Skip main navigation

Microthrombi as a Major Cause of Cardiac Injury in COVID-19

A Pathologic Study
Originally publishedhttps://doi.org/10.1161/CIRCULATIONAHA.120.051828Circulation. 2021;143:1031–1042

Abstract

Background:

Cardiac injury is common in patients who are hospitalized with coronavirus disease 2019 (COVID-19) and portends poorer prognosis. However, the mechanism and the type of myocardial damage associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remain uncertain.

Methods:

We conducted a systematic pathological analysis of 40 hearts from hospitalized patients dying of COVID-19 in Bergamo, Italy, to determine the pathological mechanisms of cardiac injury. We divided the hearts according to presence or absence of acute myocyte necrosis and then determined the underlying mechanisms of cardiac injury.

Results:

Of the 40 hearts examined, 14 (35%) had evidence of myocyte necrosis, predominantly of the left ventricle. Compared with subjects without necrosis, subjects with necrosis tended to be female, have chronic kidney disease, and have shorter symptom onset to admission. The incidence of severe coronary artery disease (ie, >75% cross-sectional narrowing) was not significantly different between those with and without necrosis. Three of 14 (21.4%) subjects with myocyte necrosis showed evidence of acute myocardial infarction, defined as ≥1 cm2 area of necrosis, whereas 11 of 14 (78.6%) showed evidence of focal (>20 necrotic myocytes with an area of ≥0.05 mm2 but <1 cm2) myocyte necrosis. Cardiac thrombi were present in 11 of 14 (78.6%) cases with necrosis, with 2 of 14 (14.2%) having epicardial coronary artery thrombi, whereas 9 of 14 (64.3%) had microthrombi in myocardial capillaries, arterioles, and small muscular arteries. We compared cardiac microthrombi from COVID-19–positive autopsy cases to intramyocardial thromboemboli from COVID-19 cases as well as to aspirated thrombi obtained during primary percutaneous coronary intervention from uninfected and COVID-19–infected patients presenting with ST-segment–elevation myocardial infarction. Microthrombi had significantly greater fibrin and terminal complement C5b-9 immunostaining compared with intramyocardial thromboemboli from COVID-19–negative subjects and with aspirated thrombi. There were no significant differences between the constituents of thrombi aspirated from COVID-19–positive and –negative patients with ST-segment–elevation myocardial infarction.

Conclusions:

The most common pathological cause of myocyte necrosis was microthrombi. Microthrombi were different in composition from intramyocardial thromboemboli from COVID-19–negative subjects and from coronary thrombi retrieved from COVID-19–positive and –negative patients with ST-segment–elevation myocardial infarction. Tailored antithrombotic strategies may be useful to counteract the cardiac effects of COVID-19 infection.

Clinical Perspective

What Is New?

  • Cardiac injury in coronavirus disease 2019 (COVID-19) infection is not uncommon, yet the mechanism by which it occurs remains uncertain.

  • In this autopsy examination of 40 hearts from subjects dying of COVID-19 infection, 14 (35%) had evidence of myocyte necrosis, predominantly of the left ventricle.

  • The major cause of myocyte necrosis was microthrombi occurring in 9 of 14 (64%) cases that were distinct in composition (ie, greater fibrin and c5b-9 complement) compared with intramyocardial thromboemboli from COVID-19–negative subjects and to coronary thrombi retrieved from COVID-19–positive and –negative patients with ST-segment–elevation myocardial infarction.

What Are the Clinical Implications?

  • Clinicians should be aware of the possibility of microthrombi, which may not be detectable clinically as a cause of cardiac injury in subjects with COVID-19 infection.

  • The use of tailored antithrombotic strategies to counteract the effects of microthrombi on the heart may be useful and should be examined.

Myocardial injury is a common phenomenon in hospitalized patients with coronavirus disease 2019 (COVID-19) and is associated with worse outcomes. In 1 study in which troponin I was measured within 24 hours of admission to assess myocardial damage, 36% of patients had elevated troponin I concentrations.1 After adjusting for disease severity and relevant risk factor differences, even small amounts of myocardial injury were associated with increased risk of death.

The cause of myocardial injury in patients with COVID-19 has not been previously elucidated in a systematic manner. Various pathophysiological mechanisms have been hypothesized, including direct viral invasion of the heart or immune-mediated cardiac injury causing myocarditis, stress-cardiomyopathy, myocardial supply-demand mismatch leading to type II myocardial infarction, cytokine storm and hypercoagulability resulting in either increased risk of epicardial coronary thrombosis, pulmonary emboli, and microvascular thrombi.2 We recently reported a case of a young woman dying of ST-segment–elevation myocardial infarction (STEMI) and cardiogenic shock who was found to have multiple myocardial microthrombi at autopsy, but the prevalence of such findings among hospitalized patients with COVID-19 has not been reported.3 Understanding the exact nature of cardiac injury in patients with COVID-19 may affect public health strategies, diagnostic testing, and new therapeutic trials for those infected with COVID-19.

Here, we report the analysis of 40 cardiac autopsies conducted on hospitalized patients dying of COVID-19 during the height of the pandemic in February 2020, in Bergamo, Italy. Our goals were to systematically determine (1) the frequency of cardiac injury in hospitalized patients as assessed by autopsy findings of myocyte necrosis, (2) the major causes and risk factors for myocardial injury, and (3) the pathophysiology of cardiac necrosis in subjects infected with COVID-19.

Methods

The current analysis was performed in the frame of a systematic pathology study to assess cardiac injury by COVID-19 infection. The data that support the findings of this study are available from the corresponding author on reasonable request.

The hearts of 40 unselected patients dying from COVID-19 at Ospedale Papa Giovanni XXIII in Bergamo, Italy, and undergoing autopsy were collected and sent to CVPath Institute (Gaithersburg, MD) for detailed pathological analysis. The study protocol was reviewed and approved by the ethical committee at Ospedale Papa Giovanni XXIII, Bergamo, Italy (2020-0056), and by the CVPath Institute Institutional Review Board (RP0112), Gaithersburg, MD, and registered at https://clinicaltrials.gov; Unique identifier: NCT04367792.

All patients had a laboratory-confirmed diagnosis of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection either by nasal swab test on admission (39/40 patients) or by postmortem reverse transcription polymerase chain reaction. Demographic data, medical history, clinical presentation, treatment, and in-hospital course were derived from medical records. All patients were managed according to local clinical practice.

The specimens were anonymized before shipping, and the examining pathologist was blinded to the clinical details. All tissue specimens were fixed in 10% buffered formalin for at least 72 to 96 hours before shipment. Whole hearts and lungs (either paraffin blocks or tissues) were sent to the CVPath Institute for pathological examination.

