Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
2.
J Immunol Methods ; 499: 113174, 2021 12.
Article in English | MEDLINE | ID: covidwho-1487843

ABSTRACT

INTRODUCTION: As in other viral infections, anti-nuclear antibodies (ANAs) are observed in SARS-CoV-2 infection. We investigated the presence of autoantibodies in acute COVID-19 and the association with early laboratory findings. MATERIALS AND METHODS: We examined 50 sera (>18 years, 25 Female) from patients with acute COVID-19. ANAs (HEp-20-10 liver biochip), anti-neutrophil cytoplasmic antibody (ANCA, Europlus Granulocyte Mosaic 32) and anti-double stranded DNA were investigated with product of Euroimmune AG (Luebeck, Germany) by indirect immunofluorescence (IIF) method. Also, antibody against cyclic citrullinated peptide (anti-CCP) was examined by a chemiluminisens assay (Euroimmun AG, Luebeck, Germany). Samples from 50 blood bank donors collected before the COVID-19 pandemic were used as controls. RESULTS: The IIF-ANA test was positive in 18% (N = 9/50) of the patients. The median time of sample collection was 7 days (range: 1-28 days) after diagnosis. ANA was positive in only one (2%) control sample. Five (55.5%) patients were ANA positive with a strong titer (3+). There was no relationship between antibody titration and time of sample collection (p = 0,55). Anti-CCP was detected in a nucleolar (3+) positive patient (2%). ANA was detected in 14.28% (N = 1/7, rods-rings (±), p = 0,78) of patients in the intensive care unit(ICU). Patients treated in the clinic have more and higher titers of ANA, mostly in nucleolar patterns, than ICU patients. CONCLUSIONS: The variety of antibodies detected in acute COVID-19 and the uncertainty of how long they persist can lead to confusion, especially in the diagnosis of systemic autoimmune rheumatic diseases for IIF-ANA testing in immunology laboratories. Improvements in cell lines and methods will facilitate the diagnostic process.


Subject(s)
Antibodies, Antinuclear/analysis , COVID-19/diagnosis , Clinical Laboratory Techniques , Fluorescent Antibody Technique, Indirect , Acute Disease , Adult , Aged , Aged, 80 and over , Antibodies, Antinuclear/immunology , COVID-19/immunology , Female , Humans , Male , Middle Aged , Pandemics , Young Adult
3.
Nat Commun ; 12(1): 5417, 2021 09 14.
Article in English | MEDLINE | ID: covidwho-1410404

ABSTRACT

COVID-19 is associated with a wide range of clinical manifestations, including autoimmune features and autoantibody production. Here we develop three protein arrays to measure IgG autoantibodies associated with connective tissue diseases, anti-cytokine antibodies, and anti-viral antibody responses in serum from 147 hospitalized COVID-19 patients. Autoantibodies are identified in approximately 50% of patients but in less than 15% of healthy controls. When present, autoantibodies largely target autoantigens associated with rare disorders such as myositis, systemic sclerosis and overlap syndromes. A subset of autoantibodies targeting traditional autoantigens or cytokines develop de novo following SARS-CoV-2 infection. Autoantibodies track with longitudinal development of IgG antibodies recognizing SARS-CoV-2 structural proteins and a subset of non-structural proteins, but not proteins from influenza, seasonal coronaviruses or other pathogenic viruses. We conclude that SARS-CoV-2 causes development of new-onset IgG autoantibodies in a significant proportion of hospitalized COVID-19 patients and are positively correlated with immune responses to SARS-CoV-2 proteins.


