wahli, W.; Pettersson, S. The gut microbiota influences skeletal muscle mass and
function in mice. Sci. Transl. Med., 2019, 11, eaan5662.
http://dx.doi.org/10.1126/scitranslmed.aan5662 PMID: 31341063
[43] Reza, M.M.; Finlay, B.B.; Pettersson, S. Gut microbes, ageing & organ function: a chameleon in
modern biology? EMBO Mol. Med., 2019, 11, 9872.
http://dx.doi.org/10.15252/emmm.201809872 PMID: 31410991
[44] Gupta, V.; Kumar, R.; Sood, U.; Singhvi, N. Reconciling Hygiene and Cleanliness: A New
Perspective from Human Microbiome. Indian J. Microbiol., 2020, 60, 37-44.
http://dx.doi.org/10.1007/s12088-019-00839-5 PMID: 32089572
[45] Singhvi, N.; Gupta, V.; Gaur, M.; Sharma, V.; Puri, A.; Singh, Y.; Pandey, G.D.; Lal, R. Interplay of
Human Gut Microbiome in Health and Wellness. Indian J. Microbiol., 2020, 60, 26–36.
http://dx.doi.org/10.1007/s12088-019-00825-x PMID: 32089571
[46] Zhao, L.P.; Zhang, F.; Ding, X.Y.; Wu, G.; Lam, Y.Y.; Wang, X.; Fu, H.; Xue, X.; Lu, C.; Ma, J.;
Yu, L.; Xu, C.; Ren, Z.; Xu, Y.; Xu, S.; Shen, H.; Zhu, X.; Shi, Y.; Shen, Q.; Dong, W.; Liu, R.;
Ling, Y.; Zeng, Y.; Wang, X.; Zhang, Q.; Wang, J.; Wang, L.; Wu, Y.; Zeng, B.; Wei, H.; Zhang,
M.; Peng, Y.; Zhang, C. Gut bacteria selectively promoted by dietary fibers alleviate type 2 diabetes.
Science, 2018, 359, 1151-1156.
http://dx.doi.org/10.1126/science.aao5774 PMID: 29590046
[47] Schuijt, T.J.; Lankelma, J.M.; Scicluna, B.P.; de Sousa e Melo, F.; Roelofs, J.J.; de Boer, J.D.;
Hoogendijk, A.J.; de Beer, R.; de Vos, A.; Belzer, C.; de Vos, W.M.; van der Poll, T.; Wiersinga,
W.J. The gut microbiota plays a protective role in the host defence against pneumococcal
pneumonia. Gut, 2016, 65, 575-583.
http://dx.doi.org/10.1136/gutjnl-2015-309728 PMID: 26511795
[48] Biagi, E.; Franceschi, C.; Rampelli, S.; Severgnini, M.; Ostan, R.; Turroni, S.; Consolandi, C.;
Quercia, S.; Scurti, M.; Monti, D.; Capri, M.; Brigidi, P.; Candela, M. Gut Microbiota and Extreme
Longevity. Curr. Biol., 2016, 26, 1480-1485.
http://dx.doi.org/10.1016/j.cub.2016.04.016 PMID: 27185560
[49] Candela, M.; Biagi, E.; Brigidi, P.; O'Toole, P.W.; De Vos, W.M. Maintenance of a healthy
trajectory of the intestinal microbiome during aging: A dietary approach. Mech. Ageing Dev., 2016,
136, 75-70 .
http://dx.doi.org/10.1016/j.mad.2013.12.004 PMID: 24373997
[50] Claesson, M.J.; Jeffery, I.B.; Conde, S.; Power, S.E.; O'Connor, E.M.; Cusack, S.; Harris, H.M.;
Coakley, M.; Lakshminarayanan, B.; O'Sullivan, O.; Fitzgerald, G.F.; Deane, J.; O'Connor, M.;
SARS-CoV-2 Infection, Inflammation, Immunonutrition, and Pathogenesis of COVID-19 Current Medicinal Chemistry, 2023, Vol. 0, No. 0 9
Harnedy, N.; O'Connor, K.; O'Mahony, D.; van Sinderen, D.; Wallace, M.; Brennan, L.; Stanton, C.;
Marchesi, J.R.; Fitzgerald, A.P.; Shanahan, F.; Hill, C.; Ross, R.P.; O'Toole, P.W. Gut microbiota
composition correlates with diet and health in the elderly. Nature, 2012, 488, 184-178 .
http://dx.doi.org/10.1038/nature11319 PMID: 22797518
[51] Yu, B.X.; Yu, B.W.; Yu, L.G. Commentary: Reconciling Hygiene and Cleanliness: A New
Perspective from Human Microbiome. Indian J. Microbiol., 2020, 60, 259–261.
http://dx.doi.org/10.1007/s12088-020-00863-w PMID: 32255860
[52] Yu, L.G. Restoring Good Health in Elderly with Diverse Gut Microbiome and Food Intake
Restriction to Combat COVID-19. Indian J. Microbiol, , 2021, 61, 104–107.
http://dx.doi.org/10.1007/s12088-020-00913-3 PMID: 33424043
[53] Kumar, R.; Sood, U.; Gupta, V.; Singh, M.; Scaria, J.; Lal, R. Recent Advancements in the
Development of Modern Probiotics for Restoring Human Gut Microbiome Dysbiosis. Indian J.
Microbiol., 2020, 60, 12–25.
http://dx.doi.org/10.1007/s12088-019-00808-y PMID: 32089570
[54] Rath, S.; Rud, T.; Karch, A.; Pieper, D.H.; Vital, M. Pathogenic functions of host microbiota.
Microbiome, 2018, 6, 174.
http://dx.doi.org/10.1186/s40168-018-0542-0 PMID: 30266099
[55] Martens, E.C.; Neumann, M.; Desai, M.S. Interactions of commensal and pathogenic
microorganisms with the intestinal mucosal barrier. Nat. Rev. Microbiol., 2018, 16(8), 457-470.
http://dx.doi.org/10.1038/s41579-018-0036-x PMID: 29904082
[56] Hornef, M. Pathogens, Commensal Symbionts, and Pathobionts: Discovery and Functional Effects
on the Host. ILAR J., 2015, 56(2), 159-162.
http://dx.doi.org/10.1093/ilar/ilv007 PMID: 26323625
[57] Proença, J.T.; Barral, D.C. & Gordo, I. Commensal-to-pathogen transition: One-single transposon
insertion results in two pathoadaptive traits in Escherichia coli -macrophage interaction. Sci. Rep.,
2017, 7, 4504.
http://dx.doi.org/10.1038/s41598-017-04081-1 PMID: 28674418
[58] Yu, B.X.; Yu, L.G. & Klionsky, D.J. Nutrition Acquisition by Human Immunity, Transient
Overnutrition and the Cytokine Storm in Severe Cases of COVID-19. Med. Hypotheses, 2021, 155,
110668.
http://dx.doi.org/10.1016/j.mehy.2021.110668 PMID: 34467856
[59] Dickson, R.P.; Martinez, F.J.; Huffnagle, G.B. The role of the microbiome in exacerbations of
chronic lung diseases. Lancet, 2014, 384, 702-691 .
10 Current Medicinal Chemistry, 2022, Vol. 0, No. 0 Yu et al.
http://dx.doi.org/10.1016/S0140-6736(14)61136-3 PMID: 25152271
[60] Sokol, H.; Seksik, P. The intestinal microbiota in inflammatory bowel diseases: time to connect with
the host. Curr. Opin. Gastroenterol., 2010, 26, 331-327 .
http://dx.doi.org/10.1097/MOG.0b013e328339536b PMID: 20445446
[61] Zuo, T.; Zhang, F.; Lui, G.C.Y.; Yeoh, Y.K.; Li, A.Y.L.; Zhan, H.; Wan, Y.; Chung, A.C.K.;
Cheung, C.P.; Chen, N.; Lai, C.K.C.; Chen, Z.; Tso, E.Y.K.; Fung, K.S.C.; Chan, V.; Ling, L.;
Joynt, G.; Hui, D.S.C.; Chan, F.K.L.; Chan, P.K.S.; Ng, S.C. Alterations in Gut Microbiota of
Patients With COVID-19 During Time of Hospitalization. Gastroenterology, 2020, 159, 944-955.e8.
http://dx.doi.org/10.1053/j.gastro.2020.05.048 PMID: 32442562
[62] Dhar, D.; Mohanty, A. Gut microbiota and Covid-19-possible link and implications. Virus Res.,
2020, 285, 198018.
http://dx.doi.org/10.1016/j.virusres.2020.198018 PMID: 32430279
[63] van der Lelie, D.; Taghavi, S. COVID-19 and the Gut Microbiome: More than a Gut Feeling.
mSystems, 2020, 5, 00453-20.
http://dx.doi.org/10.1128/mSystems.00453-20 PMID: 32694127
[64] Kalantar-Zadeh, K.; Ward, S.A.; Kalantar-Zadeh, K.; El-Omar, E.M. Considering the Effects of
Microbiome and Diet on SARS-CoV-2 Infection: Nanotechnology Roles. ACS Nano, 2020, 14,
5179-5182.
http://dx.doi.org/10.1021/acsnano.0c03402 PMID: 32356654
[65] Metcalf, J.L.; Xu, Z.Z.; Weiss, S.; Lax, S.; Van Treuren, W.; Hyde, E.R.; Song, S.J.; Amir, A.;
Larsen, P.; Sangwan, N.; Haarmann, D.; Humphrey, G.C.; Ackermann, G.; Thompson, L.R.; Lauber,
C.; Bibat, A.; Nicholas, C.; Gebert, M.J.; Petrosino, J.F.; Reed, S.C.; Gilbert, J.A.; Lynne, A.M.;
Bucheli, S.R.; Carter, D.O.; Knight, R. Microbial community assembly and metabolic function
during mammalian corpse decomposition. Science, 2016, 351(6269), 158-162.
http://dx.doi.org/10.1126/science.aad2646 PMID: 26657285
[66] Eberl, G. A new vision of immunity: homeostasis of the superorganism. Mucosal Immunol., 2010, 3,
450–460
http://dx.doi.org/10.1038/mi.2010.20 PMID: 20445502
[67] Ricklin, D.; Hajishengallis, G.; Yang, K.; Lambris, J.D. Complement: a key system for immune
surveillance and homeostasis. Nat. Immunol., 2010, 11(9), 785–797.
http://dx.doi.org/10.1038/ni.1923 PMID: 20720586
[68] Davies, L.C.; Jenkins, S.J.; Allen, J.E.; Taylor, P.R. Tissue-resident macrophages. Nat. Immunol.,
2013, 14(10), 986-95.