Pathological Analysis

Hearts were radiographed, and coronary arteries with significant calcification were removed from the heart, radiographed, decalcified as necessary before dehydration, and sectioned at 3- to 4-mm intervals at the time of embedding in paraffin to determine the extent of luminal narrowing. Coronary arteries without any significant calcification were cut on the heart at 3- to 4-mm intervals and sections submitted with the maximal narrowing from the 4 major coronary arteries (left main, left anterior descending, left circumflex, and right coronary arteries). If no significant narrowing was observed, sections from the proximal 4 major coronary arteries were submitted. A total of 14 transmural sections of myocardium (anterior, inferior, and lateral left ventricle [LV], ventricular septum, anterior, and inferior wall of the right ventricle [RV], at mid and apex, and 1 each from the left and right atrium) were dehydrated, embedded in paraffin, sectioned at 4- to 6-μm intervals, and stained with hematoxylin and eosin and Masson’s trichrome stain for histological evaluation. Acute myocardial infarction was defined as areas of myocyte necrosis ≥1 cm2, whereas focal myocyte necrosis was defined as necrosis of >20 myocytes with an area of necrosis ≥0.05 mm2 but <1 cm2 (Figure 1).

Figure 1.

Figure 1. Patterns of myocardial necrosis. A and E, Schematic illustration of myocardial necrosis patterns caused by different types of thrombi. There is a difference in the pattern of necrosis between epicardial coronary artery thrombi and microthrombi. Epicardial coronary artery thrombi are associated with confluent transmural infarction (necrosis area [≥1 cm2]) as illustrated in A, which can be hemorrhagic when reperfused, as in this case. In contrast, microthrombi cause small areas of focal necrosis (ie, >20 myocytes showing necrosis occupying an area ≥0.05 mm2 but <1 cm2), as illustrated in E. B and F, Gross images of the transverse slice of the right and left ventricles from cases 1 (B) and 9 (F) showing of acute transmural myocardial infarction in B and no macroscopic necrosis in F. C, Histology images of myocardium stained with hematoxylin and eosin shows typical acute transmural hemorrhagic myocardial infarction (black double arrow), which is characterized by transmural confluent areas of myocytes necrosis, hemorrhage, neutrophil infiltration, and edema. D, High-power image of the boxed area in C showing hemorrhage and acute inflammation in focal areas of the myocardial necrosis and edema. G, Histology image of focal myocardial necrosis caused by microthrombi shows smaller foci of myocardial necrosis (black circles indicate areas of focal necrosis; Masson trichrome stain). H, Black boxed area from G shows an area of focal myocyte necrosis outlined by the dotted black line (Masson trichrome stain). LV indicates left ventricle; and RV, right ventricle.

Additional sections from the slides with areas of interest were cut and stained by immunoperoxidase stains with anti-CD61 (prediluted, 760–4247, Ventana Medical Systems, Inc, Oro Valley, AZ), anti–von Willebrand Factor (dilution 1:4000, A0082, Agilent Technologies, Santa Clara, CA), anti–fibrin II (dilution 1:100, NYBT2G1, Accurate Chemicals, Westbury, NY), anti–d-dimer (dilution 1:100, NBP1-05045, Novus Biologicals), and anticomplement C5b-9 (dilution 1:800, ab66768, abcam, Cambridge, MA). Area of percent positive staining was quantified by ZEN (Zeiss) or HALO software (Indica Labs) as previously described.4

Total RNA was extracted from myocardial tissue and coronary artery samples. Concentration and quality of RNA samples were measured. Quantitative reverse transcription polymerase chain reaction was performed using specifically designed primers for SARS-CoV-2 (N1 and N2 from CDC EUA assay, Integrated DNA Technologies, Coralville, IA). Ribonuclease P was used as a control, and all RNA samples from the lung and heart had a cycle threshold ≤40. The detection of SARS-CoV-2 RNA was determined by the amplification at cycle threshold ≤40.5

To detect SARS-CoV-2 within formalin-fixed paraffin-embedded tissue sections, RNA scope in situ hybridization (Advanced Cell Diagnostics, Hayward, CA) was visualized by confocal microscopy (LSM800, Zeiss, Oberkochen, Germany). SARS-CoV-2 viral RNA–positive cells were visualized by the colocalization of SARS-CoV-2 positive-sense (genomic; violet) and negative-sense (replicative intermediate; turquoise) probes. Immunofluorescence staining for CD61/CD42b and vascular endothelial cadherin was performed using the same or adjacent sections as in situ hybridization.

For transmission electronic microscopy (TEM), heart samples were fixed with 2.5% glutaraldehyde in 0.1 M sodium phosphate buffer at 4°, and then 1% osmium tetroxide was used for postfixation. Tissues were dehydrated by graded alcohol and embedded in EPON Resin. Ultrathin sections were cut at 80 nm with a microtome. Specimens were assessed with the TEM (Hitachi H-7650, Hitachi Science Systems Ltd, Tokyo, Japan).

Analysis of Coronary Artery Aspirates

A comparative analysis of the constituents of thrombus was performed between microthrombi derived from autopsy cases from subjects with COVID-19 (n=5), intramyocardial thromboemboli from COVID-19–negative subjects (n=5) obtained at random from the CVPath Sudden Cardiac Death Registry, and thrombus aspirates retrieved from the culprit lesion of coronary arteries in COVID-19–positive and COVID-19–negative STEMI cases (5 of each) presenting with STEMI, and undergoing primary percutaneous coronary intervention in the same time span of the pathology data collection. Manual thrombus aspiration was performed according to clinical practice (indication and procedure). Patients with an occluded culprit coronary artery TIMI (Thrombolysis in Myocardial Infarction) flow rate 0 to 1 at baseline coronary angiography, or patients with an open artery (TIMI 2–3) but with large filling defect or haziness in a segment reachable by a thrombectomy catheter, were selected for the study. Aspirates were fixed in 10% formalin and shipped to CVPath for analysis. This protocol was reviewed and approved by the ethical committee at Ospedale Papa Giovanni XXIII, Bergamo, Italy (2020-082), and by the CVPath Institute Institutional Review Board, Gaithersburg, MD (RP0112). Aspirates were embedded in paraffin, cut, and stained with the indicated antibodies as described earlier.

Statistical Analysis

Normality of data was checked using the Shapiro-Wilk test. For normally distributed data, ANOVA or Student t test was used to compare data. For multiple comparisons testing, Tukey-Kramer post hoc testing was used. Categorical values were analyzed by the χ2 or Fisher exact test as appropriate. For nonnormally distributed data, the Wilcoxon rank-sum test and post hoc Steel-Dwass tests were applied. A P value <0.05 was considered significant. JMP (version 14, SAS, Cary, NC) or GraphPad Prism (GraphPad Software version 8.2.1, La Jolla, CA) software was used for statistical analysis as appropriate.

Results

Clinical characteristics of the 40 autopsy cases examined are shown in Table 1. The average age was 74 years, with the majority of patients (29/40, 72.5%) being male. Most (90%) were admitted for severe respiratory failure (median Pao2/fraction of inspired oxygen ratio, 123), whereas 4 patients (10%) presented with a cardiovascular emergency (3 STEMI, 1 stroke). Except for the patients admitted with STEMI, no other cases of clinical myocardial infarction were recorded during hospitalization. However, 20% of electrocardiograms in the overall study group showed abnormalities suggestive of ischemia (ie, ST-segment–elevation/depression >0.1 mV, new left bundle-branch block, or inverted T wave), although not associated with chest pain.