Subject(s)
Autoantibodies/immunology , COVID-19/immunology , Immunoglobulin G/immunology , SARS-CoV-2/immunology , Aged , Antibodies, Antinuclear/blood , Antibodies, Antinuclear/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Autoantibodies/blood , Autoantigens/immunology , Connective Tissue Diseases/immunology , Cytokines/immunology , Female , Hospitalization , Humans , Immunoglobulin G/blood , Male , Middle Aged , SARS-CoV-2/pathogenicity , Viral Proteins/immunology
4.
Front Immunol ; 12: 724047, 2021.
Article in English | MEDLINE | ID: covidwho-1405412

ABSTRACT

Objectives: Impact of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic on individuals with arthritis has been highlighted whereas data on other rheumatic diseases, e.g., systemic lupus erythematosus (SLE), are scarce. Similarly to SLE, severe SARS-CoV-2 infection includes risks for thromboembolism, an unbalanced type I interferon response, and complement activation. Herein, SARS-CoV-2 antibodies in longitudinal samples collected prior to vaccination were analyzed and compared with SLE progression and antinuclear antibody (ANA) levels. Methods: One hundred patients (83 women) with established SLE and a regular visit to the rheumatologist (March 2020 to January 2021) were included. All subjects donated blood and had done likewise prior to the pandemic. SARS-CoV-2 antibody isotypes (IgG, IgA, IgM) to the cell receptor-binding S1-spike outer envelope protein were detected by ELISA, and their neutralizing capacity was investigated. IgG-ANA were measured by multiplex technology. Results: During the pandemic, 4% had PCR-confirmed infection but 36% showed SARS-CoV-2 antibodies of ≥1 isotype; IgA was the most common (30%), followed by IgM (9%) and IgG (8%). The antibodies had low neutralizing capacity and were detected also in prepandemic samples. Plasma albumin (p = 0.04) and anti-dsDNA (p = 0.003) levels were lower in patients with SARS-CoV-2 antibodies. Blood group, BMI, smoking habits, complement proteins, daily glucocorticoid dose, use of hydroxychloroquine, or self-reported coronavirus disease 2019 (COVID-19) symptoms (except fever, >38.5°C) did not associate with SARS-CoV-2 antibodies. Conclusion: Our data from early 2021 indicate that a large proportion of Swedish SLE patients had serological signs of exposure to SARS-CoV-2 but apparently with a minor impact on the SLE course. Use of steroids and hydroxychloroquine showed no distinct effects, and self-reported COVID-19-related symptoms correlated poorly with all antibody isotypes.


Subject(s)
Antibodies, Viral/immunology , COVID-19/epidemiology , COVID-19/immunology , Lupus Erythematosus, Systemic/complications , Lupus Erythematosus, Systemic/immunology , Adult , Aged , Aged, 80 and over , Antibodies, Antinuclear/blood , Antibodies, Antinuclear/immunology , Antibodies, Viral/blood , Female , Humans , Immunoglobulin Isotypes/blood , Immunoglobulin Isotypes/immunology , Immunosuppressive Agents/therapeutic use , Lupus Erythematosus, Systemic/drug therapy , Male , Middle Aged , SARS-CoV-2
5.
Life Sci Alliance ; 4(11)2021 11.
Article in English | MEDLINE | ID: covidwho-1404295

ABSTRACT

High levels of autoimmune antibodies are observed in COVID-19 patients but their specific contribution to disease severity and clinical manifestations remains poorly understood. We performed a retrospective study of 115 COVID-19 hospitalized patients with different degrees of severity to analyze the generation of autoimmune antibodies to common antigens: a lysate of erythrocytes, the lipid phosphatidylserine (PS) and DNA. High levels of IgG autoantibodies against erythrocyte lysates were observed in a large percentage (up to 36%) of patients. Anti-DNA and anti-PS antibodies determined upon hospital admission correlated strongly with later development of severe disease, showing a positive predictive value of 85.7% and 92.8%, respectively. Patients with positive values for at least one of the two autoantibodies accounted for 24% of total severe cases. Statistical analysis identified strong correlations between anti-DNA antibodies and markers of cell injury, coagulation, neutrophil levels and erythrocyte size. Anti-DNA and anti-PS autoantibodies may play an important role in the pathogenesis of COVID-19 and could be developed as predictive biomarkers for disease severity and specific clinical manifestations.


