Review
Published: 2023-11-29
download
PDF

COVID-19. Biology, pathophysiology, and immunology: a pathologist view

Department of Pathology, Pederzoli Hospital, Peschiera del Garda, Italy
Department of Pathology, San Raffaele Scientific Institute. Milan, Italy
Department of Diseases of the Thorax, Ospedale GB Morgagni, Forlì, Italy
Department of Diseases of the Thorax, Ospedale GB Morgagni, Forlì, Italy
Department of Pathology, Ospedale GB Morgagni, Forlì, Italy
Department of Pathology, Pederzoli Hospital, Peschiera del Garda, Italy
Department of Diseases of the Thorax, Ospedale GB Morgagni, Forlì, Italy; Department of Pathology, Ospedale GB Morgagni, Forlì, Italy
SARS-CoV-2 COVID-19 long-COVID post-COVID pathogenesis ng biopsy

Abstract

Even if the SARS-CoV-2 pandemic has been declared over, several risks and clinical problems remain to be faced, including long-COVID sequelae and possible outbreaks of pathogenic variants. Intense research on COVID-19 has provided in these few years a striking amount of data covering different fields and disciplines, which can help to provide a knowledge shield against new potential infective spreads, and may also potentially be applied to other fields of medicine, including oncology and neurology. Nevertheless, areas of uncertainty still remain regarding the pathogenic mechanisms that subtend the multifaceted manifestations of the disease. To better clarify the pathogenesis of the disease, a systematic multidisciplinary evaluation of the many mechanisms involved in COVID-19 is mandatory, including clinical, physiological, radiological, immunological and pathological studies. In COVID-19 syndrome the pathological studies have been mainly performed on autopsy cases, and only a few studies are available on biopsies. Nevertheless, these studies have provided relevant information that can substantially contribute to decipher the complex scenario characterizing the different forms of COVID-19 and long-COVID-19. In this review the data provided by pathological investigations are recapitulated and discussed, in the light of different hypothesis and data provided by clinical, physiological and immunological data. 

COVID-19

Coronavirus disease 2019 (COVID-19) is characterized by a profound variability in clinical presentation and pathological features with a large majority of patients developing mild symptoms and a minority experiencing an interstitial pneumonia that can rapidly progress to severe life threatening respiratory failure requiring mechanical ventilation or even extra corporeal membrane oxygenation (ECMO) 1. It is now widely accepted that SARS-CoV-2 infection can trigger a hyper-inflammatory response in susceptible individuals (also termed “cytokine storm” or cytokine release syndrome) 2, and several hypothetic schemes have been proposed to describe the pathogenic role of different cell types and mechanisms leading to different disease phases and endotypes. The heterogeneity of clinical presentations is likely conditioned by the viral burden, the efficacy of innate and adaptive immune responses, a genetic predisposition, and the occurrence and severity of pre-existing comorbidities (such as older age, obesity, hypertension, diabetes) 3. The complex interactions of SARS-CoV-2 with human cells and tissues have been explained at the cellular and molecular levels, revealing how viral ligands (mostly the spike protein) and receptors on different cell types can allow viral entry 4. Interactions between the pathogen and host defenses are mediated by a variety of viral gene products that interfere with normal development of innate immunity, particularly focusing on the cGAS/Sting pathway 5-8.

The cGAS/Sting pathway and COVID-19

The cGAS-STING pathway is a pattern recognition receptor involved in the detection of DNA in the cell cytosol where it triggers a robust type I interferon response against viral and bacterial infections, as well as self DNA released in damaged and senescent cells 9,10. Upon DNA binding within the cytoplasm cGAS (Cyclic GMP-AMP synthase) is able to generate the second messenger cyclic cGAMP necessary to activate Sting (Stimulator of INF Genes) with eventual transcriptional induction of type I interferons 11. Paradoxically, the activation of cGAS/Sting is able to effectively block the viral infection, but its activation is the main driver of the cytokine storm in severe disease 12-15. These findings suggest that the activation of innate immunity may be relevant in viral clearance in healthy people, but is a severe pathogenic driver in predisposed individuals. In line with this hypothesis is the experimental observation that in bats, where the cGAS/Sting pathway is partially defective and IFN production is dampened, SARS-CoV-2 infection is not accompanied by disease manifestation 16. The impairment of IFN production may represent an early key pathogenic factor in COVID-19 17-19, and several immune mechanisms and involved cell types have been investigated to clarify the role of this impairment in triggering the cytokine storm 3,20-24. Among the critical issues is how SARS-CoV-2, a single-strand RNA virus, can induce the activation of the DNA sensor cGAS/Sting 25,26. A genetic basis has also been recently demonstrated in asymptomatic patients, providing robust early defense against the viral infection 27.

The search of factors involved in predisposition to severe disease has produced a striking amount of information, and several lines of evidence have been provided regarding the critical role of conditions and comorbidities in increasing the risk of severe complication (old age and age related diseases, diabetes, neurodegenerative disorders, obesity, hypertension, etc.) 28. In these conditions a chronic low-grade systemic inflammation (inflammaging) related to progressive dysregulation of cGas/Sting axis is often present 29-33.