Hearts were examined blindly at CVPath Institute without knowledge of the clinical finding except that all subjects had died of COVID-19 (as confirmed by laboratory testing during hospitalization). An analysis revealed the presence of myocyte necrosis in 14 out of the 40 (35%) cases. Of these 14 cases, 3 (21.4%) had evidence of acute myocardial infarction defined as an area of necrosis ≥1 cm2, whereas 11 (78.6%) had focal myocyte necrosis (ie, ≥0.05 mm2 but <1 cm2 contiguous area). There was no difference in the presence of severe coronary artery disease (defined as epicardial coronary stenosis >75% cross-sectional area narrowing) between cases with and without myocardial necrosis. Details of the pathological findings of the heart and lungs are shown in Table 2. Overall, heart weight, LV cavity size, and LV and RV wall thickness were not significantly different between those with and without myocardial necrosis (Figure I in the Data Supplement). Similarly, lung pathology such as diffuse alveolar damage, pulmonary artery thrombi, or microthrombi in alveolar septa were not different between cases with and without necrosis (Table 2). None of the hearts examined had evidence of myocarditis as defined by the European Society of Cardiology.6 Other significant cardiovascular findings such as the presence of amyloid, hypertensive heart disease (ie, hypertrophy), and valvular disease are listed in Table 2.

Table 1. Overall Clinical Characteristics of the 40 Autopsy Cases and According to Presence or Absence of Myocardial Necrosis

Clinical characteristics Total (n=40) Myocardial necrosis (n=14) No necrosis (n=26) P value
Age, y 74.00 (65.00–81.00) 74.50 (69.00–81.25) 73.50 (61.25–81.00) 0.43
Male sex 29 (72.5) 7 (50.0) 22 (84.6) 0.03
Body mass index 27.78 (24.72–29.32) 25.14 (23.11–32.88) 27.83 (25.28–29.50) 0.46
Hypertension 29 (72.5) 10 (71.4) 19 (73.1) 1.00
Dyslipidemia 10 (25.0) 3 (21.4) 7 (26.9) 1.00
Diabetes 11 (27.5) 4 (28.6) 7 (26.9) 0.70
Known history of coronary artery disease 8 (20.0) 3 (21.4) 5 (19.2) 1.00
Previous myocardial infarction 3 (7.5) 1 (7.1) 2 (7.7) 1.00
Peripheral artery disease 6 (15.0) 1 (7.1) 5 (19.2) 0.40
Valvular heart disease 1 (2.5) 1 (7.1) 0 (0) 0.35
Chronic obstructive pulmonary disease 3 (7.5) 0 (0.0) 3 (11.5) 0.54
Chronic kidney disease 7 (17.5) 5 (35.7) 2 (7.7) 0.04
Autoimmune disorder 3 (7.5) 0 (0.0) 3 (11.5) 0.54
Immunosuppression state
 Active 8 (20.0) 5 (35.7) 3 (11.5) 0.10
 Leukemia/lymphoma 5 (12.5) 3 (21.4) 2 (7.7) 0.32
 Previous transplantation 3 (7.5) 2 (14.3) 1 (3.9) 0.28
Therapy at home
 Acetylsalicylic acid 18 (45.0) 6 (42.9) 12 (46.2) 1.00
 Angiotensin-converting enzyme inhibitors 10 (25.0) 4 (28.6) 6 (23.1) 0.72
 Angiotensin receptor blockers 3 (7.5) 0 (0.0) 3 (11.5) 0.54
 Statin 8 (20.0) 2 (14.3) 6 (23.1) 0.69
 Chronic oral anticoagulant 3 (7.5) 2 (14.3) 1 (3.9) 0.28
Recent history
 Cause of admission
  Respiratory failure 36 (90.0) 11 (78.6) 25 (96.2) 0.04
  ST-segment–elevation myocardial infarction 3 (7.5) 3 (21.4) 0 (0.0)
  Stroke 1 (2.5) 0 (0.0) 1 (3.9)
 Interval symptom onset–admission (days) 5.00 (4.00–10.00) 4.00 (0.50–6.00) 6.50 (4.00–10.00) 0.02

Values shown are n (%) or median (interquartile range).

Table 2. Pathological Findings of the Heart and Lung in Subjects With and Without Myocardial Necrosis

Total (n=40) Myocardial necrosis (n=14) No necrosis (n=26) P value
Myocardial necrosis
 Acute myocardial infarction (≥1 cm2) 3 (7.5) 3 (21.4) 0 (0) 0.014
 Focal myocyte necrosis (≥0.05 mm2 but <1 cm2) 11 (27.5) 11 (78.6) 0 (0) 0.001
Thrombus
 Epicardial coronary artery thrombus 3 (7.5) 2 (14.2) 1 (3.8)* 0.23
 Microthrombi 9 (22.5) 9 (64.3) 0 (0) 0.001
Coronary artery disease
 Coronary stent 6 (15) 3 (21.4) 3 (11.5) 0.40
 Single vessel disease (>75% cross-sectional area) 9 (22.5) 3 (21.4) 6 (23.1) 0.91
 Multivessel disease (>75% cross-sectional area in ≥2 epicardial vessels) 9 (22.5) 3 (21.4) 6 (23.1) 0.91
Other cardiac findings
 Myocarditis 0 (0) 0 (0) 0 (0)
 Hypertrophy of myocardium 29 (72.5) 10 (71.4) 19 (73.1) 0.91
 Valvular heart disease 2 (5.0) 2 (14.3)† 0 (0) 0.21
 Cardiac amyloidosis 6 (14.3) 2 (14.3) 4 (15.4) 0.93
Lung findings
 Diffuse alveolar damage 36 (92.3) 12 (92.3) 24 (88.9) 0.51
 Pulmonary artery thrombus 18 (46.2) 5 (38.5) 13 (48.1) 0.39
 Microthrombi in alveolar septa 10 (25.6) 5 (38.5) 5 (18.5) 0.25

Values shown are n (%).

*Sudden cardiac death.

†Status postaortic valve replacement (n=1), moderate aortic stenosis (n=1).

Once the pathological findings were characterized, the clinical and laboratory findings were unblinded and divided according to the presence and absence of myocardial necrosis as shown in Tables 1 and 3. Overall, subjects with necrosis were more frequently female compared with the patients without necrosis (50% versus 84.6%, P=0.03), had a greater prevalence of chronic kidney disease (35.7% versus 7.7%, P=0.04), had a higher rate of STEMI at presentation (21.4% versus 0%, P=0.04), and had a shorter interval between symptom onset and hospital admission (4.0 versus 6.5 days, P=0.02). Subjects with myocardial necrosis had lower values of hemoglobin and C-reactive protein, greater high-sensitivity troponin I values (23 386 versus 226, P=0.03), and a higher rate of ischemic EKG changes (42.9% versus 8.3%, P=0.03; Table 3). No difference in the degree of respiratory impairment was detected, as confirmed by the similar values of Pao2/Fio2 ratio and similar degrees of ventilatory support. In-hospital therapy and rate of adverse events were also similar between the subjects with versus without myocardial necrosis (Table I in the Data Supplement). Molecular analysis of RNA extracted from the lungs and various areas of the heart revealed the presence of virus in the lungs as detected by polymerase chain reaction in 34 of the 40 cases (85%), but it was detectable in the heart in only 8 cases (20%) (Table II in the Data Supplement). There was no difference in virus presence in cases of necrosis versus no necrosis.

