201
|
Bryce C, Grimes Z, Pujadas E, Ahuja S, Beasley MB, Albrecht R, Hernandez T, Stock A, Zhao Z, AlRasheed MR, Chen J, Li L, Wang D, Corben A, Haines GK, Westra WH, Umphlett M, Gordon RE, Reidy J, Petersen B, Salem F, Fiel MI, El Jamal SM, Tsankova NM, Houldsworth J, Mussa Z, Veremis B, Sordillo E, Gitman MR, Nowak M, Brody R, Harpaz N, Merad M, Gnjatic S, Liu WC, Schotsaert M, Miorin L, Aydillo Gomez TA, Ramos-Lopez I, Garcia-Sastre A, Donnelly R, Seigler P, Keys C, Cameron J, Moultrie I, Washington KL, Treatman J, Sebra R, Jhang J, Firpo A, Lednicky J, Paniz-Mondolfi A, Cordon-Cardo C, Fowkes ME. Pathophysiology of SARS-CoV-2: the Mount Sinai COVID-19 autopsy experience. Mod Pathol 2021; 34:1456-1467. [PMID: 33795830 PMCID: PMC8015313 DOI: 10.1038/s41379-021-00793-y] [Citation(s) in RCA: 172] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 02/07/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its associated clinical syndrome COVID-19 are causing overwhelming morbidity and mortality around the globe and disproportionately affected New York City between March and May 2020. Here, we report on the first 100 COVID-19-positive autopsies performed at the Mount Sinai Hospital in New York City. Autopsies revealed large pulmonary emboli in six cases. Diffuse alveolar damage was present in over 90% of cases. We also report microthrombi in multiple organ systems including the brain, as well as hemophagocytosis. We additionally provide electron microscopic evidence of the presence of the virus in our samples. Laboratory results of our COVID-19 cohort disclose elevated inflammatory markers, abnormal coagulation values, and elevated cytokines IL-6, IL-8, and TNFα. Our autopsy series of COVID-19-positive patients reveals that this disease, often conceptualized as a primarily respiratory viral illness, has widespread effects in the body including hypercoagulability, a hyperinflammatory state, and endothelial dysfunction. Targeting of these multisystemic pathways could lead to new treatment avenues as well as combination therapies against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Clare Bryce
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zachary Grimes
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Sadhna Ahuja
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Randy Albrecht
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Aryeh Stock
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zhen Zhao
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Joyce Chen
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Li Li
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Diane Wang
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adriana Corben
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | | | | | - Jason Reidy
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bruce Petersen
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fadi Salem
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | | | | | - Zarmeen Mussa
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | | | - Michael Nowak
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rachel Brody
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Noam Harpaz
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sacha Gnjatic
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Wen-Chun Liu
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Lisa Miorin
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | | | - Ryan Donnelly
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Calvin Keys
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | | | | | - Robert Sebra
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jeffrey Jhang
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adolfo Firpo
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | | | - Mary E Fowkes
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
202
|
Tardiolo G, Brianti P, Sapienza D, dell’Utri P, Dio VD, Rao G, Calabrò RS. Are We Paving the Way to Dig Out of the "Pandemic Hole"? A Narrative Review on SARS-CoV-2 Vaccination: From Animal Models to Human Immunization. Med Sci (Basel) 2021; 9:53. [PMID: 34449681 PMCID: PMC8395838 DOI: 10.3390/medsci9030053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 01/08/2023] Open
Abstract
The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is a new pathogen agent causing the coronavirus infectious disease (COVID-19). This novel virus originated the most challenging pandemic in this century, causing economic and social upheaval internationally. The extreme infectiousness and high mortality rates incentivized the development of vaccines to control this pandemic to prevent further morbidity and mortality. This international scenario led academic scientists, industries, and governments to work and collaborate strongly to make a portfolio of vaccines available at an unprecedented pace. Indeed, the robust collaboration between public systems and private companies led to resolutive actions for accelerating therapeutic interventions and vaccines mechanism. These strategies contributed to rapidly identifying safe and effective vaccines as quickly and efficiently as possible. Preclinical research employed animal models to develop vaccines that induce protective and long-lived immune responses. A spectrum of vaccines is worldwide under investigation in various preclinical and clinical studies to develop both individual protection and safe development of population-level herd immunity. Companies employed and developed different technological approaches for vaccines production, including inactivated vaccines, live-attenuated, non-replicating viral vector vaccines, as well as acid nucleic-based vaccines. In this view, the present narrative review provides an overview of current vaccination strategies, taking into account both preclinical studies and clinical trials in humans. Furthermore, to better understand immunization, animal models on SARS-CoV-2 pathogenesis are also briefly discussed.