SARS-CoV-2 Infection, Inflammation, Immunonutrition, and Pathogenesis of COVID-19 Current Medicinal Chemistry, 2023, Vol. 0, No. 0 11
http://dx.doi.org/10.1038/ni.2705 PMID: 24048120
[69] Desgeorges, T.; Caratti, G.; Mounier, R.; Tuckermann, J.; Chazaud, B. Glucocorticoids shape
macrophage phenotype for tissue repair. Front. Immunol., 2019, 10, 1591.
http://dx.doi.org/10.3389/fimmu.2019.01591 PMID: 31354730
[70] Davis, L.E.; Oyer, R.; Beckham, J.D.; Tyler, K.L. Elevated CSF Cytokines in the Jarisch-
Herxheimer Reaction of General Paresis. JAMA Neurol., 2013, 70(8), 1060-1064.
http://dx.doi.org/10.1001/jamaneurol.2013.2120 PMID: 23732875
[71] Matzinger, P. Tolerance, danger, and the extended family. Annu. Rev. Immunol., 1994, 12, 991–1045
http://dx.doi.org/10.1146/annurev.iy.12.040194.005015 PMID: 8011301
[72] Cunliffe, J. A proliferation of pathogens through the 20th century. Scand. J. Immunol., 2008, 68(2),
120-128.
http://dx.doi.org/10.1111/j.1365-3083.2008.02130.x PMID: 18544150
[73] Cunliffe, J. Intentional pathogen killing - or denial of substrate? Scand. J. Immunol., 2007, 66(6),
604-609.
http://dx.doi.org/10.1111/j.1365-3083.2007.02017.x PMID: 17949408
[74] Cunliffe, J. Tissue homeostasis and immunity - More on models. Scand. J. Immunol., 2006, 64(3),
172-176.
http://dx.doi.org/10.1111/j.1365-3083.2006.01814.x PMID: 16918683
[75] Cunliffe, J. From terra firma to terra plana - danger is shaking the foundations: deconstructing the
'immune system'. Med. Hypotheses, 1999, 52(3), 213-219.
http://dx.doi.org/10.1054/mehy.1997.0645 PMID: 10362280
[76] Cunliffe, J. Morphostasis: an evolving perspective. Med. Hypotheses, 1997, 49(6), 449-459.
http://dx.doi.org/10.1016/S0306-9877(97)90062-1 PMID: 9466367
[77] Cunliffe, J. Morphostasis and Immunity. Med. Hypotheses, 1995, 44(2), 89-96.
http://dx.doi.org/10.1016/0306-9877(95)90076-4 PMID: 7596312
[78] Pradeu, T.; Jaeger, S.; Vivier, E. The speed of change: towards a discontinuity theory of immunity?
Nat. Rev. Immunol., 2013, 13, 764–769.
http://dx.doi.org/10.1038/nri3521 PMID: 23995627
[79] Pradeu, T. and Vivier, E. The discontinuity theory of immunity. Sci. Immunol., 2016, 1, aag0479
http://dx.doi.org/10.1126/sciimmunol.aag0479 PMID: 28239677
[80] Eberl, G.; Pradeu, T. Towards a General Theory of Immunity? Trends Immunol., 2018, 39(4), 261-
263.
http://dx.doi.org/10.1016/j.it.2017.11.004 PMID: 29229264
12 Current Medicinal Chemistry, 2022, Vol. 0, No. 0 Yu et al.
[81] Oltz, E.M. Regulation of antigen receptor gene assembly in lymphocytes. Immunol. Res., 2001,
23(2-3), 121-33.
http://dx.doi.org/10.1385/IR:23:2-3:121 PMID: 11444378
[82] Thomas, L.R.; Cobb, R.M.; Oltz, E.M. Dynamic regulation of antigen receptor gene assembly. Adv.
Exp. Med. Biol., 2009, 650, 103-15.
http://dx.doi.org/10.1007/978-1-4419-0296-2_9 PMID: 19731805
[83] Calder, P.C.; Kew, S. The immune system: a target for functional foods? Br. J. Nutr., 2002, 88 Suppl
2, S165-77.
http://dx.doi.org/10.1079/BJN2002682 PMID: 12495459
[84] Martinez, F.O.; Sica, A.; Mantovani, A.; Locati, M. Macrophage Activation and Polarization. Front.
Biosci., 2008, 13, 453–61.
http://dx.doi.org/10.2741/2692 PMID: 17981560
[85] Mantovani, A.; Sica, A.; Sozzani, S.; Allavena, P.; Vecchi, A.; Locati, M. The chemokine system in
diverse forms of macrophage activation and polarization. Trends Immunol., 2004, 25, 677- 686.
http://dx.doi.org/10.1016/j.it.2004.09.015 PMID: 15530839
[86] Shi, F.D.; Ljunggren, H.G. & Sarvetnick, N. Innate immunity and autoimmunity: from self-
protection to self-destruction. Trends Immunol., 2001, 22(2), 97–101.
http://dx.doi.org/10.1016/S1471-4906(00)01821-4 PMID: 11286711
[87] Viorritto, I.C.B.; Nikolov, N.P.; Siegel, R.M. Autoimmunity versus tolerance: can dying cells tip the
balance? Clin. Immunol., 2007, 122, 125-134.
http://dx.doi.org/10.1016/j.clim.2006.07.012 PMID: 17029966
[88] Hartl, W.H. Metabolic Self-Destruction in Critically Ill Patients (Part I): Origins, Mechanisms and
Biologic Sense. Aktuel. Ernahrungsmed., 2016, 41(1), 40-44.
http://dx.doi.org/10.1055/s-0041-111343
[89] Hartl, W.H. Metabolic Self-Destruction in Critically Ill Patients (Part II): The Importance of Modern
Medical Care and Therapeutic Consequences. Aktuel. Ernahrungsmed., 2016, 41(2), 113-117.
http://dx.doi.org/10.1055/s-0042-102160
[90] Wildbaum, G.; Nahir, M.A.; Karin, N. Beneficial autoimmunity to proinflammatory mediators
restrains the consequences of self-destructive immunity. Immunity, 2003, 19, 679-688.
http://dx.doi.org/10.1016/S1074-7613(03)00291-7 PMID: 14614855
[91] Sender R and Milo, R. The distribution of cellular turnover in the human body. Nat. Med, , 2021, 27,
45-48.
http://dx.doi.org/10.1038/s41591-020-01182-9 PMID: 33432173
SARS-CoV-2 Infection, Inflammation, Immunonutrition, and Pathogenesis of COVID-19 Current Medicinal Chemistry, 2023, Vol. 0, No. 0 13
[92] Han, C.Z. and Ravichandran, K.S. Metabolic connections during apoptotic cell engulfment. Cell,
2011, 147, 1442-1445.
http://dx.doi.org/10.1016/j.cell.2011.12.006 PMID: 22196723
[93] Singh, R.; Letai, A.; Sarosiek, K. Regulation of apoptosis in health and disease: The balancing act of
BCL-2 family proteins. Nat. Rev. Mol. Cell Biol., 2019, 20, 175–193.
http://dx.doi.org/10.1038/s41580-018-0089-8 PMID: 30655609
[94] Ravichandran, K.S.; Lorenz, U. Engulfment of apoptotic cells: signals for a good meal. Nat. Rev.
Immunol., 2007, 7, 964–974.
http://dx.doi.org/10.1038/nri2214 PMID: 18037898
[95] Henson, P.M.; Hume, D.A. Apoptotic cell removal in development and tissue homeostasis. Trends
Immunol., 2006, 27, 244–250.
http://dx.doi.org/10.1016/j.it.2006.03.005 PMID: 16584921
[96] Jorgensen, I.; Rayamajhi, M.; Miao, E.A. Programmed cell death as a defence against infection. Nat.
