Skip to main content
Erschienen in: Seminars in Immunopathology 3/2022

Open Access 12.04.2022 | COVID-19 | Review

COVID-19 Vasculitis and vasculopathy-Distinct immunopathology emerging from the close juxtaposition of Type II Pneumocytes and Pulmonary Endothelial Cells

verfasst von: Sami Giryes, Nicola Luigi Bragazzi, Charles Bridgewood, Gabriele De Marco, Dennis McGonagle

Erschienen in: Seminars in Immunopathology | Ausgabe 3/2022

Abstract

The SARS-CoV-2 virus ACE-2 receptor utilization for cellular entry and the defined ACE-2 receptor role in cardiovascular medicine hinted at dysregulated endothelial function or even direct viral endotheliitis as the key driver of severe COVID-19 vascular immunopathology including reports of vasculitis. In this article, we critically review COVID-19 immunopathology from the vasculitis perspective and highlight the non-infectious nature of vascular endothelial involvement in severe COVID-19. Whilst COVID-19 lung disease pathological changes included juxta-capillary and vascular macrophage and lymphocytic infiltration typical of vasculitis, we review the evidence reflecting that such “vasculitis” reflects an extension of pneumonic inflammatory pathology to encompass these thin-walled vessels. Definitive, extrapulmonary clinically discernible vasculitis including cutaneous and cardiac vasculitis also emerged- namely a dysregulated interferon expression or “COVID toes” and an ill-defined systemic Kawasaki-like disease. These two latter genuine vasculitis pathologies were not associated with severe COVID-19 pneumonia. This was distinct from cutaneous vasculitis in severe COVID-19 that demonstrated pauci-immune infiltrates and prominent immunothrombosis that appears to represent a novel immunothrombotic vasculitis mimic contributed to by RNAaemia or potentially diffuse pulmonary venous tree thrombosis with systemic embolization with small arteriolar territory occlusion, although the latter remains unproven. Herein, we also performed a systematic literature review of COVID-19 vasculitis and reports of post-SARS-CoV-2 vaccination related vasculitis with respect to the commonly classified pre-COVID vasculitis groupings. Across the vasculitis spectrum, we noted that Goodpasture’s syndrome was rarely linked to natural SARS-CoV-2 infection but not vaccines. Both the genuine vasculitis in the COVID-19 era and the proposed vasculitis mimic should advance the understanding of both pulmonary and systemic vascular immunopathology.
Hinweise
This article is a contribution to the special issue on: Inflammation in vascular diseases - Guest Editors: Mariana Kaplan & Peter Grayson

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction- The Scope of SARS-CoV-2 Related Vascular Pathology

The novel highly transmissible SARS-CoV-2 virus resulted in fatal pneumonia in a subset of patients and quickly garnered great interest in the cardiovascular and rheumatology arenas, because of the prominent vascular immunopathology. The most striking pathological feature was extensive viral alveolitis but also vascular thrombosis and reports of vascular wall inflammation [1, 2]. Compelling evidence for extrapulmonary vasculitis and vasculitis mimics also emerged during the COVID-19 pandemic which are described in in this article. We also review the extant literature by performing a systematic literature review up until September 2021 that has reported genuine vasculitis in SARS-CoV-2 infection, and we also review post-COVID-19 vaccinations for an emergent vasculitis signal given that most available SARS-CoV-2 vaccines are directed to the spike protein which engages the ACE2 receptor that is known to be expressed on endothelial cells[3]. We also cover the totality of systemic vascular complications—whether vasculopathy, vasculitis or vasculitis mimics and describe how these appear to be independent of productive infection of vascular endothelial cells. We will also focus on extrapulmonary vascular pathology including genuine autoimmune vasculitis (referred also as true, bespoke, or bona fide vasculitis in this text) and a novel potential vasculitis mimic related to diffuse immunothrombosis outside the pulmonary territory that has been reported in SARS-CoV-2.

ACE2 Centric Vasculopathy model including vascular infection

The SARS-CoV-2 virus employs the ACE2 receptor for cellular entry with this receptor having a widespread upper and lower respiratory tract distribution from nasal epithelium to the alveoli, most notably alveolar type 2 pneumocytes[4, 5] but the ACE2 receptor also has a well-established role in the cardiovascular system[6, 7]. Also, the comparatively minuscule SARS epidemic at the turn of the millennium had already provided substantial information about this related structurally close beta-coronavirus that also used the ACE2 receptor [4]. Beyond the lower respiratory tract symptoms of COVID-19 pneumonia explained by ACE2 receptor expression, upper respiratory tract symptoms of anosmia and pharyngitis could also be linked to high local ACE2 receptor expression levels[8]. Prior data also showed ACE-2 receptor expression on other cell types including endothelial cells and cardiomyocytes[4, 9] and provided potential pointers to the novel emergent cardiovascular pathology in the initial COVID-19 wave. Independently, of these observations in SARS, the role of ACE2 receptor as a regulator of cardiovascular system including hypertension, myocardial injury, obesity and diabetes was reported in several models systems, but to variable extents[10, 11]. Cardiac enzyme elevations and lung and systemic vasculopathy quickly cemented the notion that ACE-2 dysfunction in the cardiovascular system was a key mortality driver in emergent COVID-19 pneumonia and associated vascular pathology [12, 13]. Accordingly, early in the pandemic, there was great interest in the influence of cardiovascular drugs that modulated ACE2 expression including Renin–Angiotensin inhibitors, nonsteroidal anti-inflammatory, and many others [3, 1416].
The prior SARS epidemic was associated with both pulmonary capillary and larger vessel thrombosis and also reports of viral myocarditis and in common with SARS-CoV-2, the SARS virus also utilized the ACE2 receptor [1719]. Given that recombinant ACE-2 mitigates against experimental pneumocyte injury, and since SARS-CoV-2 spike protein can downregulate ACE-2[20], it has been considered that SARS-CoV-2 derived spike protein without actual infection might trigger endothelial cell dysregulation and immune activation[21]. A third beta coronavirus termed Middle East respiratory syndrome coronavirus (MERS-CoV) has been another twenty-first century emergent coronavirus and limited pathological literature supports the idea of an identical immunopathology with pneumonia and reported immunothrombosis and an increased mortality in subjects with cardiovascular risk factors[2224]. As this latter virus shows an identical immunopathology but does not target the ACE2 receptor [23, 25] and recent studies showing low or even absent endothelial cells ACE2 expression [2628], then it is likely that novel beta coronaviruses are capable of mediating immunopathology, including vasculitis, independently of the ACE-2 receptor and that factors extrinsic to ACE-2 appear to be critical to COVID-19 related vasculopathy, which is the focus of this paper.

Histological Reports of Pulmonary Vasculitis in COVID-19 and what it means

Pathology from the primary lung target organ in COVID-19 have consistently shown that viral alveolitis is accompanied by peri-capillary myeloid and lymphoid cell infiltration [29, 30]. Pulmonary capillary, pulmonary arteriolar and pulmonary venular vessel wall inflammation has been associated with extensive and pervasive vascular luminal thrombosis[2, 31]. Initially, it was considered that endothelial inflammation due to direct viral infection or “viral endotheliitis” accounted for the immunothrombosis [29, 32]. However, it has since emerged that endothelial cells have comparatively low ACE2 expression and are also fairly resistant to productive viral infection supporting the concept that a “genuine vasculitis” rather than active infection, may thus account for the SARS-CoV-2 vascular pathology (Fig. 1) [2628]. Indeed, it has likewise emerged that direct endothelial infection with productive SARS-CoV-2 viral replication does not occur in humans [27, 33, 34]. Also, in the prior SARS outbreak, reports of direct endothelial infection were never conclusively shown [35, 36].
Pathological pulmonary changes described as vasculitis have been reported in COVID-19 pneumonia. In one study, around a quarter of subjects had perivascular lymphocyte cuffing or capping, that was described as compatible with vasculitis[31]. In another postmortem study, 4 of 11 cases had predominant macrophage infiltration into the pulmonary arterial wall, and also CD4, CD8 T-cells and B cells were reported, with these histological features being designated as arteritis[37]. One more pathological report described an endarteritis obliterans in conjunction with C5-9 complement pathway activation at the site of vasculitic change[38]. However, the pervasive impact of severe alveolitis with associated inflammation in the juxta-capillary and thin-walled pulmonary vascular system may account for the extensive lung vascular pathology (Fig. 2). We suspect that studies demonstrating juxta-capillary lymphocytic infiltration that have been attributed to a “viral endotheliitis” may reflect lymphocyte migration or infiltration either to or from the closely juxtaposed pneumonic alveolar territory (Fig. 2) [1, 29, 39]. Rather than representing a primary pulmonary vasculitis in the later phases where the SARS-CoV-2 virus is cleared, such microscopic “vasculitis” may be part of the so-called Virchow's triad where a severe extra-vascular alveolitis leading to vascular wall inflammation is linked to vessel wall immune cell infiltration and luminal thrombosis (Fig. 2).
Severe alveolar centric inflammation without actual endothelial infection likely leads to extensive immunothrombosis by a myriad of mechanisms including pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) related endothelial activation[40, 41]. Factors including tissue factor released by activated immune cells in the lungs or systemically also likely trigger coagulation[40]. Viral RNA access to the capillary lumen consequent to damage of the sub-micrometer alveolar–capillary barrier may also contribute to Factor XII and X activation with local coagulation cascade activation [4244]. Presently, the understanding of the COVID-19 lung vascular immunopathology is conceptualized in terms of severe immunothrombosis that constrains SARS-CoV-2 to the alveolar territory[45, 46]. The severity and magnitude of the alveolar centric inflammation leads to profound endothelial cell damage and even endothelial cell death captured under the umbrella term of endothelialopathy[47, 48].

What Did We Know Before The COVID-19 Era About Infection and Vasculitis?

The question of vasculitis as a manifestation of infection disease is not a unique to COVID-19 and was well described phenomenon with the oldest pathogens know for humankind such as syphilis and tuberculosis. In most of the cases, this infectious vasculitis is a consequence of direct invasion, extension of localized focus or septic embolization of bacterial, viral, fungal, or parasitic infections to the endothelial cells and vascular wall which is usually accompanied by intense inflammatory response to the vessel wall and other symptoms related to the main infection and rarely confused with bona fide vasculitis [49, 50]. For example, in syphilis, treponema pallidum invasion of endothelial cells and endothelial barriers such retina, placenta, and blood–brain barrier is believed to be one of the main bacteria virulence factors; hence direct syphilis infection may cause different vasculitic syndromes from central nervous system vasculitis, retinal vasculitis and aortitis [5153]. Other pathogens have been studied as a possible mechanistic triggers in the development of bona fide vasculitis, such as staphylococcus aureus in ANCA associated vasculitis and streptococcal infection in Henoch–Schonlein purpura (HSP), but the causality in these cases is still controversial[54, 55]. Only for a few pathogens has causality been established, such as Hepatitis C Virus (HCV) related cryoglobulinemic vasculitis, which is mediated mainly by the binding of HCV viral particles to IgM with rheumatoid factor activity resulting in the production of cold-precipitable immune complexes, which binds to endothelial cells activating the complement system and inflammatory response [5658].
Vaccinations including influenza vaccine, hepatitis B, Bacille Calmette-Guerin (BCG), and human papillomavirus vaccines, all of which lack live microbes with the exception of BCG, have been associated with occasional vasculitis development [59]. The commonest patterns of vasculitis from the same study were HSP and Kawasaki disease. As most of these vaccines do not contain viable replicating pathogens, these findings suggest a non-specific activation of immunity in susceptible individuals that results in vasculitis.

