1
|
Dolna-Michno J, Kopiński P, Przybylski G, Wypasek E, Szymańska M, Wędrowska E, Mikołajczyk K, Senderek T, Gagat M. Cytoimmunological Profile of Lower Airways in Post-COVID-19 Syndrome (PCS): Predictive Value of Bronchoalveolar Lavage. J Clin Med 2025; 14:3361. [PMID: 40429360 PMCID: PMC12112653 DOI: 10.3390/jcm14103361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 04/24/2025] [Accepted: 04/30/2025] [Indexed: 05/29/2025] Open
Abstract
Background: It has yet to be determined whether the immunocytological profile of the bronchoalveolar lavage (BAL) in respiratory post-COVID syndrome (PCS) reflects the risk of persistent interstitial lung disease (ILD), including pulmonary fibrosis. In this study, we aimed to assess the prognostic value of the BAL cytoimmunologic profile in PCS-related ILD. Materials and Methods: We enrolled 58 non-smoking patients with a history of COVID-19 and new-onset ILD, divided into PCS remission and PCS persistence groups based on clinical data, including repeated computed tomography and pulmonary function tests. We phenotyped BAL major T cell subsets, immune checkpoints (including programmed cell death-1, PD1), and markers of Th1/Th2/Th17 polarization. Results: The PCS groups compared to the control showed increased total cell, lymphocyte, and neutrophil counts and a high BAL neutrophil:lymphocyte ratio (NLR). PCS persistence compared to the controls presented an increased neutrophil count (26 [17-36] vs. 2.6 [1.9-5.4] 103/mL, median [Q1-Q3], p < 0.001) and percentage, BAL NLR (0.77 [0.26-1.63] vs. 0.21 [0.17-0.31], p < 0.0001), CD8+PD1+ cell percentage (43.5 [34-60.5] vs. 24.5 [22-44]%, p = 0.045), and a decreased CD4:CD8 ratio. A high percentage of CD4+CD196+CD183 cells (relevant to Th17 activity, 6.2 [2.0-9.4] vs. 1.2 [0.7-2.7]%, p = 0.02) and increased BAL supernatant elevated IL-8 levels (62.5 [16-243] vs. 10.9 [3.44-32] pg/mL, p = 0.002) were found in the PCS persistence vs. control groups. In the total PCS group, predicted values of Vital Capacity (VC) [16-243] and Diffusing Lung Capacity for CO (DLCO) correlated negatively with BAL NLR; VC correlated negatively with BAL CD8+PD1+; and DLCO correlated positively with the CD4:CD8 ratio. Conclusions: Worse prognosis in PCS is associated with higher BAL NLR, BAL neutrophilia, an elevated percentage of CD8+PD1+ lymphocytes, and a decline in the CD4:CD8 ratio. Th17 cells and IL-8 participate in lung PCS persistence.
Collapse
Affiliation(s)
- Justyna Dolna-Michno
- Department of Molecular Biology, John Paul II Hospital, 31-202 Kraków, Poland; (P.K.); (E.W.); (M.S.)
- Department of Physiology and Pathophysiology, Medical College, Andrzej Frycz Modrzewski Krakow University, 31-216 Kraków, Poland;
| | - Piotr Kopiński
- Department of Molecular Biology, John Paul II Hospital, 31-202 Kraków, Poland; (P.K.); (E.W.); (M.S.)
- Department of Lung Diseases, Cancer and Tuberculosis, Collegium Medicum, Nicolaus Copernicus University, 85-067 Bydgoszcz, Poland; (G.P.); (E.W.)
| | - Grzegorz Przybylski
- Department of Lung Diseases, Cancer and Tuberculosis, Collegium Medicum, Nicolaus Copernicus University, 85-067 Bydgoszcz, Poland; (G.P.); (E.W.)
| | - Ewa Wypasek
- Department of Molecular Biology, John Paul II Hospital, 31-202 Kraków, Poland; (P.K.); (E.W.); (M.S.)
- Department of Physiology and Pathophysiology, Medical College, Andrzej Frycz Modrzewski Krakow University, 31-216 Kraków, Poland;
| | - Magdalena Szymańska
- Department of Molecular Biology, John Paul II Hospital, 31-202 Kraków, Poland; (P.K.); (E.W.); (M.S.)
| | - Ewelina Wędrowska
- Department of Lung Diseases, Cancer and Tuberculosis, Collegium Medicum, Nicolaus Copernicus University, 85-067 Bydgoszcz, Poland; (G.P.); (E.W.)
| | - Klaudia Mikołajczyk
- Department of Histology and Embryology, Collegium Medicum, Nicolaus Copernicus University, 85-067 Bydgoszcz, Poland; (K.M.); (M.G.)
| | - Tomasz Senderek
- Department of Physiology and Pathophysiology, Medical College, Andrzej Frycz Modrzewski Krakow University, 31-216 Kraków, Poland;
| | - Maciej Gagat
- Department of Histology and Embryology, Collegium Medicum, Nicolaus Copernicus University, 85-067 Bydgoszcz, Poland; (K.M.); (M.G.)
| |
Collapse
|
2
|
Tsukida S, Hongo M, Akasaki K, Sone T, Nishi K. COVID-19 Pneumonia Diagnosed by Bronchoalveolar Lavage Fluid, With CD4 T-cell Depletion Contributing to Prolonged Infection: Two Case Reports. Cureus 2024; 16:e74380. [PMID: 39723265 PMCID: PMC11669300 DOI: 10.7759/cureus.74380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
Irrespective of the underlying disease, patients treated with cluster of differentiation 20 (CD20) antibodies have a higher risk of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) long or severe infection, and there are pitfalls in this diagnosis. We herein report two patients with COVID-19 pneumonia diagnosed by bronchoalveolar lavage fluid (BALF) during lymphoma remission. Nasopharyngeal swabs (NSs) were polymerase chain reaction (PCR)-negative for SARS-CoV-2, and the virus was only detectable in the lungs. In patients with B-cell depletion, the early performance of bronchoalveolar lavage (BAL) is important for diagnosing COVID-19 pneumonia and ruling out opportunistic infections when any evidence of suspected viral pneumonia is observed on computed tomography (CT), even if the NS specimens are PCR-negative and they have no upper respiratory symptoms. In addition, blood tests with lymphocytopenia, BALF with decreased CD4/CD8 ratio, and increased neutralizing antibody titer suggested that not only low humoral immune responses but also CD4 T-cell depletion by bendamustine were associated with virus clearance. Even if neutralizing antibodies are adequate, we must be careful of prolonged COVID-19 due to CD4 T-cell depletion and low humoral immune responses.
Collapse
Affiliation(s)
- Saya Tsukida
- Internal Medicine, Keiju Medical Center, Nanao, JPN
| | - Masato Hongo
- Internal Medicine, Fukui-ken Saiseikai Hospital, Fukui, JPN
| | - Kyota Akasaki
- Internal Medicine, Ishikawa Prefectural Central Hospital, Kanazawa, JPN
| | - Takashi Sone
- Internal Medicine, Ishikawa Prefectural Central Hospital, Kanazawa, JPN
| | - Koichi Nishi
- Internal Medicine, Ishikawa Prefectural Central Hospital, Kanazawa, JPN
| |
Collapse
|
3
|
Bartolini A, Morecchiato F, Antonelli A, Malentacchi F, Rossolini GM, Pollini S. Evaluation of STANDARD™ M10 SARS-CoV-2 from bronchoalveolar lavage samples. Diagn Microbiol Infect Dis 2024; 110:116466. [PMID: 39128208 DOI: 10.1016/j.diagmicrobio.2024.116466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/08/2024] [Accepted: 07/22/2024] [Indexed: 08/13/2024]
Abstract
Detection of SARS-CoV-2 in bronchoalveolar lavage (BAL) is considered as a promising alternative method to detect COVID-19 infection. STANDARD™ M10 SARS-CoV-2 assay on 150 negative and 50 positives BAL samples for SARS-CoV-2 showed 96 % sensitivity, 100 % specificity compared to Allplex™ SARS-CoV-2 assay and a 31.25 genomic copies/mL limit of detection.