Collapse
Affiliation(s)
- Giuseppe Tardiolo
- Department of Veterinary Sciences, University of Messina, Via Palatucci snc, 98168 Messina, Italy;
| | - Pina Brianti
- Unit of Dermatology, IRCCS San Raffaele Hospital, Via Olgettina 60, 20132 Milan, Italy;
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Piazza Pugliatti 1, 98122 Messina, Italy;
| | - Daniela Sapienza
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Piazza Pugliatti 1, 98122 Messina, Italy;
| | - Pia dell’Utri
- IRCCS Centro Neurolesi “Bonino Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (P.d.); (V.D.D.); (G.R.)
| | - Viviane Di Dio
- IRCCS Centro Neurolesi “Bonino Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (P.d.); (V.D.D.); (G.R.)
| | - Giuseppe Rao
- IRCCS Centro Neurolesi “Bonino Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (P.d.); (V.D.D.); (G.R.)
| | - Rocco Salvatore Calabrò
- IRCCS Centro Neurolesi “Bonino Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (P.d.); (V.D.D.); (G.R.)
| |
Collapse
|
203
|
Ye J, Hua M, Zhu F. Machine Learning Algorithms are Superior to Conventional Regression Models in Predicting Risk Stratification of COVID-19 Patients. Risk Manag Healthc Policy 2021; 14:3159-3166. [PMID: 34349576 PMCID: PMC8328384 DOI: 10.2147/rmhp.s318265] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/22/2021] [Indexed: 01/13/2023] Open
Abstract
Background It is very important to determine the risk of patients developing severe or critical COVID-19, but most of the existing risk prediction models are established using conventional regression models. We aim to use machine learning algorithms to develop predictive models and compare predictive performance with logistic regression models. Methods The medical record of 161 COVID-19 patients who were diagnosed January-April 2020 were retrospectively analyzed. The patients were divided into two groups: asymptomatic-moderate group (132 cases) and severe or above group (29 cases). The clinical features and laboratory biomarkers of these two groups were compared. Machine learning algorithms and multivariate logistic regression analysis were used to construct two COVID-19 risk stratification prediction models, and the area under the curve (AUC) was used to compare the predictive efficacy of these two models. Results A machine learning model was constructed based on seven characteristic variables: high sensitivity C-reactive protein (hs-CRP), procalcitonin (PCT), age, neutrophil count (Neuc), hemoglobin (HGB), percentage of neutrophils (Neur), and platelet distribution width (PDW). The AUC of the model was 0.978 (95% CI: 0.960-0.996), which was significantly higher than that of the logistic regression model (0.827; 95% CI: 0.724-0.930) (P=0.002). Moreover, the machine learning model's sensitivity, specificity, and accuracy were better than those of the logistic regression model. Conclusion Machine learning algorithms improve the accuracy of risk stratification in patients with COVID-19. Using detection algorithms derived from these techniques can enhance the identification of critically ill patients.