Human endogenous retroviruses and COVID-19

It has been widely demonstrated that in severe and critical patients a highly impaired (IFN) type I response occurs, characterized by no IFN-β and low IFN-α production and activity 34. In a proportion of cases the deficiency can be ascribed to genetic inborn errors in genes that regulate interferon production 35-38. Another possible cause of innate immune deficiency in COVID-19 is the presence of anti-interferon auto-antibodies 39-41. Interestingly, these auto-antibodies are found in severe COVID-19 patients together with human endogenous retroviruses (HERV-W-env) auto-antibodies 42. Endogenous retroviruses (HERVs) are ancient integrations of exogenous viruses into the germ cells of mammalian ancestors (between 100 and 40 million years ago), and they now occupy approximately 8% of the human genome 43,44. HERV genes are defective and are not able to produce infectious viral particles, but some HERV can produce retroviral transcripts and proteins, which are pathologically related to various diverse conditions including autoimmune, infectious, neurological and oncologic diseases and also represent promising therapeutic targets 45-51. Abnormal levels of HERV transcripts have been demonstrated in COVID-19 and post-COVID-19, and a direct correlation has been demonstrated between the severity of COVID-19 and HERV expression 52-55. A complex modulation of HERVs characterizes different COVID-19 endotypes as revealed by high-throughput analysis of HERV loci expression and this diversity may have an impact on the immune-pathogenesis and clinical manifestations and outcome of the disease 56. In particular, the in vitro exposure to SARS-CoV-2 is able to activate the expression of the HERV-W pro-inflammatory envelope protein (ENV) in peripheral blood mononuclear cells 57,58. When abnormally expressed, HERV transcripts can profoundly alter the innate immunity 59, and their “awakening” is considered a key factor in triggering the cGAS/Sting and other pro-inflammatory pathways with eventual development of cytokine storm and severe disease evolution in predisposed individuals 59. HERV-mediated cGAS/Sting triggering of innate inflammation in nervous tissues is a major pathogenic mechanism in neurological and psychiatric diseases and may be also involved in the neurological complications observed in COVID-19 and long-COVID-19 49, 60-62. Most HERVs are epigenetically silenced, a process ensuring genomic stability that is progressively reduced in aging and toxic or infection-related stress, all conditions representing also risk factors for severe COVID-19 disease 63-65. Retroviral-like particles are abnormally induced in senescent cells and their repression may alleviate tissue degeneration and organismal aging thus representing a potential therapeutic target 66.

The contribute of Pathology

From the beginning of the pandemic a large number of post-mortem studies have been performed worldwide, providing relevant information regarding the organ damages occurring in severe cases 67-69. The most frequent pathological pattern observed in autoptic lung is “diffuse alveolar damage” (DAD), with hyaline membranes, alveolar fibrinous edema and type-II alveolar-epithelial cell (AECII) hyperplasia. In many studies the occurrence of thrombotic events was reported, varying from small capillary clots to thrombosis in the larger vessels 70-72. Nevertheless, these post-mortem studies did not explain the clinical heterogeneity of COVID-19, as revealed by immunological, radiological, and clinical studies, accounting for the evidence of different disease phenotypes 73,74. A possible explanation of this heterogeneity has been provided by studies of lung biopsies of patients with early/mild COVID-19 pneumonia as defined by typical lung opacities at CT scan, variable degree of hypoxemia but with no needs of intubation and mechanical ventilation 75,76. In this series a common and peculiar pattern of lung modifications was observed, characterized by acute lung injury but without the typical features of the DAD pattern. Hyaline membranes were in fact absent and interstitial fibrosis was either focal or absent. Alveolar epithelial type II cells (AECII) hyperplasia was heterogeneous and characterized by an unusual “patchy” distribution, with AECII clusters ranging from isolated small aggregates to wide proliferation of micronodular and/or pseudo-papillary sprouts, interposed to normally looking type-I pneumocytes. An unexpected finding was the occurrence of a diffuse enlargement of pulmonary interstitial blood vessels (both capillaries and venules). At immunohistochemical analysis abnormal phenotypes were demonstrated in both the epithelial (AECII) and vascular components, with robust expression of molecules involved in the STAT3 pathway (pSTAT3, IL-6). An interesting finding was the strong and diffuse expression of molecules related to innate-immune activation (PDL1 and Ido1) in interstitial blood vessels 75,76.

Indoleamine 2,3-dioxygenase (Ido1 and Ido2) in COVID-19

Indoleamine 2,3-dioxygenase enzyme activity is the rate-limiting step of the aminoacid tryptophan (Trp) degradation in extra-hepatic sites. IDO1 expression in normal tissues is negligible, but inflammatory stimuli can trigger its expression, mainly mediated by IFN-gamma 77-79. Ido1 has a relevant role in immune regulation suppressing effector T cell functions and favoring the development of regulatory T cells by Trp depletion at the local site of inflammation and the production of immunosuppressive Trp metabolites (kynurenine, kynurenic acid, xanthurenic acid). The only tissue where Ido1 is constitutively expressed and functional is the human placenta, where the enzyme is able to maintain feto-maternal immune-tolerance and antimicrobial functions. Ido1 is constitutively expressed in chorionic vascular endothelium, with highest levels in the microvasculature, where it regulates vascular tone and placental perfusion, thus providing a regular blood flux to the growing fetus 80-82. Accordingly, Ido1 deficiency is related to pregnancy disorders such in intrauterine growth restriction (IUGR) and pre-eclampsia 82-86. Critical levels of endothelial and/or perivascular concentrations of vasoactive Trp metabolites may be necessary for effective control of the vascular tone 87. Interestingly, the activity of endothelial Ido1/kynurenine axis on vascular tone is likely more effective in organs characterized by peculiar circulatory systems such as the placenta and lung, both characterized by reduced blood pressure and both constitutively expressing Ido1 88, 89. Accordingly, Ido1 protects against development of pulmonary hypertension 90. An endothelial protective role of Ido1 has been also demonstrated in experimental ischemia-reperfusion, atherosclerosis and acute lung allograft injury, thus suggesting a role for the TKP activation in contrasting vascular dysfunction 91-95. Vascular dysfunction (vascular inflammation, disruption of the endothelial homeostasis, edema, and life-threatening coagulation abnormalities) is a distinct feature of severe COVID-19 and is common in conditions predisposing to severe COVID-19 (diabetes, obesity, older age, etc.) 96-99.

Two Ido paralogs exist (Ido1 and Ido2) characterized by distinct expression patterns and roles in immune and vascular tone regulation 100-102. Ido2 is prevalently expressed in severe COVID-19 pneumonia 103, and a pathogenic mechanism based on Ido1/ Ido2 imbalance has been hypothesized in COVID-19, switching from protective vasodilatation to vascular dysfunction and hypertansion 104. The concurrent immunosuppressive and vasodilator activity of Ido1 in COVID-19 early/mild pneumonia may have a negative role in inducing vasoplegia, ventilation/perfusion mismatch (accounting for the hypoxia occurring in COVID-19) and lymphocyte depletion, but might help in contrasting the life threatening consequences of hypertension and vascular dysplasia as observed in severe COVID-19 pneumonia. It is worthwile to note that the concurrent promotion of pulmonary hypertension can be exerted in COVID-19 by up-regulation of HERV-K 105.