Collapse
Affiliation(s)
- Andrea Bartolini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; Microbiology and Virology Unit, Careggi University Hospital, Florence, Italy
| | - Fabio Morecchiato
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alberto Antonelli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; Microbiology and Virology Unit, Careggi University Hospital, Florence, Italy; NARR Joint Laboratory for antimicrobial resistance research and control, University of Florence-IRCCS Don Gnocchi Foundation, Florence, Italy
| | | | - Gian Maria Rossolini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; Microbiology and Virology Unit, Careggi University Hospital, Florence, Italy; NARR Joint Laboratory for antimicrobial resistance research and control, University of Florence-IRCCS Don Gnocchi Foundation, Florence, Italy
| | - Simona Pollini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; Microbiology and Virology Unit, Careggi University Hospital, Florence, Italy; NARR Joint Laboratory for antimicrobial resistance research and control, University of Florence-IRCCS Don Gnocchi Foundation, Florence, Italy.
| |
Collapse
|
4
|
Nardi C, Magnini A, Rastrelli V, Zantonelli G, Calistri L, Lorini C, Luzzi V, Gori L, Ciani L, Morecchiato F, Simonetti V, Peired AJ, Landini N, Cavigli E, Yang G, Guiot J, Tomassetti S, Colagrande S. Laboratory data and broncho-alveolar lavage on Covid-19 patients with no intensive care unit admission: Correlation with chest CT features and clinical outcomes. Medicine (Baltimore) 2024; 103:e39028. [PMID: 39029011 PMCID: PMC11398758 DOI: 10.1097/md.0000000000039028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 07/01/2024] [Indexed: 07/21/2024] Open
Abstract
Broncho-alveolar lavage (BAL) is indicated in cases of uncertain diagnosis but high suspicion of Sars-Cov-2 infection allowing to collect material for microbiological culture to define the presence of coinfection or super-infection. This prospective study investigated the correlation between chest computed tomography (CT) findings, Covid-19 Reporting and Data System score, and clinical outcomes in Coronavirus disease 2019 (Covid-19) patients who underwent BAL with the aim of predicting outcomes such as lung coinfection, respiratory failure, and hospitalization length based on chest CT abnormalities. Study population included 34 patients (range 38-90 years old; 20 males, 14 females) with a positive nucleic acid amplification test for Covid-19 infection, suitable BAL examination, and good quality chest CT scan in the absence of lung cancer history. Pulmonary coinfections were found in 20.6% of patients, predominantly caused by bacteria. Specific correlations were found between right middle lobe involvement and pulmonary co-infections. Severe lung injury (PaO2/FiO2 ratio of 100-200) was associated with substantial involvement of right middle, right upper, and left lower lobes. No significant correlation was found between chest CT findings and inflammatory markers (C-reactive protein, procalcitonin) or hospitalization length of stay. Specific chest CT patterns, especially in right middle lobe, could serve as indicators for the presence of co-infections and disease severity in noncritically ill Covid-19 patients, aiding clinicians in timely interventions and personalized treatment strategies.
Collapse
Affiliation(s)
- Cosimo Nardi
- Department of Experimental and Clinical Biomedical Sciences, Radiodiagnostic Unit n. 2, University of Florence - Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Andrea Magnini
- Department of Experimental and Clinical Biomedical Sciences, Radiodiagnostic Unit n. 2, University of Florence - Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Vieri Rastrelli
- Department of Experimental and Clinical Biomedical Sciences, Radiodiagnostic Unit n. 2, University of Florence - Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Giulia Zantonelli
- Department of Experimental and Clinical Biomedical Sciences, Radiodiagnostic Unit n. 2, University of Florence - Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Linda Calistri
- Department of Experimental and Clinical Biomedical Sciences, Radiodiagnostic Unit n. 2, University of Florence - Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Chiara Lorini
- Department of Health Science, University of Florence, Florence, Italy
| | - Valentina Luzzi
- Department of Clinical and Experimental Medicine, Interventional Pulmonology Unit, Careggi University Hospital, Florence, Italy
| | - Leonardo Gori
- Department of Clinical and Experimental Medicine, Interventional Pulmonology Unit, Careggi University Hospital, Florence, Italy
| | - Luca Ciani
- Department of Clinical and Experimental Medicine, Interventional Pulmonology Unit, Careggi University Hospital, Florence, Italy
| | - Fabio Morecchiato
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Clinical Microbiology and Virology Unit, Florence Careggi University Hospital, Florence, Italy
| | | | - Anna Julie Peired
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
| | - Nicholas Landini
- Department of Radiological, Oncological and Pathological Sciences, Policlinico Umberto I Hospital, “Sapienza” Rome University, Rome, Italy
| | - Edoardo Cavigli
- Department of Radiology, Careggi University Hospital, Florence, Italy
| | - Guang Yang
- Bioengineering Department and Imperial-X, Imperial College London, London, UK
| | - Julien Guiot
- Department of Respiratory Medicine, University Hospital of Liège, Liège, Belgium
| | - Sara Tomassetti
- Department of Clinical and Experimental Medicine, Interventional Pulmonology Unit, Careggi University Hospital, Florence, Italy
| | - Stefano Colagrande
- Department of Experimental and Clinical Biomedical Sciences, Radiodiagnostic Unit n. 2, University of Florence - Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| |
Collapse
|
5
|
Akasov RA, Chepikova OE, Pallaeva TN, Gorokhovets NV, Siniavin AE, Gushchin VA, Savvateeva LV, Vinokurov IA, Khochenkov DA, Zamyatnin AA, Khaydukov EV. Evaluation of molecular mechanisms of riboflavin anti-COVID-19 action reveals anti-inflammatory efficacy rather than antiviral activity. Biochim Biophys Acta Gen Subj 2024; 1868:130582. [PMID: 38340879 DOI: 10.1016/j.bbagen.2024.130582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/03/2024] [Accepted: 02/06/2024] [Indexed: 02/12/2024]
Abstract
BACKGROUND Riboflavin (vitamin B2) is one of the most important water-soluble vitamins and a coenzyme involved in many biochemical processes. It has previously been shown that adjuvant therapy with flavin mononucleotide (a water-soluble form of riboflavin) correlates with normalization of clinically relevant immune markers in patients with COVID-19, but the mechanism of this effect remains unclear. Here, the antiviral and anti-inflammatory effects of riboflavin were investigated to elucidate the molecular mechanisms underlying the riboflavin-induced effects. METHODS Riboflavin was evaluated for recombinant SARS-CoV-2 PLpro inhibition in an enzyme kinetic assay and for direct inhibition of SARS-CoV-2 replication in Vero E6 cells, as well as for anti-inflammatory activity in polysaccharide-induced inflammation models, including endothelial cells in vitro and acute lung inflammation in vivo. RESULTS For the first time, the ability of riboflavin at high concentrations (above 50 μM) to inhibit SARS-CoV-2 PLpro protease in vitro was demonstrated; however, no inhibition of viral replication in Vero E6 cells in vitro was found. At the same time, riboflavin exerted a pronounced anti-inflammatory effect in the polysaccharide-induced inflammation model, both in vitro, preventing polysaccharide-induced cell death, and in vivo, reducing inflammatory markers (IL-1β, IL-6, and TNF-α) and normalizing lung histology. CONCLUSIONS It is concluded that riboflavin reveals anti-inflammatory rather than antiviral activity for SARS-CoV-2 infection. GENERAL SIGNIFICANCE Riboflavin could be suggested as a promising compound for the therapy of inflammatory diseases of broad origin.
Collapse
Affiliation(s)
- Roman A Akasov
- Federal Scientific Research Center "Crystallography and Photonics" of the Russian Academy of Sciences, Moscow 119333, Russia; Institute of Molecular Theranostics, Sechenov First Moscow State Medical University, Moscow 119991, Russia; Moscow State Pedagogical University, Moscow 119435, Russia.
| | - Olga E Chepikova
- Institute of Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, Moscow 119991, Russia; Research Center for Translational Medicine, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Tatiana N Pallaeva
- Federal Scientific Research Center "Crystallography and Photonics" of the Russian Academy of Sciences, Moscow 119333, Russia; Research Center for Translational Medicine, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Neonila V Gorokhovets
- Institute of Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Andrei E Siniavin
- Federal State Budget Institution "National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N F Gamaleya" of the Ministry of Health of the Russian Federation, Moscow 123098, Russia; Department of Molecular Neuroimmune Signalling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Vladimir A Gushchin
- Federal State Budget Institution "National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N F Gamaleya" of the Ministry of Health of the Russian Federation, Moscow 123098, Russia; Department of Virology, Biological Faculty, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Lyudmila V Savvateeva
- Institute of Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Ivan A Vinokurov
- Petrovsky National Research Center of Surgery, Moscow 119991, Russia
| | - Dmitry A Khochenkov
- N.N. Blokhin National Medical Research Center of Oncology, Moscow 115478, Russia; Togliatti State University, Togliatti 445020, Russia
| | - Andrey A Zamyatnin
- Research Center for Translational Medicine, Sirius University of Science and Technology, Sochi 354340, Russia; Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow 119234, Russia; Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Evgeny V Khaydukov
- Federal Scientific Research Center "Crystallography and Photonics" of the Russian Academy of Sciences, Moscow 119333, Russia; Moscow State Pedagogical University, Moscow 119435, Russia
| |
Collapse
|
6
|
Gheban-Roșca IA, Gheban BA, Pop B, Mironescu DC, Siserman VC, Jianu EM, Drugan T, Bolboacă SD. Immunohistochemical and Morphometric Analysis of Lung Tissue in Fatal COVID-19. Diagnostics (Basel) 2024; 14:914. [PMID: 38732328 PMCID: PMC11082993 DOI: 10.3390/diagnostics14090914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/15/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
The primary targets of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in the lungs are type I pneumocytes, macrophages, and endothelial cells. We aimed to identify lung cells targeted by SARS-CoV-2 using viral nucleocapsid protein staining and morphometric features on patients with fatal COVID-19. We conducted a retrospective analysis of fifty-one autopsy cases of individuals who tested positive for SARS-CoV-2. Demographic and clinical information were collected from forensic reports, and lung tissue was examined for microscopic lesions and the presence of specific cell types. Half of the evaluated cohort were older than 71 years, and the majority were male (74.5%). In total, 24 patients presented diffuse alveolar damage (DAD), and 50.9% had comorbidities (56.9% obesity, 33.3% hypertension, 15.7% diabetes mellitus). Immunohistochemical analysis showed a similar pattern of infected macrophages, infected type I pneumocytes, and endothelial cells, regardless of the presence of DAD (p > 0.5). The immunohistochemical reactivity score (IRS) was predominantly moderate but without significant differences between patients with and without DAD (p = 0.633 IRS for type I pneumocytes, p = 0.773 IRS for macrophage, and p = 0.737 for IRS endothelium). The nucleus/cytoplasm ratio shows lower values in patients with DAD (median: 0.29 vs. 0.35), but the difference only reaches a tendency for statistical significance (p = 0.083). Our study confirms the presence of infected macrophages, type I pneumocytes, and endothelial cells with a similar pattern in patients with and without diffuse alveolar damage.