Table 3. Overall Laboratory Characteristics of the 40 Autopsy Cases and According to Presence or Absence of Myocardial Necrosis

Laboratory values Available (n) Total (n=40) Myocardial necrosis (n=14) No necrosis (n=26) P value
Hemoglobin (g/dL) 40 13.15 (11.03–14.60) 11.65 (9.70–13.55) 13.45 (11.63–15.13) 0.04
Total white blood cell count (n/mm3) 40 9415 (6128–13 063) 9655 (6215–12 867) 9415 (5990–13 557) 0.10
Neutrophil
 Count (n/mm3) 40 6884 (4275–9713) 7830 (5533–11 370) 6050 (4145–9427) 0.22
 Relative (%) 40 84.15 (70.53–89.93) 89.60 (70.88–93.05) 82.30 (69.90–86.63) 0.10
Lymphocytes
 Count (n/mm3) 36 725 (520–1078) 500 (170–1100) 750 (558–1059) 0.19
 Relative (%) 36 10.10 (5.08–15.78) 4.40 (2.70–24.10) 10.20 (6.55–14.85) 0.34
Platelets (n/mm3) 40 208 500 (136 250–292 750) 215 500 (132 500–315 750) 207 000 (134 500–255 750) 0.49
C-reactive protein (mg/dL) 40 15.45 (9.18–26.20) 11.15 (3.90–17.85) 18.90 (14.38–28.93) 0.02
Creatinine (mg/dL) 40 1.10 (0.86–1.78) 0.98 (0.75–1.82) 1.17 (0.91–1.82) 0.35
Serum glutamic oxaloacetic transaminase (U/L) 39 47.00 (33.00–86.00) 42.00 (26.00–81.50) 53.50 (38.75–87.25) 0.23
Serum glutamic pyruvic transaminase (U/L) 40 40.00 (28.25–67.50) 38.50 (20.00–80.00) 40.00 (29.75–66.50) 0.74
Troponin I (ng/L)* 15 419 (73–1000) 23 386 (334–115 786) 226 (39–526) 0.03
D-dimer (ng/mL) 26 5059 (1709–16 158) 5117 (1418–15 471) 5000 (1887–17 145) 0.73
Prothrombin time international normalized ratio 40 1.10 (1.04–1.18) 1.10 (1.04–1.31) 1.10 (1.02–1.17) 0.73
Activated partial thromboplastin time 40 1.12 (1.00–1.22) 1.11 (0.98–1.29) 1.12 (1.01–1.21) 0.79
Interleukin-6 (pg/mL) 22 108.10 (68.33–210.75) 125.00 (61.05–208.50) 96.20 (66.80–265.50) 0.97
Pao2/Fio2 ratio at presentation 36 123.00 (92.00–176.88) 112.22 (93.00–181.00) 140.0 (86.00–179.17) 0.73
Ischemic ECG changes† 38 8 (20.0) 6 (42.9) 2 (8.3) 0.03
Abnormal cardiac ultrasound findings 14 8 (57.1) 5 (83.3) 3 (37.5) 0.14
 Regional wall motion impairment 3 (21.4) 3 (50.0) 0 (0.0) 0.05
 Impaired right ventricle 5 (35.7) 2 (33.3) 3 (37.5) 1.00

Values shown are n (%) or median (interquartile range).

*High-sensitivity troponin I analysis was performed based on clinical needs, according to judgment of the treating physicians. Median interval from admission to troponin evaluation was 3 days (interquartile range, 1.5–7.5).

†Ischemic ECG changes defined as ST-segment–elevation/depression >0.1 mV, new left bundle-branch block, inverted T wave.

Table 4 lists the pathological findings in the 14 subjects with myocyte necrosis. Two cases had evidence of epicardial coronary thrombosis in the setting of severe coronary atherosclerosis and underwent percutaneous coronary intervention. Four of 14 (28.6%) cases had evidence of RV strain as indicated by RV necrosis (Table 4). Of the 11 cases with focal myocyte necrosis, 8 (72.7%) had microthrombi, whereas 3 (27.3%) showed no microthrombi. Microthrombi were observed in 9 of 14 (64.3%) cases with myocyte necrosis, 8 with focal necrosis, and 1 with acute myocardial infarction. We measured the distribution and extent of focal myocyte necrosis and microvessel thrombosis in subjects with COVID-19. Overall, focal myocardial necrosis was more common in the LV inferior and lateral wall and ventricular septum as follows: inferior 55%, lateral 36%, septum 36%, inferior RV 36%, anterior LV 18%, atria 9%, and anterior RV 0% (Figure II in the Data Supplement). The distribution pattern of microthrombi were similar to focal myocyte necrosis (inferior LV 67%, lateral LV 56%, ventricular septum 44%, inferior RV 44%, anterior LV 22%, anterior RV 11%, and atria 11%) (Figure II in the Data Supplement). Focal necrosis was found in 13% of histology sections (22/154 sections from 11 patients). Similarly, microthrombi were observed in 27.7% of histology sections (31/126 sections from 9 patients). The presence of microthrombi was significantly associated with the presence of focal necrosis in a section-based analysis (P<0.01, Fisher exact test, Figure III in the Data Supplement). A cumulative distribution curve showing the percentage area of microthrombi per 1 mm2 myocardial tissue is shown in Figure IV in the Data Supplement. Only 1 case of microthrombi was associated with acute myocardial infarction. In this case, the subject presented with STEMI involving the inferior and lateral regions of the LV and posterior wall of the RV accompanied by profound cardiogenic shock causing global myocyte necrosis as described previously.3 Coronary artery disease was generally mild to moderate in almost all cases of microthrombi. None of the cases with myocyte necrosis had an unstable coronary plaque phenotype.