Vascular dilatation is also evident at CT scan in early/mild COVID-19 pneumonia (described as “vascular enlargement pattern”) 106-108, and this pattern is likely responsible of the peculiar pulmonary hemodynamic profile, perfusion abnormalities, and hypoxia (“happy hypoxia”) characterizing mild COVID-19 pneumonia 109-111.

Cross-talk of Idos and HERVs in COVID-19

Several links exist between HERV-mediated mechanisms and the Ido1/Trp/Kyn pathway. HERVs can act as proximal regulatory elements in promoting interferon responses and the expression of Ido enzymes strictly depends on IFNs 79,112. Both Ido1 and HERVs are physiologically expressed in the placenta and exert important concurrent roles in mammalian placental development and functions 113. Ido1 enzyme activity is in fact necessary to provide sufficient vascular perfusion and to avoid immune rejection of the fetus. HERVs are also necessary for placental morphology and trophoblast invasiveness, inducing syncytialization through the synthesis of syncitins, highly fusogenic env-like glycoproteins expressed at high levels in human placenta 47,114. Syncitins help maintain trophoblast stem cell proliferation, placental angiogenesis and contribute to maternal immune system suppression and tolerance toward the fetus 115-118. In addition, syncytins have a role in amino acid transport and allow protection against viral infection 119. In line with this evidence, a decrease in syncytin levels has been demonstrated in fetal growth restriction and pre-eclampsia 120-124. It is possible to speculate that in COVID-19 an abnormal fusogenic activity of syncitins may be related to the observed formation of AECII clusters in mild pneumonia 75, and epithelial multinucleated syncitia in severe pneumonia 125. When abnormally activated in infected lungs these molecular mechanisms likely determine perturbations of immune responses and vascular tone control with eventual triggering of autoinflammatory responses and vascular dysplasia in aging and susceptible individuals.

Long-COVID-19 (PACS)

A proportion of patients may suffer from post-acute sequelae experiencing complications affecting different organs (a condition defined as “long COVID-19”, or “post-acute COVID syndrome” (PACS) 126-133. Most common symptoms in PACS include systemic manifestations (fatigue, asthenia, poor concentration, wandering fever), pulmonary functional impairment (dyspnea, cough, reduced DLCO), neuropsychiatric manifestations (sleep disturbances, cognitive dysfunction, depression, mood changes, anxiety, headache, taste, and/or smell loss), and cardiac manifestations (chest pain, palpitations, tachycardia, dysrhytmias), as well as a variety of muscle-skeletal, renal, dermatological, and gastrointestinal manifestations 131. In a series of lung biopsies the most frequent findings were similar to those observed in acute early/mild COVID-19 pneumonias (vascular enlargement and abnormal endothelial expression of Ido1, PD-L1 and STAT3) 75,134. The persistence of Ido1 activity in pulmonary vessels may be involved in vasoplagia and hypoxia. In both COVID-19 and PACS, dysregulation of the Tryptophan/Kynurenine pathway is likely to be involved in the development of neurological complications by decreasing the availability of Trp (necessary for conversion to 5-HT and melatonin, and by altering the physiological proportions of neurotoxic (quinolinic acid, 3-hydroxykynurenine) versus neuroprotective (kynurenic acid, picolinic acid, and the essential cofactor NAD+) Trp metabolites 135-138. Persistence of HERV transcription products may be involved in PACS inducing long-lasting symptoms of chronic inflammation in different organs 49,54,139.

Conclusions

In conclusion, a pathogenic scenario can be hypothesized where a cascade of events follow SARS-CoV-2 infection of ACEII expressing epithelial cells. Central in this scheme is the production of a variety of HERV products that can interfere with the functions of non-infected cell types (monocytes, myeloid-derived suppressor cells, dendritic cells, lymphocytes, endothelial cells, etc.) in different tissue compartments. The clinical development and severity of the disease (asymptomatic, mild, severe) is likely determined by genetic background and physiopathogical status (age, comorbidities) interfering with the many biological mechanisms and pathways regulating immunity and cardio-vascular and pulmonary systems. A rational catalog of chronology and relevance of these mechanisms is needed to obtain a meaningful comprehension of COVID-19 pathogenesis.

CONFLICTS OF INTEREST

The authors report no competing interests.

FUNDING

This study did not receive external funding.

AUTHORS’ CONTRIBUTIONS

MC, CD and VP contributed to the design of the work; MC, CD, VP, LS and CR have drafted the work; MC, CD, CR, SP, AD, LS and VP have revised the manuscript. All authors have read and agreed to the published version of the manuscript

ETHICAL CONSIDERATION

Not applicable.