Collapse
Affiliation(s)
- Ioana-Andreea Gheban-Roșca
- Department of Medical Informatics and Biostatistics, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania; (I.-A.G.-R.); (T.D.)
- Clinical Hospital for Infectious Diseases, 400348 Cluj-Napoca, Romania
| | - Bogdan-Alexandru Gheban
- County Emergency Clinical Hospital, 400006 Cluj-Napoca, Romania
- Department of Histology, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania;
| | - Bogdan Pop
- The Oncology Institute “Prof. Dr. Ion Chiricuță”, 400015 Cluj-Napoca, Romania;
- Department of Anatomic Pathology, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Daniela-Cristina Mironescu
- Forensic Institute, 400006 Cluj-Napoca, Romania; (D.-C.M.); (V.C.S.)
- Department of Forensic Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Vasile Costel Siserman
- Forensic Institute, 400006 Cluj-Napoca, Romania; (D.-C.M.); (V.C.S.)
- Department of Forensic Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Elena Mihaela Jianu
- Department of Histology, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania;
| | - Tudor Drugan
- Department of Medical Informatics and Biostatistics, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania; (I.-A.G.-R.); (T.D.)
| | - Sorana D. Bolboacă
- Department of Medical Informatics and Biostatistics, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania; (I.-A.G.-R.); (T.D.)
| |
Collapse
|
7
|
Tomassetti S, Ciani L, Luzzi V, Gori L, Trigiani M, Giuntoli L, Lavorini F, Poletti V, Ravaglia C, Torrego A, Maldonado F, Lentz R, Annunziato F, Maggi L, Rossolini GM, Pollini S, Para O, Ciurleo G, Casini A, Rasero L, Bartoloni A, Spinicci M, Munavvar M, Gasparini S, Comin C, Cerinic MM, Peired A, Henket M, Ernst B, Louis R, Corhay JL, Nardi C, Guiot J. Utility of bronchoalveolar lavage for COVID-19: a perspective from the Dragon consortium. Front Med (Lausanne) 2024; 11:1259570. [PMID: 38371516 PMCID: PMC10869531 DOI: 10.3389/fmed.2024.1259570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 01/09/2024] [Indexed: 02/20/2024] Open
Abstract
Diagnosing COVID-19 and treating its complications remains a challenge. This review reflects the perspective of some of the Dragon (IMI 2-call 21, #101005122) research consortium collaborators on the utility of bronchoalveolar lavage (BAL) in COVID-19. BAL has been proposed as a potentially useful diagnostic tool to increase COVID-19 diagnosis sensitivity. In both critically ill and non-critically ill COVID-19 patients, BAL has a relevant role in detecting other infections or supporting alternative diagnoses and can change management decisions in up to two-thirds of patients. BAL is used to guide steroid and immunosuppressive treatment and to narrow or discontinue antibiotic treatment, reducing the use of unnecessary broad antibiotics. Moreover, cellular analysis and novel multi-omics techniques on BAL are of critical importance for understanding the microenvironment and interaction between epithelial cells and immunity, revealing novel potential prognostic and therapeutic targets. The BAL technique has been described as safe for both patients and healthcare workers in more than a thousand procedures reported to date in the literature. Based on these preliminary studies, we recognize that BAL is a feasible procedure in COVID-19 known or suspected cases, useful to properly guide patient management, and has great potential for research.
Collapse
Affiliation(s)
- Sara Tomassetti
- Interventional Pulmonology Unit, Department of Experimental and Clinical Medicine, Careggi University Hospital, Florence, Italy
| | - Luca Ciani
- Interventional Pulmonology Unit, Department of Experimental and Clinical Medicine, Careggi University Hospital, Florence, Italy
| | - Valentina Luzzi
- Interventional Pulmonology Unit, Department of Experimental and Clinical Medicine, Careggi University Hospital, Florence, Italy
| | - Leonardo Gori
- Pulmonology Unit, Department of Experimental and Clinical Medicine, Careggi University Hospital, Florence, Italy
| | - Marco Trigiani
- Interventional Pulmonology Unit, Department of Experimental and Clinical Medicine, Careggi University Hospital, Florence, Italy
| | - Leonardo Giuntoli
- Interventional Pulmonology Unit, Department of Experimental and Clinical Medicine, Careggi University Hospital, Florence, Italy
| | - Federico Lavorini
- Pulmonology Unit, Department of Experimental and Clinical Medicine, Careggi University Hospital, Florence, Italy
| | - Venerino Poletti
- Department of Diseases of the Thorax, GB Morgagni Hospital, Forlì, Italy
| | - Claudia Ravaglia
- Department of Diseases of the Thorax, GB Morgagni Hospital, Forlì, Italy
| | - Alfons Torrego
- Respiratory Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Fabien Maldonado
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Thoracic Surgery, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Robert Lentz
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Thoracic Surgery, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Laura Maggi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Gian Maria Rossolini
- Department of Experimental Medicine, University of Florence, Florence, Italy
- Microbiology and Virology Unit, Florence Careggi University Hospital, Florence, Italy
| | - Simona Pollini
- Department of Experimental Medicine, University of Florence, Florence, Italy
- Microbiology and Virology Unit, Florence Careggi University Hospital, Florence, Italy
| | - Ombretta Para
- Internal Medicine Unit 1, AOU Careggi, Florence, Italy
| | - Greta Ciurleo
- Internal Medicine Unit 2, AOU Careggi, Florence, Italy
| | | | - Laura Rasero
- Department of Health Science, Clinical Innovations and Research Unit, Careggi University Hospital, Florence, Italy
| | - Alessandro Bartoloni
- Infectious and Tropical Diseases Unit, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Michele Spinicci
- Infectious and Tropical Diseases Unit, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Mohammed Munavvar
- School of Biological Sciences, The University of Manchester, Manchester, United Kingdom
- Department of Respiratory, Lancashire Teaching Hospital NHS Foundation Trust, Preston, United Kingdom
| | - Stefano Gasparini
- Interventional Pulmonology Unit, University Hospital Riuniti di Ancona, Ancona, Italy
| | - Camilla Comin
- Department of Experimental and Clinical Medicine Section of Surgery, Histopathology, and Molecular Pathology, University of Florence, Florence, Italy
| | - Marco Matucci Cerinic
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Anna Peired
- Department of Clinical and Experimental Biomedical Sciences, University of Florence, Florence, Italy
| | - Monique Henket
- Department of Respiratory Medicine, Universitary Hospital of Liège, Liège, Belgium
| | - Benoit Ernst
- Department of Respiratory Medicine, Universitary Hospital of Liège, Liège, Belgium
| | - Renaud Louis
- Department of Respiratory Medicine, Universitary Hospital of Liège, Liège, Belgium
| | - Jean-louis Corhay
- Department of Respiratory Medicine, Universitary Hospital of Liège, Liège, Belgium
| | - Cosimo Nardi
- Department of Experimental and Clinical Biomedical Sciences, Radiodiagnostic Unit n. 2, University of Florence, Florence, Italy
| | - Julien Guiot
- Department of Respiratory Medicine, Universitary Hospital of Liège, Liège, Belgium
| |
Collapse
|
8
|
McKendry R, Lemm NM, Papargyris L, Chiu C. Human Challenge Studies with Coronaviruses Old and New. Curr Top Microbiol Immunol 2024; 445:69-108. [PMID: 35181805 DOI: 10.1007/82_2021_247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Coronavirus infections have been known to cause disease in animals since as early as the 1920s. However, only seven coronaviruses capable of causing human disease have been identified thus far. These Human Coronaviruses (HCoVs) include the causes of the common cold, but more recent coronaviruses that have emerged (i.e. SARS-CoV, MERS-CoV and SARS-CoV-2) are associated with much greater morbidity and mortality. HCoVs have been relatively under-studied compared to other common respiratory infections, as historically they have presented with mild symptoms. This has led to a relatively limited understanding of their animal reservoirs, transmission and determinants of immune protection. To address this, human infection challenge studies with HCoVs have been performed that enable a detailed clinical and immunological analysis of the host response at specific time points under controlled conditions with standardised viral inocula. Until recently, all such human challenge studies were conducted with common cold HCoVs, with the study of SARS-CoV and MERS-CoV unacceptable due to their greater pathogenicity. However, with the emergence of SARS-CoV-2 and the COVID-19 pandemic during which severe outcomes in young healthy adults have been rare, human challenge studies with SARS-CoV-2 are now being developed. Two SARS-CoV-2 human challenge studies in the UK studying individuals with and without pre-existing immunity are underway. As well as providing a platform for testing of antivirals and vaccines, such studies will be critical for understanding the factors associated with susceptibility to SARS-CoV-2 infection and thus developing improved strategies to tackle the current as well as future HCoV pandemics. Here, we summarise the major questions about protection and pathogenesis in HCoV infection that human infection challenge studies have attempted to answer historically, as well as the knowledge gaps that aim to be addressed with contemporary models.