Table 4. Pathological Findings of the Heart in Subjects With Myocardial Necrosis

Case no. Type of myocardial injury Location of acute myocardial necrosis Epicardial coronary artery acute thrombus Coronary intervention Epicardial CA stenosis Healed MI Type of thrombus in myocardium
1 AMI (reperfused, transmural infarction) Anterior, ventricular septum, lateral, anterior RV Yes (LAD) Yes, LAD (stented acutely) 50% RCA, 10% LM, 70% LAD*, 70% LCX No PCI-related intramyocardial thrombus
2 AMI (reperfused, subendocardial infarction) Lateral, inferior, anterior LV Yes (LCX) Yes, LCX (in-stent restenosis and acute DCB treatment), RCA (previous stent, open), LAD (in-stent restenosis and acute DCB treatment) 50% RCA, 40% LM, 40% LAD, 40% LCX* Yes DCB (PCI)–related intramyocardial thrombus
3 AMI (focal areas of myocyte necrosis, transmural myocyte necrosis caused by shock) Circumferential No No 50% RCA, 50% LM, 50% LAD, 40% LCX No Microthrombus
4 Focal myocyte necrosis Lateral, inferior LV No Yes, RCA (previous stent, 30%) 80% RCA, 35% LM, 50% LAD, 50% LCX Yes Microthrombus
5 Focal myocyte necrosis Inferior LV, inferior RV No No 60% RCA, 75% LM, 75% LAD, 70% LCX Yes Microthrombus
6 Focal myocyte necrosis Inferior, anterior, lateral LV No No 65% RCA, 40% LM, 65% LAD, 50% LCX No Microthrombus
7 Focal myocyte necrosis Ventricular septum, inferior RV No No 40% RCA, 40% LM, 60% LAD, 70% LCX No Microthrombus
8 Focal myocyte necrosis Inferior LV No No 75% RCA, 30% LM, 65% LAD, 40% LCX No Microthrombus
9 Focal myocyte necrosis Ventricular septum No No 100% CTO RCA, 50% LM, 80% LAD, 70% LCX No Microthrombus
10 Focal myocyte necrosis Lateral LV, right atrium No No 40% RCA, 20% LM, 30% LAD, 50% LCX No Microthrombus
11 Focal myocyte necrosis Anterior, ventricular septum No No 50% RCA, 20% LM, 50% LAD, 0% LCX No Microthrombus
Lateral, inferior LV
12 Focal myocyte necrosis Ventricular septum, inferior RV No No 25% RCA, 25% LM, 25% LAD, 25% LCX No No
13 Focal myocyte necrosis Inferior RV No No 15% RCA, 30% LM, 30% LAD, 25% LCX Yes No
14 Focal myocyte necrosis Inferior LV No No 20% RCA, 5% LM, 30% LAD, 20% LCX No No

AMI indicates acute myocardial infarction; CA, coronary artery; CAD, coronary artery disease; CTO, chronic total occlusion; DCB, drug-coated balloon; LAD, left anterior descending artery; LCX, left circumflex artery; LM, left main trunk; LV, left ventricle; MI, myocardial infarction; PCI, percutaneous coronary intervention; RCA, right coronary artery; and RV, right ventricle.

*Culprit vessel of acute myocardial infarction.

Because recent reports have suggested that SARS-CoV-2 may be found in endothelial cells in the hearts of subjects dying of COVID-19, we conducted in situ hybridization and indirect immunofluorescence as well as TEM to look for virus presence in COVID-19 hearts with evidence of microthrombi. In the 3 hearts examined by in situ hybridization, rare virus presence could be found in cardiac myocytes (without associated inflammation) but not in microvascular endothelial cells with evidence of microthrombi (Figure 2). Similarly, by TEM, no evidence of virus particles could be found within endothelial cells in vessels with and without microthrombi in the 5 hearts examined (Figure 2). These data suggest direct endothelial infection is likely not a major mechanism of cardiac microthrombi formation in subjects with COVID-19 infection.

Figure 2.

Figure 2. In situ hybridization fluorescence microscopy and TEM to detect SARS-CoV-2 in endothelial cells in cases with cardiac microthrombi. A through C, Pathology of COVID-19 autopsy lung (80-year-old female). A, Hematoxylin and eosin–stained section shows thickened alveolar wall with inflammatory cells, including multinuclear giant cells and the formation of hyaline membranes, consistent with exudative phase of diffuse alveolar damage. B, Corresponding low-power image of SARS CoV-2 RNA scope in situ hybridization. Multiple double-positive cells (ie, viral RNA positive-sense [violet] and viral RNA negative-sense [turquoise]) were observed. C, High-power image in white rectangle area in B shows infected alveolar pneumocyte. D through I, Pathology of COVID-19 autopsy heart with myocardial necrosis caused by microthrombi (43-year-old female). D, Representative hematoxylin and eosin images showing microthrombi (black arrows) with surrounding necrosis are observed (black dotted area). E, Small focal lesions of myocardial necrosis were observed (black dotted area; Masson trichrome). F, Corresponding low-power image from E showing immunostaining against VE-cad (endothelial marker, green), CD61+42b (platelet marker, red), and DAPI (nuclear stain). Multiple microvessels with platelet thrombi were observed around areas of myocyte necrosis. G, High-power image in white rectangular area in F shows a microvessel with occlusive thrombus. H, Representative viral RNA scope image of area shown in G, which is also stained against VE-cad (endothelial marker, green). No evidence of viral infection (ie, viral RNA positive-sense [violet] and viral RNA negative-sense [turquoise]) in vascular endothelium (shown by white arrows) was observed. I, TEM image of cardiac microthrombus. A microvessel is filled by a fibrin-rich thrombus. There was no evidence of viral particles within endothelial cells and thrombus. J and K, TEM image obtained from another case of COVID-19 autopsy heart with myocardial necrosis but no microthrombi (66-year-old male). Low-power (J) and high-power images (K) failed to show any viral particles in microvascular endothelium. BM indicates basement membrane; COVID-19, coronavirus disease 2019; Cyto, cytoplasm; DAPI, 4’,6-diamidino-2-phenylindole; Myo, myocyte; Nu, nucleus, RBC, red blood cell; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2; TEM, transmission electron microscopy; Th, thrombus; and VE-cad, vascular endothelial cadherin.

We compared the constituents of thrombi between coronary thrombus aspirates retrieved from the culprit lesions of a separate group of COVID-19–positive and COVID-19–negative STEMI cases (5 of each) treated during the height of the pandemic to 5 of the autopsy cases with myocardial microthrombi already described and to 5 additional cases from COVID-19–negative patients with evidence of intramyocardial thromboemboli (associated with epicardial coronary thrombosis in all cases) from the CVPath Sudden Coronary Death Registry. Clinical characteristics of the 4 groups are shown in Table III in the Data Supplement. Given the previous connection between COVID-19 and hypercoagulability, we examined factors known to be involved with clotting using antibodies against platelets (CD61), fibrin II, von Willebrand Factor, D-dimer, and terminal complement complex C5b-9, also known as the membrane attack complex. Microthrombi from subjects with COVID-19 were richer in fibrin II compared with both myocardial thromboemboli from COVID-19–negative autopsies and aspirates from patients with STEMI (both non–COVID-19 and COVID-19) (fibrin II % area: microthrombi from COVID-19–positive subjects, 71.8 [interquartile range, 57.4–85.1]; non–COVID-19 thromboemboli, 35.6 [interquartile range, 10.1–58.7]; non–COVID-19 STEMI thrombus aspirates, 21.3 [interquartile range, 12.3–24.0]; COVID-19 STEMI thrombus aspirates, 36.7 [interquartile range, 12.4–52.1]; overall P<0.0001). Microthrombi from COVID-19–positive subjects were also richer in C5b-9 compared with non–COVID-19 myocardial thromboemboli and with COVID-19–negative and COVID-19–positive STEMI thrombus aspirates (C5b-9 % area: microthrombi from COVID-19–positive subjects, 19.3 [interquartile range, 6.7–39.2]; non–COVID-19 thromboemboli, 0.001 [interquartile range, 0–0.047]; non–COVID-19 STEMI thrombus aspirates, 0.22 [interquartile range, 0.036–0.28]; and COVID-19 STEMI thrombus aspirates, 0.027 [interquartile range, 0.014–8.5]; overall P<0.0001) (Figure 3). However, thromboemboli associated with epicardial coronary thrombosis from COVID-19–negative cases had significantly greater percent area of CD61 (platelet) staining (overall P=0.0002) compared with the 3 other groups of COVID-19–positive and –negative thrombus aspirated from patients with STEMI and COVID-19–positive microthrombi. Constituents of thrombi aspirated from COVID-19 and non–COVID-19 patients with STEMI were numerically similar.