References

  1. Huang C, Wang Y, Li X. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020; 395:497-506. DOI
  2. Fajgenbaum DC, June CH. Cytokine Storm. N Engl J Med. 2020; 383:2255-2573. DOI
  3. Zhou Y, Fu B, Zheng X. Pathogenic T-cells and inflammatory monocytes incite inflammatory storms in severe COVID-19 patients. Natl Sci Rev. 2020; 7:998-1002. DOI
  4. Han L, Zhuang MW, Deng J. SARS-CoV-2 ORF9b antagonizes type I and III interferons by targeting multiple components of the RIG-I/MDA-5-MAVS, TLR3-TRIF, and cGAS-STING signaling pathways. J Med Virol. 2021; 93:5376-5389. DOI
  5. Han L, Zheng Y, Deng J. SARS-CoV-2 ORF10 antagonizes STING-dependent interferon activation and autophagy. J Med Virol. 2022; 94:5174-5188. DOI
  6. Deng J, Zheng Y, Zheng SN. SARS-CoV-2 NSP7 inhibits type I and III IFN production by targeting the RIG-I/MDA5, TRIF, and STING signaling pathways. J Med Virol. 2023; 95:e28561. DOI
  7. Cai S, Zhang C, Zhuang Z. Phase-separated nucleocapsid protein of SARS-CoV-2 suppresses cGAS-DNA recognition by disrupting cGAS-G3BP1 complex. Signal Transduct Target Ther. 2023; 8:170. DOI
  8. Shu C, Li X, Li P. The mechanism of double-stranded DNA sensing through the cGAS-STING pathway. Cytokine Growth Factor Rev. 2014; 25:641-648. DOI
  9. Zeng PH, Yin WJ. The cGAS/STING signaling pathway: a cross-talk of infection, senescence and tumors. Cell Cycle. 2023; 22:38-56. DOI
  10. Hornung V, Hartmann R, Ablasser A. OAS proteins and cGAS: unifying concepts in sensing and responding to cytosolic nucleic acids. Nat Rev Immunol. 2014; 14:521-528. DOI
  11. Liu W, Reyes HM, Yang JF. Activation of STING Signaling Pathway Effectively Blocks Human Coronavirus Infection. J Virol. 2021; 95:e00490-21. DOI
  12. Neufeldt CJ, Cerikan B, Cortese M. SARS-CoV-2 infection induces a pro-inflammatory cytokine response through cGAS-STING and NF-κB. Commun Biol. 2022; 5:45. DOI
  13. Di Domizio JD, Gulen MF, Saidoune F. The cGAS-STING pathway drives type I IFN immunopathology in COVID-19. Nature. 2022; 603:145-151. DOI
  14. Andreakos E. STINGing type I IFN-mediated immunopathology in COVID-19. Nat Immunol. 2022; 23:478-480. DOI
  15. Xie J, Li Y, Shen X. Dampened STING-Dependent Interferon Activation in Bats. Cell Host Microbe. 2018; 23:297-301.e4. DOI
  16. Sa Ribero M, Jouvenet N, Dreux M. Interplay between SARS-CoV-2 and the type I interferon response. PLoS Pathog. 2020; 16:e1008737. DOI
  17. Xia H, Cao Z, Xie X. Evasion of Type I Interferon by SARS-CoV-2. Cell Rep. 2020; 33:108234. DOI
  18. Yuen CK, Lam JY, Wong WM. SARS-CoV-2 nsp13, nsp14, nsp15 and orf6 function as potent interferon antagonists. Emerg Microbes Infect. 2020; 9:1418-1428. DOI
  19. Wen W, Su W, Tang H. Immune cell profiling of COVID-19 patients in the recovery stage by single-cell sequencing. Cell Discov. 2020; 6:31. DOI
  20. Trombetta AC, Farias GB, Gomes AMC. Severe COVID-19 Recovery Is Associated with Timely Acquisition of a Myeloid Cell Immune-Regulatory Phenotype. Front Immunol. 2021; 12:691725. DOI
  21. Ait-Belkacem I, Cartagena García C, Millet-Wallisky E. SARS-CoV-2 spike protein induces a differential monocyte activation that may contribute to age bias in COVID-19 severity. Sci Rep. 2022; 12:20824. DOI
  22. Bost P, De Sanctis F, Canè S. Deciphering the state of immune silence in fatal COVID-19 patients. Nat Commun. 2021; 12:1428. DOI
  23. Falck-Jones S, Österberg B, Smed-Sörensen A. Respiratory and systemic monocytes, dendritic cells, and myeloid-derived suppressor cells in COVID-19: Implications for disease severity. J Intern Med. 2023; 293:130-143. DOI
  24. Fan YM, Zhang YL, Luo H. Crosstalk between RNA viruses and DNA sensors: Role of the cGAS-STING signalling pathway. Rev Med Virol. 2022; 32:e2343. DOI
  25. Amurri L, Horvat B, Iampietro M. Interplay between RNA viruses and cGAS/STING axis in innate immunity. Front Cell Infect Microbiol. 2023; 13:1172739. DOI
  26. Augusto DG, Murdolo LD, Chatzileontiadou DSM. A common allele of HLA is associated with asymptomatic SARS-CoV-2 infection. Nature. 2023; 620:128-136. DOI
  27. Zhou F, Yu T, Du R. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet. 2020; 395:1054-1062. DOI
  28. Bai J, Cervantes C, Liu J. DsbA-L prevents obesity-induced inflammation and insulin resistance by suppressing the mtDNA release-activated cGAS-cGAMP-STING pathway. Proc Natl Acad Sci U S A. 2017; 114:12196-12201. DOI
  29. Schmitz CRR, Maurmann RM, Guma FTCR. cGAS-STING pathway as a potential trigger of immunosenescence and inflammaging. Front Immunol. 2023; 14:1132653. DOI
  30. Zheng W, Feng D, Xiong X. The Role of cGAS-STING in Age-Related Diseases from Mechanisms to Therapies. Aging Dis. 2023; 14:1145-1165. DOI
  31. Elzinga SE, Henn R, Murdock BJ. cGAS/STING and innate brain inflammation following acute high-fat feeding. Front Immunol. 2022; 13:1012594. DOI
  32. Hu H, Zhao R, He Q. cGAS-STING mediates cytoplasmic mitochondrial-DNA-induced inflammatory signal transduction during accelerated senescence of pancreatic β-cells induced by metabolic stress. FASEB J. 2022; 36:e22266. DOI
  33. Zang N, Cui C, Guo X. cGAS-STING activation contributes to podocyte injury in diabetic kidney disease. iScience. 2022; 25:105145. DOI
  34. Hadjadj J, Yatim N, Barnabei L. Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients. Science. 2020; 369:718-724. DOI
  35. Zhang Q, Bastard P, Liu Z. Inborn errors of type I IFN immunity in patients with life-threatening COVID-19. Science. 2020; 370:eabd4570. DOI
  36. Zhang Q, Bastard P, COVID Human Genetic Effort. Human genetic and immunological determinants of critical COVID-19 pneumonia. Nature. 2022; 603:587-598. DOI
  37. Matuozzo D, Talouarn E, Marchal A. Rare predicted loss-of-function variants of type I IFN immunity genes are associated with life-threatening COVID-19. Genome Med. 2023; 15:22. DOI
  38. Casanova JL, Anderson MS. Unlocking life-threatening COVID-19 through two types of inborn errors of type I IFNs. J Clin Invest. 2023; 133:e166283. DOI