COVID-19 Pulmonary Pathology versus Behcet's Pulmonary Vasculitis

It is useful to compare the COVID-19 pulmonary vasculopathy with the genuine pulmonary vasculitis in Behcet's Disease (BD) that is usually characterized by an absence of interstitial pathology but pulmonary vascular wall inflammation with neutrophilic inflammation and aneurysmal dilatation[60]. The BD vascular centric inflammation is associated with both superficial and deep immunothrombosis and neutrophilic inflammation[60, 61]. Likewise, neutrophils play a key role in the immunothrombosis associated with COVID-19 with netotic material, platelets and other immune cells forming a key part of the clot in COVID-19 disease [62, 63]. Moreover, in critical COVID-19 patients there is evidence of extensive immune cell activation in the peripheral blood that also includes neutrophils[64].
An interesting facet pointing to shared features between BD and COVID-19 pulmonary vascular involvement is anti-coagulation inefficacy in BD immunothrombosis and likewise in critical COVID-19 pneumonia where full dose anti-coagulation has no benefit and may be potentially detrimental[65]. Therapeutically, corticosteroids may be beneficial in deep venous thrombosis (DVT) and other severe manifestations of BD as in severe COVID-19 patients [66]. Although an element of pulmonary haemorrhage is a histological feature of severe COVID-19, this may be dysregulation perfusion rather than the aneurysmal rupture as seen in BD. In keeping with the fact that the genuine vasculitis associated with BD is completely distinct from the COVID-19 immunopathology is the limited data suggesting exacerbation of BD related vascular pathology being linked to COVID-19 infection or vasculitis. In one report, of ten patients with BD during COVID-19 infection, one patient with long-standing BD and central nervous system involvement reported having a DVT, which was unusual to his BD course and responded to steroids without anticoagulation [67]. We have noted occasional BD flares following SARS-CoV-2 vaccination, but these were restricted to mucocutaneous disease and arthritis[68].

Bona Fide Vasculitis in COVID-19

We performed two separate systemic literature reviews of COVID-19 related vasculitis using the following search engines; PubMed/MEDLINE, Scopus, Google Scholar and Embase. The searches were conducted to include articles until September 2021. In the first search, we looked for reported cases of vasculitis after SARS-CoV-2 infection and the second after COVID-19 vaccination. We used this strategy as we reasoned that systemic infection with severe lung damage may predispose to vasculitis compared to the controlled inoculum strategies of vaccination, where the lung tissue is spared severe damage.
For this purpose, we used the following search string in the first search: (SARS-CoV-2 OR COVID-19 OR "novel Coronavirus" OR "emerging Coronavirus") AND (vasculitis OR "Giant cell arteritis" OR Takayasu OR “large-vessel vasculitis” OR Cogan OR "Polyarteritis nodosa" OR Buerger OR "ANCA-associated vasculitis" OR (“antineutrophil cytoplasmic antibodies” AND vasculitis) OR Wegener OR polyangiitis OR Churg-Strauss OR "Urticarial vasculitis" OR Goodpasture OR “anti-glomerular basement membrane disease” OR “cutaneous small-vessel vasculitis” OR “cutaneous vasculitis” OR “IgA vasculitis” OR “leukocytoclastic vasculitis” OR “Henoch-Schönlein Purpura”).
For the second search strategy in SARS-CoV-2 vaccinated subjects, we used the same search string, but we added the vaccine-related component: (vaccine OR vaccination OR immunisation OR immunization). Medical subheadings (MeSH) terms and wild card truncated words option were used when necessary.
Clinical case reports and case series providing sufficient details were included (for instance, case reports/case series reporting only clinical illustrations/image were excluded). Reports of vasculitis like features including thrombosis and associated complement activation within vessels were not defined as vasculitis but considered as immunothrombotic vasculitis mimics and were not included in the vasculitis groups. Articles were excluded if the vasculitis could be linked to a drug, rather than directly to the SARS-CoV-2 infection or vaccination or if the vasculitis relapsed and not newly diagnosed. Epidemiological surveys computing incidence/prevalence rates but not providing sufficient clinical details were not retained in the analysis. Extensive cross-referencing was applied, to ensure the maximum coverage and to increase the chance of including all relevant articles. Already existing reviews (in particular, systematic reviews) were scanned to include already abstracted data and eventually update them. We did not search for “COVID toes” or Kawasaki Disease (KD) that is linked to Multisystem Inflammatory Syndrome (MIS). Our focus was on previously known systemic vasculitides rather than the two recently defined vasculitis entities.

Vasculitis in SARS-CoV-2 Infection

Excluding Kawasaki disease like vasculitis associated with the MIS spectrum and “COVID toes”, only forty-eight vasculitis cases were described (Table 1) [6994]. These are described starting with large vessel vasculitis.
Table 1
Reported cases of genuine vasculitis in SARS-CoV-2 infection
 
N.of cases
Gender
Age
Underlying conditions
Symptoms
Workup
Timing
Treatment
Outcome
Cutaneous:
CSVV /LCKV[6975] / UV[7682]
CSVV/LCKV:6
UV: 9
7 X M
8 X F
1.5–83
Only in three patients:
1. HTN, TIA, AF, CKD
2. HTN; DM
3. HTN, MI, HF, COPD
One pregnant (a case of UV)
CSVV/LCKV: chilblain-like, ulcerative lesions; arthralgia; constitutional symptoms
UV: Urticarial rash; pruritic rash; constitutional symptoms; arthralgia; abdominal tenderness; conjunctival erythema with periorbital edema; acral nonpitting edema; diarrhea; hypotension
Clinical; Neutrophilia; Lymphopenia; anemia hypoalbuminemia with albuminuria; C3-; LDH + CRP + ESR + ; Cr + ; auto immune serology + ; SARS-Cov-2 serology; skin biopsy; immunolabelled SARS-CoV-2 antigens in skin biopsies;chest CT scans/ HRCT
5 days- 3 weeks after COVID-19 symptoms onset; sometimes, COVID-19 diagnosed after vasculitis onset
CSVV: Paracetamol; steroids
UV:
Antihistamines, steroids, Colchicine, HCQ, heparin if needed
Two deaths (a case of UV and a case of CSVV/LCKV)
ANCA-associated vasculitis[8387]
6
2XF
4XM
25–64
Only in Two patients:
1.DM
2.DM and scleroderma
Fever, respiratory symptoms; GI symptoms
Clinical; ANCA + (MPO in 3 cases, PR3 in 3 cases); Cr + ; Proteinuria; Haematuria; Skin Biopsy;
Renal biopsy; Chest CT scan
Simultaneously or shortly after COVID-19 diagnosis
Glucocorticoids; CYC; PEX; HCQ; rituximab, if needed
Improvement / Resolution Some with end organ damage
IgAV/HSP[8890]
16
13XM
3XF
 ~ 1–78
HTN; Alcohol consumption; HyperL; aortic Stenosis; bladder cancer; Hirschprung disease; Crohn disease;
none or not reported in 10 cases
Fever; rhinorrhoea, cough; chills; dyspnea; myalgia, fatigue; headache; pruritic rash; maculopapular rash, arthralgia or arthritis; GI symptoms (nonbloody diarrhea, hematochezia, vomiting); lower limbs pitting edema; HTN
Clinical; CRP + ; ESR + ; C3-; leucocytosis; Anaemia; thrombocytosis or thrombocytopenia; albumin-; proteinuria; haematuria; Cr + ; hyaline casts; renal biopsy and electron microscopy; skin biopsy; IF
Simultaneously-37 days after COVID-19
Steroids; ABX; anticoagulation; antivirals; NSAIDS; statins; Rituximab, anti-helminthics
One patient died
LVV/GCA[91, 92]
2
2XM
47–50
None
Headache; temporal thickening; paracentral acute middle maculopathy in one case
Clinical; Doppler ultrasound of the right temporal artery; FDG PET-CT scan
Simultaneously (one case);
2 months after COVID-19
N/A in one case; Steroids in the second case
Full resolution
Goodpasture[90, 93, 94]
9
2XM
7xF
27–73
HTN; rheumatic HD; COPD; SLE; asthma; bronchiectasis;
None in two cases
Fever; fatigue; myalgia; GI symptoms; haemoptysis; epistaxis; petechial rash; dyspnoea; ARDS
Clinical; WBC + , lymphocytes-, Cr + and BUN + ; CRP + , ESR + ; proteinuria; chest CT scan
Simultaneously
ABX; steroids; CYC; Rituximab; PEX; Haemodialysis
One patient died
Abbreviation: CSVV- Cutaneous small-vessel vasculitis, LKCV- Leukocytoclastic vasculitis, UV- Urticarial vasculitis ,IgAV- IgA Vasculitis, HSP- Henoch-Schönlein Purpura, ANCA -Antineutrophil cytoplasmic antibodies, LVV – Large vessel vasculitis , (+) - positive/elevated, (-)- Decrease, F- Female, M- Male, HTN- Hypertension ,HyperL- Hyperlipidaemia ,IF- immunofluorescence, AF- atrial fibrillation, TIA- transient ischemic attack, DM- Diabetes mellitus, GI- gastro- intestinal, CKD-Chronic kidney disease ,MI- myocardial infarction, HF- heart failure, COPD- chronic obstructive pulmonary disease, Cr – Creatinine , ARDS- Acute respiratory distress syndrome, HRCT- High-resolution computer tomography, CYC- cyclophosphamide, PEX- plasma exchange or Plasmapheresis , HCQ- Hydroxychloroquine, ABX – Antibiotics.
Large Vessel Vasculitis
There have been occasional reports of large vessel vasculitis including central nervous system vasculitis occurring with severe COVID-19 where imaging demonstrated a pattern resembling GCA, but these were uncommon[91, 92, 95]. We found no cases of Takayasu’s vasculitis following COVID-19.
Medium and Small Vessel Vasculitis- ANCA associated vasculitis
The diagnosis of new ANCA associated vasculitis may be challenging in the context of severe COVID-19 infection due to shared anatomical territories of infection and inflammation. Also, the multisystem nature of severe COVID-19 infection and the fact that a multiplicity of autoantibodies, including anti-neutrophil cytoplasmic antibodies (ANCA) may appear during infection may complicate the diagnosis[96]. One potential mechanism for ANCA development and other autoantibody development in general in the context of COVID-19 infection is the presence of extensive neutrophil infiltration and neutrophil extracellular traps (NETs) at sites of immunothrombosis and tissue necrosis that could contribute to tissue tolerance failure with antibodies formation[9698]. In one report about the prevalence of ANCA in hospitalized COVID-19 patients, 57% of randomly selected patients sera confirmed positive for ANCAs, of which 72% were C-ANCA, and 28% were P-ANCA, but only one sample was myeloperoxidase positive[99]. In the same report, there was a positive association between C-ANCA status with intensive care unit admission but not death. Whether these ANCA autoantibodies play a true role in the pathogenesis and severity of COVID-19 still need to be determined. Also, a plethora of other antibodies have been reported in severe COVID-19 and may reflect tissue tolerance breakdown or occasionally perhaps play a role in pathology[100, 101].
Allowing for these caveats, to the best of our knowledge, there are only six described cases of ANCA associated vasculitis with temporal association with COVID-19 infection; most patients were previously healthy, two females and four males, with an age range from 25 until 64 years. All described cases were ANCA positive, and the diagnosis made simultaneously or shortly after the COVID-19 diagnosis. All patients survived after treatment with immunosuppressive medications with some accruing irreversible end organ damage [8387].
Immunoglobulin A Vasculitis (IgAV)/Henoch-Schönlein Purpura
IgAV is typically a childhood and early adolescent disease and is linked strongly to triggering environmental factors with subsequent immune-complex deposition and complement activation [102]. Sixteen cases of IgAV were described in correlation with COVID-19 infections, some of the cases presented simultaneously with COVID-19, but others appeared more than one month after the infection, the majority were males, and the age range was between 1 to 78 years. A high percentage of the patients suffered from purpuric rash and gastrointestinal symptoms. In some cases, the diagnosis was confirmed by skin or renal biopsy; skin biopsies revealed the classical finding of leukocytoclastic vasculitis, but some lacked positive IgA staining [89]. A Kidney biopsy was reported in six patients, all kidney biopsies were positive for IgA on direct immunofluorescence staining, usually in the context of a mesangial proliferative pathology [8890]. Electron microscopy showed mesangial and subendothelial immune deposits to podocyte effacement in IgAV [8890].
Goodpasture's syndrome
Given that the alveolus and the endothelium are prominent targets in severe COVID-19, then a genuine concern exists that severe SARS-CoV-2 infection with pulmonary basement membrane damage could lead to tolerance failure and the development of autoimmunity (Fig. 3). Indeed, autoimmunity to a plethora of antigens including cytokines and endothelial molecules and others are well reported in severe or critical COVID-19[103, 104]. An interesting case report supplemented by epidemiological data trends in the London region of the UK showed cases of Goodpasture's syndrome increasing following COVID-19 [93]. Thankfully, Goodpasture's syndrome cases in the literature have totalled only nine cases, and it is interesting why this is comparatively underrepresented given the target tissue and the multiplicity of other reported autoantibodies.