Figure 3.

Figure 3. Analysis of thrombi aspirated from culprit lesions of COVID-19–positive and –negative STEMI cases, myocardial thromboemboli from COVID-19–negative autopsy subjects, and cardiac microthrombi from COVID-19–positive autopsy subjects. A through L, The histology images of thrombus aspirates were obtained from the same patient of non-COVID-19 (A through F) and COVID-19 (G through L) during primary PCI stained for indicated targets. Thrombus is composed primarily of platelets and fibrin but terminal complement is not present. M through R: The histology images of intramyocardial thromboemboli associated with epicardial coronary thrombosis from non-COVID-19 autopsy cases. S through X: Histology images of COVID-19 microthrombi. Microthrombi immunostaining for the same targets shows they are strongly positive for CD61, vWF, fibrin II, D-dimer, and C5b-9. Y, Quantitative analyses of thrombus components among epicardial coronary arteries thrombus aspirates obtained from STEMI (both non–COVID-19 [n=5] and COVID-19 [n=5]) patients, intramyocardial thromboemboli associated with epicardial coronary thrombosis from non–COVID-19 autopsy cases (n=5), and microthrombus in COVID-19 autopsy cases (n=5). Overall P value for each analysis is shown (Wilcoxon rank sum test); data are expressed as median with interquartile range. C indicates complement; COVID-19, coronavirus disease 2019; H&E, hematoxylin and eosin stain; STEMI, ST-segment–elevation myocardial infarction, and vWF, von Willebrand factor.

Discussion

COVID-19 infection continues to be a major cause of mortality throughout the world. Evidence of cardiac injury as indicated by elevated levels of cardiac troponin is not uncommon in hospitalized patients and increases risk of death. In 1 case series from New York City involving 18 patients with confirmed diagnosis of COVID-19 and ST-segment–elevation, 44% received a diagnosis of acute coronary thrombosis causing myocardial infarction, whereas 56% had evidence of noncoronary myocardial injury (defined as nonobstructive disease on coronary angiography).7 Of the patients with noncoronary myocardial injury, 90% died, suggesting a desperate need for better understanding its pathogenesis and how best to treat them. Here, we report the first systematic analysis of the causes of cardiac injury in subjects dying of COVID-19 infection. In our series, 35% of subjects had evidence of cardiac injury as indicated by myocardial necrosis at autopsy. The most common cause of necrosis was microthrombi, found in 64% of cases with myocyte necrosis. Microthrombi were distinctly different in composition compared with epicardial coronary thrombus aspirates from STEMI cases, consisting of higher levels of fibrin and terminal complement.

Although many studies have focused on pulmonary findings of COVID-19, few pathology studies have been conducted specifically examining the effects of COVID-19 on the heart, and most of these did not describe specific findings related to myocardial injury. In the largest series of autopsies conducted in a New York hospital, hearts from 25 cases were evaluated. Most hearts showed evidence of preexisting atherosclerotic or hypertensive heart disease, with 60% of cases showing nonspecific patchy mild interstitial chronic inflammation within the myocardium without associated myonecrosis.8 More recently, Basso et al reported the findings from 21 autopsy cases collected from 4 hospitals around the world and reported a 14.2% (3/21 cases) incidence of myocarditis.9 Other pathological series have documented rare findings of lymphocytic myocarditis without clear clinical sequelae, whereas clinical case reports have described myocardial injury presumed to be consistent with myocarditis but without actual tissue diagnosis.10,11 Indeed, after quantification of viral load in 39 consecutive autopsy cases from Germany, SARS-CoV-2 could be documented in 24 of 39 (61.5%), with 26 of 39 (41%) having copy numbers >1000 copies per microgram RNA.12 There was no difference in inflammatory infiltrate or leukocyte numbers between individuals with and without cardiac infection. We were able to recover viral RNA in only 20% of hearts studied despite it being detectable in lungs in the vast majority of cases (85%). In our series, there were no differences in the percentage of cases with viral RNA detected in the heart with and without myocardial necrosis (14.3% versus 23.1%, P=0.51), suggesting direct viral invasion of the heart does not play a major role in the development of necrosis.

Although direct infection of the lungs with resulting multifocal pneumonia is thought to be the major cause of death in patients with COVID-19, inflammatory cytokine syndrome may also be an important cause of morbidity and mortality. Li et al reported significantly increased levels of interleukin (IL)–8, IL-6, tumor necrosis factor–α, macrophage chemotactic protein-1, and regulated upon activation, normal T cell expressed and presumably secreted in severe COVID-19 cases, and IL-6 and IL-8 were associated with disease progression.13 It is thought that severely ill patients with COVID-19 are at an increased risk for thromboembolic events, including pulmonary microthrombi as well as venous thrombosis, perhaps resulting from cytokine storm. We show the presence of microthrombi in the hearts of COVID-19–related deaths as the leading cause of cardiac injury. An autopsy case series of 32 cases from New York City reported 77% had elevated troponin I. Pathologically, 1 case showed epicardial coronary artery thrombi, and 19% had intramyocardial small vessel thrombi.14 Furthermore, Bois et al found nonocclusive microthrombi in the small intramyocardial vasculature in 80% (12/15) of COVID-19 autopsy cases.15 Of note, they reported that only 2 out of 15 patients (13.3%) had acute ischemic injury. A multicenter autopsy study by Basso et al reported acute myocyte injury in the RV and thrombi in small vessels of myocardium in 4 out of 21 cases (19% in both cases).9 However, their distribution and association with necrosis were not described. We had previously reported 1 case of microthrombi in a COVID-19–infected young female patient who died of acute myocardial infarction and cardiogenic shock, but now we have expanded our findings in a larger series of cases (n=40) and found that microthrombi are the leading cause of cardiac injury (defined as myocyte necrosis). Rapkiewicz et al also reported autopsy findings of 7 cases with COVID-19, with 5 of the 7 being hospitalized at the time of death and 2 having sudden cardiac death at home.16 In all 7 cases, fibrin microthrombi were identified in the heart; however, the extent of microthrombi and their location were not specifically identified, nor was the extent of myocardial necrosis. Although staining for complement (C4d) was performed to rule out complement-mediated myocyte damage, and was negative in all cases tested, complement in microthrombi was not specifically examined.

Cardiac microthrombi would not be detectable clinically because no laboratory test can specifically detect microthrombi, but future studies should be directed toward developing methods and laboratory testing to diagnose this type of injury. Overall, there was a significant difference in ischemic EKG changes in those with versus without myocyte necrosis, although sensitivity of such findings was poor, because only 6 of the 14 (42.9%) cases showed myocardial injury by EKG. Clinical presentation may also be misleading, because chest pain may be absent or significantly underreported, especially in patients with from severe respiratory impairment.