Collapse
Affiliation(s)
- Richard McKendry
- Department of Infectious Disease, Imperial College London, London, UK
| | - Nana-Marie Lemm
- Department of Infectious Disease, Imperial College London, London, UK
| | - Loukas Papargyris
- Department of Infectious Disease, Imperial College London, London, UK
| | - Christopher Chiu
- Department of Infectious Disease, Imperial College London, London, UK.
| |
Collapse
|
9
|
Notz Q, Hermann J, Muellenbach RM, Lotz C. [Pathophysiology of Acute Respiratory Distress Syndrome]. Anasthesiol Intensivmed Notfallmed Schmerzther 2024; 59:12-22. [PMID: 38190822 DOI: 10.1055/a-2043-8602] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
Acute respiratory distress syndrome (ARDS) is a common condition in intensive care medicine. Various intra- and extrapulmonal causes may trigger an epithelial and endothelial permeability increase, which leads to impaired gas exchange due to fluid overload of the alveoli and transmigration of leukocytes. This results in hypoxemia and hypercapnia, as well as deleterious consequences for the macro- and microcirculation with the risk of multi-organ failure and high mortality. This review summarizes ARDS pathophysiology and clinical consequences.
Collapse
|
10
|
Singh SJ, Baldwin MM, Daynes E, Evans RA, Greening NJ, Jenkins RG, Lone NI, McAuley H, Mehta P, Newman J, Novotny P, Smith DJF, Stanel S, Toshner M, Brightling CE. Respiratory sequelae of COVID-19: pulmonary and extrapulmonary origins, and approaches to clinical care and rehabilitation. THE LANCET. RESPIRATORY MEDICINE 2023; 11:709-725. [PMID: 37216955 PMCID: PMC10198676 DOI: 10.1016/s2213-2600(23)00159-5] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/17/2023] [Accepted: 04/17/2023] [Indexed: 05/24/2023]
Abstract
Although the exact prevalence of post-COVID-19 condition (also known as long COVID) is unknown, more than a third of patients with COVID-19 develop symptoms that persist for more than 3 months after SARS-CoV-2 infection. These sequelae are highly heterogeneous in nature and adversely affect multiple biological systems, although breathlessness is a frequently cited symptom. Specific pulmonary sequelae, including pulmonary fibrosis and thromboembolic disease, need careful assessment and might require particular investigations and treatments. COVID-19 outcomes in people with pre-existing respiratory conditions vary according to the nature and severity of the respiratory disease and how well it is controlled. Extrapulmonary complications such as reduced exercise tolerance and frailty might contribute to breathlessness in post-COVID-19 condition. Non-pharmacological therapeutic options, including adapted pulmonary rehabilitation programmes and physiotherapy techniques for breathing management, might help to attenuate breathlessness in people with post-COVID-19 condition. Further research is needed to understand the origins and course of respiratory symptoms and to develop effective therapeutic and rehabilitative strategies.
Collapse
Affiliation(s)
- Sally J Singh
- Institute for Lung Health, NIHR Leicester Biomedical Research Centre-Respiratory and Infectious Diseases, Leicester, UK.
| | - Molly M Baldwin
- Institute for Lung Health, NIHR Leicester Biomedical Research Centre-Respiratory and Infectious Diseases, Leicester, UK
| | - Enya Daynes
- Institute for Lung Health, NIHR Leicester Biomedical Research Centre-Respiratory and Infectious Diseases, Leicester, UK
| | - Rachael A Evans
- Institute for Lung Health, NIHR Leicester Biomedical Research Centre-Respiratory and Infectious Diseases, Leicester, UK
| | - Neil J Greening
- Institute for Lung Health, NIHR Leicester Biomedical Research Centre-Respiratory and Infectious Diseases, Leicester, UK
| | - R Gisli Jenkins
- Imperial College London National Heart and Lung Institute, London, UK
| | - Nazir I Lone
- Department of Anaesthesia, Critical Care and Pain Medicine, Usher Institute, The University of Edinburgh, Edinburgh, UK
| | - Hamish McAuley
- Institute for Lung Health, NIHR Leicester Biomedical Research Centre-Respiratory and Infectious Diseases, Leicester, UK
| | - Puja Mehta
- Centre for Inflammation and Tissue Repair, Division of Medicine, University College London, London, UK
| | - Joseph Newman
- Royal Papworth Hospital NHS Foundation Trust, Cambridge, UK
| | - Petr Novotny
- Institute for Lung Health, NIHR Leicester Biomedical Research Centre-Respiratory and Infectious Diseases, Leicester, UK
| | | | - Stefan Stanel
- Manchester University NHS Foundation Trust, Manchester, UK
| | - Mark Toshner
- NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Christopher E Brightling
- Institute for Lung Health, NIHR Leicester Biomedical Research Centre-Respiratory and Infectious Diseases, Leicester, UK
| |
Collapse
|
11
|
Haslbauer JD, Savic Prince S, Stalder AK, Matter MS, Zinner CP, Jahn K, Obermann E, Hanke J, Leuzinger K, Hirsch HH, Tzankov A. Differential Gene Expression of SARS-CoV-2 Positive Bronchoalveolar Lavages: A Case Series. Pathobiology 2023; 91:158-168. [PMID: 37490884 PMCID: PMC10997241 DOI: 10.1159/000532057] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/12/2023] [Indexed: 07/27/2023] Open
Abstract
BACKGROUND Transcriptomic data on bronchoalveolar lavage (BAL) from COVID-19 patients are currently scarce. OBJECTIVES This case series seeks to characterize the intra-alveolar immunopathology of COVID-19. METHOD BALs were performed on 14 patients (5 COVID-19, of which 3 mild and 2 largely asymptomatic, 9 controls). Controls included asthma (n = 1), unremarkable BALs (n = 3), infections with respiratory syncytial virus (n = 1), influenza B (n = 1), and infections with other coronaviruses (n = 3). SARS-CoV-2 RNA load was measured by quantitative nucleic acid testing, while the detection of other pathogens was performed by immunofluorescence or multiplex NAT. RESULTS Gene expression profiling showed 71 significantly downregulated and 5 upregulated transcripts in SARS-CoV-2-positive lavages versus controls. Downregulated transcripts included genes involved in macrophage development, polarization, and crosstalk (LGALS3, MARCO, ERG2, BTK, RAC1, CD83), and genes involved in chemokine signaling and immunometabolism (NUPR1, CEBPB, CEBPA, PECAM1, CCL18, PPARG, ALOX5, ALOX5AP). Upregulated transcripts featured genes involved in NK-T cell signaling (GZMA, GZMH, GNLY, PRF1, CD3G). Patients with mild COVID-19 showed a significant upregulation of genes involved in blood mononuclear cell/leukocyte function (G0S2, ANXA6, FCGR2B, ADORA3), coagulation (von Willebrand factor [VWF]), interferon response (IFRD1, IL12RB2), and a zinc metalloprotease elevated in asthma (CPA3) compared to asymptomatic cases. In-silico comparison of the 5 COVID-19 BAL cases to a published cohort of lethal COVID-19 showed a significant upregulation of "antigen processing and presentation" and "lysosome" pathways in lethal cases. CONCLUSIONS These data underscore the heterogeneity of immune response in COVID-19. Further studies with a larger dataset are required to gain a better understanding of the hallmarks of SARS-CoV-2 immunological response.