  39. Bastard P, Rosen LB, Zhang Q. Autoantibodies against type I IFNs in patients with life-threatening COVID-19. Science. 2020; 370:eabd4585. DOI
  40. Zhou W, Wang W. Auto-antibodies against type I IFNs are associated with severe COVID-19 pneumonia. Signal Transduct Target Ther. 2021; 6:96. DOI
  41. Philippot Q, Fekkar A, Gervais A. J Clin Immunol. 2023; 43:1093-1103. DOI
  42. Simula ER, Manca MA, Noli M. Increased Presence of Antibodies against Type I Interferons and Human Endogenous Retrovirus W in Intensive Care Unit COVID-19 Patients. Microbiol Spectr. 2022; 10:e0128022. DOI
  43. Tokuyama M, Kong Y, Song E. ERVmap analysis reveals genome-wide transcription of human endogenous retroviruses. Proc Natl Acad Sci U S A. 2018; 115:12565-12572. DOI
  44. Perl A. Role of endogenous retroviruses in autoimmune diseases. Rheum Dis Clin North Am. 2003; 29:123-43. DOI
  45. Perl A, Nagy G, Koncz A. Molecular mimicry and immunomodulation by the HRES-1 endogenous retrovirus in SLE. Autoimmunity. 2008; 41:287-97. DOI
  46. Volkman HE, Stetson DB. The enemy within: endogenous retroelements and autoimmune disease. Nat Immunol. 2014; 15:415-422. DOI
  47. Stein RA, DePaola RV. Human endogenous retroviruses: our genomic fossils and companions. Physiol Genomics. 2023; 55:249-258. DOI
  48. Dolei A. The aliens inside us: HERV-W endogenous retroviruses and multiple sclerosis. Mult Scler. 2018; 24:42-47. DOI
  49. Gruchot J, Herrero F, Weber-Stadlbauer U. Interplay between activation of endogenous retroviruses and inflammation as common pathogenic mechanism in neurological and psychiatric disorders. Brain Behav Immun. 2023; 107:242-252. DOI
  50. Hartung HP, Derfuss T, Cree BA. Efficacy and safety of temelimab in multiple sclerosis: Results of a randomized phase 2b and extension study. Mult Scler. 2022; 28:429-440. DOI
  51. Ng KW, Boumelha J, Enfield KSS. Antibodies against endogenous retroviruses promote lung cancer immunotherapy. Nature. 2023; 616:563-573. DOI
  52. Balestrieri E, Minutolo A, Petrone V. Evidence of the pathogenic HERV-W envelope expression in T lymphocytes in association with the respiratory outcome of COVID-19 patients. EBioMedicine. 2021; 66:103341. DOI
  53. Temerozo JR, Fintelman-Rodrigues N, Dos Santos MC. Human endogenous retrovirus K in the respiratory tract is associated with COVID-19 physiopathology. Microbiome. 2022; 10:65. DOI
  54. Giménez-Orenga K, Pierquin J, Brunel J. HERV-W ENV antigenemia and correlation of increased anti-SARS-CoV-2 immunoglobulin levels with post-COVID-19 symptoms. Front Immunol. 2022; 13:1020064. DOI
  55. Petrone V, Fanelli M, Giudice M. Expression profile of HERVs and inflammatory mediators detected in nasal mucosa as a predictive biomarker of COVID-19 severity. Front Microbiol. 2023; 14:1155624. DOI
  56. Grandi N, Erbì MC, Scognamiglio S. Human Endogenous Retrovirus (HERV) Transcriptome Is Dynamically Modulated during SARS-CoV-2 Infection and Allows Discrimination of COVID-19 Clinical Stages. Microbiol Spectr. 2023; 11:e0251622. DOI
  57. Grandi N, Tramontano E. Type W Human Endogenous Retrovirus (HERV-W) Integrations and Their Mobilization by L1 Machinery: Contribution to the Human Transcriptome and Impact on the Host Physiopathology. Viruses. 2017; 9:162. DOI
  58. Charvet B, Brunel J, Pierquin J. SARS-CoV-2 awakens ancient retroviral genes and the expression of proinflammatory HERV-W envelope protein in COVID-19 patients. iScience. 2023; 26:106604. DOI
  59. Grandi N, Tramontano E. Human Endogenous Retroviruses Are Ancient Acquired Elements Still Shaping Innate Immune Responses. Front Immunol. 2018; 9:2039. DOI
  60. Li X, Wu X, Li W. HERV-W ENV Induces Innate Immune Activation and Neuronal Apoptosis via linc01930/cGAS Axis in Recent-Onset Schizophrenia. Int J Mol Sci. 2023; 24:3000. DOI
  61. Sankowski R, Strohl JJ, Huerta TS. Endogenous retroviruses are associated with hippocampus-based memory impairment. Proc Natl Acad Sci U S A. 2019; 116:25982-25990. DOI
  62. Bao H, Yan J, Huang J. Activation of endogenous retrovirus triggers microglial immuno-inflammation and contributes to negative emotional behaviors in mice with chronic stress. J Neuroinflammation. 2023; 20:37. DOI
  63. Hurst TP, Magiorkinis G. Epigenetic Control of Human Endogenous Retrovirus Expression: Focus on Regulation of Long-Terminal Repeats (LTRs). Viruses. 2017; 9:130. DOI
  64. Cardelli M. The epigenetic alterations of endogenous retroelements in aging. Mech Ageing Dev. 2018; 174:30-46. DOI
  65. Di Giorgio E, Xodo LE. Endogenous Retroviruses (ERVs): Does RLR (RIG-I-Like Receptors)-MAVS Pathway Directly Control Senescence and Aging as a Consequence of ERV De-Repression?. Front Immunol. 2022; 13:917998. DOI
  66. Liu X, Liu Z, Wu Z. Resurrection of endogenous retroviruses during aging reinforces senescence. Cell. 2023; 186:287-304.e26. DOI
  67. Polak SB, Van Gool IC, Cohen D. A systematic review of pathological findings in COVID-19: a pathophysiological timeline and possible mechanisms of disease progression. Mod Pathol. 2020; 33:2128-2138. DOI
  68. Mohanty SK, Satapathy A, Naidu MM. Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and coronavirus disease 19 (COVID-19) - anatomic pathology perspective on current knowledge. Diagn Pathol. 2020; 15:103. DOI
  69. Xu Z, Shi L, Wang Y. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir Med. 2020; 8:420-422. DOI
  70. Carsana L, Sonzogni A, Nasr A. Pulmonary post-mortem findings in a series of COVID-19 cases from northern Italy: a two-centre descriptive study. Lancet Infect Dis. 2020; 20:1135-1140. DOI
  71. Ackermann M, Verleden SE, Kuehnel M. Pulmonary Vascular Endothelialitis, Thrombosis, and Angiogenesis in Covid-19. N Engl J Med. 2020; 383:120-128. DOI
  72. Menter T, Haslbauer JD, Nienhold R. Postmortem examination of COVID-19 patients reveals diffuse alveolar damage with severe capillary congestion and variegated findings in lungs and other organs suggesting vascular dysfunction. Histopathology. 2020; 77:198-209. DOI