Rev. Immunol., 2017, 17(3), 151-64.
http://dx.doi.org/10.1038/nri.2016.147 PMID: 28138137
[97] Kanduc, D.; Mittelman, A.; Serpico, R.; Kanduc, D.; Mittelman, A.; Serpico, R.; Sinigaglia, E.;
Sinha, A.A.; Natale, C.; Santacroce, R.; Di Corcia, M.G.; Lucchese, A.; Dini, L.; Pani, P.;
Santacroce, S.; Simone, S.; Bucci, R.; Farber, E. Cell death: Apoptosis versus necrosis (review). Int.
J. Oncol., 2002, 21, 165-170.
http://dx.doi.org/10.3892/ijo.21.1.165 PMID: 12063564
[98] Broderick, N.A. A common origin for immunity and digestion. Front. Immunol., 2015, 6, 72.
http://dx.doi.org/10.3389/fimmu.2015.00072 PMID: 25745424
[99] Seeberg, J.C.; Loibl, M.; Moser, F.; Schwegler, M.; Büttner-Herold, M.; Daniel, C.; Engel, F.B.;
Hartmann, A.; Schlötzer-Schrehardt, U.; Goppelt-Struebe, M.V.; Schellerer, V.; Naschberger, E.;
Ganzleben, I.; Heinzerling, L.; Fietkau, R.; Distel, L.V. Non-professional phagocytosis: A general
feature of normal tissue cells. Sci. Rep., 2019, 9, 11875.
http://dx.doi.org/10.1038/s41598-019-48370-3 PMID: 31417141
[100] Schwegler, M.; Wirsing, A.M.; Dollinger, A.J.; Abendroth, B.; Putz, F.; Fietkau, R.; Distel, L.V.
Clearance of primary necrotic cells by non-professional phagocytes. Biol. Cell, 2015, 107(10), 372–
87.
http://dx.doi.org/10.1111/boc.201400090 PMID: 26032600
[101] Arandjelovic, S.; Ravichandran, K.S. Phagocytosis of apoptotic cells in homeostasis. Nat. Immunol.,
2015, 16, 907–917.
14 Current Medicinal Chemistry, 2022, Vol. 0, No. 0 Yu et al.
http://dx.doi.org/10.1038/ni.3253 PMID: 26287597
[102] Green, D.; Oguin T & Martinez, J. The clearance of dying cells: table for two. Cell Death Differ.,
2016, 23, 915–926.
http://dx.doi.org/10.1038/cdd.2015.172 PMID: 26990661
[103] Klionsky, D.J.; Abdel-Aziz, A.K.; Abdelfatah, S.; Abdellatif, M.; Abdoli, A.; Abel, S.; Abeliovich,
H.; Abildgaard, M.H.; Abudu, Y.P.; Acevedo-Arozena, A.; Adamopoulos, I.E.; Adeli, K.; Adolph,
T.E.; Adornetto, A.; Aflaki, E.; Agam, G.; Agarwal, A.; Aggarwal, B.B.; Agnello, M.; Agostinis, P.
and 2909 more. Guidelines for the use and interpretation of assays for monitoring autophagy (4th
edition), Autophagy, 2021, 17(1), 1-382.
http://dx.doi.org/10.1080/15548627.2020.1797280 PMID: 33634751
[104] van Niekerk, G.; Loos, B.; Nell, T.; Engelbrecht, A.M. Autophagy - a free meal in sickness-
associated anorexia. Autophagy, 2016, 12, 727–734.
http://dx.doi.org/10.1080/15548627.2016.1147672 PMID: 27050464
[105] Haq, S.; Grondin, J.; Banskota, S.; Khan, W.I. Autophagy: roles in intestinal mucosal homeostasis
and inflammation. J. Biomed. Science, 2019, 19, 26.
http://dx.doi.org/10.1186/s12929-019-0512-2 PMID: 30764829
[106] Deretic, V.; Levine, B. Autophagy, Immunity, and Microbial Adaptations. Cell Host Microbe, 2009,
5, 527-549.
http://dx.doi.org/10.1016/j.chom.2009.05.016 PMID: 19527881
[107] Benjamin, J.L.; Sumpter, R. Jr.; Levine, B.; Hooper LV. Intestinal Epithelial Autophagy Is Essential
for Host Defense against Invasive Bacteria. Cell Host Microbe, 2013, 13, 734-723 .
http://dx.doi.org/10.1016/j.chom.2013.05.004 PMID: 23768496
[108] Cuervo, A.M.; Macian, F. Autophagy, nutrition and immunology. Mol. Aspects Med., 2012, 33: 13-2 .
http://dx.doi.org/10.1016/j.mam.2011.09.001 PMID: 21982744
[109] Singh, R.; Cuervo, A.M. Autophagy in the cellular energetic balance. Cell Metab., 2011, 13, 495–
504.
http://dx.doi.org/10.1016/j.cmet.2011.04.004 PMID: 21531332
[110] Huett, A.; Goel, G.; Xavier, R.J. A systems biology viewpoint on autophagy in health and disease.
Curr Opin Gastroenterol., 2010, 26, 309-302 .
http://dx.doi.org/10.1097/MOG.0b013e32833ae2ed PMID: 20571384
[111] Kuma, A.; Hatano, M.; Matsui, M.; Yamamoto, A.; Nakaya, H.; Yoshimori, T.; Ohsumi, Y.;
Tokuhisa, T.; Mizushima, N. The role of autophagy during the early neonatal starvation period.
Nature, 2004, 432, 1032-1036.
SARS-CoV-2 Infection, Inflammation, Immunonutrition, and Pathogenesis of COVID-19 Current Medicinal Chemistry, 2023, Vol. 0, No. 0 15
http://dx.doi.org/10.1038/nature03029 PMID: 15525940
[112] Kheloufi, M.; Boulanger, C.M.; Durand, F.; Rautou, P.E. Liver Autophagy in Anorexia Nervosa and
Acute Liver Injury. Biomed. Res. Int., 2014, 2014, 701064.
http://dx.doi.org/10.1155/2014/701064 PMID: 25250330
[113] Zhi, X.Y.; Feng, W.Z.; Rong, Y.G.; Liu, R. Anatomy of autophagy: from the beginning to the end.
Cell Mol. Life Sci., 2018, 75, 815-831.
http://dx.doi.org/10.1007/s00018-017-2657-z PMID: 28939950
[114] Eskelinen, E.L.; Saftig, P. Autophagy: A lysosomal degradation pathway with a central role in health
and disease. Biochim. Biophys. Acta, 2009, 1793, 664-673.
http://dx.doi.org/10.1016/j.bbamcr.2008.07.014 PMID: 18706940
[115] Levine, B.; Kroemer, G. Autophagy in the pathogenesis of disease. Cell, 2008, 132, 27-42.
http://dx.doi.org/10.1016/j.cell.2007.12.018 PMID: 18191218
[116] Kuballa, P.; Nolte, W.M.; Castoreno, A.B.; Xavier, R.J. Autophagy and the Immune System. Ann.
Rev. Immunol., 2012, 30, 611-646.
http://dx.doi.org/10.1146/annurev-immunol-020711-074948 PMID: 22449030
[117] Jo, E.K.; Yuk, J.M.; Shin, D.M.; Sasakawa, C. Roles of autophagy in elimination of intracellular
bacterial pathogens. Front. Immunol., 2013, 4, 97.
http://dx.doi.org/10.3389/fimmu.2013.00097 PMID: 23653625
[118] Gomes, L.C.; Dikic, I. Autophagy in Antimicrobial Immunity. Mol. Cell, 2014, 54, 224-33.
http://dx.doi.org/10.1016/j.molcel.2014.03.009 PMID: 24766886
[119] Randall-Demllo, S.; Chieppa, M.; Eri, R. Intestinal epithelium and autophagy: Partners in gut
homeostasis. Front. Immunol., 2013, 4, 301.
http://dx.doi.org/10.3389/fimmu.2013.00301 PMID: 24137160
[120] Kabat, A.M.; Pott, J.; Maloy, K.J. The mucosal immune system and its regulation by autophagy.
Front. Immunol., 2016, 7, 240.
http://dx.doi.org/10.3389/fimmu.2016.00240 PMID: 27446072
[121] Ghartey-Kwansah, G; Adu-Nti, F; Aboagye, B; Ankobil, A; Essuman, E.E; Opoku, Y.K; Abokyi, S;
Abu, E.K; Boampong, J.N. Autophagy in the control and pathogenesis of parasitic infections. Cell
Biosci., 2020, 10(1), 101.
http://dx.doi.org/.10.1186/s13578-020-00464-6 PMID: 32944216
[122] Bergsbaken, T.; Fink, S.L.; Cookson, B.T. Pyroptosis: host cell death and inflammation. Nat. Rev.
Microbiol., 2009, 7(2), 99–109.
http://dx.doi.org/10.1038/nrmicro2070 PMID: 19148178
16 Current Medicinal Chemistry, 2022, Vol. 0, No. 0 Yu et al.
[123] Yu, P.; Zhang, X.; Liu, N.; Tang, L.; Peng, C.; Chen, X. Pyroptosis: mechanisms and diseases. Sig.
Transduct. Target. Ther., 2021, 6, 128.
http://dx.doi.org/10.1038/s41392-021-00507-5 PMID: 33776057
[124] Wimmer, K.; Sachet, M.; Oehler, R. Circulating biomarkers of cell death. Clin. Chim. Acta, 2020,
500, 87–97.
http://dx.doi.org/10.1016/j.cca.2019.10.003 PMID: 31655053
[125] Mitteldorf, J. How evolutionary thinking affects people's ideas about aging interventions.