Collapse
Affiliation(s)
- Jasmin D Haslbauer
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland,
| | - Spasenija Savic Prince
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Anna K Stalder
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Matthias S Matter
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Carl P Zinner
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Kathleen Jahn
- Clinic of Respiratory Medicine and Pulmonary Cell Research, University Hospital Basel, Basel, Switzerland
| | - Ellen Obermann
- Institute of Pathology, Cantonal Hospital Lucerne, Lucerne, Switzerland
| | - Jasmin Hanke
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Karoline Leuzinger
- Transplantation and Clinical Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
- Clinical Virology, University Hospital Basel, Basel, Switzerland
| | - Hans H Hirsch
- Transplantation and Clinical Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
- Clinical Virology, University Hospital Basel, Basel, Switzerland
- Infectious Diseases and Hospital Epidemiology, Department of Acute Medicine, University Hospital Basel, Basel, Switzerland
| | - Alexandar Tzankov
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| |
Collapse
|
12
|
Santinelli L, Lazzaro A, Sciarra F, Maddaloni L, Frasca F, Fracella M, Moretti S, Borsetti A, Bugani G, Alessandri F, Zullino V, Ruberto F, Pugliese F, Sorrentino L, Gianfrilli D, Isidori A, Venneri MA, Mastroianni CM, Ceccarelli G, d’Ettorre G. Cellular Immune Profiling of Lung and Blood Compartments in Patients with SARS-CoV-2 Infection. Pathogens 2023; 12:pathogens12030442. [PMID: 36986364 PMCID: PMC10057444 DOI: 10.3390/pathogens12030442] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/07/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023] Open
Abstract
Background: SARS-CoV-2 related immunopathology may be the driving cause underlying severe COVID-19. Through an immunophenotyping analysis on paired bronchoalveolar lavage fluid (BALF) and blood samples collected from mechanically ventilated patients with COVID-19-associated Acute Respiratory Distress Syndrome (CARDS), this study aimed to evaluate the cellular immune responses in survivors and non-survivors of COVID-19. Methods: A total of 36 paired clinical samples of bronchoalveolar lavage fluid (BALF) mononuclear cells (BALF-MC) and peripheral blood mononuclear cells (PBMC) were collected from 18 SARS-CoV-2-infected subjects admitted to the intensive care unit (ICU) of the Policlinico Umberto I, Sapienza University Hospital in Rome (Italy) for severe interstitial pneumonia. The frequencies of monocytes (total, classical, intermediate and non-classical) and Natural Killer (NK) cell subsets (total, CD56bright and CD56dim), as well as CD4+ and CD8+ T cell subsets [naïve, central memory (TCM) and effector memory (TEM)], and those expressing CD38 and/or HLADR were evaluated by multiparametric flow cytometry. Results: Survivors with CARDS exhibited higher frequencies of classical monocytes in blood compared to non-survivors (p < 0.05), while no differences in the frequencies of the other monocytes, NK cell and T cell subsets were recorded between these two groups of patients (p > 0.05). The only exception was for peripheral naïve CD4+ T cells levels that were reduced in non-survivors (p = 0.04). An increase in the levels of CD56bright (p = 0.012) and a decrease in CD56dim (p = 0.002) NK cell frequencies was also observed in BALF-MC samples compared to PBMC in deceased COVID-19 patients. Total CD4+ and CD8+ T cell levels in the lung compartment were lower compared to blood (p = 0.002 and p < 0.01, respectively) among non-survivors. Moreover, CD38 and HLA-DR were differentially expressed by CD4+ and CD8+ T cell subsets in BALF-MC and in PBMC among SARS-CoV-2-infected patients who died from COVID-19 (p < 0.05). Conclusions: These results show that the immune cellular profile in blood and pulmonary compartments was similar in survivors and non-survivors of COVID-19. T lymphocyte levels were reduced, but resulted highly immune-activated in the lung compartment of patients who faced a fatal outcome.
Collapse
Affiliation(s)
- Letizia Santinelli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy
| | - Alessandro Lazzaro
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy
| | - Francesca Sciarra
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Luca Maddaloni
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy
| | - Federica Frasca
- Laboratory of Virology, Department of Molecular Medicine, Affiliated to Istituto Pasteur Italia, Sapienza University, 00185 Rome, Italy
| | - Matteo Fracella
- Laboratory of Virology, Department of Molecular Medicine, Affiliated to Istituto Pasteur Italia, Sapienza University, 00185 Rome, Italy
| | - Sonia Moretti
- National HIV/AIDS Research Center, Italian Institute of Health, 00161 Rome, Italy
- Correspondence: (S.M.); (G.C.); Tel.: +39-0649903591 (S.M.); +39-0649970311 (G.C.)
| | - Alessandra Borsetti
- National HIV/AIDS Research Center, Italian Institute of Health, 00161 Rome, Italy
| | - Ginevra Bugani
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy
| | - Francesco Alessandri
- Department of General and Specialistic Surgery, Sapienza University of Rome, 00185 Rome, Italy
| | - Veronica Zullino
- Department of General and Specialistic Surgery, Sapienza University of Rome, 00185 Rome, Italy
| | - Franco Ruberto
- Department of General and Specialistic Surgery, Sapienza University of Rome, 00185 Rome, Italy
| | - Francesco Pugliese
- Department of General and Specialistic Surgery, Sapienza University of Rome, 00185 Rome, Italy
| | - Leonardo Sorrentino
- Laboratory of Virology, Department of Molecular Medicine, Affiliated to Istituto Pasteur Italia, Sapienza University, 00185 Rome, Italy
| | - Daniele Gianfrilli
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Andrea Isidori
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Mary Anna Venneri
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Claudio M. Mastroianni
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy
| | - Giancarlo Ceccarelli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy
- Azienda Ospedaliero Universitaria Policlinico Umberto I, 00185 Rome, Italy
- Correspondence: (S.M.); (G.C.); Tel.: +39-0649903591 (S.M.); +39-0649970311 (G.C.)
| | - Gabriella d’Ettorre
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
13
|
Dillard JA, Martinez SA, Dearing JJ, Montgomery SA, Baxter AK. Animal Models for the Study of SARS-CoV-2-Induced Respiratory Disease and Pathology. Comp Med 2023; 73:72-90. [PMID: 36229170 PMCID: PMC9948904 DOI: 10.30802/aalas-cm-22-000089] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Emergence of the betacoronavirus SARS-CoV-2 has resulted in a historic pandemic, with millions of deaths worldwide. An unprecedented effort has been made by the medical, scientific, and public health communities to rapidly develop and implement vaccines and therapeutics to prevent and reduce hospitalizations and deaths. Although SARS-CoV-2 infection can lead to disease in many organ systems, the respiratory system is its main target, with pneumonia and acute respiratory distress syndrome as the hallmark features of severe disease. The large number of patients who have contracted COVID-19 infections since 2019 has permitted a detailed characterization of the clinical and pathologic features of the disease in humans. However, continued progress in the development of effective preventatives and therapies requires a deeper understanding of the pathogenesis of infection. Studies using animal models are necessary to complement in vitro findings and human clinical data. Multiple animal species have been evaluated as potential models for studying the respiratory disease caused by SARSCoV-2 infection. Knowing the similarities and differences between animal and human responses to infection is critical for effective translation of animal data into human medicine. This review provides a detailed summary of the respiratory disease and associated pathology induced by SARS-CoV-2 infection in humans and compares them with the disease that develops in 3 commonly used models: NHP, hamsters, and mice. The effective use of animals to study SARS-CoV-2-induced respiratory disease will enhance our understanding of SARS-CoV-2 pathogenesis, allow the development of novel preventatives and therapeutics, and aid in the preparation for the next emerging virus with pandemic potential.
Collapse
Key Words
- ace2, angiotensin-converting enzyme 2
- agm, african green monkey
- ali, acute lung injury
- ards, acute respiratory distress syndrome
- balf, bronchoalveolar lavage fluid
- cards, covid-19-associated acute respiratory distress syndrome
- dad, diffuse alveolar damage
- dpi, days postinfection
- ggo, ground glass opacities
- s, spike glycoprotein
Collapse
Affiliation(s)
- Jacob A Dillard
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Sabian A Martinez
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Justin J Dearing
- Biological and Biomedical Sciences Program, Office of Graduate Education, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Stephanie A Montgomery
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Andvictoria K Baxter
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina;,
| |
Collapse
|
14
|
Gao CA, Pickens CI, Morales-Nebreda L, Wunderink RG. Clinical Features of COVID-19 and Differentiation from Other Causes of CAP. Semin Respir Crit Care Med 2023; 44:8-20. [PMID: 36646082 DOI: 10.1055/s-0042-1759889] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Community-acquired pneumonia (CAP) is a significant cause of morbidity and mortality, one of the most common reasons for infection-related death worldwide. Causes of CAP include numerous viral, bacterial, and fungal pathogens, though frequently no specific organism is found. Beginning in 2019, the COVID-19 pandemic has caused incredible morbidity and mortality. COVID-19 has many features typical of CAP such as fever, respiratory distress, and cough, and can be difficult to distinguish from other types of CAP. Here, we highlight unique clinical features of COVID-19 pneumonia such as olfactory and gustatory dysfunction, lymphopenia, and distinct imaging appearance.