Subject(s)
Antibodies, Antinuclear/immunology , Autoantibodies/immunology , COVID-19/immunology , COVID-19/metabolism , Adult , Aged , Aged, 80 and over , Antibodies, Antinuclear/blood , Autoantibodies/blood , Biomarkers , DNA/chemistry , DNA/immunology , Erythrocytes/immunology , Female , Humans , Male , Middle Aged , Phosphatidylserines/immunology , Prognosis , Retrospective Studies , SARS-CoV-2/isolation & purification , Severity of Illness Index
6.
J Autoimmun ; 123: 102706, 2021 09.
Article in English | MEDLINE | ID: covidwho-1309270

ABSTRACT

Autoimmune phenomena and clinically apparent autoimmune diseases, including autoimmune hepatitis, are increasingly been reported not only after natural infection with the SARS-CoV-2 virus, but also after vaccination against it. We report the case of a 63-year old man without a history of autoimmunity or SARS-CoV-2 natural infection who experienced acute severe autoimmune-like hepatitis seven days after the first dose of the mRNA-1273 SARS-CoV-2 vaccine. Liver histology showed inflammatory portal infiltrate with interface hepatitis, lobular and centrilobular inflammation with centrilobular necrosis, in absence of fibrosis and steatosis. Serum immunoglobulin G was slightly elevated. Autoimmune liver serology showed an indirect immunofluorescence pattern on triple rodent tissue compatible with anti-mitochondrial antibody (AMA), but, unexpectedly, this pattern was not mirrored by positivity for primary biliary cholangitis (PBC)-specific molecular tests, indicating that this antibody is different from classical AMA. Anti-nuclear antibody (ANA) was also positive with a rim-like indirect immunofluorescence pattern on liver and HEp2 cell substrates, similar to PBC-specific ANA; however, anti-gp210 and a large panel of molecular-based assays for nuclear antigens were negative, suggesting a unique ANA in our patient. He carries the HLA DRB1*11:01 allele, which is protective against PBC. Response to prednisone treatment was satisfactory. The clinical significance of these novel specificities needs to be further evaluated in this emerging condition.


Subject(s)
Autoantibodies/immunology , COVID-19 Vaccines/adverse effects , COVID-19/prevention & control , HLA-DRB1 Chains/immunology , Hepatitis, Autoimmune/etiology , Mitochondria/immunology , SARS-CoV-2/immunology , Vaccination/adverse effects , 2019-nCoV Vaccine mRNA-1273 , Animals , Antibodies, Antinuclear/immunology , Antibody Specificity , Autoantigens/immunology , Cell Line , Fluorescent Antibody Technique, Indirect , Hepatitis, Autoimmune/drug therapy , Hepatitis, Autoimmune/immunology , Hepatitis, Autoimmune/pathology , Humans , Immunosuppressive Agents/therapeutic use , Liver/immunology , Liver/pathology , Male , Middle Aged , Prednisone/therapeutic use , Rosuvastatin Calcium/adverse effects , Rosuvastatin Calcium/therapeutic use
7.
Eur J Immunol ; 51(4): 893-902, 2021 04.
Article in English | MEDLINE | ID: covidwho-986037

ABSTRACT

The aim of this study is to evaluate the blood level of anti-heart antibodies (AHA) and its correlation with clinical outcomes in patients with severe and moderate coronavirus disease 2019 (COVID-19). The study included 34 patients (23 males; mean age 60.2 ± 16.6 years) with COVID-19 pneumonia. Besides standard medical examination, the AHA blood levels were observed, including antinuclear antibodies, antiendothelial cell antibodies, anti-cardiomyocyte antibodies (AbC), anti-smooth muscle antibodies (ASMA), and cardiac conducting tissue antibodies. Median hospital length of stay was 14 [13; 18] days. AHA levels were increased in 25 (73.5%) patients. Significant correlation (p < 0.05) of AHA levels with cardiovascular manifestations (r = 0.459) was found. AbC levels correlated with pneumonia severity (r = 0.472), respiratory failure (r = 0.387), need for invasive ventilation (r = 0.469), chest pain (r = 0.374), low QRS voltage (r = 0.415), and levels of C-reactive protein (r = 0.360) and lactate dehydrogenase (r = 0.360). ASMA levels were found to correlate with atrial fibrillation (r = 0.414, p < 0.05). Antinuclear antibodies and AbC levels correlated with pericardial effusion (r = 0.721 and r = 0.745, respectively, p < 0.05). The lethality rate was 8.8%. AbC and ASMA levels correlated significantly with lethality (r = 0.363 and r = 0.426, respectively, p < 0.05) and were prognostically important. AHA can be considered as part of the systemic immune and inflammatory response in COVID-19. Its possible role in the inflammatory heart disease requires further investigation.