We found that microthrombi were distinct in composition compared with epicardial thrombus aspirates from patients with STEMI with and without COVID-19 infection with higher levels of fibrin and terminal complement. A recent study of SARS-CoV, which is closely related to SARS-CoV-2, found disease exacerbation was related to the activation of complement C3.17 Others have reported that SARS-CoV-2 autoactivates MASP-2 (mannan-binding lectin-associated serine protease 2), the primary enzymatic initiator of the lectin pathway.18 MASP-2 activation leads to generation of C3 convertase and activation of the membrane attack complex (C5b-9).19 Moreover, alteration of the MASP-2–binding motif, either by Masp2 deletion or blocking the MASP-2–N protein interaction, attenuated lung injury.18 Immunohistochemistry analysis of pulmonary autopsy samples revealed MASP-2 and C5b-9 deposition localized in interalveolar septa. These data, along with human proteomic studies, suggest that coronavirus infections are associated with the activation of multiple complement pathways.20–22

Moreover, inhibiting complement pathway seems to have a therapeutic effect, at least in experimental models.17,21 C3-deficient mice infected with SARS-CoV exhibited less respiratory problems despite similar viral loads in the lung, with lower levels of cytokines found in the lung and serum.17 The reduction in lung neutrophils reduced intrapulmonary and plasma IL-6 levels. At present, C3 blockade with agents such as AMY-101 are undergoing clinical trials in patients infected with COVID-19 (NCT04395456).

We should also address some of the limitations of this study. Autopsy material from subjects dying of COVID-19 infection may have its own biases and may not be reflective of cardiac findings in those who survive COVID-19 infection. Thus, the true incidence of cardiac injury and microthrombi may be different in subjects who survive COVID-19 infection. Moreover, the lack of troponin levels in all subjects also may limit the clinical translation of myocardial necrosis detected at autopsy. Although we surveyed all regions of the heart at multiple levels, small areas of necrosis and microthrombi may have been missed because of sampling. Nonetheless, we believe this study reveals important and novel insights about the nature of cardiac injury in subjects dying of COVID-19 infection.

In conclusion, our study is the first to examine systematically the causes of cardiac injury in patients dying of COVID-19 infection. Here, we report that 35% of subjects dying with COVID-19 had evidence of cardiac injury as identified by the presence of myocyte necrosis, with the majority (78.6%) having focal myocyte necrosis. The major cause of myocyte necrosis was cardiac microthrombi, occurring in 64.3% of those with myocyte necrosis. Microthrombi were different in thrombus constituents with richer fibrin and complement compared with intramyocardial thrombi from COVID-19–negative subjects and from thrombi aspirated from coronary arteries of both COVID-19–positive and –negative STEMI cases. Our data suggest that microvascular thrombosis should be entertained as a likely cause of cardiac injury in hospitalized patients with COVID-19 and that further investigation of antiplatelet, anticoagulant, and anticomplement therapies that specifically target microthrombi should be examined in clinical trials.

Supplemental Materials

Data Supplement Figures I–IV

Data Supplement Tables I–III

Disclosures The CVPath Institute has received institutional research support from the Leducq Foundation (grant no. R01 HL141425); 480 Biomedical; 4C Medical; 4Tech; Abbott; Accumedical; Amgen; Biosensors; Boston Scientific; Cardiac Implants; Celonova; Claret Medical; Concept Medical; Cook; CSI; DuNing, Inc; Edwards LifeSciences; Emboline; Endotronix; Envision Scientific; Lutonix/Bard; Gateway; Lifetech; Limflo; MedAlliance; Medtronic; Mercator; Merill; Microport Medical; Microvention; Mitraalign; Mitrassist; NAMSA; Nanova; Neovasc; NIPRO; Novogate; Occulotech; OrbusNeich Medical; Phenox; Profusa; Protembis; Qool; Recor; Senseonics; Shockwave; Sinomed; Spectranetics; Surmodics; Symic; Vesper; WL Gore; and Xeltis. A.V.F. has received honoraria from Abbott Vascular, Biosensors, Boston Scientific, Celonova, Cook Medical, CSI, Lutonix Bard, Sinomed, and Terumo Corporation; and is a consultant to Amgen, Abbott Vascular, Boston Scientific, Celonova, Cook Medical, Lutonix Bard, and Sinomed. A.C. receives research grants from University Hospital RWTH Aachen. R.V. has received honoraria from Abbott Vascular, Biosensors, Boston Scientific, Celonova, Cook Medical, Cordis, CSI, Lutonix Bard, Medtronic, OrbusNeich Medical, CeloNova, SINO Medical Technology, ReCore, Terumo Corporation, WL Gore, and Spectranetics; and is a consultant Abbott Vascular, Boston Scientific, Celonova, Cook Medical, Cordis, CSI, Edwards Lifescience, Lutonix Bard, Medtronic, OrbusNeich Medical, ReCore, Sinomededical Technology, Spectranetics, Surmodics, Terumo Corporation, WL Gore, and Xeltis. The other authors report no conflict.

Footnotes

*Drs Pellegrini and Kawakami contributed equally.

†Drs Sakamoto and Kawai contributed equally.

Sources of Funding, see page 1041

The Data Supplement, podcast, and transcript are available with this article at https://www.ahajournals.org/doi/suppl/10.1161/CIRCULATIONAHA.120.051828.

Aloke V. Finn, MD, CVPath Institute Inc, 19 Firstfield Road, Gaithersburg, MD 20878. Email