Collapse
Affiliation(s)
- Catherine A Gao
- Division of Pulmonary and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Chiagozie I Pickens
- Division of Pulmonary and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Luisa Morales-Nebreda
- Division of Pulmonary and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Richard G Wunderink
- Division of Pulmonary and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
15
|
Rocha-Hasler M, Müller L, Wagner A, Tu A, Stanek V, Campion NJ, Bartosik T, Zghaebi M, Stoshikj S, Gompelmann D, Zech A, Mei H, Kratochwill K, Spittler A, Idzko M, Schneider S, Eckl-Dorna J. Using mass cytometry for the analysis of samples of the human airways. Front Immunol 2022; 13:1004583. [PMID: 36578479 PMCID: PMC9791368 DOI: 10.3389/fimmu.2022.1004583] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 11/16/2022] [Indexed: 12/14/2022] Open
Abstract
Mass cytometry (MC) is a powerful method for mapping complex cellular systems at single-cell levels, based on the detection of cellular proteins. Numerous studies have been performed using human blood, but there is a lack of protocols describing the processing and labeling of bronchoalveolar lavage fluid (BALF) and nasal polyps (NP) for acquisition by MC. These specimens are essential in the investigation of immune cell characteristics in airway diseases such as asthma and chronic rhinosinusitis with NP (CRSwNP). Here we optimized a workflow for processing, labeling, and acquisition of BALF and NP cells by MC. Among three methods tested for NP digestion, combined enzymatic/mechanical processing yielded maximum cell recovery, viability and labeling patterns compared to the other methods. Treatment with DNAse improved sample acquisition by MC. In a final step, we performed a comparison of blood, BALF and NP cell composition using a 31-marker MC antibody panel, revealing expected differences between the different tissue but also heterogeneity among the BALF and NP samples. We here introduce an optimized workflow for the MC analysis of human NP and BALF, which enables comparative analysis of different samples in larger cohorts. A deeper understanding of immune cell characteristics in these samples may guide future researchers and clinicians to a better disease management.
Collapse
Affiliation(s)
- Marianne Rocha-Hasler
- Allergology and Sinusitis Research Lab, Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria
| | - Lena Müller
- Core Facility Flow Cytometry & Department of Surgery, Research Lab, Medical University of Vienna, Vienna, Austria
| | - Anja Wagner
- Core Facility Proteomics, Medical University of Vienna, Vienna, Austria,Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Aldine Tu
- Allergology and Sinusitis Research Lab, Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria
| | - Victoria Stanek
- Allergology and Sinusitis Research Lab, Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria
| | - Nicholas James Campion
- Allergology and Sinusitis Research Lab, Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria
| | - Tina Bartosik
- Allergology and Sinusitis Research Lab, Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria
| | - Mohammed Zghaebi
- Allergology and Sinusitis Research Lab, Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria
| | - Slagjana Stoshikj
- Division of Pulmonology, Department of Internal Medicine II, Medical University Vienna, Vienna, Austria
| | - Daniela Gompelmann
- Division of Pulmonology, Department of Internal Medicine II, Medical University Vienna, Vienna, Austria
| | - Andreas Zech
- Division of Pulmonology, Department of Internal Medicine II, Medical University Vienna, Vienna, Austria
| | - Henrik Mei
- German Rheumatism Research Center Berlin, Berlin, Germany
| | - Klaus Kratochwill
- Core Facility Proteomics, Medical University of Vienna, Vienna, Austria,Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Andreas Spittler
- Core Facility Flow Cytometry & Department of Surgery, Research Lab, Medical University of Vienna, Vienna, Austria
| | - Marco Idzko
- Division of Pulmonology, Department of Internal Medicine II, Medical University Vienna, Vienna, Austria
| | - Sven Schneider
- Allergology and Sinusitis Research Lab, Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria,*Correspondence: Sven Schneider,
| | - Julia Eckl-Dorna
- Allergology and Sinusitis Research Lab, Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
16
|
Singh G, Martin Rumende C, Sharma SK, Rengganis I, Amin Z, Loho T, Hermiyanti E, Harimurti K, Wibowo H. Low BALF CD4 T cells count is associated with extubation failure and mortality in critically ill covid-19 pneumonia. Ann Med 2022; 54:1894-1905. [PMID: 35786088 PMCID: PMC9258432 DOI: 10.1080/07853890.2022.2095012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Critically ill COVID-19 pneumonia is one of the main causes of extubation failure and mortality. Understanding clinical characteristics, laboratory profiles and bronchoalveolar lavage fluid (BALF) immunopathology may help improve outcomes in critically ill COVID-19 pneumonia. We aimed to describe clinical characteristics, laboratory profiles and BALF immunopathology based on lung severity in critically ill COVID-19 pneumonia patients. MATERIALS AND METHODS Forty critically ill severe pneumonia patients requiring invasive mechanical ventilation in Cipto Mangunkusumo General (National Tertiary Referral Hospital), Indonesia within November 2020-January 2021 were enrolled in this study. Early BALF collection was performed after patients' intubation. Clinical characteristics, laboratory profiles and BALF biomarkers (sTREM-1, alveolar macrophage amount and function, IL-6, IL-17, CD4 T-cells, Tregs, SP-A and Caspase-3) were observed and analysed. Outcomes were measured based on extubation failure (within 19 days) and 28-days mortality. Univariate and bivariate analyses were performed. RESULTS Early bronchoscopy was performed in an average of 4 h (SD = 0.82) after patients' intubation. Twenty-three and twenty-two patients had extubation failure (within 19 days) and 28-days mortality, respectively. In the baseline clinical characteristics of critically ill COVID-19 patients, we found no significant differences in the extubation and mortality status groups. In the laboratory profiles of critically ill COVID-19 patients, we found no significant differences in the extubation status groups. In critically ill COVID-19 pneumonia patients, there was a significant high D-dimer levels in survived group (p = .027), a significant low BALF CD4 T-cells count in the right lung (p = .001) and a significant low BALF CD4 T-cells count (p = .010 and p = .018) in severely affected lung with extubation failure and mortality. CONCLUSIONS BALF CD4 T-cells count evaluation of severely affected lung is associated with early extubation failure and mortality in critically ill COVID-19 pneumonia patients. KEY MESSAGEFew studies have been conducted during the peak COVID-19 period analysing combined bronchoalveolar lavage fluid (BALF) immunopathology biomarkers within four hours of intubation to assess extubation failure and mortality. In this study, we reported eight BALF immunopathology biomarkers (sTREM-1, alveolar macrophage, IL-6, IL-17, CD4 T-cells, Tregs, SP-A and Caspase-3).We found significantly low BALF CD4 T-cells count in the right lung, and low BALF CD4 T-cells count in severely affected lung of critically ill COVID-19 pneumonia patients in extubation failure and mortality.
Collapse
Affiliation(s)
- Gurmeet Singh
- Department of Internal Medicine, Faculty of Medicine, Division of Respirology and Critical Illness, Universitas Indonesia - Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Cleopas Martin Rumende
- Department of Internal Medicine, Faculty of Medicine, Division of Respirology and Critical Illness, Universitas Indonesia - Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Surendra K Sharma
- Department of Molecular Medicine, Jamia Hamdard Institute of Molecular Medicine, Hamdard University, New Delhi, India.,Department of General Medicine & Pulmonary Medicine, JNMC, Datta Meghe Institute of Medical Science, New Delhi, India
| | - Iris Rengganis
- Department of Internal Medicine, Faculty of Medicine, Division of Allergy and Clinical Immunology, Universitas Indonesia, Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Zulkifli Amin
- Department of Internal Medicine, Faculty of Medicine, Division of Respirology and Critical Illness, Universitas Indonesia - Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Tonny Loho
- Department of Clinical Pathology, Faculty of Medicine, Universitas Indonesia - Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Emmy Hermiyanti
- Department of Internal Medicine, Faculty of Medicine, Division of Respirology and Critical Illness, Universitas Padjadjaran, Dr Hasan Sadikin Hospital Bandung, Bandung, Indonesia
| | - Kuntjoro Harimurti
- Department of Internal Medicine, Faculty of Medicine, Division of Geriatrics, Universitas Indonesia - Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Heri Wibowo
- Head of Integrated Laboratory, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| |
Collapse
|
17
|
Oliveira CBSD, Andrade JMDA, Oliveira JIN. Are atypical lymphocytes a new predictive factor in the development of COVID-19? Rev Soc Bras Med Trop 2022; 55:e0154. [PMID: 36134860 PMCID: PMC9549947 DOI: 10.1590/0037-8682-0154-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 07/06/2022] [Indexed: 01/08/2023] Open
Affiliation(s)
| | | | - Jonas Ivan Nobre Oliveira
- Universidade Federal do Rio Grande do Norte, Departamento de Biofísica e Farmacologia, Natal, RN, Brasil
| |
Collapse
|
18
|
Hastak PS, Andersen CR, Kelleher AD, Sasson SC. Frontline workers: Mediators of mucosal immunity in community acquired pneumonia and COVID-19. Front Immunol 2022; 13:983550. [PMID: 36211412 PMCID: PMC9539803 DOI: 10.3389/fimmu.2022.983550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/08/2022] [Indexed: 11/13/2022] Open
Abstract
The current COVID-19 pandemic has highlighted a need to further understand lung mucosal immunity to reduce the burden of community acquired pneumonia, including that caused by the SARS-CoV-2 virus. Local mucosal immunity provides the first line of defence against respiratory pathogens, however very little is known about the mechanisms involved, with a majority of literature on respiratory infections based on the examination of peripheral blood. The mortality for severe community acquired pneumonia has been rising annually, even prior to the current pandemic, highlighting a significant need to increase knowledge, understanding and research in this field. In this review we profile key mediators of lung mucosal immunity, the dysfunction that occurs in the diseased lung microenvironment including the imbalance of inflammatory mediators and dysbiosis of the local microbiome. A greater understanding of lung tissue-based immunity may lead to improved diagnostic and prognostic procedures and novel treatment strategies aimed at reducing the disease burden of community acquired pneumonia, avoiding the systemic manifestations of infection and excess morbidity and mortality.