The SARS-CoV-2 Virus Bespoke Vasculitides

Although not included in our systemic literature review, “COVID toes” and KD like vasculitis were the most frequently reported vasculitides with COVID-19 infection and discussed separately below. Otherwise, healthy people presented during the first phase of the COVID-19 outbreak without respiratory symptoms and SARS-CoV-2 PCR test negativity but with chilblain lesions, of erythema or dusky discolouration of the digits in particular[105107]. Histological evaluation of these lesions confirmed a small vessel vasculitis with lymphocytic infiltration and interferon pathway activation within tissue as evidenced by MX-1 staining[108] suggesting a interferonpathy disease mechanism akin to monogenic systemic lupus erythematosus variants as first defined by Crow et al.[109, 110]. These lesions were identical to the typical chilblain pattern of autoimmune chilblain lesions exhibiting both a similar pattern of lymphocytic infiltration and interferon staining[111, 112].
A severe inflammatory reaction termed MIS has been reported in children and encompasses cardiac aneurysmal involvement [113116], and it seems to be the most common reported vasculitis syndrome after SARS-CoV-2 infection with estimated incidence of 316 person per 1,000,000 SARS-CoV-2 infection in person younger than 21 years and most closely resembles KD[117, 118]. Prior to the emergence of SARS-CoV-2 this vasculitic pathology was a typical feature of KD in children under five of age but the emergent SARS-CoV-2 related pathology affects older children who develop MIS [119, 120]. Unlike COVID-19 pneumonia, young subjects who develop MIS may be a PCR negative for SARS-CoV-2 but usually antibody positive [113, 118, 120122], suggesting some sort of immune hypersensitivity reaction to SARS-CoV-2. Beyond the KD-like vasculitis, patients may also experience prominent abdominal symptoms[121] and different mechanisms have been advocated to explain this genuine vasculitis related pathology. Given that KD itself is poorly understood, it remains unclear how KD like disease develops with different theories including potentially a superantigen mediated disease related to persistent spike protein antigen [123].

SARS-CoV-2 Vaccination Reports of Vasculitis.

There is a well-recognized association between natural infection or vaccination and the occasional precipitation or activation of either autoinflammatory or autoimmune diseases, but causality is not certain. Given the known role of spike protein potentially downregulating ACE in experimental settings then a theoretical concern exists around spike protein-based coronavirus vaccines and vasculitis pathology [20, 124]. This is mitigated against by very low level of systemic protein expression with vaccines based on RNA backbones[125]. A few well documented rare immune reactions have been described following SARS-CoV-2 vaccination, most notably vaccine-induced immune thrombotic thrombocytopenia (VITT) after DNA vaccination and myocarditis following RNA vaccination [126130]. Furthermore, spike protein-based vaccines permit investigation of whether a specific viral component, namely spike protein, might be linked to recognized immunopathology such as KD like disease or other vasculitis.
As of September 2021, there were fifty cases [131151] of reported vasculitis with temporal association with COVID-19 vaccination. Of these cases, only twenty-three peer-reviewed cases are newly diagnosed vasculitis (Table 2) [131147]. Cutaneous vasculitides were the most common type of reported vasculitis, with a total of ten patients described in the literature[131138], and noting that we excluded “COVID toes” from our search these included cutaneous small-vessel vasculitis, leukocytoclastic vasculitis and urticarial vasculitis. This was followed by ANCA associated vasculitis with total of five patients[139142, 144] and then with IgA vasculitis or HSP with four reported cases[143146]. Eight cases had underlying comorbidities. Only three of these cases had prior SARS-CoV-2 infection. Symptom onset occurred from a few hours to four weeks after COVID-19 vaccination, all patients had a good outcome, and full recovery was generally reported.
Table 2
Reported cases of genuine vasculitis after COVID-19 vaccination
 
N.of cases
Gender
Age
Underlying conditions
Symptoms
Workup
Type of vaccine
Treatment
Cutaneous:
CSVV [131134]/LCKV[135137]/ UV[136, 138]
CSVV:4
LCKV: 4
UA: 2
3 X M
5 X F
2 × N/A
31–83
Only in two patients:
1.HTN; HyperL; mechanical AVR (on warfarin); Algy to ibuprofen (mild rash)
2.HTN; Hypothyroidism
CSVV: Purpuric rash; fever; Itchy maculo-papular rash; pitting oedema
LCKV: Purpura
UV: Urticarial rash; fever; arthralgia
Clinical; CRP + ESR + ; auto immune serology + ; skin biopsy
1 X Jan
1 X Oxf
3X Pfi
1X COVAXIN®;
2XMod
1X Whole Virion inactivated
1XN/A
CSVV: Systemic or topical steroids /anti Histamine
LCKV: NA in two cases. Sys ABX and Topical Steroids in one case
UV: Oral indomethacin, topical calamine lotion, levocetirizine Antihistamines, steroids, dapsone
ANCA-associated vasculitis[139142, 144]
5
2XF
3XM
37–81
Only in two patients:
1. Graves’ disease
2. T2DM; HTN;PAF
Flu-like; Anorexia; Rash; fever; pain; Haemoptysis; GI symptoms
Clinical; ANCA + ; CRP + ; Cr + ; Proteinuria; Haematuria; Skin Biopsy
Renal biopsy (in two cases); Chest CT scan; [18F]FDG-PET/CT (increased uptake in middle-sized vessels)
2X Pfi
2X Oxf
1XMod
Sys steroids, oral CYC/
Rituximab/plasmapheresis if needed
IgAV [143, 144]/HSP[145, 146]
IgAV:2
HSP:2
IgAV:2XM
HSP:2XF
39–72
HTN; MI;T2DM; obesity; asthma. Osteosarcoma; intercostal shingles; tonsillectomy
Hashimoto’s thyroiditis; Assisted reproductive therapy
Flu-like illness; Purpura; Arthralgia; Fever; macroscopic haematuria
IgAV: Clinical; CRP + ; Cr + ; Skin biopsy one case; Renal biopsy in one case;
HSP: Clinical; CRP + ; ANA + , RF + ; Microscopic haematuria
2XOxf
1XMod
1XPfi
IgAV: Sys steroids, CYC when needed
HSP: Sys steroids
Vasculitis NOS[135]
3
N/A
N/A
N/A
N/A
N/A
2xMod
1XPfi
N/A
LVV [147]
1
F
78
None
Cephalalgia, Osteomyalgia
[18F] FDG-PET/CT + (large arteries of the legs); CRP + , ESR + 
Mod
N/A
Abbreviation: CSVV- Cutaneous small-vessel vasculitis, LKCV- Leukocytoclastic vasculitis, UV- Urticarial vasculitis ,IgAV- IgA Vasculitis, HSP- Henoch-Schönlein Purpura, ANCA -Antineutrophil cytoplasmic antibodies, LVV – Large vessel vasculitis , + - positive/elevated, F- Female, M- Male, HTN- Hypertension ,HyperL- Hyperlipidemia ,AVR -Aortic valve replacement ,Algy – Allergy ,Mod -Moderna COVID-19 vaccine (mRNA-1273),Jan- Janssen Ad26.COV2.S, Oxf- Oxford-AstraZeneca, Pfi- BNT162B2/Pfizer, Sys- systemic ,PAF- paroxysmal atrial fibrillation, T2DM- Type 2 Diabetes mellitus, GI- gastro- intestinal ,Cr – Creatinine, RF- rheumatoid factor, CT- computer tomography, CYC- cyclophosphamide , ABX – Antibiotics, N/A-not available, NOS-not otherwise specified
Despite anecdotal reports of COVID-19 vaccine-induced MIS, larger cohorts have not confirmed as association. In one report of 20 patients with MIS, seven reported to have MIS after COVID-19 vaccination; all patients had evidence of previous COVID-19 infection, so COVID-19 induced MIS was not ruled out in these cases[152]. In another report of 107 pediatric patients admitted to intensive care units in France within two months period because of MIS, 33 of them were eligible for vaccination but none were fully vaccinated, and only 7 received one dose [135, 153]. It remains uncertain whether spike protein vaccines alone can trigger MIS, thus incriminating this antigen that MIS.
The cutaneous “COVID toes” has been reported in temporal association with COVID-19 vaccination, which likely reflects poorly understood excessive type 1 interferon response to spike protein encoding nucleic acid and is reminiscent of SARS-CoV-2 where replicating virus is not usually detectable in COVID toes [154, 155].
As stated earlier, Goodpasture syndrome was associated with the COVID-19 pandemic[93] With respect to relevant vaccination, only two cases of questionable anti-GBM nephritis without pulmonary involvement were reported. In the first case, anti-GBM antibodies were negative, and the diagnosis was suspected based on histology[156]. In the putative second case anti-GBM nephritis was accompanied by mesangial IgA deposit [157]. In some way, this difference in Goodpasture frequency may be explained by the absence of tissue damage after the COVID-19 vaccination as opposed to SARS-CoV-2 infection in which basement membrane damage provokes the production of anti-basement membrane antibodies but case numbers are too small to be definitive (Fig. 3).