Subject(s)
Antibodies, Antinuclear/blood , COVID-19/immunology , COVID-19/pathology , Myocytes, Cardiac/immunology , SARS-CoV-2/immunology , Adult , Aged , Aged, 80 and over , Antibodies, Antinuclear/immunology , Atrial Fibrillation/pathology , Autoantibodies/blood , Autoantibodies/immunology , C-Reactive Protein/analysis , Endothelial Cells/immunology , Female , Heart/physiopathology , Humans , L-Lactate Dehydrogenase/blood , Male , Middle Aged , Muscle, Smooth/immunology , Myocardium/immunology , Pericardial Effusion/pathology , Young Adult
8.
Medicina (Kaunas) ; 56(10)2020 Oct 05.
Article in English | MEDLINE | ID: covidwho-982923

ABSTRACT

The clinical spectrum of novel coronavirus infection appears to be wide, encompassing asymptomatic infection, mild upper respiratory tract illness, and severe viral pneumonia, with respiratory failure and even death. Autoantibodies, especially antiphospholipid antibodies, can occur in severe infections. Other autoantibodies are seldom reported. Here, a 60-year-old female patient without dry-mouth symptoms detected positive for anti-60 kDa SSA/Ro antibodies on day 43 after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. To investigate this unique clinical case of SARS-CoV-2 infection, immunological characteristics of this case were detected by using flow cytometry and were compared to the other three groups of patients-health subjects, 2019 novel coronavirus disease (COVID-19) recovery patients, and Sjögren's syndrome (SS) patients. Monitoring the autoantibody level and the development of subsequently related autoimmune diseases are warranted after SARS-CoV-2 infection.


Subject(s)
Antibodies, Antinuclear/immunology , Antibodies, Viral/immunology , Betacoronavirus/immunology , Coronavirus Infections/immunology , Immunophenotyping , Pneumonia, Viral/immunology , COVID-19 , Female , Flow Cytometry , Humans , Middle Aged , Pandemics , SARS-CoV-2 , Sjogren's Syndrome
9.
Eur Rev Med Pharmacol Sci ; 24(22): 11960-11963, 2020 11.
Article in English | MEDLINE | ID: covidwho-962031

ABSTRACT

Though the exact etiology of autoimmune diseases still remains not completely known, there are various factors which are known to contribute to be trigger of autoimmune diseases. Viral infection is known to be among the other. It is known as the infection from severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV) can be an autoimmune trigger, so, we suppose that SARS-Coronavirus (SARS-CoV-2) could be as well. Several authors have highlighted the temporal consequence between SARS-CoV-2 and autoimmune diseases. In this case report we described a patient admitted for COVID-19 pneumonia with completely negative autoimmunity at admission who developed major pulmonary interstitial disease. During the hospitalization the weaning difficulties from oxygen led us to the repetition of autoimmunity pattern which became positive (both during hospitalization then after two months from dismission) with marked positivity for specific antibodies for myositis even after the patient's infectious healing. In the follow-up, the patient continued to have asthenia and muscle weakness despite steroid therapy. She is still in follow-up and will be further evaluated over time. Can we therefore think that in this case the development of autoimmunity can persist beyond the infectious phase and determine over time the development of a real autoimmune myositis?