Collapse
Affiliation(s)
- Priyanka S. Hastak
- The Kirby Institute, Immunovirology and Pathogenesis Program, University of New South Wales, Sydney, NSW, Australia
| | - Christopher R. Andersen
- The Kirby Institute, Immunovirology and Pathogenesis Program, University of New South Wales, Sydney, NSW, Australia
- Intensive Care Unit, Royal North Shore Hospital, Sydney, NSW, Australia
- Critical Care and Trauma Division, The George Institute for Global Health, Sydney, NSW, Australia
| | - Anthony D. Kelleher
- The Kirby Institute, Immunovirology and Pathogenesis Program, University of New South Wales, Sydney, NSW, Australia
| | - Sarah C. Sasson
- The Kirby Institute, Immunovirology and Pathogenesis Program, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
19
|
Erjefält JS, de Souza Xavier Costa N, Jönsson J, Cozzolino O, Dantas KC, Clausson CM, Siddhuraj P, Lindö C, Alyamani M, Lombardi SCFS, Mendroni Júnior A, Antonangelo L, Faria CS, Duarte-Neto AN, de Almeida Monteiro RA, Rebello Pinho JR, Gomes-Gouvêa MS, Verciano Pereira R, Monteiro JS, Setubal JC, de Oliveira EP, Theodoro Filho J, Sanden C, Orengo JM, Sleeman MA, da Silva LFF, Saldiva PHN, Dolhnikoff M, Mauad T. Diffuse alveolar damage patterns reflect the immunological and molecular heterogeneity in fatal COVID-19. EBioMedicine 2022; 83:104229. [PMID: 36027872 PMCID: PMC9398470 DOI: 10.1016/j.ebiom.2022.104229] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 07/20/2022] [Accepted: 08/01/2022] [Indexed: 11/25/2022] Open
Abstract
Background Severe COVID-19 lung disease exhibits a high degree of spatial and temporal heterogeneity, with different histological features coexisting within a single individual. It is important to capture the disease complexity to support patient management and treatment strategies. We provide spatially decoded analyses on the immunopathology of diffuse alveolar damage (DAD) patterns and factors that modulate immune and structural changes in fatal COVID-19. Methods We spatially quantified the immune and structural cells in exudative, intermediate, and advanced DAD through multiplex immunohistochemistry in autopsy lung tissue of 18 COVID-19 patients. Cytokine profiling, viral, bacteria, and fungi detection, and transcriptome analyses were performed. Findings Spatial DAD progression was associated with expansion of immune cells, macrophages, CD8+ T cells, fibroblasts, and (lymph)angiogenesis. Viral load correlated positively with exudative DAD and negatively with disease/hospital length. In all cases, enteric bacteria were isolated, and Candida parapsilosis in eight cases. Cytokines correlated mainly with macrophages and CD8+T cells. Pro-coagulation and acute repair were enriched pathways in exudative DAD whereas intermediate/advanced DAD had a molecular profile of elevated humoral and innate immune responses and extracellular matrix production. Interpretation Unraveling the spatial and molecular immunopathology of COVID-19 cases exposes the responses to SARS-CoV-2-induced exudative DAD and subsequent immune-modulatory and remodeling changes in proliferative/advanced DAD that occur side-by-side together with secondary infections in the lungs. These complex features have important implications for disease management and the development of novel treatments. Funding CNPq, Bill and Melinda Gates Foundation, HC-Convida, FAPESP, Regeneron Pharmaceuticals, and the Swedish Heart & Lung Foundation.
Collapse
Affiliation(s)
- Jonas S Erjefält
- Unit of Airway inflammation, Department of Experimental Medicine Sciences, Lund University, Sweden; Department of Allergology and Respiratory Medicine, Lund University, Sweden
| | - Natália de Souza Xavier Costa
- Departamento de Patologia, LIM-05 Laboratório de Patologia Ambiental, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | | | - Olga Cozzolino
- Unit of Airway inflammation, Department of Experimental Medicine Sciences, Lund University, Sweden
| | - Katia Cristina Dantas
- Departamento de Patologia, LIM-05 Laboratório de Patologia Ambiental, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Carl-Magnus Clausson
- Unit of Airway inflammation, Department of Experimental Medicine Sciences, Lund University, Sweden
| | - Premkumar Siddhuraj
- Unit of Airway inflammation, Department of Experimental Medicine Sciences, Lund University, Sweden
| | | | - Manar Alyamani
- Unit of Airway inflammation, Department of Experimental Medicine Sciences, Lund University, Sweden
| | - Suzete Cleusa Ferreira Spina Lombardi
- Divisão de Pesquisa & Medicina Transfusional, Fundação Pró-Sangue Hemocentro de São Paulo, São Paulo, Brazil; Laboratório Investigação Médica em Patogênese e Terapia dirigida em Onco-Imuno-Hematologia (LIM-31), Departamento de Hematologia, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Alfredo Mendroni Júnior
- Divisão de Pesquisa & Medicina Transfusional, Fundação Pró-Sangue Hemocentro de São Paulo, São Paulo, Brazil; Laboratório Investigação Médica em Patogênese e Terapia dirigida em Onco-Imuno-Hematologia (LIM-31), Departamento de Hematologia, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Leila Antonangelo
- Laboratório de Investigação Médica (LIM03), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil; Divisão de Patologia Clínica - Departamento de Patologia, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Caroline Silvério Faria
- Laboratório de Investigação Médica (LIM03), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Amaro Nunes Duarte-Neto
- Departamento de Patologia, LIM-05 Laboratório de Patologia Ambiental, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | | | - João Renato Rebello Pinho
- Laboratório de Investigação Médica (LIM03), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil; Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Michele Soares Gomes-Gouvêa
- Departamento de Gastroenterologia (LIM-07), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Roberta Verciano Pereira
- Laboratório de Investigação Médica (LIM03), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | | | - João Carlos Setubal
- Departamento de Bioquímica, Instituto de Química Universidade de São Paulo, São Paulo, Brazil
| | - Ellen Pierre de Oliveira
- Departamento de Cardiopneumologia, Instituto do Coração, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Jair Theodoro Filho
- Departamento de Patologia, LIM-05 Laboratório de Patologia Ambiental, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | | | | | | | - Luiz Fernando Ferraz da Silva
- Departamento de Patologia, LIM-05 Laboratório de Patologia Ambiental, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil; Serviço de Verificação de Óbitos da Capital, Universidade de São Paulo, São Paulo, Brazil
| | - Paulo Hilário Nascimento Saldiva
- Departamento de Patologia, LIM-05 Laboratório de Patologia Ambiental, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Marisa Dolhnikoff
- Departamento de Patologia, LIM-05 Laboratório de Patologia Ambiental, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Thais Mauad
- Departamento de Patologia, LIM-05 Laboratório de Patologia Ambiental, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.
| |
Collapse
|
20
|
Mullender C, da Costa KAS, Alrubayyi A, Pett SL, Peppa D. SARS-CoV-2 immunity and vaccine strategies in people with HIV. OXFORD OPEN IMMUNOLOGY 2022; 3:iqac005. [PMID: 36846557 PMCID: PMC9452103 DOI: 10.1093/oxfimm/iqac005] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/24/2022] [Accepted: 08/02/2022] [Indexed: 12/15/2022] Open
Abstract
Current severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) vaccines, based on the ancestral Wuhan strain, were developed rapidly to meet the needs of a devastating global pandemic. People living with Human Immunodeficiency Virus (PLWH) have been designated as a priority group for SARS-CoV-2 vaccination in most regions and varying primary courses (two- or three-dose schedule) and additional boosters are recommended depending on current CD4+ T cell count and/or detectable HIV viraemia. From the current published data, licensed vaccines are safe for PLWH, and stimulate robust responses to vaccination in those well controlled on antiretroviral therapy and with high CD4+ T cell counts. Data on vaccine efficacy and immunogenicity remain, however, scarce in PLWH, especially in people with advanced disease. A greater concern is a potentially diminished immune response to the primary course and subsequent boosters, as well as an attenuated magnitude and durability of protective immune responses. A detailed understanding of the breadth and durability of humoral and T cell responses to vaccination, and the boosting effects of natural immunity to SARS-CoV-2, in more diverse populations of PLWH with a spectrum of HIV-related immunosuppression is therefore critical. This article summarizes focused studies of humoral and cellular responses to SARS-CoV-2 infection in PLWH and provides a comprehensive review of the emerging literature on SARS-CoV-2 vaccine responses. Emphasis is placed on the potential effect of HIV-related factors and presence of co-morbidities modulating responses to SARS-CoV-2 vaccination, and the remaining challenges informing the optimal vaccination strategy to elicit enduring responses against existing and emerging variants in PLWH.