Potential COVID Vasculitis Mimic Related to Small Vessel Thrombosis

As distinct from the vasculitis occurring in mild COVID-19 disease that is IFN associated, a second clinical pattern of cutaneous vasculitis has been reported in severe or critical COVID-19. It is possible that some of the aforementioned diagnosed vasculitides may in fact fall within the vasculitis mimic category, that is linked to primary immunothrombotic mechanisms. Retiform purpura, livedo reticularis and purpuric lesions have a quite distinct immunopathology and are characterized by vessel lumen thrombosis and pauci-immune infiltration and an absence of IFN pathway protein expression in the tissue[105, 108, 158, 159].
Akin to lung pathology in COVID-19 pneumonia, there is a lack of evidence for direct endothelial infection. However, immunochemistry has shown staining for SARS-CoV-2 related proteins in the vascular lumen[32, 159161]. It has been suggested that such pulmonary shedding of SARS-CoV-2 non-replicating viral fragments or “pseudo-virus” (spike, envelope, membrane proteins with or without viral RNA) may lead to ACE-2 engagement with resultant endothelial disturbance triggering local inflammation and then the wider thrombotic and vasculitic pathology[162]. It has also emerged that viral protein antigenaemia is strongly linked to systemic inflammatory responses and severity of COVID-19 pneumonia. These pauci immune lesions have been associated with complement C5-9 terminal pathway activation. Whether complement is playing a role driver role in such thrombotic immunopathology is unclear as vascular ischemia damage may also activate the complement system[40, 159, 163166].
We have proposed two simple mechanism for this vasculitis mimic immunopathology[167]. First, physiological immunothrombosis contains the SARS-CoV-2 virus in the pulmonary territory but breakdown of the alveolar-vascular barrier with translocation of viral RNA and debris to the systemic circulation including small arteriolar, capillary, and venous circulation is associated with RNAaemia activation of immunothrombosis at sites distant from the lungs (Fig. 4). Given that the extensive pulmonary immunothrombosis is also evident in the pulmonary venular territory in postmortem studies and occasionally thrombosis in the pulmonary vein root on computed tomography pulmonary angiogram studies, we proposed that embolization from this territory distal to the alveolar-capillary clot filtration system offered a novel hitherto unappreciated source of embolization[168, 169]. Further detailed postmortem pathological studies of lung tissue and distal organs are needed to confirm this theory.
Critical COVID-19 pneumonia is associated with organ ischaemia in many territories including the brain where cryptogenic strokes are commonly reported[170, 171]. Some of these ischemic complications, especially pulmonary embolism and venous thrombosis are much commoner with COVID-19 compared to influenza A pneumonia and likely reflects the differential alveolar versus bronchial tissue tropism[172174]. These lesions do not show typical histological features of vasculitis and are pauci immune. Haematuria is reported in over 40% of cases with renal involvement in severe COVID-19 pneumonia but histologically the changes are of ischaemic injury rather than vasculitis[175, 176]. Likewise, intestinal ischemia is a common manifestation and histology shows diffuse ischemic change rather than vasculitis[177]

Conclusions

The massive alveolar-capillary territory juxtaposition forms the key insight to understanding the physiological role of vascular immunity in containing infection within the alveolar space. This is associated with extensive non-infectious endotheliitis or endothelialopathy that appears to be predominantly cytokine and inflammatory mediator driven and dysregulation of this pathway leads to local and systemic immunothrombosis that serves as a vasculitis mimic. Despite the initial obvious link with the ACE2 receptor, it appears that the vasculitis mimic mechanism is not strongly linked to this. Occasional vasculitis does occur with COVID-19 and with the exception of the rare MIS pathology the vasculitis is usually self-limiting and furthermore both KD like vasculitis and “COVID toes” do not appear to be linked to productive viral infection in airways or the lungs. Mechanistically, COVID toes have been best conceptualized in relationship to excessive interferon driven response in the skin with strong similarities to the monogenic type 1 interferonopathies that caused chilblain lupus[108]. MIS exhibits features of KD in some patients including coronary vasculitis, but the mechanism remains unclear as KD itself is not well understood. There is no evidence that SARS-CoV-2 vaccine strategies, many of which encode for spike protein, have contributed to the bone fide vasculitis pathology apart from occasional reports of “COVID toes” following vaccination. Overall, the COVID-19 pandemic has served a highly informative model for a refined understanding of vascular immunopathology mechanism following the global emergence of a novel RNA virus.

Declarations

Ethics declarations

Additional declarations for articles in life science journals that report the results of studies involving humans and /or animals.
Not applicable.

Ethics approval

Not applicable.
Not applicable.
Not applicable.