Rejuvenation Res., 2006, 9, 346-350.
http://dx.doi.org/10.1089/rej.2006.9.346 PMID: 16706667
[126] Exton, M.S. Infection-induced anorexia: Active host defence strategy. Appetite, 1997, 29, 369-383.
http://dx.doi.org/10.1006/appe.1997.0116 PMID: 9468766
[127] van Niekerk, G.; Isaacs, A.W.; Nell, T.; Engelbrecht, A.M. Sickness-Associated Anorexia: Mother
Nature's Idea of Immunonutrition? Mediators Inflamm., 2016, 2016, 8071539.
http://dx.doi.org/10.1155/2016/8071539 PMID: 27445441
[128] Nilsson, A. Mechanisms Behind Illness-Induced Anorexia. PhD Thesis, Linköping University
Medical Dissertations, No. 1549. 2016.
https://liu.diva-portal.org/smash/get/diva2:1047669/FULLTEXT01.pdf (Accessed Dec 01 2021)
[129] Garbarino, J.; Sturley, S.L. Saturated with fat: new perspectives on lipotoxicity. Curr. Opin. Clin.
Nutr. Metab. Care, 2009, 12, 110–116.
http://dx.doi.org/10.1097/MCO.0b013e32832182ee PMID: 19202381
[130] Chen, L.; Deng, H.; Cui, H.; Fang, J.; Zuo, Z.; Deng, J.; Li, Y.; Wang, X.; Zhao, L. Inflammatory
Responses and Inflammation-Associated Diseases in Organs. Oncotarget., 2018, 9, 7204-7218.
http://dx.doi.org/10.18632/oncotarget.23208 PMID: 29467962
[131] Medzhitov, R. Inflammation 2010: new adventures of an old flame. Cell, 2010, 140, 771-776.
http://dx.doi.org/10.1016/j.cell.2010.03.006 PMID: 20303867
[132] Ferrero-Miliani, L.; Nielsen, O.; Andersen, P.; Girardin, S. Chronic inflammation: importance of
NOD2 and NALP3 in interleukin-1β generation. Clin. Exp. Immunol., 2007, 147, 227-235.
http://dx.doi.org/10.1111/j.1365-2249.2006.03261.x PMID: 17223962
[133] Costantini, S.; Sharma, A. and Colonna, G. The Value of the Cytokinome Profile. In: Inflammatory
Diseases - A Modern Perspective, Amit Nagal, Ed.; IntechOpen. 2011.
http://dx.doi.org/10.5772/25707
[134] Virtue, S.; Vidal-Puig, A. Adipose tissue expandability, lipotoxicity and the Metabolic Syndrome-an
allostatic perspective. Biochim. Biophys. Acta, 2010, 1801, 338-49.
SARS-CoV-2 Infection, Inflammation, Immunonutrition, and Pathogenesis of COVID-19 Current Medicinal Chemistry, 2023, Vol. 0, No. 0 17
http://dx.doi.org/10.1016/j.bbalip.2009.12.006 PMID: 20056169
[135] Posey, K.A.; Clegg, D.J.; Printz, R.L.; Byun, J.; Morton, G.J.; VivekanandanGiri, A.; Pennathur, S.;
Baskin, D.G.; Heinecke, J.W.; Woods, S.C.; Schwartz, M.W.; Niswender, K.D. Hypothalamic
proinflammatory lipid accumulation, inflammation, and insulin resistance in rats fed a high-fat diet.
Am. J. Physiol. Endocrinol. Metab., 2009, 296, E1003-E1012.
http://dx.doi.org/10.1152/ajpendo.90377.2008 PMID: 19116375
[136] Stahlschmidt, Z.R; Acker, M; Kovalko, I; Adamo. S.A. The double-edged sword of immune defence
and damage control: do food availability and immune challenge alter the balance? Funct. Ecol.,
2015, 29(11), 1445-1452.
http://dx.doi.org/10.1111/1365-2435.12454
[137] van Niekerk, G.; du Toit, A.; Loos, B.; Engelbrecht, A.M. Nutrient excess and autophagic
deficiency: explaining metabolic diseases in obesity. Metabolism, 2018, 82, 14-21.
http://dx.doi.org/10.1016/j.metabol.2017.12.007 PMID: 29289514
[138] Chang, H.R.; Bistrian, B. The role of cytokines in the catabolic consequences of infection and injury.
J. Parenter. Enteral. Nutr., 1998, 22, 156-166.
http://dx.doi.org/10.1177/0148607198022003156 PMID: 9586794
[139] Demling, R.H.; De Santi, L. Effect of a Catabolic State with Involuntary Weight Loss on Acute and
Chronic Respiratory Disease. Medscape, 2002, https://www.medscape.org/viewarticle/432384
(Accessed Jul 18, 2022)
[140] Romijn, J.A. Substrate metabolism in the metabolic response to injury. Proc. Nutr. Soc., 2000, 59,
447-449.
http://dx.doi.org/10.1017/S0029665100000616 PMID: 10997672
[141] Akner, G.; Cederholm, T. Treatment of protein-energy malnutrition in chronic nonmalignant
disorders. Am. J. Clin. Nutr., 2001, 74, 6-24.
http://dx.doi.org/10.1093/ajcn/74.1.6 PMID: 11451713
[142] Brunelli S and Rovere-Querini, P. The immune system and the repair of skeletal muscle. Pharmacol.
Res., 2008, 58, 117–121.
http://dx.doi.org/10.1016/j.phrs.2008.06.008 PMID: 18639637
[143] Berardi, E.; Madaro, L.; Lozanoska-Ochser, B.; Adamo, S.; Thorrez, L.; Bouche, M.; Coletti, D. A
Pound of Flesh: What Cachexia Is and What It Is Not. Diagnostics, 2021, 11, 116.
http://dx.doi.org/10.3390/diagnostics11010116 PMID: 33445790
[144] Arabi, Y.M.; Reintam, B.A.; Preiser, J.C. Less is more in nutrition: critically ill patients are starving
but not hungry. Intensive Care Med., 2019, 45, 1629-1631.
18 Current Medicinal Chemistry, 2022, Vol. 0, No. 0 Yu et al.
http://dx.doi.org/10.1007/s00134-019-05765-0 PMID: 31531714
[145] Omodei, D.; Pucino, V.; Labruna, G.; Procaccini, C.; Galgani, M.; Perna, F.; Pirozzi, D.; De Caprio,
C.; Marone, G.; Fontana, L.; Contaldo, F.; Pasanisi, F.; Matarese, G.; Sacchetti, L. Immune-
metabolic profiling of anorexic patients reveals an anti-oxidant and anti-inflammatory phenotype.
Metabolism, 2015, 64, 396-405.
http://dx.doi.org/10.1016/j.metabol.2014.10.025 PMID: 25500208
[146] Nova, E.; Samartín, S.; Gómez, S.; Morandé, G.; Marcos, A. The adaptive response of the immune
system to the particular malnutrition of eating disorders. Eur. J. Clin. Nutr., 2002, 56 Suppl 3, S34-
S37.
http://dx.doi.org/10.1038/sj.ejcn.1601482 PMID: 12142959
[147] Barrea, L.; Muscogiuri, G.; Frias-Toral, E.; Laudisio, D.; Pugliese, G.; Castellucci, B.; Garcia-
Velasquez, E.; Savastano, S.; Colao, A. Nutrition and immune system: from the Mediterranean diet
to dietary supplementary through the microbiota. Crit. Rev. Food. Sci. Nutr., 2021, 61(18), 3066-
3090.
http://dx.doi.org/10.1080/10408398.2020.1792826 PMID: 32691606
[148] Gombart, A.F.; Pierre, A.; Maggini, S. A Review of Micronutrients and the Immune System-
Working in Harmony to Reduce the Risk of Infection. Nutrients, 2020, 12, 236.
http://dx.doi.org/10.3390/nu12010236 PMID: 31963293
[149] Allen, A.; Snary, D. The structure and function of gastric mucus. Progress report. Gut, 1972, 13,
666-672.
http://dx.doi.org/10.1136/gut.13.8.666 PMID: 4562023
[150] Forstner JF: Intestinal Mucins in Health and Disease. Digestion, 1978, 17, 234-263.
http://dx.doi.org/10.1159/000198115 PMID: 25218
[151] Higashizono, K.; Fukatsu, K.; Watkins, A.; Watanabe, T.; Noguchi, M.; Tominaga, E.; Ri, M.;
Murakoshi, S.; Yasuhara, H.; Seto, Y. Effects of short-term fasting on gut-associated lymphoid
tissue and intestinal morphology in mice. Clin. Nutr. Exp., 2018, 18, 6-14.
http://dx.doi.org/10.1016/j.yclnex.2017.12.002
[152] Papavramidis, T.S.; Kaidoglou, K.; Grosomanidis, V.; Kazamias, P.; Anagnostopoulos, T.H.;
Paramythiotis, D.; Kotzampassi K: Short-term fasting-induced jejunal mucosa atrophy in rats –the
role of probiotics during refeeding. Ann. Gastroenterol., 2009, 22, 268-274.