Subject(s)
Autoantibodies/immunology , Autoimmune Diseases/immunology , COVID-19/immunology , Lung Diseases, Interstitial/immunology , Muscle Weakness/immunology , Myositis/immunology , Aged , Antibodies, Antineutrophil Cytoplasmic/immunology , Antibodies, Antinuclear/immunology , Antigens, Nuclear/immunology , Asthenia/immunology , Autoimmune Diseases/drug therapy , Autoimmune Diseases/etiology , Autoimmune Diseases/physiopathology , COVID-19/complications , COVID-19/physiopathology , COVID-19/therapy , Female , Humans , Ku Autoantigen/immunology , Mi-2 Nucleosome Remodeling and Deacetylase Complex/immunology , Myositis/drug therapy , Myositis/etiology , Myositis/physiopathology
10.
Clin Rheumatol ; 39(11): 3171-3175, 2020 Nov.
Article in English | MEDLINE | ID: covidwho-730357

ABSTRACT

We treated two patients with severe respiratory failure due to coronavirus disease 2019 (COVID-19). Case 1 was a 73-year-old woman, and Case 2 was a 65-year-old-man. Neither of them had a history of autoimmune disease. Chest computed tomography scans before the antiviral therapy showed bilateral multiple patchy ground-glass opacities (GGO) consistent with COVID-19 pneumonia. The GGO regressed over the course of the antiviral treatment; however, new non-segmental patchy consolidations emerged, which resembled those of interstitial lung disease (ILD), specifically collagen vascular disease-associated ILD. We tested the patients' sera for autoantibodies and discovered that both patients had high anti-SSA/Ro antibody titers. In Case 1, the patient recovered with antiviral therapy alone. However, in Case 2, the patient did not improve with antiviral therapy alone but responded well to corticosteroid therapy (methylprednisolone) and made a full recovery. The relationship between some immunological responses and COVID-19 pneumonia exacerbation has been discussed previously; our discovery of the elevation of anti-SSA/Ro antibodies suggests a contribution from autoimmunity functions of the immune system. Although it is unclear whether the elevation of anti-SSA/Ro antibodies was a cause or an outcome of aggravated COVID-19 pneumonia, we hypothesize that both patients developed aggravated the COVID-19 pneumonia due to an autoimmune response. In COVID-19 lung injury, there may be a presence of autoimmunity factors in addition to the known effects of cytokine storms. In patients with COVID-19, a high level of anti-SSA/Ro52 antibodies may be a surrogate marker of pneumonia severity and poor prognosis.


Subject(s)
Antibodies, Antinuclear/immunology , Coronavirus Infections/immunology , Lung Diseases, Interstitial/immunology , Pneumonia, Viral/immunology , Respiratory Insufficiency/immunology , Aged , Amides/therapeutic use , Antiviral Agents/therapeutic use , Benzamidines , Betacoronavirus , COVID-19 , Coronavirus Infections/complications , Coronavirus Infections/diagnostic imaging , Coronavirus Infections/drug therapy , Female , Glucocorticoids/therapeutic use , Guanidines/therapeutic use , Humans , Hydroxychloroquine/therapeutic use , Lung Diseases, Interstitial/diagnostic imaging , Lung Diseases, Interstitial/drug therapy , Lung Diseases, Interstitial/etiology , Male , Methylprednisolone/therapeutic use , Pandemics , Pneumonia, Viral/complications , Pneumonia, Viral/diagnostic imaging , Pneumonia, Viral/drug therapy , Pregnenediones/therapeutic use , Pyrazines/therapeutic use , Recovery of Function , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/immunology , Respiratory Insufficiency/etiology , SARS-CoV-2 , Severity of Illness Index , Tomography, X-Ray Computed
11.
Clin Rheumatol ; 39(9): 2811-2815, 2020 Sep.
Article in English | MEDLINE | ID: covidwho-679749