  73. Gattinoni L, Coppola S, Cressoni M. COVID-19 Does Not Lead to a “Typical” Acute Respiratory Distress Syndrome. Am J Respir Crit Care Med. 2020; 201:1299-1300. DOI
  74. Maley JH, Winkler T, Hardin CC. Heterogeneity of Acute Respiratory Distress Syndrome in COVID-19: “Typical” or Not?. Am J Respir Crit Care Med. 202:618-619. DOI
  75. Doglioni C, Ravaglia C, Chilosi M. Covid-19 Interstitial Pneumonia: Histological and Immunohistochemical Features on Cryobiopsies. Respiration. 2021; 100:488-498. DOI
  76. Chilosi M, Poletti V, Ravaglia C. The pathogenic role of epithelial and endothelial cells in early-phase COVID-19 pneumonia: victims and partners in crime. Mod Pathol. 2021; 34:1444-1455. DOI
  77. Pfefferkorn ER, Rebhun S, Eckel M. Characterization of an indoleamine 2,3-dioxygenase induced by gamma-interferon in cultured human fibroblasts. J Interferon Res. 1986; 6:267-279. DOI
  78. Taylor MW, Feng GS. Relationship between interferon-gamma, indoleamine 2,3-dioxygenase, and tryptophan catabolism. FASEB J. 1991; 5:2516-2522.
  79. Yeung AW, Terentis AC, King NJ. Role of indoleamine 2,3-dioxygenase in health and disease. Clin Sci (Lond). 2015; 129:601-672. DOI
  80. Sedlmayr P, Blaschitz A. Placental expression of indoleamine 2,3-dioxygenase. Wien Med Wochenschr. 2012; 162:214-219. DOI
  81. Wang Y, Liu H, McKenzie G. Kynurenine is an endothelium-derived relaxing factor produced during inflammation. Nat Med. 2010; 16:279-285. DOI
  82. Zardoya-Laguardia P, Blaschitz A, Hirschmugl B. Endothelial indoleamine 2,3-dioxygenase-1 regulates the placental vascular tone and is deficient in intrauterine growth restriction and pre-eclampsia. Sci Rep. 2018; 8:5488. DOI
  83. Nishizawa H, Pryor-Koishi K, Kato T. Microarray analysis of differentially expressed fetal genes in placental tissue derived from early and late onset severe pre-eclampsia. Placenta. 2007; 28:487-497. DOI
  84. Nishizawa H, Ota S, Suzuki M. Comparative gene expression profiling of placentas from patients with severe pre-eclampsia and unexplained fetal growth restriction. Reprod Biol Endocrinol. 2011; 9:107. DOI
  85. Santillan MK, Pelham CJ, Ketsawatsomkron P. Pregnant mice lacking indoleamine 2,3-dioxygenase exhibit preeclampsia phenotypes. Physiol Rep. 2015; 3:e12257. DOI
  86. Broekhuizen M, Hitzerd E, van den Bosch TPP. The Placental Innate Immune System Is Altered in Early-Onset Preeclampsia, but Not in Late-Onset Preeclampsia. Front Immunol. 2021; 12:780043. DOI
  87. Watts SW, Shaw S, Burnett R. Indoleamine 2,3-diooxygenase in periaortic fat: mechanisms of inhibition of contraction. Am J Physiol Heart Circ Physiol. 2011; 301:H1236-47. DOI
  88. Struijk PC, Mathews VJ, Loupas T. Blood pressure estimation in the human fetal descending aorta. Ultrasound Obstet Gynecol. 2008; 32:673-681. DOI
  89. Théate I, van Baren N, Pilotte L. Extensive profiling of the expression of the indoleamine 2,3-dioxygenase 1 protein in normal and tumoral human tissues. Cancer Immunol Res. 2015; 3:161-172. DOI
  90. Xiao Y, Christou H, Liu L. Endothelial indoleamine 2,3-dioxygenase protects against development of pulmonary hypertension. Am J Respir Crit Care Med. 2013; 188:482-491. DOI
  91. Liu H, Liu L, Visner GA. Nonviral gene delivery with indoleamine 2,3-dioxygenase targeting pulmonary endothelium protects against ischemia-reperfusion injury. Am J Transplant. 2007; 7:2291-300. DOI
  92. Cole JE, Astola N, Cribbs AP. Indoleamine 2,3-dioxygenase-1 is protective in atherosclerosis and its metabolites provide new opportunities for drug development. Proc Natl Acad Sci U S A. 2015; 112:13033-8. DOI
  93. Forteza MJ, Polyzos KA, Baumgartner R. Activation of the Regulatory T-Cell/Indoleamine 2,3-Dioxygenase Axis Reduces Vascular Inflammation and Atherosclerosis in Hyperlipidemic Mice. Front Immunol. 2018; 9:950. DOI
  94. Wolowczuk I, Hennart B, Leloire A. Tryptophan metabolism activation by indoleamine 2,3-dioxygenase in adipose tissue of obese women: an attempt to maintain immune homeostasis and vascular tone. Am J Physiol Regul Integr Comp Physiol. 2012; 303:R135-43. DOI
  95. Lee SM, Park HY, Suh YS. Inhibition of acute lethal pulmonary inflammation by the IDO-AhR pathway. Proc Natl Acad Sci U S A. 2017; 114:E5881-E5890. DOI
  96. Evans PC, Rainger GE, Mason JC. Endothelial dysfunction in COVID-19: a position paper of the ESC Working Group for Atherosclerosis and Vascular Biology, and the ESC Council of Basic Cardiovascular Science. Cardiovasc Res. 2020; 116:2177-2184. DOI
  97. Potus F, Mai V, Lebret M. Novel insights on the pulmonary vascular consequences of COVID-19. Am J Physiol Lung Cell Mol Physiol. 2020; 319:L277-L288. DOI
  98. Bonaventura A, Vecchié A, Dagna L. Endothelial dysfunction and immunothrombosis as key pathogenic mechanisms in COVID-19. Nat Rev Immunol. 2021; 21:319-329. DOI
  99. Mejia-Renteria H, Travieso A, Sagir A. In-vivo evidence of systemic endothelial vascular dysfunction in COVID-19. Int J Cardiol. 2021; 345:153-155. DOI
  100. Merlo LMF, Pigott E, DuHadaway JB. IDO2 is a critical mediator of autoantibody production and inflammatory pathogenesis in a mouse model of autoimmune arthritis. J Immunol. 2014; 192:2082-2090. DOI
  101. Merlo LM, Mandik-Nayak L. IDO2: A Pathogenic Mediator of Inflammatory Autoimmunity. Clin Med Insights Pathol. 2016; 9:21-28. DOI
  102. Fatokun AA, Hunt NH, Ball HJ. Indoleamine 2,3-dioxygenase 2 (IDO2) and the kynurenine pathway: characteristics and potential roles in health and disease. Amino Acids. 2013; 45:1319-1329. DOI
  103. Guo L, Schurink B, Roos E. Indoleamine 2,3-dioxygenase (IDO)-1 and IDO-2 activity and severe course of COVID-19. J Pathol. 2022; 256:256-261. DOI
  104. Chilosi M, Doglioni C, Ravaglia C. Unbalanced IDO1/IDO2 Endothelial Expression and Skewed Keynurenine Pathway in the Pathogenesis of COVID-19 and Post-COVID-19 Pneumonia. Biomedicines. 2022; 10:1332. DOI