Conflict of interest

The authors declare no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Fox SE et al (2020) Pulmonary and cardiac pathology in African American patients with COVID-19: an autopsy series from New Orleans. Lancet Respir Med 8(7):681–686PubMedPubMedCentralCrossRef Fox SE et al (2020) Pulmonary and cardiac pathology in African American patients with COVID-19: an autopsy series from New Orleans. Lancet Respir Med 8(7):681–686PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat Milross L et al (2022) Post-mortem lung tissue: the fossil record of the pathophysiology and immunopathology of severe COVID-19. Lancet Respir Med 10(1):95–106PubMedCrossRef Milross L et al (2022) Post-mortem lung tissue: the fossil record of the pathophysiology and immunopathology of severe COVID-19. Lancet Respir Med 10(1):95–106PubMedCrossRef
3.
Zurück zum Zitat Zhang H et al (2020) Angiotensin-converting enzyme 2 (ACE2) as a SARS-CoV-2 receptor: molecular mechanisms and potential therapeutic target. Intensive Care Med 46(4):586–590PubMedPubMedCentralCrossRef Zhang H et al (2020) Angiotensin-converting enzyme 2 (ACE2) as a SARS-CoV-2 receptor: molecular mechanisms and potential therapeutic target. Intensive Care Med 46(4):586–590PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Hamming I et al (2004) Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. J Pathol 203(2):631–7PubMedPubMedCentralCrossRef Hamming I et al (2004) Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. J Pathol 203(2):631–7PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Hoffmann M et al (2020) SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 181(2):271-280.e8PubMedPubMedCentralCrossRef Hoffmann M et al (2020) SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 181(2):271-280.e8PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Crackower MA et al (2002) Angiotensin-converting enzyme 2 is an essential regulator of heart function. Nature 417(6891):822–828PubMedCrossRef Crackower MA et al (2002) Angiotensin-converting enzyme 2 is an essential regulator of heart function. Nature 417(6891):822–828PubMedCrossRef
7.
Zurück zum Zitat Zhong J et al (2010) Angiotensin-Converting Enzyme 2 Suppresses Pathological Hypertrophy, Myocardial Fibrosis, and Cardiac Dysfunction. Circulation 122(7):717–728PubMedCrossRef Zhong J et al (2010) Angiotensin-Converting Enzyme 2 Suppresses Pathological Hypertrophy, Myocardial Fibrosis, and Cardiac Dysfunction. Circulation 122(7):717–728PubMedCrossRef
8.
Zurück zum Zitat Ziegler CG et al (2020) SARS-CoV-2 receptor ACE2 is an interferon-stimulated gene in human airway epithelial cells and is detected in specific cell subsets across tissues. Cell 181(5):1016-1035e19PubMedPubMedCentralCrossRef Ziegler CG et al (2020) SARS-CoV-2 receptor ACE2 is an interferon-stimulated gene in human airway epithelial cells and is detected in specific cell subsets across tissues. Cell 181(5):1016-1035e19PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Nicin L et al (2020) Cell type-specific expression of the putative SARS-CoV-2 receptor ACE2 in human hearts. Eur Heart J 41(19):1804–1806PubMedCrossRef Nicin L et al (2020) Cell type-specific expression of the putative SARS-CoV-2 receptor ACE2 in human hearts. Eur Heart J 41(19):1804–1806PubMedCrossRef
11.
Zurück zum Zitat Oudit GY et al (2003) The role of ACE2 in cardiovascular physiology. Trends Cardiovasc Med 13(3):93–101PubMedCrossRef Oudit GY et al (2003) The role of ACE2 in cardiovascular physiology. Trends Cardiovasc Med 13(3):93–101PubMedCrossRef
13.
Zurück zum Zitat Rivellese F, Prediletto E (2020) ACE2 at the centre of COVID-19 from paucisymptomatic infections to severe pneumonia. Autoimmun Rev 19(6):102536–102536PubMedPubMedCentralCrossRef Rivellese F, Prediletto E (2020) ACE2 at the centre of COVID-19 from paucisymptomatic infections to severe pneumonia. Autoimmun Rev 19(6):102536–102536PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Semenzato L et al (2021) Antihypertensive Drugs and COVID-19 Risk. Hypertension 77(3):833–842PubMedCrossRef Semenzato L et al (2021) Antihypertensive Drugs and COVID-19 Risk. Hypertension 77(3):833–842PubMedCrossRef
17.
Zurück zum Zitat Oudit G et al (2009) SARS-coronavirus modulation of myocardial ACE2 expression and inflammation in patients with SARS. Eur J Clin Invest 39(7):618–625PubMedPubMedCentralCrossRef Oudit G et al (2009) SARS-coronavirus modulation of myocardial ACE2 expression and inflammation in patients with SARS. Eur J Clin Invest 39(7):618–625PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Chong PY et al (2004) Analysis of deaths during the severe acute respiratory syndrome (SARS) epidemic in Singapore: challenges in determining a SARS diagnosis. Arch Pathol Lab Med 128(2):195–204PubMedCrossRef Chong PY et al (2004) Analysis of deaths during the severe acute respiratory syndrome (SARS) epidemic in Singapore: challenges in determining a SARS diagnosis. Arch Pathol Lab Med 128(2):195–204PubMedCrossRef
19.
Zurück zum Zitat Nicholls JM et al (2003) Lung pathology of fatal severe acute respiratory syndrome. The Lancet 361(9371):1773–1778CrossRef Nicholls JM et al (2003) Lung pathology of fatal severe acute respiratory syndrome. The Lancet 361(9371):1773–1778CrossRef
20.
21.
Zurück zum Zitat Nuovo GJ et al (2021) Endothelial cell damage is the central part of COVID-19 and a mouse model induced by injection of the S1 subunit of the spike protein. Ann Diagn Pathol 51:151682–151682PubMedCrossRef Nuovo GJ et al (2021) Endothelial cell damage is the central part of COVID-19 and a mouse model induced by injection of the S1 subunit of the spike protein. Ann Diagn Pathol 51:151682–151682PubMedCrossRef
23.
Zurück zum Zitat Memish ZA et al (2020) Middle East respiratory syndrome. The Lancet 395(10229):1063–1077CrossRef Memish ZA et al (2020) Middle East respiratory syndrome. The Lancet 395(10229):1063–1077CrossRef
24.
Zurück zum Zitat Giannis D, Ziogas IA, Gianni P (2020) Coagulation disorders in coronavirus infected patients: COVID-19, SARS-CoV-1, MERS-CoV and lessons from the past. J Clin Virol 127:104362PubMedPubMedCentralCrossRef Giannis D, Ziogas IA, Gianni P (2020) Coagulation disorders in coronavirus infected patients: COVID-19, SARS-CoV-1, MERS-CoV and lessons from the past. J Clin Virol 127:104362PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Nicosia RF et al (2021) COVID-19 vasculopathy: mounting evidence for an indirect mechanism of endothelial injury. Am J Pathol 191(8):1374–1384 Nicosia RF et al (2021) COVID-19 vasculopathy: mounting evidence for an indirect mechanism of endothelial injury. Am J Pathol 191(8):1374–1384
27.
Zurück zum Zitat McCracken IR et al (2021) Lack of evidence of angiotensin-converting enzyme 2 expression and replicative infection by SARS-CoV-2 in human endothelial cells. Circulation 143(8):865–868PubMedPubMedCentralCrossRef McCracken IR et al (2021) Lack of evidence of angiotensin-converting enzyme 2 expression and replicative infection by SARS-CoV-2 in human endothelial cells. Circulation 143(8):865–868PubMedPubMedCentralCrossRef
28.
30.
Zurück zum Zitat Polak SB, et al (2020) A systematic review of pathological findings in COVID-19: a pathophysiological timeline and possible mechanisms of disease progression. Modern pathology : an official journal of the United States and Canadian Academy of Pathology, Inc 33(11) 2128–2138 Polak SB, et al (2020) A systematic review of pathological findings in COVID-19: a pathophysiological timeline and possible mechanisms of disease progression. Modern pathology : an official journal of the United States and Canadian Academy of Pathology, Inc 33(11) 2128–2138
31.
Zurück zum Zitat Bussani R, et al (2020) Persistence of viral RNA, pneumocyte syncytia and thrombosis are hallmarks of advanced COVID-19 pathology. EBioMedicine 61 Bussani R, et al (2020) Persistence of viral RNA, pneumocyte syncytia and thrombosis are hallmarks of advanced COVID-19 pathology. EBioMedicine 61
33.
Zurück zum Zitat Goldsmith CS et al (2020) Electron microscopy of SARS-CoV-2: a challenging task. The Lancet 395(10238):e99CrossRef Goldsmith CS et al (2020) Electron microscopy of SARS-CoV-2: a challenging task. The Lancet 395(10238):e99CrossRef
34.
Zurück zum Zitat Schimmel L et al (2021) Endothelial cells are not productively infected by SARS-CoV-2. Clinical & translational immunology 10(10):e1350CrossRef Schimmel L et al (2021) Endothelial cells are not productively infected by SARS-CoV-2. Clinical & translational immunology 10(10):e1350CrossRef
35.
Zurück zum Zitat Nicholls JM et al (2006) Time course and cellular localization of SARS-CoV nucleoprotein and RNA in lungs from fatal cases of SARS. PLoS Medicine 3(2):e27PubMedPubMedCentralCrossRef Nicholls JM et al (2006) Time course and cellular localization of SARS-CoV nucleoprotein and RNA in lungs from fatal cases of SARS. PLoS Medicine 3(2):e27PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat To K, Lo AW (2004) Exploring the pathogenesis of severe acute respiratory syndrome (SARS): the tissue distribution of the coronavirus (SARS-CoV) and its putative receptor, angiotensin-converting enzyme 2 (ACE2). J Pathol 203(3):740–743PubMedPubMedCentralCrossRef To K, Lo AW (2004) Exploring the pathogenesis of severe acute respiratory syndrome (SARS): the tissue distribution of the coronavirus (SARS-CoV) and its putative receptor, angiotensin-converting enzyme 2 (ACE2). J Pathol 203(3):740–743PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Calabrese F, et al (2020) Pulmonary pathology and COVID-19: lessons from autopsy. The experience of European Pulmonary Pathologists. Virchows Archiv: an international journal of pathology 477(3) 359–372 Calabrese F, et al (2020) Pulmonary pathology and COVID-19: lessons from autopsy. The experience of European Pulmonary Pathologists. Virchows Archiv: an international journal of pathology 477(3) 359–372
40.
Zurück zum Zitat Loo J, Spittle DA, Newnham M (2021) COVID-19, immunothrombosis and venous thromboembolism: biological mechanisms. Thorax 76(4):412–420PubMedCrossRef Loo J, Spittle DA, Newnham M (2021) COVID-19, immunothrombosis and venous thromboembolism: biological mechanisms. Thorax 76(4):412–420PubMedCrossRef
41.
Zurück zum Zitat Land WG (2021) Role of DAMPs in respiratory virus-induced acute respiratory distress syndrome—with a preliminary reference to SARS-CoV-2 pneumonia. Genes Immun 22(3):141–160PubMedPubMedCentralCrossRef Land WG (2021) Role of DAMPs in respiratory virus-induced acute respiratory distress syndrome—with a preliminary reference to SARS-CoV-2 pneumonia. Genes Immun 22(3):141–160PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Fei Y et al (2020) Coagulation DysfunctionA Hallmark in COVID-19. Arch Pathol Lab Med 144(10):1223–1229PubMedCrossRef Fei Y et al (2020) Coagulation DysfunctionA Hallmark in COVID-19. Arch Pathol Lab Med 144(10):1223–1229PubMedCrossRef
43.
Zurück zum Zitat Englert H, et al (2021) Defective NET clearance contributes to sustained FXII activation in COVID-19-associated pulmonary thrombo-inflammation. EBioMedicine 67 Englert H, et al (2021) Defective NET clearance contributes to sustained FXII activation in COVID-19-associated pulmonary thrombo-inflammation. EBioMedicine 67
44.
Zurück zum Zitat Wang J et al (2021) SARS-CoV-2 infection induces the activation of tissue factor–mediated coagulation via activation of acid sphingomyelinase. Blood 138(4):344–349 Wang J et al (2021) SARS-CoV-2 infection induces the activation of tissue factor–mediated coagulation via activation of acid sphingomyelinase. Blood 138(4):344–349
45.
Zurück zum Zitat McGonagle D et al (2020) Immune mechanisms of pulmonary intravascular coagulopathy in COVID-19 pneumonia. The Lancet Rheumatology 2(7):e437–e445PubMedPubMedCentralCrossRef McGonagle D et al (2020) Immune mechanisms of pulmonary intravascular coagulopathy in COVID-19 pneumonia. The Lancet Rheumatology 2(7):e437–e445PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat McGonagle D, Bridgewood C, Meaney JF (2021) A tricompartmental model of lung oxygenation disruption to explain pulmonary and systemic pathology in severe COVID-19. Lancet Respir Med 9(6):665–672 McGonagle D, Bridgewood C, Meaney JF (2021) A tricompartmental model of lung oxygenation disruption to explain pulmonary and systemic pathology in severe COVID-19. Lancet Respir Med 9(6):665–672