Collapse
Affiliation(s)
- Claire Mullender
- Centre for Clinical Research in Infection and Sexual Health, Institute for Global Health, University College London Institute for Global Health, London, UK
| | - Kelly A S da Costa
- Division of Infection and Immunity, University College London, London, UK
| | - Aljawharah Alrubayyi
- Division of Infection and Immunity, University College London, London, UK
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Sarah L Pett
- Centre for Clinical Research in Infection and Sexual Health, Institute for Global Health, University College London Institute for Global Health, London, UK
- Medical Research Council Clinical Trials Unit, Institute of Clinical Trials and Methodology, London, UK
| | - Dimitra Peppa
- Division of Infection and Immunity, University College London, London, UK
| |
Collapse
|
21
|
Tariq H. Marked polytypic proliferation of immunoblasts, plasmacytoid lymphocytes, and plasma cells in bronchoalveolar lavage fluid in SARS-CoV-2 infection. Int J Lab Hematol 2022; 44:974-975. [PMID: 35338579 PMCID: PMC9111654 DOI: 10.1111/ijlh.13833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/18/2022] [Accepted: 03/10/2022] [Indexed: 11/30/2022]
Affiliation(s)
- Hamza Tariq
- Rush University Medical Center, Chicago, Illinois, USA
| |
Collapse
|
22
|
A systematic review and metanalysis of diagnostic yield of BAL for detection of SARS-CoV-2. Heart Lung 2021; 52:95-105. [PMID: 34929538 PMCID: PMC8666306 DOI: 10.1016/j.hrtlng.2021.11.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 11/24/2021] [Accepted: 11/30/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND The gold standard for diagnosing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is microbiological confirmation by reverse transcriptase-polymerase chain reaction (RT-PCR)1 most commonly done using oropharyngeal (OP) and nasopharyngeal swabs (NP). But in suspected cases, where these samples are false-negative, bronchoalveolar lavage (BAL) may prove diagnostic. OBJECTIVES Hence, the diagnostic yield of BAL for detection of SARS-CoV-2 in cases of non-diagnostic upper respiratory tract samples is reviewed. METHODS Databases such as MEDLINE, Scopus, and Google Scholar were searched using a systematic search strategy. The current study has been in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines and has been registered with the International Prospective Registry of Systematic Reviews (CRD42020224088). RESULTS 911 records were identified at initial database extraction, of which 317 duplicates were removed and, 596 records were screened for inclusion eligibility. We included total 19 studies in the systematic review, and 17 were included in metanalysis. The pooled estimate of SARS-CoV-2 positivity in BAL was 11% (95%CI: 0.01-0.24). A sensitivity analysis also showed that the results appear to be robust and minimal risk of bias amongst the studies. CONCLUSION The current study demonstrates that BAL can be used to diagnose additional cases primary disease and superadded infections in patients with severe COVID-19 lower respiratory tract infection.
Collapse
|
23
|
Sugihara J, Shibata S, Doi M, Shimmura T, Inoue S, Matsumoto O, Suzuki H, Makino A, Miyazaki Y. Atypical lymphocytes in the peripheral blood of COVID-19 patients: A prognostic factor for the clinical course of COVID-19. PLoS One 2021; 16:e0259910. [PMID: 34767614 PMCID: PMC8589156 DOI: 10.1371/journal.pone.0259910] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 10/28/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Clinical observations have shown that there is a relationship between coronavirus disease 2019 (COVID-19) and atypical lymphocytes in the peripheral blood; however, knowledge about the time course of the changes in atypical lymphocytes and the association with the clinical course of COVID-19 is limited. OBJECTIVE Our purposes were to investigate the dynamics of atypical lymphocytes in COVID-19 patients and to estimate their clinical significance for diagnosis and monitoring disease course. MATERIALS AND METHODS We retrospectively identified 98 inpatients in a general ward at Kashiwa Municipal Hospital from May 1st, 2020, to October 31st, 2020. We extracted data on patient demographics, symptoms, comorbidities, blood test results, radiographic findings, treatment after admission and clinical course. We compared clinical findings between patients with and without atypical lymphocytes, investigated the behavior of atypical lymphocytes throughout the clinical course of COVID-19, and determined the relationships among the development of pneumonia, the use of supplemental oxygen and the presence of atypical lymphocytes. RESULTS Patients with atypical lymphocytes had a significantly higher prevalence of pneumonia (80.4% vs. 42.6%, p < 0.0001) and the use of supplemental oxygen (25.5% vs. 4.3%, p = 0.0042). The median time to the appearance of atypical lymphocytes after disease onset was eight days, and atypical lymphocytes were observed in 16/98 (16.3%) patients at the first visit. Atypical lymphocytes appeared after the confirmation of lung infiltrates in 31/41 (75.6%) patients. Of the 13 oxygen-treated patients with atypical lymphocytes, approximately two-thirds had a stable or improved clinical course after the appearance of atypical lymphocytes. CONCLUSION Atypical lymphocytes frequently appeared in the peripheral blood of COVID-19 patients one week after disease onset. Patients with atypical lymphocytes were more likely to have pneumonia and to need supplemental oxygen; however, two-thirds of them showed clinical improvement after the appearance of atypical lymphocytes.
Collapse
Affiliation(s)
- Jun Sugihara
- Department of Respiratory Medicine, Kashiwa Municipal Hospital, Kashiwa, Chiba, Japan
- Department of Respiratory Medicine, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
- * E-mail:
| | - Sho Shibata
- Department of Respiratory Medicine, Kashiwa Municipal Hospital, Kashiwa, Chiba, Japan
- Department of Respiratory Medicine, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Masafumi Doi
- Department of Respiratory Medicine, Kashiwa Municipal Hospital, Kashiwa, Chiba, Japan
| | - Takuya Shimmura
- Department of Respiratory Medicine, Kashiwa Municipal Hospital, Kashiwa, Chiba, Japan
| | - Shinichiro Inoue
- Department of Respiratory Medicine, Kashiwa Municipal Hospital, Kashiwa, Chiba, Japan
| | - Osamu Matsumoto
- Clinical Laboratory, Kashiwa Municipal Hospital, Kashiwa, Chiba, Japan
| | - Hiroyuki Suzuki
- Clinical Laboratory, Kashiwa Municipal Hospital, Kashiwa, Chiba, Japan
| | - Ayaka Makino
- Clinical Laboratory, Kashiwa Municipal Hospital, Kashiwa, Chiba, Japan
| | - Yasunari Miyazaki
- Department of Respiratory Medicine, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
24
|
Pickens CO, Gao CA, Cuttica MJ, Smith SB, Pesce LL, Grant RA, Kang M, Morales-Nebreda L, Bavishi AA, Arnold JM, Pawlowski A, Qi C, Budinger GRS, Singer BD, Wunderink RG. Bacterial Superinfection Pneumonia in Patients Mechanically Ventilated for COVID-19 Pneumonia. Am J Respir Crit Care Med 2021; 204:921-932. [PMID: 34409924 PMCID: PMC8534629 DOI: 10.1164/rccm.202106-1354oc] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Rationale: Current guidelines recommend patients with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pneumonia receive empirical antibiotics for suspected bacterial superinfection on the basis of weak evidence. Rates of ventilator-associated pneumonia (VAP) in clinical trials of patients with SARS-CoV-2 pneumonia are unexpectedly low. Objectives: We conducted an observational single-center study to determine the prevalence and etiology of bacterial superinfection at the time of initial intubation and the incidence and etiology of subsequent bacterial VAP in patients with severe SARS-CoV-2 pneumonia. Methods: Bronchoscopic BAL fluid samples from all patients with SARS-CoV-2 pneumonia requiring mechanical ventilation were analyzed using quantitative cultures and a multiplex PCR panel. Actual antibiotic use was compared with guideline-recommended therapy. Measurements and Main Results: We analyzed 386 BAL samples from 179 patients with SARS-CoV-2 pneumonia requiring mechanical ventilation. Bacterial superinfection within 48 hours of intubation was detected in 21% of patients. Seventy-two patients (44.4%) developed at least one VAP episode (VAP incidence rate = 45.2/1,000 ventilator days); 15 (20.8%) initial VAPs were caused by difficult-to-treat pathogens. The clinical criteria did not distinguish between patients with or without bacterial superinfection. BAL-based management was associated with significantly reduced antibiotic use compared with guideline recommendations. Conclusions: In patients with SARS-CoV-2 pneumonia requiring mechanical ventilation, bacterial superinfection at the time of intubation occurs in <25% of patients. Guideline-based empirical antibiotic management at the time of intubation results in antibiotic overuse. Bacterial VAP developed in 44% of patients and could not be accurately identified in the absence of microbiologic analysis of BAL fluid.
Collapse
Affiliation(s)
| | - Catherine A Gao
- Division of Pulmonary and Critical Care, Department of Medicine
| | | | - Sean B Smith
- Division of Pulmonary and Critical Care, Department of Medicine
| | | | - Rogan A Grant
- Division of Pulmonary and Critical Care, Department of Medicine
| | - Mengjia Kang
- Division of Pulmonary and Critical Care, Department of Medicine
| | | | - Avni A Bavishi
- Division of Pulmonary and Critical Care, Department of Medicine
| | - Jason M Arnold
- Division of Pulmonary and Critical Care, Department of Medicine
| | | | - Chao Qi
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | | | | | | |
Collapse
|