[153] Grundy, S.M. Adipose tissue and metabolic syndrome: too much, too little or neither. Eur. J. Clin.
Invest., 2015, 45, 1209-1217.
http://dx.doi.org/10.1111/eci.12519 PMID: 26291691
SARS-CoV-2 Infection, Inflammation, Immunonutrition, and Pathogenesis of COVID-19 Current Medicinal Chemistry, 2023, Vol. 0, No. 0 19
[154] Saklayen, M.G. The Global Epidemic of the Metabolic Syndrome. Curr. Hypertens. Rep., 2018, 20,
12.
http://dx.doi.org/10.1007/s11906-018-0812-z PMID: 29480368
[155] Humphries, D.L.; Scott, M.E.; Vermund, S.H. Pathways Linking Nutritional Status and Infectious
Disease: Causal and Conceptual Frameworks. In: Nutrition and Infectious Diseases. Nutrition and
Health. Humphries, D.L.; Scott, M.E.; Vermund, S.H.; Ed., Humana, Cham. 2021.
http://dx.doi.org/10.1007/978-3-030-56913-6_1
[156] Poon, I.K.; Lucas, C.D.; Rossi, A.G.; Ravichandran, K.S. Apoptotic cell clearance: basic biology
and therapeutic potential. Nat. Rev. Immunol., 2014, 14(3), 166-180.
http://dx.doi.org/10.1038/nri3607 PMID: 24481336
[157] Fazeli, G.; Wehman, A.M. Safely removing cell debris with LC3-associated phagocytosis. Biol Cell,
2017, 109, 355-63.
http://dx.doi.org/10.1111/boc.201700028 PMID: 28755428
[158] Roos, W.; Thomas, A.; Kaina, B. DNA damage and the balance between survival and death in
cancer biology. Nat. Rev. Cancer, 2016, 16, 20-33.
http://dx.doi.org/10.1038/nrc.2015.2 PMID: 26678314
[159] Tamang, J.P.; Shin, D.H.; Jung, S.J.; Chae, S.W. Functional Properties of Microorganisms in
Fermented Foods. Front. Microbiol., 2016, 7, 578.
http://dx.doi.org/10.3389/fmicb.2016.00578 PMID: 27199913
[160] Nair, M.R.B.; Chouhan, D.; Gupta, S.S.; Chattopadhyay, S. Fermented Foods: Are They Tasty
Medicines for Helicobacter pylori Associated Peptic Ulcer and Gastric Cancer? Front. Microbiol.,
2016, 7, 1148.
http://dx.doi.org/10.3389/fmicb.2016.01148 PMID: 27504109
[161] Mota de Carvalho, N.; Costa, E.M.; Silva, S.; Pimentel, L.; Fernandes, T.H.; Estevez Pintado, M.
Fermented foods and beverages in human diet and their influence on gut microbiota and health.
Fermentation, 2018, 4, 90.
http://dx.doi.org/10.3390/fermentation4040090
[162] Yu, B.W.; Yu, B.X.; Yu, L.G. Restore gut homeostasis and healthy weight for an anorexia nervosa
Patient by the Luigi Cornaro diet – a case report. Institute of Materials (East Asia), Singapore,
2018. ISBN 978-981-14-0181-7.
https://www.researchgate.net/publication/329915614
[163] van Ommen, B.; Wopereis, S.; van Empelen, P.; van Keulen, H.M.; Otten, W.; Kasteleyn, M.;
Molema, J.J.W.; de Hoogh, I.M.; Chavannes, N.H.; Numans, M.E.; Evers, A.W.M and Pijl, H. From
20 Current Medicinal Chemistry, 2022, Vol. 0, No. 0 Yu et al.
Diabetes Care to Diabetes Cure—The Integration of Systems Biology, eHealth, and Behavioral
Change. Front. Endocrinol., 2018, 8, 381.
http://dx.doi.org/10.3389/fendo.2017.00381 PMID: 29403436
[164] Ahn, A.C.; Tewari, M.; Poon, C.S.; Phillips, R.S. The clinical applications of a systems approach.
PLoS Med., 2006, 3(7), 209.
http://dx.doi.org/10.1371/journal.pmed.0030209 PMID: 16683861
[165] Ahn, A.C.; Tewari, M.; Poon, C.-S.; Phillips, R.S. The Limits of Reductionism in Medicine: Could
Systems Biology Offer an Alternative? PLoS Med., 2006, 3(6), e208.
http://dx.doi.org/10.1371/journal.pmed.0030208 PMID: 16681415
[166] Baxter, A.J.; Coyne, T.; Mcclintock, C. Dietary patterns and metabolic syndrome - a review of
epidemiologic evidence. Asia Pac. J. Clin. Nutr., 2006, 15(2), 134-142. PMID: 16672196
[167] Hayden, M.R. An immediate and long-term complication of COVID-19 may be type 2 diabetes
mellitus: The central role of beta-cell dysfunction, apoptosis and exploration of possible
mechanisms. Cells, 2020, 9(11), 2475.
http://dx.doi.org/10.3390/cells9112475 PMID: 33202960
[168] Rocca, E.; Anjum, R.L. Complexity, Reductionism and the Biomedical Model. In: Rethinking
Causality, Complexity and Evidence for the Unique Patient; Anjum, R., Copeland, S.; Rocca, E.;
Ed., Springer, Cham., 2020.
http://dx.doi.org/10.1007/978-3-030-41239-5_5
[169] DeFronzo, R.A. Insulin resistance, lipotoxicity, type 2 diabetes and atherosclerosis: the missing
links. The Claude Bernard Lecture 2009. Diabetologia, 2010, 53(7), 1270-1287.
http://dx.doi.org/10.1007/s00125-010-1684-1 PMID: 20361178
[170] Tomita, T. Apoptosis in pancreatic beta-islet cells in type 2 diabetes. Bosn. J. Basic Med. Sci. 2016,
16(3), 162-179.
http://dx.doi.org/10.17305/bjbms.2016.919 PMID: 27209071
[171] Sutton, E.F.; Beyl, R.; Early, K.S.; Cefalu, W.T.; Ravussin, E.; Peterson, C.M. Early Time-
Restricted Feeding Improves Insulin Sensitivity, Blood Pressure, and Oxidative Stress Even without
Weight Loss in Men with Prediabetes. Cell Metab., 2018, 27(6), 1212-1221.
http://dx.doi.org/10.1016/j.cmet.2018.04.010 PMID: 29754952
[172] Chaix, A.; Manoogian, E.N.C.; Melkani, G.C.; Panda, S. Time-Restricted Eating to Prevent and
Manage Chronic Metabolic Diseases. Annu. Rev. Nutr. (Book Series, Stover, P.J., Balling, R., Ed.),
2019, 39, 291-315.
http://dx.doi.org/10.1146/annurev-nutr-082018-124320 PMID: 31180809
SARS-CoV-2 Infection, Inflammation, Immunonutrition, and Pathogenesis of COVID-19 Current Medicinal Chemistry, 2023, Vol. 0, No. 0 21
[173] Hutchison, A.T.; Regmi, P.; Manoogian, E.N.C.; Fleischer, J.G.; Wittert, G.A.; Panda, S.;
Heilbronn, L.K. Time-Restricted Feeding Improves Glucose Tolerance in Men at Risk for Type 2
Diabetes: A Randomized Crossover Trial. Obesity, 2019, 27(5), 724-732.
http://dx.doi.org/10.1002/oby.22449 PMID: 31002478
[174] Yang, J.S.; Lu, C.C.; Kuo, S.C.; Hsu, Y.M.; Tsai, S.C.; Chen, S.Y.; Chen, Y.T.; Lin, Y.J.; Huang,
Y.C.; Chen, C.J.; Lin, W.D.; Liao, W.L.; Lin, W.Y.; Liu, Y.H.; Sheu, J.C.; Tsai, F.J. Autophagy and
its link to type II diabetes mellitus. BioMed., 2017, 7(2), 1–12.
http://dx.doi.org/10.1051/bmdcn/2017070201 PMID: 28612706
[175] Dhurandhar, N.V. Infectobesity: Obesity of Infectious Origin. J. Nutr., 2001, 131, 2794S-2797S.
http://dx.doi.org/10.1093/jn/131.10.2794S PMID: 11584109
[176] van Ginneken, V.; Sitnyakowsky, L.; Jeffery, J.E. Infectobesity: Viral infections (especially with
human adenovirus-36: Ad-36) may be a cause of obesity. Med. Hypotheses, 2009, 72, 383-388.
http://dx.doi.org/10.1016/j.mehy.2008.11.034 PMID: 19138827
[177] Na, H.N.; Nam, J.H. Infectobesity: a New Area for Microbiological and Virological Research. J.
Bacteriol. Virol., 2011, 41, 65.
http://dx.doi.org/10.4167/jbv.2011.41.2.65
[178] Valiquette, L.; Sirard, S.; Laupland, K. A microbiological explanation for the obesity pandemic?
Can. J. Infect. Dis. Med. Microbiol., 2014, 25, 294-295.
http://dx.doi.org/10.1155/2014/464162 PMID: 25587289
[179] Patterson, S. The Perils of Germaphobia. Smithsonian, 2013, 44, 8-8.