Collapse
Affiliation(s)
- Jiru Ye
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Meng Hua
- Department of Respiratory and Critical Care Medicine, Wuxi Fifth People's Hospital, Wuxi, 214000, China
| | - Feng Zhu
- Department of Respiratory and Critical Care Medicine, Wuxi Fifth People's Hospital, Wuxi, 214000, China
| |
Collapse
|
204
|
A Fragile Balance: Does Neutrophil Extracellular Trap Formation Drive Pulmonary Disease Progression? Cells 2021; 10:cells10081932. [PMID: 34440701 PMCID: PMC8394734 DOI: 10.3390/cells10081932] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/22/2021] [Accepted: 07/28/2021] [Indexed: 12/13/2022] Open
Abstract
Neutrophils act as the first line of defense during infection and inflammation. Once activated, they are able to fulfil numerous tasks to fight inflammatory insults while keeping a balanced immune response. Besides well-known functions, such as phagocytosis and degranulation, neutrophils are also able to release "neutrophil extracellular traps" (NETs). In response to most stimuli, the neutrophils release decondensed chromatin in a NADPH oxidase-dependent manner decorated with histones and granule proteins, such as neutrophil elastase, myeloperoxidase, and cathelicidins. Although primarily supposed to prevent microbial dissemination and fight infections, there is increasing evidence that an overwhelming NET response correlates with poor outcome in many diseases. Lung-related diseases especially, such as bacterial pneumonia, cystic fibrosis, chronic obstructive pulmonary disease, aspergillosis, influenza, and COVID-19, are often affected by massive NET formation. Highly vascularized areas as in the lung are susceptible to immunothrombotic events promoted by chromatin fibers. Keeping this fragile equilibrium seems to be the key for an appropriate immune response. Therapies targeting dysregulated NET formation might positively influence many disease progressions. This review highlights recent findings on the pathophysiological influence of NET formation in different bacterial, viral, and non-infectious lung diseases and summarizes medical treatment strategies.
Collapse
|
205
|
Yuan S, Jiang SC, Zhang ZW, Fu YF, Hu J, Li ZL. The Role of Alveolar Edema in COVID-19. Cells 2021; 10:cells10081897. [PMID: 34440665 PMCID: PMC8391241 DOI: 10.3390/cells10081897] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 02/06/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) has spread over the world for more than one year. COVID-19 often develops life-threatening hypoxemia. Endothelial injury caused by the viral infection leads to intravascular coagulation and ventilation-perfusion mismatch. However, besides above pathogenic mechanisms, the role of alveolar edema in the disease progression has not been discussed comprehensively. Since the exudation of pulmonary edema fluid was extremely serious in COVID-19 patients, we bring out a hypothesis that severity of alveolar edema may determine the size of poorly-ventilated area and the blood oxygen content. Treatments to pulmonary edema (conservative fluid management, exogenous surfactant replacements and ethanol–oxygen vapor therapy hypothetically) may be greatly helpful for reducing the occurrences of severe cases. Given that late mechanical ventilation may cause mucus (edema fluid) to be blown deep into the small airways, oxygen therapy should be given at the early stages. The optimal time and blood oxygen saturation (SpO2) threshold for oxygen therapy are also discussed.
Collapse
Affiliation(s)
- Shu Yuan
- College of Resources, Sichuan Agricultural University, Chengdu 611130, China; (Z.-W.Z.); (Y.-F.F.)
- Correspondence:
| | - Si-Cong Jiang
- Chengdu Kang Hong Pharmaceutical Group Comp. Ltd., Chengdu 610036, China;
| | - Zhong-Wei Zhang
- College of Resources, Sichuan Agricultural University, Chengdu 611130, China; (Z.-W.Z.); (Y.-F.F.)
| | - Yu-Fan Fu
- College of Resources, Sichuan Agricultural University, Chengdu 611130, China; (Z.-W.Z.); (Y.-F.F.)