References

  • 1. Lala A, Johnson KW, Januzzi JL, Russak AJ, Paranjpe I, Richter F, Zhao S, Somani S, Van Vleck T, Vaid A, et al ; Mount Sinai COVID Informatics Center. Prevalence and impact of myocardial injury in patients hospitalized with COVID-19 infection. J Am Coll Cardiol. 2020; 76:533–546. doi: 10.1016/j.jacc.2020.06.007CrossrefMedlineGoogle Scholar
  • 2. Hendren NS, Drazner MH, Bozkurt B, Cooper LT . Description and proposed management of the acute COVID-19 cardiovascular syndrome. Circulation. 2020; 141:1903–1914. doi: 10.1161/CIRCULATIONAHA.120.047349LinkGoogle Scholar
  • 3. Guagliumi G, Sonzogni A, Pescetelli I, Pellegrini D, Finn AV . Microthrombi and ST-segment-elevation myocardial infarction in COVID-19. Circulation. 2020; 142:804–809. doi: 10.1161/CIRCULATIONAHA.120.049294LinkGoogle Scholar
  • 4. Torii S, Jinnouchi H, Sakamoto A, Romero ME, Kolodgie FD, Virmani R, Finn AV . Comparison of biologic effect and particulate embolization after femoral artery treatment with three drug-coated balloons in healthy swine model. J Vasc Interv Radiol. 2019; 30:103–109. doi: 10.1016/j.jvir.2018.07.025CrossrefMedlineGoogle Scholar
  • 5. Ramlall V, Thangaraj PM, Meydan C, Foox J, Butler D, Kim J, May B, De Freitas JK, Glicksberg BS, Mason CE, et al . Immune complement and coagulation dysfunction in adverse outcomes of SARS-CoV-2 infection. Nat Med. 2020; 26:1609–1615. doi: 10.1038/s41591-020-1021-2CrossrefMedlineGoogle Scholar
  • 6. Caforio AL, Pankuweit S, Arbustini E, Basso C, Gimeno-Blanes J, Felix SB, Fu M, Heliö T, Heymans S, Jahns R, et al ; European Society of Cardiology Working Group on Myocardial and Pericardial Diseases. . Current state of knowledge on aetiology, diagnosis, management, and therapy of myocarditis: a position statement of the European Society of Cardiology Working Group on Myocardial and Pericardial Diseases. Eur Heart J. 2013; 34:2636–48, 2648a. doi: 10.1093/eurheartj/eht210CrossrefMedlineGoogle Scholar
  • 7. Bangalore S, Sharma A, Slotwiner A, Yatskar L, Harari R, Shah B, Ibrahim H, Friedman GH, Thompson C, Alviar CL, et al . ST-segment elevation in patients with Covid-19 - acase series. N Engl J Med. 2020; 382:2478–2480. doi: 10.1056/NEJMc2009020CrossrefMedlineGoogle Scholar
  • 8. Bryce C, Grimes Z, Pujadas E, Ahuja S, Beasley MB, Albrecht R, Hernandez T, Stock A, Zhao Z, Al Rasheed M, et al . Pathophysiology of SARS-CoV-2: targeting of endothelial cells renders a complex disease with thrombotic microangiopathy and aberrant immune response. The Mount Sinai COVID-19 autopsy experience [published online May 22, 2020].medRxiv. https://www.medrxiv.org/content/10.1101/2020.05.18.20099960v1. doi: 10.1101/2020.05.18.20099960Google Scholar
  • 9. Basso C, Leone O, Rizzo S, De Gaspari M, van der Wal AC, Aubry MC, Bois MC, Lin PT, Maleszewski JJ, Stone JR . Pathological features of COVID-19-associated myocardial injury: a multicentre cardiovascular pathology study. Eur Heart J. 2020; 41:3827–3835. doi: 10.1093/eurheartj/ehaa664CrossrefMedlineGoogle Scholar
  • 10. Schaller T, Hirschbühl K, Burkhardt K, Braun G, Trepel M, Märkl B, Claus R . Postmortem examination of patients with COVID-19. JAMA. 2020; 323:2518–2520. doi: 10.1001/jama.2020.8907CrossrefMedlineGoogle Scholar
  • 11. Schurink B, Roos E, Radonic T, Barbe E, Bouman CSC, de Boer HH, de Bree GJ, Bulle EB, Aronica EM, Florquin S, et al . Viral presence and immunopathology in patients with lethal COVID-19: a prospective autopsy cohort study. Lancet Microbe. 2020; 1:e290–e299. doi: 10.1016/S2666-5247(20)30144-0CrossrefMedlineGoogle Scholar
  • 12. Lindner D, Fitzek A, Bräuninger H, Aleshcheva G, Edler C, Meissner K, Scherschel K, Kirchhof P, Escher F, Schultheiss HP, et al . Association of cardiac infection with SARS-CoV-2 in confirmed COVID-19 autopsy cases. JAMA Cardiol. 2020; 5:1281–1285. doi: 10.1001/jamacardio.2020.3551CrossrefMedlineGoogle Scholar
  • 13. Li S, Jiang L, Li X, Lin F, Wang Y, Li B, Jiang T, An W, Liu S, Liu H, et al . Clinical and pathological investigation of patients with severe COVID-19.JCI Insight. 2020; 5:e138070. doi: 10.1172/jci.insight.138070CrossrefMedlineGoogle Scholar
  • 14. Elsoukkary SS, Mostyka M, Dillard A, Berman DR, Ma LX, Chadburn A, Yantiss RK, Jessurun J, Seshan SV, Borczuk AC, et al . Autopsy findings in 32 patients with COVID-19: a single-institution experience. Pathobiology. 2020; 88:1–13. doi: 10.1159/000511325Google Scholar
  • 15. Bois MC, Boire NA, Layman AJ, Aubry MC, Alexander MP, Roden AC, Hagen CE, Quinton RA, Larsen C, Erben Y, et al . COVID-19-associated nonocclusive fibrin microthrombi in the heart. Circulation. 2021; 143:230–243. doi: 10.1161/CIRCULATIONAHA.120.050754LinkGoogle Scholar
  • 16. Rapkiewicz AV, Mai X, Carsons SE, Pittaluga S, Kleiner DE, Berger JS, Thomas S, Adler NM, Charytan DM, Gasmi B, et al . Megakaryocytes and platelet-fibrin thrombi characterize multi-organ thrombosis at autopsy in COVID-19: a case series. EClinicalMedicine. 2020; 24:100434. doi: 10.1016/j.eclinm.2020.100434CrossrefMedlineGoogle Scholar
  • 17. Gralinski LE, Sheahan TP, Morrison TE, Menachery VD, Jensen K, Leist SR, Whitmore A, Heise MT, Baric RS . Complement activation contributes to severe acute respiratory syndrome coronavirus pathogenesis. mBio. 2018; 9:01753-18.CrossrefGoogle Scholar
  • 18. Gao T, Hu M, Zhang X, Li H, Zhu L, Liu H, Dong Q, Zhang Z, Wang Z, Hu Y, et al . Highly pathogenic coronavirus N protein aggravates lung injury by MASP-2-mediated complement over-activation. medRxiv. 2020. https://www.medrxiv.org/content/10.1101/2020.03.29.20041962v3. doi: 10.1101/2020.03.29.20041962Google Scholar
  • 19. Noris M, Benigni A, Remuzzi G . The case of complement activation in COVID-19 multiorgan impact. Kidney Int. 2020; 98:314–322. doi: 10.1016/j.kint.2020.05.013CrossrefMedlineGoogle Scholar
  • 20. Shen B, Yi X, Sun Y, Bi X, Du J, Zhang C, Quan S, Zhang F, Sun R, Qian L, et al . Proteomic and metabolomic characterization of COVID-19 patient sera. Cell. 2020; 182:59–72.e15. doi: 10.1016/j.cell.2020.05.032CrossrefMedlineGoogle Scholar
  • 21. Carvelli J, Demaria O, Vély F, Batista L, Chouaki Benmansour N, Fares J, Carpentier S, Thibult ML, Morel A, Remark R, et al ; Explore COVID-19 IPH Group; Explore COVID-19 Marseille Immunopole Group. . Association of COVID-19 inflammation with activation of the C5a-C5aR1 axis. Nature. 2020; 588:146–150. doi: 10.1038/s41586-020-2600-6CrossrefMedlineGoogle Scholar
  • 22. Polycarpou A, Howard M, Farrar CA, Greenlaw R, Fanelli G, Wallis R, Klavinskis LS, Sacks S . Rationale for targeting complement in COVID-19. EMBO Mol Med. 2020; 12:e12642. doi: 10.15252/emmm.202012642CrossrefMedlineGoogle Scholar

eLetters(0)

eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.

Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.