47.
Zurück zum Zitat Goshua G et al (2020) Endotheliopathy in COVID-19-associated coagulopathy: evidence from a single-centre, cross-sectional study. The Lancet Haematology 7(8):e575–e582PubMedPubMedCentralCrossRef Goshua G et al (2020) Endotheliopathy in COVID-19-associated coagulopathy: evidence from a single-centre, cross-sectional study. The Lancet Haematology 7(8):e575–e582PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Pagnoux C, Cohen P, Guillevin L (2006) Vasculitides secondary to infections. Clin Exp Rheumatol 24(2):S71PubMed Pagnoux C, Cohen P, Guillevin L (2006) Vasculitides secondary to infections. Clin Exp Rheumatol 24(2):S71PubMed
50.
52.
Zurück zum Zitat Kakumani PL, Hajj-Ali RA (2009) A forgotten cause of central nervous system vasculitis. J Rheumatol 36(3):655–655PubMedCrossRef Kakumani PL, Hajj-Ali RA (2009) A forgotten cause of central nervous system vasculitis. J Rheumatol 36(3):655–655PubMedCrossRef
53.
Zurück zum Zitat Lithgow KV et al (2020) Identification of the Neuroinvasive Pathogen Host Target, LamR, as an Endothelial Receptor for the Treponema pallidum Adhesin Tp0751. Msphere 5(2):e00195-e220PubMedPubMedCentralCrossRef Lithgow KV et al (2020) Identification of the Neuroinvasive Pathogen Host Target, LamR, as an Endothelial Receptor for the Treponema pallidum Adhesin Tp0751. Msphere 5(2):e00195-e220PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Kallenberg CG (2011) Pathogenesis of ANCA-associated vasculitides. Ann Rheum Dis 70(Suppl 1):i59–i63PubMedCrossRef Kallenberg CG (2011) Pathogenesis of ANCA-associated vasculitides. Ann Rheum Dis 70(Suppl 1):i59–i63PubMedCrossRef
55.
Zurück zum Zitat Wang JJ et al (2020) Association of the infectious triggers with childhood Henoch-Schonlein purpura in Anhui province, China. J Infect Public Health 13(1):110–117PubMedCrossRef Wang JJ et al (2020) Association of the infectious triggers with childhood Henoch-Schonlein purpura in Anhui province, China. J Infect Public Health 13(1):110–117PubMedCrossRef
57.
Zurück zum Zitat Muñoz-Grajales C, Pineda JC (2015) Pathophysiological relationship between infections and systemic vasculitis. Autoimmune diseases 2015 Muñoz-Grajales C, Pineda JC (2015) Pathophysiological relationship between infections and systemic vasculitis. Autoimmune diseases 2015
58.
Zurück zum Zitat Dammacco F, Sansonno D (2013) Therapy for hepatitis C virus–related cryoglobulinemic vasculitis. N Engl J Med 369(11):1035–1045PubMedCrossRef Dammacco F, Sansonno D (2013) Therapy for hepatitis C virus–related cryoglobulinemic vasculitis. N Engl J Med 369(11):1035–1045PubMedCrossRef
59.
Zurück zum Zitat Bonetto C et al (2016) Vasculitis as an adverse event following immunization–systematic literature review. Vaccine 34(51):6641–6651PubMedCrossRef Bonetto C et al (2016) Vasculitis as an adverse event following immunization–systematic literature review. Vaccine 34(51):6641–6651PubMedCrossRef
60.
Zurück zum Zitat Greco A et al (2018) Behçet’s disease: New insights into pathophysiology, clinical features and treatment options. Autoimmun Rev 17(6):567–575PubMedCrossRef Greco A et al (2018) Behçet’s disease: New insights into pathophysiology, clinical features and treatment options. Autoimmun Rev 17(6):567–575PubMedCrossRef
61.
Zurück zum Zitat Le Joncour A et al (2019) Critical role of neutrophil extracellular traps (NETs) in patients with Behcet’s disease. Ann Rheum Dis 78(9):1274–1282PubMedCrossRef Le Joncour A et al (2019) Critical role of neutrophil extracellular traps (NETs) in patients with Behcet’s disease. Ann Rheum Dis 78(9):1274–1282PubMedCrossRef
62.
Zurück zum Zitat Barnes BJ et al (2020) Targeting potential drivers of COVID-19: Neutrophil extracellular traps. The Journal of experimental medicine 217(6):e20200652PubMedPubMedCentralCrossRef Barnes BJ et al (2020) Targeting potential drivers of COVID-19: Neutrophil extracellular traps. The Journal of experimental medicine 217(6):e20200652PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Middleton EA et al (2020) Neutrophil extracellular traps contribute to immunothrombosis in COVID-19 acute respiratory distress syndrome. Blood 136(10):1169–1179PubMedCrossRef Middleton EA et al (2020) Neutrophil extracellular traps contribute to immunothrombosis in COVID-19 acute respiratory distress syndrome. Blood 136(10):1169–1179PubMedCrossRef
64.
Zurück zum Zitat Zuo Y, et al (2020) Neutrophil extracellular traps in COVID-19. JCI insight 5(11). Zuo Y, et al (2020) Neutrophil extracellular traps in COVID-19. JCI insight 5(11).
65.
Zurück zum Zitat REMAP-CAP A.-a., A. Investigators (2021) Therapeutic anticoagulation with heparin in critically ill patients with Covid-19. N Engl J Med 385(9):777–789 REMAP-CAP A.-a., A. Investigators (2021) Therapeutic anticoagulation with heparin in critically ill patients with Covid-19. N Engl J Med 385(9):777–789
66.
Zurück zum Zitat Group RC (2021) Dexamethasone in hospitalized patients with Covid-19. N Engl J Med 384(8):693–704 Group RC (2021) Dexamethasone in hospitalized patients with Covid-19. N Engl J Med 384(8):693–704
67.
Zurück zum Zitat Yurttaş B et al (2020) Characteristics and outcomes of Behçet’s syndrome patients with Coronavirus Disease 2019: a case series of 10 patients. Intern Emerg Med 15(8):1567–1571PubMedPubMedCentralCrossRef Yurttaş B et al (2020) Characteristics and outcomes of Behçet’s syndrome patients with Coronavirus Disease 2019: a case series of 10 patients. Intern Emerg Med 15(8):1567–1571PubMedPubMedCentralCrossRef
68.
Zurück zum Zitat Watad A et al (2021) Immune-Mediated Disease Flares or New-Onset Disease in 27 Subjects Following mRNA/DNA SARS-CoV-2 Vaccination. Vaccines 9(5):435PubMedPubMedCentralCrossRef Watad A et al (2021) Immune-Mediated Disease Flares or New-Onset Disease in 27 Subjects Following mRNA/DNA SARS-CoV-2 Vaccination. Vaccines 9(5):435PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Mayor-Ibarguren A et al (2020) Cutaneous small vessel vasculitis secondary to COVID-19 infection: a case report. J Eur Acad Dermatol Venereol 34(10):e541–e542 Mayor-Ibarguren A et al (2020) Cutaneous small vessel vasculitis secondary to COVID-19 infection: a case report. J Eur Acad Dermatol Venereol 34(10):e541–e542
72.
Zurück zum Zitat Dominguez-SantasM et al (2020) Cutaneous small-vessel vasculitis associated with novel 2019 coronavirus SARS-CoV-2 infection (COVID-19). J Eur Acad Dermatol Venereol 34(10):e536–e537 Dominguez-SantasM et al (2020) Cutaneous small-vessel vasculitis associated with novel 2019 coronavirus SARS-CoV-2 infection (COVID-19). J Eur Acad Dermatol Venereol 34(10):e536–e537
73.
Zurück zum Zitat Tahir A et al (2020) Widespread cutaneous small vessel vasculitis secondary to COVID-19 infection. Int J Dermatol 59(10):1278–1279 Tahir A et al (2020) Widespread cutaneous small vessel vasculitis secondary to COVID-19 infection. Int J Dermatol 59(10):1278–1279
74.
Zurück zum Zitat da Cruz Gouveia PA et al (2021) Exuberant bullous vasculitis associated with SARS-CoV-2 infection. IDCases 23:e01047CrossRef da Cruz Gouveia PA et al (2021) Exuberant bullous vasculitis associated with SARS-CoV-2 infection. IDCases 23:e01047CrossRef
75.
Zurück zum Zitat Negrini S et al (2020) An unusual case of bullous haemorrhagic vasculitis in a COVID-19 patient. J Eur Acad Dermatol Venereol 34(11):e675–e676 Negrini S et al (2020) An unusual case of bullous haemorrhagic vasculitis in a COVID-19 patient. J Eur Acad Dermatol Venereol 34(11):e675–e676
76.
Zurück zum Zitat de Perosanz-Lobo D et al (2020) Urticarial vasculitis in COVID-19 infection: a vasculopathy-related symptom? J Eur Acad Dermatol Venereol 34(10):e566–e568 de Perosanz-Lobo D et al (2020) Urticarial vasculitis in COVID-19 infection: a vasculopathy-related symptom? J Eur Acad Dermatol Venereol 34(10):e566–e568
77.
Zurück zum Zitat ShahidiDadras M et al (2021) SARS-CoV-2 infection as a potential triggering factor for urticarial vasculitis during pregnancy: A case report. Clinical Case Reports 9(6):e04323 ShahidiDadras M et al (2021) SARS-CoV-2 infection as a potential triggering factor for urticarial vasculitis during pregnancy: A case report. Clinical Case Reports 9(6):e04323
78.
Zurück zum Zitat Mohta A, Mehta RD, Ghiya BC (2021) Multisystem Inflammatory Syndrome in Children Related to COVID-19 With Urticarial Vasculitis—A Double Whammy! Indian Pediatrics 58(9):894PubMedPubMedCentralCrossRef Mohta A, Mehta RD, Ghiya BC (2021) Multisystem Inflammatory Syndrome in Children Related to COVID-19 With Urticarial Vasculitis—A Double Whammy! Indian Pediatrics 58(9):894PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Najafzadeh M et al (2020) Urticaria (angioedema) and COVID-19 infection. J Eur Acad Dermatol Venereol 34(10):e568–e570 Najafzadeh M et al (2020) Urticaria (angioedema) and COVID-19 infection. J Eur Acad Dermatol Venereol 34(10):e568–e570
80.
Zurück zum Zitat Nasiri S et al (2020) Urticarial vasculitis in a COVID-19 recovered patient. Int J Dermatol 59(10):1285–1286 Nasiri S et al (2020) Urticarial vasculitis in a COVID-19 recovered patient. Int J Dermatol 59(10):1285–1286
81.
Zurück zum Zitat Criado PR et al (2021) Urticarial vasculitis revealing immunolabelled nucleocapsid protein of SARS-CoV-2 in two Brazilian asymptomatic patients: the tip of the COVID-19 hidden iceberg? J Eur Acad Dermatol Venereol 35(9):e563–e566 Criado PR et al (2021) Urticarial vasculitis revealing immunolabelled nucleocapsid protein of SARS-CoV-2 in two Brazilian asymptomatic patients: the tip of the COVID-19 hidden iceberg? J Eur Acad Dermatol Venereol 35(9):e563–e566
82.
Zurück zum Zitat Hope L, et al (2021) The COVID rash that puts the ‘U’in GROUCH! in Baylor University Medical Center Proceedings Taylor & Francis Hope L, et al (2021) The COVID rash that puts the ‘U’in GROUCH! in Baylor University Medical Center Proceedings Taylor & Francis
83.
Zurück zum Zitat Duran TI, et al (2021) ANCA-associated vasculitis after COVID-19. Rheumatol Int 1–7 Duran TI, et al (2021) ANCA-associated vasculitis after COVID-19. Rheumatol Int 1–7
84.
85.
86.
Zurück zum Zitat Moeinzadeh F et al (2020) Newly diagnosed glomerulonephritis during COVID-19 infection undergoing immunosuppression therapy, a case report. Iran J Kidney Dis 14(3):239–242PubMed Moeinzadeh F et al (2020) Newly diagnosed glomerulonephritis during COVID-19 infection undergoing immunosuppression therapy, a case report. Iran J Kidney Dis 14(3):239–242PubMed
87.
Zurück zum Zitat Jalalzadeh M, et al (2021) Antineutrophil cytoplasmic antibody-associated glomerulonephritis in a case of scleroderma after recent diagnosis with COVID-19. Cureus 13(1) Jalalzadeh M, et al (2021) Antineutrophil cytoplasmic antibody-associated glomerulonephritis in a case of scleroderma after recent diagnosis with COVID-19. Cureus 13(1)
88.
Zurück zum Zitat Farooq H et al (2022) The pathogenesis of COVID-19-induced IgA nephropathy and IgA vasculitis: a systematic review. J Taibah Univ Med Sci 17(1):1–13 Farooq H et al (2022) The pathogenesis of COVID-19-induced IgA nephropathy and IgA vasculitis: a systematic review. J Taibah Univ Med Sci 17(1):1–13
89.
Zurück zum Zitat Jedlowski PM et al (2022) Coronavirus disease 2019-associated immunoglobulin A vasculitis/Henoch–Schönlein purpura: a case report and review. J Dermatol 49(1):190–196 Jedlowski PM et al (2022) Coronavirus disease 2019-associated immunoglobulin A vasculitis/Henoch–Schönlein purpura: a case report and review. J Dermatol 49(1):190–196
91.
Zurück zum Zitat Riera-Martí N, Romaní J, Calvet J (2021) SARS-CoV-2 infection triggering a giant cell arteritis. Medicina clinica (English ed) 156(5):253CrossRef Riera-Martí N, Romaní J, Calvet J (2021) SARS-CoV-2 infection triggering a giant cell arteritis. Medicina clinica (English ed) 156(5):253CrossRef
92.
Zurück zum Zitat Jonathan GL, Scott FM, Matthew KD (2021) A Case of Post-COVID-19–Associated Paracentral Acute Middle Maculopathy and Giant Cell Arteritis-Like Vasculitis. J Neuroophthalmol 41(3):351–355PubMedCrossRef Jonathan GL, Scott FM, Matthew KD (2021) A Case of Post-COVID-19–Associated Paracentral Acute Middle Maculopathy and Giant Cell Arteritis-Like Vasculitis. J Neuroophthalmol 41(3):351–355PubMedCrossRef