ABSTRACT

In the midst of the COVID-19 pandemic, further understanding of its complications points towards dysregulated immune response as a major component. Systemic lupus erythematosus (SLE) is also a disease of immune dysregulation leading to multisystem compromise. We present a case of new-onset SLE concomitantly with COVID-19 and development of antiphospholipid antibodies. An 18-year-old female that presented with hemodynamic collapse and respiratory failure, progressed to cardiac arrest, and had a pericardial tamponade drained. She then progressed to severe acute respiratory distress syndrome, severe ventricular dysfunction, and worsening renal function with proteinuria and hematuria. Further studies showed bilateral pleural effusions, positive antinuclear and antidouble-stranded DNA antibodies, lupus anticoagulant, and anticardiolipin B. C3 and C4 levels were low. SARS-Cov-2 PCR was positive after 2 negative tests. She also developed multiple deep venous thrombosis, in the setting of positive antiphospholipid antibodies and lupus anticoagulant. In terms of pathophysiology, COVID-19 is believed to cause a dysregulated cytokine response which could potentially be exacerbated by the shift in Th1 to Th2 response seen in SLE. Also, it is well documented that viral infections are an environmental factor that contributes to the development of autoimmunity; however, COVID-19 is a new entity, and it is not known if it could trigger autoimmune conditions. Additionally, it is possible that SARS-CoV-2, as it happens with other viruses, might lead to the formation of antiphospholipid antibodies, potentially contributing to the increased rates of thrombosis seen in COVID-19.


Subject(s)
Antiphospholipid Syndrome/immunology , Coronavirus Infections/immunology , Lupus Erythematosus, Systemic/immunology , Pneumonia, Viral/immunology , Adolescent , Anemia/etiology , Antibodies, Anticardiolipin/immunology , Antibodies, Antinuclear/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Antiphospholipid Syndrome/complications , Antiphospholipid Syndrome/diagnosis , Antiphospholipid Syndrome/therapy , Anuria/etiology , Betacoronavirus , COVID-19 , Cardiac Tamponade/diagnostic imaging , Cardiac Tamponade/etiology , Cardiac Tamponade/therapy , Complement C3/immunology , Complement C4/immunology , Coronavirus Infections/complications , Coronavirus Infections/diagnosis , Coronavirus Infections/therapy , DNA/immunology , Echocardiography , Fatal Outcome , Female , Heart Arrest/etiology , Hematuria/etiology , Humans , Lupus Coagulation Inhibitor/immunology , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/complications , Lupus Erythematosus, Systemic/diagnosis , Pandemics , Patient Positioning , Pericardiocentesis , Pneumonia, Viral/complications , Pneumonia, Viral/diagnosis , Pneumonia, Viral/therapy , Prone Position , Proteinuria/etiology , Renal Dialysis , Renal Insufficiency/etiology , Renal Insufficiency/therapy , Respiration, Artificial , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/therapy , Respiratory Insufficiency/etiology , Respiratory Insufficiency/therapy , SARS-CoV-2 , Thrombocytopenia/etiology , Venous Thrombosis/etiology , Ventricular Dysfunction, Left/diagnostic imaging
12.
Rheumatol Int ; 40(10): 1539-1554, 2020 10.
Article in English | MEDLINE | ID: covidwho-646938

ABSTRACT

The coronavirus disease-2019 (COVID-19) pandemic is likely to pose new challenges to the rheumatology community in the near and distant future. Some of the challenges, like the severity of COVID-19 among patients on immunosuppressive agents, are predictable and are being evaluated with great care and effort across the globe. A few others, such as atypical manifestations of COVID-19 mimicking rheumatic musculoskeletal diseases (RMDs) are being reported. Like in many other viral infections, severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection can potentially lead to an array of rheumatological and autoimmune manifestations by molecular mimicry (cross-reacting epitope between the virus and the host), bystander killing (virus-specific CD8 + T cells migrating to the target tissues and exerting cytotoxicity), epitope spreading, viral persistence (polyclonal activation due to the constant presence of viral antigens driving immune-mediated injury) and formation of neutrophil extracellular traps. In addition, the myriad of antiviral drugs presently being tried in the treatment of COVID-19 can result in several rheumatic musculoskeletal adverse effects. In this review, we have addressed the possible spectrum and mechanisms of various autoimmune and rheumatic musculoskeletal manifestations that can be precipitated by COVID-19 infection, its therapy, and the preventive strategies to contain the infection.