[180] Vandegrift, R.; Bateman, A.C.; Siemens, K.N.; Nguyen, M.; Wilson, H.E.; Green, J.L.; Van Den
Wymelenberg, K.G.; Hickey, R.J. Cleanliness in context: reconciling hygiene with a modern
microbial perspective. Microbiome, 2017, 5, 76.
http://dx.doi.org/10.1186/s40168-017-0294-2 PMID: 28705228
[181] Han, J.H.; Sullivan, N.; Leas, B.F.; Pegues, D.A.; Kaczmarek, J.L.; Umscheid, C.A. Cleaning
Hospital Room Surfaces to Prevent Health Care-Associated Infections A Technical Brief. Ann.
Intern. Med., 2015, 163, 598-607.
http://dx.doi.org/10.7326/M15-1192 PMID: 26258903
[182] Sherlock, O.; O'Connell, N.; Creamer, E.; Humphreys, H. Is it really clean? An evaluation of the
efficacy of four methods for determining hospital cleanliness. J. Hosp. Infect., 2009, 72, 140-146.
http://dx.doi.org/10.1016/j.jhin.2009.02.013 PMID: 19321226
[183] Ragonnaud, E., Biragyn, A. Gut microbiota as the key controllers of “healthy” aging of elderly
people. Immun. Ageing, 2021, 18(1), 2.
22 Current Medicinal Chemistry, 2022, Vol. 0, No. 0 Yu et al.
https://doi.org/10.1186/s12979-020-00213-w PMID: 33397404
[184] Lu, M.; Zhang, Z.; Xue, M.; Zhao, B.S.; Harder, O.; Li, A.; Liang, X.; Gao, T.Z.; Xu, Y.; Zhou, J.;
Feng, Z.; Niewiesk, S.; Peeples, M.E.; He, C.; Li, J. N6-methyladenosine modification enables viral
RNA to escape recognition by RNA sensor RIG-I. Nat. Microbiol., 2020, 5(4), 584-598.
http://dx.doi.org/10.1038/s41564-019-0653-9 PMID: 32015498
[185] COVIDSurg Collaborative, GlobalSurg Collaborative. SARS-CoV-2 vaccination modelling for safe
surgery to save lives: data from an international prospective cohort study. Br. J. Surg., 2021, 108(9),
1056-1063.
http://dx.doi.org/10.1093/bjs/znab101 PMID: 33761533
[186] Bianco, F.; Ranieri, A.J.; Paterniti, G.; Pata, F.; Gallo, G. Acute intestinal ischemia in a patient with
COVID-19. Tech. Coloproctol., 2020, 24(11), 1217-1218.
http://dx.doi.org/10.1007/s10151-020-02255-0 PMID: 32506344
[187] COVIDSurg Collaborative. Global guidance for surgical care during the COVID-19 pandemic. Br.
J. Surg., 2020, 107(9), 1097-1103.
http://dx.doi.org/10.1002/bjs.11646 PMID: 32293715
[188] Havervall, S.; Rosell, A.; Phillipson, M.; Mangsbo, S.M.; Nilsson, P.; Hober, S.; Thålin, C.
Symptoms and functional impairment assessed 8 months after mild COVID-19 among health care
workers. JAMA, 2021, 325, 2015.
http://dx.doi.org/10.1001/jama.2021.5612 PMID: 33825846
[189] Hayden, M.R. Hypothesis: Neuroglia Activation Due to Increased Peripheral and CNS
Proinflammatory Cytokines/Chemokines with Neuroinflammation May Result in Long COVID.
Neuroglia 2021, 2, 7-35.
https://doi.org/10.3390/neuroglia2010004
[190] Chang, L.; Wei, Y.; Hashimoto, K. Brain-gut-microbiota axis in depression: A historical overview
and future directions. Brain Res. Bull., 2022, 182, 44-56.
http://dx.doi.org/10.1016/j.brainresbull.2022.02.004 PMID: 35151796.
[191] Liu, Q.; Mak, J.W.Y.; Su, Q.; Yeoh, Y.K.; Lui, G.C.; Ng, S.S.S.; Zhang, F.; Li, A.Y.L.; Lu, W.; Hui,
D.S.; Chan, P.K.; Chan, F.K.L.; Ng, S.C. Gut microbiota dynamics in a prospective cohort of
patients with post-acute COVID-19 syndrome. Gut. 2022, 71(3), 544-552.
http://dx.doi.org/10.1136/gutjnl-2021-325989 PMID: 35082169
[192] Wang B, Zhang L, Wang Y, Dai T, Qin Z, Zhou F, Zhang L. Alterations in microbiota of patients
with COVID-19: potential mechanisms and therapeutic interventions. Signal Transduct Target Ther.
2022 Apr 29;7(1):143.
SARS-CoV-2 Infection, Inflammation, Immunonutrition, and Pathogenesis of COVID-19 Current Medicinal Chemistry, 2023, Vol. 0, No. 0 23
http://dx.doi.org/10.1038/s41392-022-00986-0 PMID: 35487886
[193] Panelli, S.; Calcaterra, V.; Verduci, E.; Comandatore, F.; Pelizzo, G.; Borghi, E.; Bandi, C.;
Zuccotti, G. Dysbiosis in Children with Neurological Impairment and Long-Term Enteral Nutrition.
Front Nutr. 2022, 9, 895046.
http://dx.doi.org/10.3389/fnut.2022.895046 PMID: 35811980
[194] Jansen van Vuren, E.; Steyn, S.F.; Brink, C.B.; Möller, M.; Viljoen, F.P.; Harvey, B.H. The
neuropsychiatric manifestations of COVID-19: Interactions with psychiatric illness and
pharmacological treatment. Biomed. Pharmacother. 2021, 135, 111200,
http://dx.doi.org/10.1016/j.biopha.2020.111200 PMID: 33421734
[195] Hayden M.R., Tyagi S.C. Impaired Folate-Mediated One-Carbon Metabolism in Type 2 Diabetes,
Late-Onset Alzheimer’s Disease and Long COVID. Medicina. 2022;58(1), 16;
https://doi.org/10.3390/medicina58010016 PMID: 35056324
... Protein intake amount, renal failure, lymphocyte and AST were independent influencing factors for COVID-19 "repositive" in malnourished older adults. Previous studies have shown that energy consumption and protein breakdown are accelerated in COVID-19 infection patients, 34 resulting in a reduction of available protein in the body, leading to a decrease in the number of functional active immunoglobulins and intestinal-associated lymphoid tissue, hindering the role of intestinal mucosal defense against infection; 35,36 another study 37 found that patients with comorbidities were more likely to have re-positive after discharge, compared with those with normal liver or kidney function, re-positive patients with liver or kidney function impairment had a 2.32-fold higher risk of developing severe cases, [38][39][40] and the tendency of severity and mortality risk increased significantly. Therefore, adequate intake of high-quality protein is essential for antibody production, 41 while for patients with liver or kidney dysfunction, protein intake should be strictly and standardized controlled. ...
Article
Full-text available
Purpose Building and validating a clinical prediction model for novel coronavirus (COVID-19) re-positive cases in malnourished older adults. Patients and Methods Malnourished older adults from January to May 2023 were retrospectively collected from the Department of Geriatrics of the Affiliated Hospital of Chengdu University of Traditional Chinese Medicine. They were divided into a “non-re-positive” group and a “re-positive” group based on the number of COVID-19 infections, and into a training set and a validation set at a 7:3 ratio. The least absolute shrinkage and selection operator (LASSO) regression analysis was used to identify predictive factors for COVID-19 re-positivity in malnourished older adults, and a nomogram was constructed. Independent influencing factors were screened by multivariate logistic regression. The model’s goodness-of-fit, discrimination, calibration, and clinical impact were assessed by Hosmer-Lemeshow test, area under the curve (AUC), calibration curve, decision curve analysis (DCA), and clinical impact curve analysis (CIC), respectively. Results We included 347 cases, 243 in the training set, and 104 in the validation set. We screened 10 variables as factors influencing the outcome. By multivariate logistic regression analysis, preliminary identified protective factors, risk factors, and independent influencing factors that affect the re-positive outcome. We constructed a clinical prediction model for COVID-19 re-positivity in malnourished older adults. The Hosmer-Lemeshow test yielded χ² =5.916, P =0.657; the AUC was 0.881; when the threshold probability was >8%, using this model to predict whether malnourished older adults were re-positive for COVID-19 was more beneficial than implementing intervention programs for all patients; when the threshold was >80%, the positive estimated value was closer to the actual number of cases. Conclusion This model can help identify the risk of COVID-19 re-positivity in malnourished older adults early, facilitate early clinical decision-making and intervention, and have important implications for improving patient outcomes. We also expect more large-scale, multicenter studies to further validate, refine, and update this model.
... COVID-19 is a highly contagious respiratory disease caused by the novel coronavirus SARS-CoV-2 [1][2][3][4][5][6]. The rapid and accurate diagnosis of COVID-19 is crucial for preventing and controlling the pandemic [7][8][9][10]. However, the conventional methods of COVID-19 detection, such as reverse transcription polymerase chain reaction (RT-PCR) and enzyme-linked immunosorbent assay (ELISA), have some limitations, such as high cost, long processing time, complex operation, and low sensitivity [11][12][13][14][15][16][17]. ...