94.
Zurück zum Zitat Nahhal S, et al (2020) Anti-glomerular basement membrane disease as a potential complication of COVID-19: a case report and review of literature. Cureus 12(12) Nahhal S, et al (2020) Anti-glomerular basement membrane disease as a potential complication of COVID-19: a case report and review of literature. Cureus 12(12)
95.
Zurück zum Zitat Dixon L et al (2021) Immunosuppression for intracranial vasculitis associated with SARS-CoV-2: therapeutic implications for COVID-19 cerebrovascular pathology. J Neurol Neurosurg Psychiatry 92(1):103–104CrossRef Dixon L et al (2021) Immunosuppression for intracranial vasculitis associated with SARS-CoV-2: therapeutic implications for COVID-19 cerebrovascular pathology. J Neurol Neurosurg Psychiatry 92(1):103–104CrossRef
96.
Zurück zum Zitat Kadkhoda K, Laurita K (2021) Antineutrophil cytoplasmic antibodies and their association with clinical outcomes in hospitalized COVID-19 patients. Cell Death Discovery 7(1):277PubMedPubMedCentralCrossRef Kadkhoda K, Laurita K (2021) Antineutrophil cytoplasmic antibodies and their association with clinical outcomes in hospitalized COVID-19 patients. Cell Death Discovery 7(1):277PubMedPubMedCentralCrossRef
97.
Zurück zum Zitat Söderberg D and Segelmark M (2016) Neutrophil Extracellular Traps in ANCA-Associated Vasculitis. Front Immunol 7(256). Söderberg D and Segelmark M (2016) Neutrophil Extracellular Traps in ANCA-Associated Vasculitis. Front Immunol 7(256).
99.
Zurück zum Zitat Kadkhoda K, Laurita K (2021) Antineutrophil cytoplasmic antibodies and their association with clinical outcomes in hospitalized COVID-19 patients. Cell death discovery 7(1):1–3CrossRef Kadkhoda K, Laurita K (2021) Antineutrophil cytoplasmic antibodies and their association with clinical outcomes in hospitalized COVID-19 patients. Cell death discovery 7(1):1–3CrossRef
100.
Zurück zum Zitat Wijst MGPVD et al (2021) Type I interferon autoantibodies are associated with systemic immune alterations in patients with COVID-19. Science Translational Medicine 13(612):eabh2624PubMedPubMedCentralCrossRef Wijst MGPVD et al (2021) Type I interferon autoantibodies are associated with systemic immune alterations in patients with COVID-19. Science Translational Medicine 13(612):eabh2624PubMedPubMedCentralCrossRef
102.
Zurück zum Zitat Heineke MH et al (2017) New insights in the pathogenesis of immunoglobulin A vasculitis (Henoch-Schönlein purpura). Autoimmun Rev 16(12):1246–1253PubMedCrossRef Heineke MH et al (2017) New insights in the pathogenesis of immunoglobulin A vasculitis (Henoch-Schönlein purpura). Autoimmun Rev 16(12):1246–1253PubMedCrossRef
104.
Zurück zum Zitat Vlachoyiannopoulos PG et al (2020) Autoantibodies related to systemic autoimmune rheumatic diseases in severely ill patients with COVID-19. Ann Rheum Dis 79(12):1661–1663PubMedCrossRef Vlachoyiannopoulos PG et al (2020) Autoantibodies related to systemic autoimmune rheumatic diseases in severely ill patients with COVID-19. Ann Rheum Dis 79(12):1661–1663PubMedCrossRef
105.
Zurück zum Zitat Galván Casas C et al (2020) Classification of the cutaneous manifestations of COVID-19: a rapid prospective nationwide consensus study in Spain with 375 cases. Br J Dermatol 183(1):71–77PubMedCrossRef Galván Casas C et al (2020) Classification of the cutaneous manifestations of COVID-19: a rapid prospective nationwide consensus study in Spain with 375 cases. Br J Dermatol 183(1):71–77PubMedCrossRef
106.
Zurück zum Zitat Freeman EE et al (2020) Pernio-like skin lesions associated with COVID-19: A case series of 318 patients from 8 countries. J Am Acad Dermatol 83(2):486–492PubMedPubMedCentralCrossRef Freeman EE et al (2020) Pernio-like skin lesions associated with COVID-19: A case series of 318 patients from 8 countries. J Am Acad Dermatol 83(2):486–492PubMedPubMedCentralCrossRef
107.
Zurück zum Zitat Kolivras A et al (2020) Coronavirus (COVID-19) infection–induced chilblains: A case report with histopathologic findings. JAAD case reports 6(6):489–492PubMedPubMedCentralCrossRef Kolivras A et al (2020) Coronavirus (COVID-19) infection–induced chilblains: A case report with histopathologic findings. JAAD case reports 6(6):489–492PubMedPubMedCentralCrossRef
108.
Zurück zum Zitat Magro C et al (2021) The skin as a critical window in unveiling the pathophysiologic principles of COVID-19. Clin Dermatol 39(6):934–965 Magro C et al (2021) The skin as a critical window in unveiling the pathophysiologic principles of COVID-19. Clin Dermatol 39(6):934–965
109.
Zurück zum Zitat Crow YJ (2011) Type I interferonopathies: a novel set of inborn errors of immunity. Ann N Y Acad Sci 1238(1):91–98PubMedCrossRef Crow YJ (2011) Type I interferonopathies: a novel set of inborn errors of immunity. Ann N Y Acad Sci 1238(1):91–98PubMedCrossRef
112.
Zurück zum Zitat Frumholtz L et al (2021) Type I interferon response and vascular alteration in chilblain-like lesions during the COVID-19 outbreak. Br J Dermatol 185(6):1176–1185 Frumholtz L et al (2021) Type I interferon response and vascular alteration in chilblain-like lesions during the COVID-19 outbreak. Br J Dermatol 185(6):1176–1185
113.
Zurück zum Zitat Feldstein LR et al (2021) Characteristics and Outcomes of US Children and Adolescents With Multisystem Inflammatory Syndrome in Children (MIS-C) Compared With Severe Acute COVID-19. JAMA 325(11):1074–1087PubMedCrossRef Feldstein LR et al (2021) Characteristics and Outcomes of US Children and Adolescents With Multisystem Inflammatory Syndrome in Children (MIS-C) Compared With Severe Acute COVID-19. JAMA 325(11):1074–1087PubMedCrossRef
114.
Zurück zum Zitat Riphagen S et al (2020) Hyperinflammatory shock in children during COVID-19 pandemic. The Lancet 395(10237):1607–1608CrossRef Riphagen S et al (2020) Hyperinflammatory shock in children during COVID-19 pandemic. The Lancet 395(10237):1607–1608CrossRef
115.
Zurück zum Zitat Viner RM, Whittaker E (2020) Kawasaki-like disease: emerging complication during the COVID-19 pandemic. The Lancet 395(10239):1741–1743CrossRef Viner RM, Whittaker E (2020) Kawasaki-like disease: emerging complication during the COVID-19 pandemic. The Lancet 395(10239):1741–1743CrossRef
116.
Zurück zum Zitat Verdoni L et al (2020) An outbreak of severe Kawasaki-like disease at the Italian epicentre of the SARS-CoV-2 epidemic: an observational cohort study. The Lancet 395(10239):1771–1778CrossRef Verdoni L et al (2020) An outbreak of severe Kawasaki-like disease at the Italian epicentre of the SARS-CoV-2 epidemic: an observational cohort study. The Lancet 395(10239):1771–1778CrossRef
117.
Zurück zum Zitat Belay ED et al (2021) Trends in geographic and temporal distribution of US children with multisystem inflammatory syndrome during the COVID-19 pandemic. JAMA Pediatr 175(8):837–845 Belay ED et al (2021) Trends in geographic and temporal distribution of US children with multisystem inflammatory syndrome during the COVID-19 pandemic. JAMA Pediatr 175(8):837–845
118.
Zurück zum Zitat Payne AB et al (2021) Incidence of Multisystem Inflammatory Syndrome in Children Among US Persons Infected With SARS-CoV-2. JAMA Netw Open 4(6):e2116420–e2116420PubMedPubMedCentralCrossRef Payne AB et al (2021) Incidence of Multisystem Inflammatory Syndrome in Children Among US Persons Infected With SARS-CoV-2. JAMA Netw Open 4(6):e2116420–e2116420PubMedPubMedCentralCrossRef
119.
120.
Zurück zum Zitat Pouletty M et al (2020) Paediatric multisystem inflammatory syndrome temporally associated with SARS-CoV-2 mimicking Kawasaki disease (Kawa-COVID-19): a multicentre cohort. Ann Rheum Dis 79(8):999–1006PubMedCrossRef Pouletty M et al (2020) Paediatric multisystem inflammatory syndrome temporally associated with SARS-CoV-2 mimicking Kawasaki disease (Kawa-COVID-19): a multicentre cohort. Ann Rheum Dis 79(8):999–1006PubMedCrossRef
121.
Zurück zum Zitat Toubiana J et al (2020) Kawasaki-like multisystem inflammatory syndrome in children during the covid-19 pandemic in Paris, France: prospective observational study. BMJ 369:m2094PubMedPubMedCentralCrossRef Toubiana J et al (2020) Kawasaki-like multisystem inflammatory syndrome in children during the covid-19 pandemic in Paris, France: prospective observational study. BMJ 369:m2094PubMedPubMedCentralCrossRef
122.
Zurück zum Zitat Dufort EM et al (2020) Multisystem Inflammatory Syndrome in Children in New York State. N Engl J Med 383(4):347–358PubMedCrossRef Dufort EM et al (2020) Multisystem Inflammatory Syndrome in Children in New York State. N Engl J Med 383(4):347–358PubMedCrossRef
123.
125.
Zurück zum Zitat Ogata AF et al (2022) Circulating severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine antigen detected in the plasma of mRNA-1273 vaccine recipients. Clin Infect Dis 74(4):715–718 Ogata AF et al (2022) Circulating severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine antigen detected in the plasma of mRNA-1273 vaccine recipients. Clin Infect Dis 74(4):715–718
126.
Zurück zum Zitat Greinacher A et al (2021) Thrombotic Thrombocytopenia after ChAdOx1 nCov-19 Vaccination. N Engl J Med 384(22):2092–2101PubMedCrossRef Greinacher A et al (2021) Thrombotic Thrombocytopenia after ChAdOx1 nCov-19 Vaccination. N Engl J Med 384(22):2092–2101PubMedCrossRef
127.
Zurück zum Zitat Schultz NH et al (2021) Thrombosis and Thrombocytopenia after ChAdOx1 nCoV-19 Vaccination. N Engl J Med 384(22):2124–2130PubMedCrossRef Schultz NH et al (2021) Thrombosis and Thrombocytopenia after ChAdOx1 nCoV-19 Vaccination. N Engl J Med 384(22):2124–2130PubMedCrossRef
128.
Zurück zum Zitat Barda N et al (2021) Safety of the BNT162b2 mRNA Covid-19 Vaccine in a Nationwide Setting. N Engl J Med 385(12):1078–1090PubMedCrossRef Barda N et al (2021) Safety of the BNT162b2 mRNA Covid-19 Vaccine in a Nationwide Setting. N Engl J Med 385(12):1078–1090PubMedCrossRef
129.
Zurück zum Zitat Mevorach D, et al (2021) Myocarditis after BNT162b2 mRNA vaccine against Covid-19 in Israel. N Engl J Med 385(23):2140–2149 Mevorach D, et al (2021) Myocarditis after BNT162b2 mRNA vaccine against Covid-19 in Israel. N Engl J Med 385(23):2140–2149
130.
Zurück zum Zitat Witberg G et al (2021) Myocarditis after Covid-19 vaccination in a large health care organization. N Engl J Med 385(23):2132–2139 Witberg G et al (2021) Myocarditis after Covid-19 vaccination in a large health care organization. N Engl J Med 385(23):2132–2139
131.
132.
Zurück zum Zitat Guzmán-Pérez L et al (2021) Small-vessel vasculitis following Oxford-AstraZeneca vaccination against SARS-CoV-2. J Eur Acad Dermatol Venereol 35(11):e741–e743 Guzmán-Pérez L et al (2021) Small-vessel vasculitis following Oxford-AstraZeneca vaccination against SARS-CoV-2. J Eur Acad Dermatol Venereol 35(11):e741–e743
133.
Zurück zum Zitat Vassallo C et al (2021) Cutaneous lymphocytic vasculitis after administration of COVID-19 mRNA vaccine. Dermatol Ther 34(5):e15076–e15076 Vassallo C et al (2021) Cutaneous lymphocytic vasculitis after administration of COVID-19 mRNA vaccine. Dermatol Ther 34(5):e15076–e15076
134.
Zurück zum Zitat Kharkar V et al (2021) Asymmetrical cutaneous vasculitis following COVID-19 vaccination with unusual eosinophil preponderance. Clin Exp Dermatol 46(8):1596–1597 Kharkar V et al (2021) Asymmetrical cutaneous vasculitis following COVID-19 vaccination with unusual eosinophil preponderance. Clin Exp Dermatol 46(8):1596–1597
135.
Zurück zum Zitat McMahon DE et al (2021) Cutaneous reactions reported after Moderna and Pfizer COVID-19 vaccination: A registry-based study of 414 cases. J Am Acad Dermatol 85(1):46–55PubMedPubMedCentralCrossRef McMahon DE et al (2021) Cutaneous reactions reported after Moderna and Pfizer COVID-19 vaccination: A registry-based study of 414 cases. J Am Acad Dermatol 85(1):46–55PubMedPubMedCentralCrossRef
136.
Zurück zum Zitat Larson V et al (2022) Clinical and histopathological spectrum of delayed adverse cutaneous reactions following COVID‐19 vaccination. J Cutan Pathol 49(1):34–41 Larson V et al (2022) Clinical and histopathological spectrum of delayed adverse cutaneous reactions following COVID‐19 vaccination. J Cutan Pathol 49(1):34–41