Subject(s)
Autoimmune Diseases/physiopathology , Coronavirus Infections/physiopathology , Musculoskeletal Diseases/physiopathology , Pneumonia, Viral/physiopathology , Rheumatic Diseases/physiopathology , Antibodies, Antinuclear/immunology , Antibodies, Antiphospholipid/immunology , Antiviral Agents/adverse effects , Arthralgia/etiology , Arthralgia/immunology , Arthralgia/physiopathology , Autoimmune Diseases/etiology , Autoimmune Diseases/immunology , Betacoronavirus , Blood Coagulation Disorders/etiology , Blood Coagulation Disorders/immunology , Blood Coagulation Disorders/physiopathology , COVID-19 , Coronavirus Infections/complications , Coronavirus Infections/drug therapy , Coronavirus Infections/immunology , Cross Reactions/immunology , Extracellular Traps/immunology , Fibrin Fibrinogen Degradation Products , Guillain-Barre Syndrome/etiology , Guillain-Barre Syndrome/immunology , Guillain-Barre Syndrome/physiopathology , Humans , Lupus Coagulation Inhibitor/immunology , Molecular Mimicry , Mucocutaneous Lymph Node Syndrome/etiology , Mucocutaneous Lymph Node Syndrome/immunology , Mucocutaneous Lymph Node Syndrome/physiopathology , Muscle Weakness/etiology , Muscle Weakness/immunology , Muscle Weakness/physiopathology , Musculoskeletal Diseases/etiology , Musculoskeletal Diseases/immunology , Myalgia/etiology , Myalgia/immunology , Myalgia/physiopathology , Myocarditis/etiology , Myocarditis/immunology , Myocarditis/physiopathology , Pandemics , Pneumonia, Viral/complications , Pneumonia, Viral/drug therapy , Pneumonia, Viral/immunology , Rheumatic Diseases/etiology , Rheumatic Diseases/immunology , SARS-CoV-2 , COVID-19 Drug Treatment
14.
J Investig Med High Impact Case Rep ; 8: 2324709620933438, 2020.
Article in English | MEDLINE | ID: covidwho-535847

ABSTRACT

In this article, we present a case of a young female patient with previously diagnosed lupus pneumonitis, now with a flare and new superimposed COVID-19 infection that was treated with intravenous steroids. On computed tomography scans, she had extensive interstitial lung fibrosis in addition to a positive COVID-19 polymerase chain reaction test requiring 6 L of oxygen via nasal cannula on admission. After administration of methylprednisolone, the patient improved and was weaned off her oxygen requirements and was discharged home.


Subject(s)
Coronavirus Infections/complications , Lupus Erythematosus, Systemic/complications , Pneumonia, Viral/complications , Pneumonia/complications , Antibodies, Antinuclear/immunology , Antirheumatic Agents/therapeutic use , Betacoronavirus , COVID-19 , Cardiac Tamponade , Complement C3/immunology , Complement C4/immunology , Coronavirus Infections/diagnostic imaging , Coronavirus Infections/drug therapy , Coronavirus Infections/therapy , DNA , Disease Progression , Enzyme Inhibitors/therapeutic use , Female , Fibrin Fibrinogen Degradation Products , Glucocorticoids/therapeutic use , Humans , Hydroxychloroquine/therapeutic use , Lung/diagnostic imaging , Lupus Erythematosus, Systemic/diagnostic imaging , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/immunology , Lupus Nephritis , Lymphopenia/etiology , Methylprednisolone/therapeutic use , Mycophenolic Acid/therapeutic use , Oxygen Inhalation Therapy , Pandemics , Pneumonia/diagnostic imaging , Pneumonia/immunology , Pneumonia/therapy , Pneumonia, Viral/diagnostic imaging , Pneumonia, Viral/therapy , SARS-CoV-2 , Tomography, X-Ray Computed , Young Adult , COVID-19 Drug Treatment
SELECTION OF CITATIONS
SEARCH DETAIL