Article
An immediate and precise diagnosis is required due to the COVID-19 outbreak. Labelfree electrochemical biosensors show promise as potentially valuable instruments for detecting COVID-19. These biosensors are distinguished by their lack of complexity, high speed, sensitivity, and relatively low cost. The precise COVID-19 biomarkers may be recognized without labeling or amplification by detecting the electrical signal created by direct contact between the target analyte and the identification element positioned on the electrode surface. This can be done by placing the electrode in contact with the target analyte, which will amplify the signal. It has been shown that using gold screen printed electrodes, also known as Au SPE, is beneficial when used as an electrode material in label-free electrochemical biosensors. This review study examines and contrasts the performance of several label-free electrochemical biosensors that use Au SPE to detect COVID-19. The merits and limitations of each biosensor will also be discussed. These biosensors use recognition components like DNA, RNA, antibody, aptamer, and MIP and depend on various indicators, such as viral RNA, viral protein, and host antibody. In addition, an analysis of the difficulties and possibilities that may present within this burgeoning subject is carried out. This includes the enhancement of sensor selectivity and stability, optimizing sensor manufacture and design, integrating the sensor with portable readout equipment, and validating the sensor's effectiveness via the use of genuine clinical samples. It can be reasoned out that label-free electrochemical biosensors that make use of gold screen-printed electrodes (Au SPE) have a significant amount of potential for the detection of COVID-19. However, further study is required to address various difficulties, improve their dependability, and broaden the range of applications for these technologies.
... Most of the EBV infected adults remain asymptomatic during their lifetime. This is because with the programmed cell death like apoptosis, the human innate immunity performs the "cell self-destruction, debris clearing and cell rebuilding" strategy to clear the infection-damaged cells (18,19). Immune dysfunction such as the dysfunction in dead cell debris clearance and apoptosis-resistance induces chronic inflammation and causing gene mutation and NPC. ...
Article
Full-text available
Background Circulating immune cells are associated with tumor development and poor prognosis in multiple solid tumors. However, the circulating immune-cell profile of nasopharyngeal carcinoma (NPC) remains largely unknown. Therefore, we aimed to determine the changes in immune status and the prognostic significance of circulating immune cells before and after chemoradiotherapy (CRT) in patients, which can provide clinicians with valuable insights to optimize treatment strategies, monitor immune function, and personalize interventions, ultimately improving patient outcomes. Methods Circulating immune cells before and after CRT in 77 patients with NPC and in 30 healthy controls were measured with flow cytometry. A thorough follow-up was conducted to assess prognosis outcomes, including local failure-free rate (LFFR), distant failure-free rate (DFFR), disease-free survival (DFS), and overall survival (OS). The differences of the subpopulation distribution in the two groups were determined by t-tests or Mann-Whitney tests. The paired t-test or Wilcoxon matched-pairs signed rank test was used to compare differences in lymphocyte subsets before and after CRT. The prognostic significance of lymphocyte subsets was evaluated by Kaplan-Meier analysis and Cox proportional hazards model. Results Compared with the control group, the NPC group showed significant decreases in the proportions of CD3⁺ cells, CD4⁺ T cells, CD8⁺CD28⁺ T cells, and CD19⁺ B cells as well as the CD4⁺:CD8⁺ ratio (P<0.05) but a significant increase in the proportion of natural killer (NK) cells (P<0.05). After CRT, the proportions of CD4⁺ cells, CD8⁺CD28⁺ T cells, and CD19⁺ B cells as well as the CD4⁺:CD8⁺ ratio were markedly decreased (P<0.05), while the proportions of CD8⁺ T cells and NK cells were significantly increased (P<0.05). Multivariate analysis showed that a lower percentage of CD19⁺ B cells [hazard ratio (HR) 6.550, 95% CI: 1.661–25.831; P=0.007] and a positive test for Epstein-Barr virus (EBV) DNA (HR 0.261, 95% CI: 0.074–0.926; P=0.038) before treatment independently predicted worse 5-year OS (P<0.05). Conclusions The disproportion of circulating immune cells was observed in patients with NPC before treatment. CRT further aggravated immune dysfunction. Notably, a lower percentage of CD19⁺ B cells and EBV DNA-positive status before treatment were independent predictors of a worse prognosis. Thus, the measurement of circulating immune cells may help elucidate immune function status and predict the outcomes of patients with NPC.
... It is important to recognize that both malnutrition/sarcopenia and hyper-inflammatory state are significantly associated with morbidity and mortality of COVID-19 patients [99] (Figure 1). The effects of immuno-nutrition approaches on COVID-19 patients can be explained by their anti-inflammatory effects, an increase in SCFA production, a consequent improvement in the metabolism of colonocytes, and the re-shaping of healthy gut microflora [100]. ...
Article
Full-text available
Critically ill patients have a hyper-inflammatory response against various offending injuries that can result in tissue damage, organ failure, and fatal prognosis. The origin of this detrimental, uncontrolled inflammatory cascade can be found also within our gut. In detail, one of the main actors is our gut microbiota with its imbalance, namely gut dysbiosis: learning about the microbiota's dysfunction and pathophysiology in the frame of critical patients is of crucial and emerging importance in the management of the systemic inflammatory response syndrome (SIRS) and the multiple organ dysfunction syndrome (MODS). Multiple pieces of evidence indicate that the bacteria that populate our gut efficiently modulate the immune response. Treatment and pretreatment with probiotics have shown promising preliminary results to attenuate systemic inflammation, especially in postoperative infections and ventilation performance. Finally, it is emerging how immunonutrition may exert a possible impact on the health status of patients in intensive care. Thus, this manuscript reviews evidence from the literature on gut microbiota composition, its derangement in critically ill patients, its pathophysiological role, and the described and emerging opportunities arising from its modulation.
... Bibliometric analysis connecting discrete studies in nasopharyngeal carcinoma and predict future research trends pyroptosis (20) and necroptosis (21), our immune system performs the "self-destroy and rebuild" strategy targeting the damaged body cells (22) to actively destroy the damaged body cells, and restoring homeostasis. The apoptotic cells need to be effectively cleared away, a process termed as efferocytosis (23). ...
Article
Full-text available
Commentary on: Xing CY, Lin MQ, Luo WT, et al. The 100 most cited papers in nasopharyngeal carcinoma between 2000 and 2019: a bibliometric study. Transl Cancer Res 2023;12:84
... Immunologists have long been puzzled by the self-destructive nature of the inflammatory response [10]. In our recent article, we proposed the "self-destroy and rebuild" strategy of inflammatory response [11]. Inflammation can be elicited by various harmful stimuli, such as microbial/viral infections, allergic reactions, chemical insults, lipotoxicity and tissue damage [12]. ...
Article
Full-text available
The main purpose of this Special Issue is to provide readers with current understandings of the interactions and causal relations among injury stimuli (including microorganism infections), immune response and overnutrition/lipotoxicity in disease pathogenesis [...]
Article
Full-text available
Most of the new emerging and re-emerging zoonotic virus outbreaks in recent years stem from close interaction with dead or alive infected animals. Since late 2019, the coronavirus disease 2019 (COVID-19) has spread into 221 countries and territories resulting in close to 300 million known infections and 5.4 million deaths in addition to a huge impact on both public health and the world economy. This paper reviews the COVID-19 prevalence in animals, raise concerns about animal welfare and discusses the role of environment in the transmission of COVID-19. Attention is drawn to the One Health concept as it emphasizes the environment in connection with the risk of transmission and establishment of diseases shared between animals and humans. Considering the importance of One Health for an effective response to the dissemination of infections of pandemic character, some unsettled issues with respect to COVID-19 are highlighted.
Article
Full-text available
Severe neurological impairment (NI) is often accompanied by the need for artificial nutritional support, normally provided enterally (enteral nutrition [EN]) to ensure growth, counteract morbidity and mortality, and improve quality of life. On the other hand, long-term EN (LTEN) may contribute to the establishment, or exacerbation, of gastrointestinal disorders that may lead to malnutrition, which in turn is associated with alterations in gut microbiota (GM) composition and functional capacities. To the best of our knowledge, we investigated, for the first time in this study, the consequences of LTEN in a pediatric population in this pathological context. Using amplicon sequencing, we compared the fecal microbiota of a pediatric population suffering from severe NI and under LTEN to that of sex- and age-matched controls. The two groups presented evident differences in GM composition and a consistent differential clustering. In general, the taxonomic picture in NI children under LTEN seemed to mirror a profound dysbiotic condition, in which anti-inflammatory taxa appear severely depleted (among others, the Clostridiales families of Lachnospiraceae and Ruminococcaceae, and, within the latter, Faecalibacterium spp. and Gemmiger spp.), while known pathobionts (Gammaproteobacteria and Klebsiella) or emerging pathogens (e.g., Synergistales, Cloacibacillus, and Fusobacterium) were significantly enriched. Our data suggest that LTEN has a significant impact on the GM taxonomic composition in NI children. Even if other factors are probably at work, such as the bidirectional interaction between gastrointestinal impairment/immaturity and the central nervous system (CNS), the assumption of drugs, and physical inactivity, these data define possible routes and targets to try to alleviate this dysbiosis, with a view to better management of these patients and an improvement in their quality of life.