  105. Wang D, Gomes MT, Mo Y. Human Endogenous Retrovirus, SARS-CoV-2, and HIV Promote PAH via Inflammation and Growth Stimulation. Int J Mol Sci. 2023; 24:7472. DOI
  106. Li Q, Huang XT, Li CH. CT features of coronavirus disease 2019 (COVID-19) with an emphasis on the vascular enlargement pattern. Eur J Radiol. 2021; 134:109442. DOI
  107. Lang M, Som A, Carey D. Pulmonary Vascular Manifestations of COVID-19 Pneumonia. Radiol Cardiothorac Imaging. 2020; 2:e200277. DOI
  108. Piciucchi S, Ravaglia C, Vizzuso A. Reversibility of venous dilatation and parenchymal changes density in Sars-Cov-2 pneumonia: toward the definition of a peculiar pattern. Pulmonology. 2021; 27:353-357. DOI
  109. Gattinoni L, Chiumello D, Caironi P. COVID-19 pneumonia: different respiratory treatments for different phenotypes?. Intensive Care Med. 2020; 46:1099-1102. DOI
  110. Busana M, Schiavone M, Lanfranchi A. Non-invasive hemodynamic profile of early COVID-19 infection. Physiol Rep. 2020; 8:e14628. DOI
  111. Oldani S, Ravaglia C, Bensai S. Pathophysiology of light phenotype SARS-CoV-2 interstitial pneumonia: from histopathological features to clinical presentations. Pulmonology. 2022; 28:333-344. DOI
  112. Wang Y, Liu M, Guo X. Endogenous Retrovirus Elements Are Co-Expressed with IFN Stimulation Genes in the JAK-STAT Pathway. Viruses. 2022; 15:60. DOI
  113. Blond JL, Besème F, Duret L. Molecular characterization and placental expression of HERV-W, a new human endogenous retrovirus family. J Virol. 1999; 73:1175-85. DOI
  114. Holder BS, Tower CL, Abrahams VM. Syncytin 1 in the human placenta. Placenta. 2012; 33:460-466. DOI
  115. Mangeney M, Renard M, Schlecht-Louf G. Placental syncytins: Genetic disjunction between the fusogenic and immunosuppressive activity of retroviral envelope proteins. Proc Natl Acad Sci U S A. 2007; 104:20534-9. DOI
  116. Qiao S, Wang F, Chen H. Inducible knockout of Syncytin-A gene leads to an extensive placental vasculature deficiency, implications for preeclampsia. Clin Chim Acta. 2017; 474:137-146. DOI
  117. Wang YN, Ye Y, Zhou D. The Role of Syncytin in Placental Angiogenesis and Fetal Growth. Front Cell Dev Biol. 2022; 10:852561. DOI
  118. Yu M, Hu X, Pan Z. Endogenous retrovirus-derived enhancers confer the transcriptional regulation of human trophoblast syncytialization. Nucleic Acids Res. 2023; 51:4745-4759. DOI
  119. Gasent Blesa J, Candel V. Cell-cell fusion as a potential target in cancer therapy. Ecancermedicalscience. 2009; 3:145. DOI
  120. Knerr I, Beinder E, Rascher W. Syncytin, a novel human endogenous retroviral gene in human placenta: evidence for its dysregulation in preeclampsia and HELLP syndrome. Am J Obstet Gynecol. 2002; 186:210-213. DOI
  121. Langbein M, Strick R, Strissel PL. Impaired cytotrophoblast cell-cell fusion is associated with reduced Syncytin and increased apoptosis in patients with placental dysfunction. Mol Reprod Dev. 2008; 75:175-183. DOI
  122. Ruebner M, Strissel PL, Ekici AB. Reduced syncytin-1 expression levels in placental syndromes correlates with epigenetic hypermethylation of the ERVW-1 promoter region. PLoS One. 2013; 8:e56145. DOI
  123. Lee X, Keith JC, Stumm N. Downregulation of placental syncytin expression and abnormal protein localization in pre-eclampsia. Placenta. 2001; 22:808-812. DOI
  124. Zhuang XW, Li J, Brost BC. Decreased expression and altered methylation of syncytin-1 gene in human placentas associated with preeclampsia. Curr Pharm Des. 2014; 20:1796-1802. DOI
  125. Braga L, Ali H, Secco I. Drugs that inhibit TMEM16 proteins block SARS-CoV-2 spike-induced syncytia. Nature. 2021; 594:88-93. DOI
  126. Logue JK, Franko NM, McCulloch DJ. Sequelae in Adults at 6 Months After COVID-19 Infection. JAMA Netw Open. 2021; 4:e210830. DOI
  127. Darcis G, Bouquegneau A, Maes N. Long-term clinical follow-up of patients suffering from moderate-to-severe COVID-19 infection: a monocentric prospective observational cohort study. Int J Infect Dis. 2021; 109:209-216. DOI
  128. Nalbandian A, Sehgal K, Gupta A. Post-acute COVID-19 syndrome. Nat Med. 2021; 27:601-615. DOI
  129. Augustin M, Schommers P, Stecher M. Post-COVID syndrome in non-hospitalised patients with COVID-19: a longitudinal prospective cohort study. Lancet Reg Health Eur. 2021; 6:100122. DOI
  130. Soriano JB, Murthy S, Marshall JC. A clinical case definition of post-COVID-19 condition by a Delphi consensus. Lancet Infect Dis. 2022; 22:e102-e107. DOI
  131. Yan Z, Yang M, Lai CL. Long COVID-19 Syndrome: A Comprehensive Review of Its Effect on Various Organ Systems and Recommendation on Rehabilitation Plans. Biomedicines. 2021; 9:966. DOI
  132. Mehandru S, Merad M. Pathological sequelae of long-haul COVID. Nat Immunol. 2022; 23:194-202. DOI
  133. Antoniou KM, Vasarmidi E, Russell AM. European Respiratory Society statement on long COVID follow-up. Eur Respir J. 2022; 60:2102174. DOI
  134. Ravaglia C, Doglioni C, Chilosi M. Clinical, radiological and pathological findings in patients with persistent lung disease following SARS-CoV-2 infection. Eur Respir J. 2022; 60:2102411. DOI
  135. Lovelace MD, Varney B, Sundaram G. Recent evidence for an expanded role of the kynurenine pathway of tryptophan metabolism in neurological diseases. Neuropharmacology. 2017; 112(Pt B):373-388. DOI
  136. Braidy N, Grant R, Adams S. Mechanism for quinolinic acid cytotoxicity in human astrocytes and neurons. Neurotox Res. 2009; 16:77-86. DOI
  137. Lugo-Huitrón R, Ugalde Muñiz P, Pineda B. Quinolinic acid: an endogenous neurotoxin with multiple targets. Oxid Med Cell Longev. 2013; 2013:104024. DOI
  138. Gietl M, Burkert F, Seiwald S. Interferon-gamma Mediated Metabolic Pathways in Hospitalized Patients During Acute and Reconvalescent COVID-19. Int J Tryptophan Res. 2023; 16:11786469231154244. DOI
  139. Apostolou E, Rizwan M, Moustardas P. Saliva antibody-fingerprint of reactivated latent viruses after mild/asymptomatic COVID-19 is unique in patients with myalgic-encephalomyelitis/chronic fatigue syndrome. Front Immunol. 2022; 13:949787. DOI