137.
Zurück zum Zitat Bostan E, Gulseren D, Gokoz O (2021) New-onset leukocytoclastic vasculitis after COVID-19 vaccine. Int J Dermatol 60(10):1305–1306 Bostan E, Gulseren D, Gokoz O (2021) New-onset leukocytoclastic vasculitis after COVID-19 vaccine. Int J Dermatol 60(10):1305–1306
138.
Zurück zum Zitat Dash S et al (2021) COVID-19 vaccine-induced urticarial vasculitis. Dermatol Ther 34(5):e15093–e15093 Dash S et al (2021) COVID-19 vaccine-induced urticarial vasculitis. Dermatol Ther 34(5):e15093–e15093
139.
Zurück zum Zitat Okuda S, Hirooka Y, Sugiyama M (2021) Propylthiouracil-Induced Antineutrophil Cytoplasmic Antibody-Associated Vasculitis after COVID-19 Vaccination. Vaccines 9(8):842PubMedPubMedCentralCrossRef Okuda S, Hirooka Y, Sugiyama M (2021) Propylthiouracil-Induced Antineutrophil Cytoplasmic Antibody-Associated Vasculitis after COVID-19 Vaccination. Vaccines 9(8):842PubMedPubMedCentralCrossRef
140.
Zurück zum Zitat Villa M et al (2021) A case of ANCA-associated vasculitis after AZD1222 (Oxford–AstraZeneca) SARS-CoV-2 vaccination: casualty or causality? Kidney Int 100(4):937–938 Villa M et al (2021) A case of ANCA-associated vasculitis after AZD1222 (Oxford–AstraZeneca) SARS-CoV-2 vaccination: casualty or causality? Kidney Int 100(4):937–938
141.
143.
Zurück zum Zitat Badier L et al (2021) IgA vasculitis in adult patient following vaccination by ChadOx1 nCoV-19. Autoimmun Rev 20(11):102951–102951 Badier L et al (2021) IgA vasculitis in adult patient following vaccination by ChadOx1 nCoV-19. Autoimmun Rev 20(11):102951–102951
144.
Zurück zum Zitat Anderegg MA et al (2021) De novo vasculitis after mRNA-1273 (Moderna) vaccination. Kidney Int 100(2):474–476 Anderegg MA et al (2021) De novo vasculitis after mRNA-1273 (Moderna) vaccination. Kidney Int 100(2):474–476
145.
Zurück zum Zitat Naitlho A et al (2021) A rare case of Henoch-Schönlein Purpura following a COVID-19 vaccine—case report. SN Compr Clin Med 3(12):2618–2621 Naitlho A et al (2021) A rare case of Henoch-Schönlein Purpura following a COVID-19 vaccine—case report. SN Compr Clin Med 3(12):2618–2621
147.
Zurück zum Zitat Schierz J-H, et al (2021) Vasculitis and bursitis on [(18)F]FDG-PET/CT following COVID-19 mRNA vaccine: post hoc ergo propter hoc? European journal of nuclear medicine and molecular imaging 1–2. Schierz J-H, et al (2021) Vasculitis and bursitis on [(18)F]FDG-PET/CT following COVID-19 mRNA vaccine: post hoc ergo propter hoc? European journal of nuclear medicine and molecular imaging 1–2.
148.
Zurück zum Zitat Conticini E et al (2021) Relapse of microscopic polyangiitis after vaccination against COVID-19: a case report. J Med Virol 93(12):6439–6441 Conticini E et al (2021) Relapse of microscopic polyangiitis after vaccination against COVID-19: a case report. J Med Virol 93(12):6439–6441
149.
Zurück zum Zitat Nastro F et al (2021) Small vessel vasculitis related to varicella-zoster virus after Pfizer-BioNTech COVID-19 vaccine. J Eur Acad Dermatol Venereol 35(11):e745–e747 Nastro F et al (2021) Small vessel vasculitis related to varicella-zoster virus after Pfizer-BioNTech COVID-19 vaccine. J Eur Acad Dermatol Venereol 35(11):e745–e747
150.
Zurück zum Zitat Cohen SR et al (2021) Leukocytoclastic vasculitis flare following the COVID‐19 vaccine. Int J Dermatol 60(8):1032–1033 Cohen SR et al (2021) Leukocytoclastic vasculitis flare following the COVID‐19 vaccine. Int J Dermatol 60(8):1032–1033
151.
152.
Zurück zum Zitat Belay ED, et al (2021) Multisystem Inflammatory Syndrome in Adults After Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Infection and Coronavirus Disease 2019 (COVID-19) Vaccination. Clinical Infectious Diseases Belay ED, et al (2021) Multisystem Inflammatory Syndrome in Adults After Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Infection and Coronavirus Disease 2019 (COVID-19) Vaccination. Clinical Infectious Diseases
153.
Zurück zum Zitat Levy M et al (2022) Multisystem inflammatory syndrome in children by COVID-19 vaccination status of adolescents in France. JAMA 327(3):281–283 Levy M et al (2022) Multisystem inflammatory syndrome in children by COVID-19 vaccination status of adolescents in France. JAMA 327(3):281–283
154.
Zurück zum Zitat Piccolo V et al (2021) BNT162b2 mRNA COVID-19 vaccine-induced chilblain-like lesions reinforces the hypothesis of their relationship with SARS-CoV-2. J Eur Acad Dermatol Venereol 35(8):e493–e494PubMedCrossRef Piccolo V et al (2021) BNT162b2 mRNA COVID-19 vaccine-induced chilblain-like lesions reinforces the hypothesis of their relationship with SARS-CoV-2. J Eur Acad Dermatol Venereol 35(8):e493–e494PubMedCrossRef
155.
156.
Zurück zum Zitat Tan HZ et al (2021) Is COVID-19 vaccination unmasking glomerulonephritis? Kidney Int 100(2):469–471 Tan HZ et al (2021) Is COVID-19 vaccination unmasking glomerulonephritis? Kidney Int 100(2):469–471
157.
158.
Zurück zum Zitat Magro C et al (2021) The differing pathophysiologies that underlie COVID-19-associated perniosis and thrombotic retiform purpura: a case series. Br J Dermatol 184(1):141–150PubMedCrossRef Magro C et al (2021) The differing pathophysiologies that underlie COVID-19-associated perniosis and thrombotic retiform purpura: a case series. Br J Dermatol 184(1):141–150PubMedCrossRef
159.
Zurück zum Zitat Magro C et al (2020) Complement associated microvascular injury and thrombosis in the pathogenesis of severe COVID-19 infection: a report of five cases. Transl Res 220:1–13PubMedPubMedCentralCrossRef Magro C et al (2020) Complement associated microvascular injury and thrombosis in the pathogenesis of severe COVID-19 infection: a report of five cases. Transl Res 220:1–13PubMedPubMedCentralCrossRef
160.
Zurück zum Zitat Becker RC (2020) COVID-19-associated vasculitis and vasculopathy. J Thromb Thrombolysis 50(3):499–511PubMedCrossRef Becker RC (2020) COVID-19-associated vasculitis and vasculopathy. J Thromb Thrombolysis 50(3):499–511PubMedCrossRef
162.
Zurück zum Zitat Magro CM et al (2021) Severe COVID-19: A multifaceted viral vasculopathy syndrome. Ann Diagn Pathol 50:151645–151645PubMedCrossRef Magro CM et al (2021) Severe COVID-19: A multifaceted viral vasculopathy syndrome. Ann Diagn Pathol 50:151645–151645PubMedCrossRef
163.
Zurück zum Zitat Conway EM, Pryzdial ELG (2020) Is the COVID-19 thrombotic catastrophe complement-connected? J Thromb Haemost 18(11):2812–2822PubMedCrossRef Conway EM, Pryzdial ELG (2020) Is the COVID-19 thrombotic catastrophe complement-connected? J Thromb Haemost 18(11):2812–2822PubMedCrossRef
164.
Zurück zum Zitat Song W-C, FitzGerald GA (2020) COVID-19, microangiopathy, hemostatic activation, and complement. J Clin Investig 130(8):3950–3953PubMedPubMedCentral Song W-C, FitzGerald GA (2020) COVID-19, microangiopathy, hemostatic activation, and complement. J Clin Investig 130(8):3950–3953PubMedPubMedCentral
168.
Zurück zum Zitat van Dam LF et al (2020) Clinical and computed tomography characteristics of COVID-19 associated acute pulmonary embolism: A different phenotype of thrombotic disease? Thromb Res 193:86–89PubMedPubMedCentralCrossRef van Dam LF et al (2020) Clinical and computed tomography characteristics of COVID-19 associated acute pulmonary embolism: A different phenotype of thrombotic disease? Thromb Res 193:86–89PubMedPubMedCentralCrossRef
169.
Zurück zum Zitat Goette A et al (2020) COVID-19-Induced Cytokine Release Syndrome Associated with Pulmonary Vein Thromboses, Atrial Cardiomyopathy, and Arterial Intima Inflammation. TH open : companion journal to thrombosis and haemostasis 4(3):e271–e279CrossRef Goette A et al (2020) COVID-19-Induced Cytokine Release Syndrome Associated with Pulmonary Vein Thromboses, Atrial Cardiomyopathy, and Arterial Intima Inflammation. TH open : companion journal to thrombosis and haemostasis 4(3):e271–e279CrossRef
170.
Zurück zum Zitat Zakeri A et al (2021) Ischemic stroke in COVID-19-positive patients: an overview of SARS-CoV-2 and thrombotic mechanisms for the neurointerventionalist. J NeuroInterv Surg 13(3):202–206PubMedCrossRef Zakeri A et al (2021) Ischemic stroke in COVID-19-positive patients: an overview of SARS-CoV-2 and thrombotic mechanisms for the neurointerventionalist. J NeuroInterv Surg 13(3):202–206PubMedCrossRef
171.
Zurück zum Zitat South K et al (2020) Preceding infection and risk of stroke: An old concept revived by the COVID-19 pandemic. International journal of stroke : official journal of the International Stroke Society 15(7):722–732CrossRef South K et al (2020) Preceding infection and risk of stroke: An old concept revived by the COVID-19 pandemic. International journal of stroke : official journal of the International Stroke Society 15(7):722–732CrossRef
172.
Zurück zum Zitat Piroth L et al (2021) Comparison of the characteristics, morbidity, and mortality of COVID-19 and seasonal influenza: a nationwide, population-based retrospective cohort study. Lancet Respir Med 9(3):251–259PubMedCrossRef Piroth L et al (2021) Comparison of the characteristics, morbidity, and mortality of COVID-19 and seasonal influenza: a nationwide, population-based retrospective cohort study. Lancet Respir Med 9(3):251–259PubMedCrossRef
173.
Zurück zum Zitat Merkler AE et al (2020) Risk of ischemic stroke in patients with coronavirus disease 2019 (COVID-19) vs patients with influenza. JAMA Neurol 77(11):1366–1372CrossRef Merkler AE et al (2020) Risk of ischemic stroke in patients with coronavirus disease 2019 (COVID-19) vs patients with influenza. JAMA Neurol 77(11):1366–1372CrossRef
174.
Zurück zum Zitat Taquet M et al (2021) 6-month neurological and psychiatric outcomes in 236 379 survivors of COVID-19: a retrospective cohort study using electronic health records. The lancet Psychiatry 8(5):416–427PubMedPubMedCentralCrossRef Taquet M et al (2021) 6-month neurological and psychiatric outcomes in 236 379 survivors of COVID-19: a retrospective cohort study using electronic health records. The lancet Psychiatry 8(5):416–427PubMedPubMedCentralCrossRef
175.
Zurück zum Zitat Chen T, et al (2020) Clinical characteristics of 113 deceased patients with coronavirus disease 2019: retrospective study. BMJ 368 Chen T, et al (2020) Clinical characteristics of 113 deceased patients with coronavirus disease 2019: retrospective study. BMJ 368
176.
Zurück zum Zitat Benedetti C, et al (2020) COVID-19 and the kidneys: an update. Front Med 7 Benedetti C, et al (2020) COVID-19 and the kidneys: an update. Front Med 7
177.
Metadaten
Titel
COVID-19 Vasculitis and vasculopathy-Distinct immunopathology emerging from the close juxtaposition of Type II Pneumocytes and Pulmonary Endothelial Cells
verfasst von
Sami Giryes
Nicola Luigi Bragazzi
Charles Bridgewood
Gabriele De Marco
Dennis McGonagle
Publikationsdatum
12.04.2022
Verlag
Springer Berlin Heidelberg
Schlagwort
COVID-19
Erschienen in
Seminars in Immunopathology / Ausgabe 3/2022
Print ISSN: 1863-2297
Elektronische ISSN: 1863-2300
DOI
https://doi.org/10.1007/s00281-022-00928-6

Weitere Artikel der Ausgabe 3/2022

Seminars in Immunopathology 3/2022 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Perioperative Checkpointhemmer-Therapie verbessert NSCLC-Prognose

28.05.2024 NSCLC Nachrichten

Eine perioperative Therapie mit Nivolumab reduziert das Risiko für Rezidive und Todesfälle bei operablem NSCLC im Vergleich zu einer alleinigen neoadjuvanten Chemotherapie um über 40%. Darauf deuten die Resultate der Phase-3-Studie CheckMate 77T.

Positiver FIT: Die Ursache liegt nicht immer im Dickdarm

27.05.2024 Blut im Stuhl Nachrichten

Immunchemischer Stuhltest positiv, Koloskopie negativ – in solchen Fällen kann die Blutungsquelle auch weiter proximal sitzen. Ein Forschungsteam hat nachgesehen, wie häufig und in welchen Lokalisationen das der Fall ist.

GLP-1-Agonisten können Fortschreiten diabetischer Retinopathie begünstigen

24.05.2024 Diabetische Retinopathie Nachrichten

Möglicherweise hängt es von der Art der Diabetesmedikamente ab, wie hoch das Risiko der Betroffenen ist, dass sich sehkraftgefährdende Komplikationen verschlimmern.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.