Article
Full-text available
The global coronavirus disease 2019 (COVID-19) pandemic is currently ongoing. It is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). A high proportion of COVID-19 patients exhibit gastrointestinal manifestations such as diarrhea, nausea, or vomiting. Moreover, the respiratory and gastrointestinal tracts are the primary habitats of human microbiota and targets for SARS-CoV-2 infection as they express angiotensin-converting enzyme-2 (ACE2) and transmembrane protease serine 2 (TMPRSS2) at high levels. There is accumulating evidence that the microbiota are significantly altered in patients with COVID-19 and post-acute COVID-19 syndrome (PACS). Microbiota are powerful immunomodulatory factors in various human diseases, such as diabetes, obesity, cancers, ulcerative colitis, Crohn’s disease, and certain viral infections. In the present review, we explore the associations between host microbiota and COVID-19 in terms of their clinical relevance. Microbiota-derived metabolites or components are the main mediators of microbiota-host interactions that influence host immunity. Hence, we discuss the potential mechanisms by which microbiota-derived metabolites or components modulate the host immune responses to SARS-CoV-2 infection. Finally, we review and discuss a variety of possible microbiota-based prophylaxes and therapies for COVID-19 and PACS, including fecal microbiota transplantation (FMT), probiotics, prebiotics, microbiota-derived metabolites, and engineered symbiotic bacteria. This treatment strategy could modulate host microbiota and mitigate virus-induced inflammation.
Article
Full-text available
Background Long-term complications after COVID-19 are common, but the potential cause for persistent symptoms after viral clearance remains unclear. Objective To investigate whether gut microbiome composition is linked to post-acute COVID-19 syndrome (PACS), defined as at least one persistent symptom 4 weeks after clearance of the SARS-CoV-2 virus. Methods We conducted a prospective study of 106 patients with a spectrum of COVID-19 severity followed up from admission to 6 months and 68 non-COVID-19 controls. We analysed serial faecal microbiome of 258 samples using shotgun metagenomic sequencing, and correlated the results with persistent symptoms at 6 months. Results At 6 months, 76% of patients had PACS and the most common symptoms were fatigue, poor memory and hair loss. Gut microbiota composition at admission was associated with occurrence of PACS. Patients without PACS showed recovered gut microbiome profile at 6 months comparable to that of non-COVID-19 controls. Gut microbiome of patients with PACS were characterised by higher levels of Ruminococcus gnavus , Bacteroides vulgatus and lower levels of Faecalibacterium prausnitzii . Persistent respiratory symptoms were correlated with opportunistic gut pathogens, and neuropsychiatric symptoms and fatigue were correlated with nosocomial gut pathogens, including Clostridium innocuum and Actinomyces naeslundii (all p<0.05). Butyrate-producing bacteria, including Bifidobacterium pseudocatenulatum and Faecalibacterium prausnitzii showed the largest inverse correlations with PACS at 6 months. Conclusion These findings provided observational evidence of compositional alterations of gut microbiome in patients with long-term complications of COVID-19. Further studies should investigate whether microbiota modulation can facilitate timely recovery from post-acute COVID-19 syndrome.
Article
Full-text available
Impaired folate-mediated one-carbon metabolism (FOCM) is associated with many pathologies and developmental abnormalities. FOCM is a metabolic network of interdependent biosynthetic pathways that is known to be compartmentalized in the cytoplasm, mitochondria and nucleus. Currently, the biochemical mechanisms and causal metabolic pathways responsible for the initiation and/or progression of folate-associated pathologies have yet to be fully established. This review specifically examines the role of impaired FOCM in type 2 diabetes mellitus, Alzheimer’s disease and the emerging Long COVID/post-acute sequelae of SARS-CoV-2 (PASC). Importantly, elevated homocysteine may be considered a biomarker for impaired FOCM, which is known to result in increased oxidative–redox stress. Therefore, the incorporation of hyperhomocysteinemia will be discussed in relation to impaired FOCM in each of the previously listed clinical diseases. This review is intended to fill gaps in knowledge associated with these clinical diseases and impaired FOCM. Additionally, some of the therapeutics will be discussed at this early time point in studying impaired FOCM in each of the above clinical disease states. It is hoped that this review will allow the reader to better understand the role of FOCM in the development and treatment of clinical disease states that may be associated with impaired FOCM and how to restore a more normal functional role for FOCM through improved nutrition and/or restoring the essential water-soluble B vitamins through oral supplementation
Article
Full-text available
Background The global spread of the novel coronavirus pneumonia is still continuing, and a new round of more serious outbreaks has even begun in some countries. In this context, this paper studies the dynamics of a type of delayed reaction-diffusion novel coronavirus pneumonia model with relapse and self-limiting treatment in a temporal-spatial heterogeneous environment. Methods First, focus on the self-limiting characteristics of COVID-19, incorporate the relapse and self-limiting treatment factors into the diffusion model, and study the influence of self-limiting treatment on the diffusion of the epidemic. Second, because the traditional Lyapunov stability method is difficult to determine the spread of the epidemic with relapse and self-limiting treatment, we introduce a completely different method, relying on the existence conditions of the exponential attractor of our newly established in the infinite-dimensional dynamic system to determine the diffusion of novel coronavirus pneumonia. Third, relapse and self-limiting treatment have led to a change in the structure of the delayed diffusion COVID-19 model, and the traditional basic reproduction number $$R_0$$ R 0 no longer has threshold characteristics. With the help of the Krein-Rutman theorem and the eigenvalue method, we studied the threshold characteristics of the principal eigenvalue and found that it can be used as a new threshold to describe the diffusion of the epidemic. Results Our results prove that the principal eigenvalue $$\uplambda ^{*}$$ λ ∗ of the delayed reaction-diffusion COVID-19 system with relapse and self-limiting treatment can replace the basic reproduction number $$R_0$$ R 0 to describe the threshold effect of disease transmission. Combine with the latest official data and the prevention and control strategies, some numerical simulations on the stability and global exponential attractiveness of the diffusion of the COVID-19 epidemic in China and the USA are given. Conclusions Through the comparison of numerical simulations, we find that self-limiting treatment can significantly promote the prevention and control of the epidemic. And if the free activities of asymptomatic infected persons are not restricted, it will seriously hinder the progress of epidemic prevention and control.
Article
Full-text available
Background: Data regarding the course of SARS-CoV-2 infection in children with primary immunodeficiency (PID) is insufficient. The purpose of the study was to evaluate the morbidity and clinical course of COVID-19 and the ability to produce anti-SARS-CoV-2 IgG antibodies in children with PID. Methods: In this retrospective study, medical records of 99 patients aged 0-18 were evaluated. The patients were divided into three groups: PID group (68.69%), control group (19.19%) and patients with ongoing or previous paediatric inflammatory multisystem syndrome (12.12%). Data such as morbidity, clinical outcome, and IgG anti-SARS-CoV-2 antibody titres were assessed. Results: A confirmed diagnosis of SARS-CoV-2 infection has been established in 26.47% of patients with PID. Among patients with PID infected with SARS-CoV-2, only three cases were hospitalised. Mortality in the PID group was 0%. Throughout an observation period of 1 year, 47.06% of patients with PID were tested positive for the anti-SARS-CoV-2 antibody. Conclusions: In the study group, in most cases the disease had a mild and self-limiting course. Remarkably, even though IgG deficiency was the most prevalent form of PID in the study group, the patients were able to respond satisfactorily to the infection in terms of anti-SARS-CoV-2 IgG.
Article
Full-text available
The COVID-19 pandemic has paralleled the great Spanish flu pandemic of 1918–1919 in the United States. Previous historical accounts have strongly suggested a post-viral syndrome and, currently, a post-COVID-19 viral syndrome is unquestionable, which shares many of the characteristics of myalgic encephalomyelitis/chronic fatigue syndrome that is present globally. The original term for this post-acute sequela of SARS-CoV-2 (PASC) was termed long haulers by those who were affected with this syndrome and it is now termed long COVID (LC) or PASC. International researchers and clinicians are desperately trying to better understand the pathobiological mechanisms possibly involved in this syndrome. This review aims to summarize many of the cumulated findings associated with LC/PASC and provides supportive and representative illustrations and transmission electron micrographic remodeling changes within brain tissues associated with a stress type of injury as occurs in the classic db/db and novel BTBR ob/ob obesity and diabetes mellitus mice models. These models are utilized to merely provide a response to metabolic stress injury wound healing mechanisms that are also present in humans. This review posits that neuroglial activation and chronic neuroinflammation may be a common denominator for the development of the complex LC/PASC syndrome following acute COVID-19 due to SARS-CoV-2.
Article
Depression is the most common mental disorder and a leading cause of disability worldwide. Despite abundant research, the precise mechanisms underlying the pathophysiology of depression remain elusive. Accumulating evidence from preclinical and clinical studies suggests that alterations in the gut microbiota, microbe-derived short-chain fatty acids, D-amino acids and metabolites play a key role in the pathophysiology of depression via the brain–gut–microbiota axis, including the neural and immune systems. Notably, the brain–gut–microbiota axis might play a crucial role in susceptibility versus resilience in rodents exposed to stress. Vagotomy is reported to block depression-like phenotypes in rodents after fecal microbiota transplantation of “depression-related” microbiome, suggesting that the vagus nerve influences depression through the brain–gut–microbiota axis. In this article, we review recent findings regarding the brain–gut–microbiota axis in depression and discuss its potential as a therapeutic target for depression.