Affiliations

$authorString->getOrcid() =>

$authorString->getFullName() => Marco Chilosi

$authorString->getUrl() =>

Marco Chilosi

Department of Pathology, Pederzoli Hospital, Peschiera del Garda, Italy
non esiste orcidID ""

$authorString->getOrcid() =>

$authorString->getFullName() => Caludio Doglioni

$authorString->getUrl() =>

Caludio Doglioni

Department of Pathology, San Raffaele Scientific Institute. Milan, Italy
non esiste orcidID ""

$authorString->getOrcid() =>

$authorString->getFullName() => Claudia Ravaglia

$authorString->getUrl() =>

Claudia Ravaglia

Department of Diseases of the Thorax, Ospedale GB Morgagni, Forlì, Italy
non esiste orcidID ""

$authorString->getOrcid() =>

$authorString->getFullName() => Sara Piciucchi

$authorString->getUrl() =>

Sara Piciucchi

Department of Diseases of the Thorax, Ospedale GB Morgagni, Forlì, Italy
non esiste orcidID ""

$authorString->getOrcid() =>

$authorString->getFullName() => Alessandra Dubini

$authorString->getUrl() =>

Alessandra Dubini

Department of Pathology, Ospedale GB Morgagni, Forlì, Italy
non esiste orcidID ""

$authorString->getOrcid() =>

$authorString->getFullName() => Lavinia Stefanizzi

$authorString->getUrl() =>

Lavinia Stefanizzi

Department of Pathology, Pederzoli Hospital, Peschiera del Garda, Italy
non esiste orcidID ""

$authorString->getOrcid() =>

$authorString->getFullName() => Venerino Poletti

$authorString->getUrl() =>

Venerino Poletti

Department of Diseases of the Thorax, Ospedale GB Morgagni, Forlì, Italy; Department of Pathology, Ospedale GB Morgagni, Forlì, Italy
non esiste orcidID ""

Copyright

© Società Italiana di Anatomia Patologica e Citopatologia Diagnostica, Divisione Italiana della International Academy of Pathology , 2023

How to Cite

[1]
Chilosi, M., Doglioni, C., Ravaglia, C., Piciucchi, S., Dubini, A., Stefanizzi, L. and Poletti, V. 2023. COVID-19. Biology, pathophysiology, and immunology: a pathologist view. Pathologica - Journal of the Italian Society of Anatomic Pathology and Diagnostic Cytopathology. 115, 5 (Nov. 2023), 248-256. DOI:https://doi.org/10.32074/1591-951X-954.
  • Abstract viewed - 531 times
  • PDF downloaded - 323 times