| | - Jing Hu
- School of Medicine, Northwest University, Xi’an 710069, China;
| | - Zi-Lin Li
- Department of Cardiovascular Surgery, Xijing Hospital, Medical University of the Air Force, Xi’an 710032, China;
| |
Collapse
|
206
|
Ray A, Jain D, Goel A, Agarwal S, Swaroop S, Das P, Arava SK, Mridha AR, Nambirajan A, Singh G, Arulselvi S, Mathur P, Kumar S, Sahni S, Nehra J, Nazneen, Bm M, Rastogi N, Mahato S, Gupta C, Bharadhan S, Dhital G, Goel P, Pandey P, Kn S, Chaudhary S, Keri VC, Chauhan VS, Mahishi N, Shahi A, R R, Gupta BK, Aggarwal R, Soni KD, Nischal N, Soneja M, Lalwani S, Sarkar C, Guleria R, Wig N, Trikha A. Clinico-pathological features in fatal COVID-19 infection: a preliminary experience of a tertiary care center in North India using postmortem minimally invasive tissue sampling. Expert Rev Respir Med 2021; 15:1367-1375. [PMID: 34227439 DOI: 10.1080/17476348.2021.1951708] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVES To study the histopathology of patients dying of COVID-19 using post-mortem minimally invasive sampling techniques. METHODS This was a single-center observational study conducted at JPNATC, AIIMS. Thirty-seven patients who died of COVID-19 were enrolled. Post-mortem percutaneous biopsies were taken from lung, heart, liver, kidney and stained with hematoxylin and eosin. Immunohistochemistry was performed using CD61 and CD163. SARS-CoV-2 virus was detected using IHC with primary antibodies. RESULTS The mean age was 48.7 years and 59.5% were males. Lung histopathology showed diffuse alveolar damage in 78% patients. Associated bronchopneumonia was seen in 37.5% and scattered microthrombi in 21% patients. Immunopositivity for SARS-CoV-2 was observed in Type II pneumocytes. Acute tubular injury with epithelial vacuolization was seen in 46% of renal biopsies. Seventy-one percent of liver biopsies showed Kupffer cell hyperplasia and 27.5% showed submassive hepatic necrosis. CONCLUSIONS Predominant finding was diffuse alveolar damage with demonstration of SARS-CoV-2 protein in the acute phase. Microvascular thrombi were rarely identified in any organ. Substantial hepatocyte necrosis, Kupffer cell hypertrophy, microvesicular, and macrovesicular steatosis unrelated to microvascular thrombi suggested that liver might be a primary target of COVID-19.
Collapse
Affiliation(s)
- Animesh Ray
- Department of Medicine, AIIMS, New Delhi, India
| | - Deepali Jain
- Department of Pathology, AIIMS, New Delhi, India
| | - Ayush Goel
- Department of Medicine, AIIMS, New Delhi, India
| | | | | | | | | | | | | | | | - S Arulselvi
- Department of Laboratory Medicine, Jpnatc, Aiims, New Delhi, India
| | - Purva Mathur
- Department of Laboratory Medicine, Jpnatc, Aiims, New Delhi, India
| | | | | | | | - Nazneen
- Department of Medicine, AIIMS, New Delhi, India
| | - Mouna Bm
- Department of Medicine, AIIMS, New Delhi, India
| | | | | | | | - S Bharadhan
- Department of Medicine, AIIMS, New Delhi, India
| | | | - Pawan Goel
- Department of Medicine, AIIMS, New Delhi, India
| | | | - Santosh Kn
- Department of Medicine, AIIMS, New Delhi, India
| | | | | | | | | | - Anand Shahi
- Department of Medicine, AIIMS, New Delhi, India
| | - Ragu R
- Department of Medicine, AIIMS, New Delhi, India
| | | | | | | | | | | | - Sanjeev Lalwani
- Division of Forensic Pathology and Molecular Laboratory, JPNATC, AIIMS, New Delhi, India
| | | | - Randeep Guleria
- Department of Pulmonary Medicine, Sleep and critical care disorders, Director, AIIMS, New Delhi, India
| | - Naveet Wig
- Department of Medicine, AIIMS, New Delhi, India
| | - Anjan Trikha
- Department of Anaesthesiology, Critical Care and Pain Medicine, JPNATC, AIIMS, New Delhi, India
| |
Collapse
|
207
|
Tohamy D, Sharaf M, Abdelazeem K, Saleh MGA, Rateb MF, Soliman W, Kedwany SM, Omar Abdelmalek M, Medhat MA, Tohamy AM, Mahmoud H. Ocular Manifestations of Post-Acute COVID-19 Syndrome, Upper Egypt Early Report. J Multidiscip Healthc 2021; 14:1935-1944. [PMID: 34326644 PMCID: PMC8315779 DOI: 10.2147/jmdh.s323582] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 07/08/2021] [Indexed: 12/14/2022] Open
Abstract
Purpose To evaluate the ocular manifestations of post-acute COVID-19 syndrome. Methods A retrospective, comparative study included 100 patients who had recovered from COVID-19 and 100 controls who were recruited by stratified randomization from hospital registration system and analyzed regarding history, full ophthalmological examination, general examination including internal medicine and neurological evaluation. Laboratory tests were done. Results Mean±SD of age were 55.5 ± 6.2 in COVID group vs 56.5 ± 5.8 in control group; P value = 0.7. In COVID group, 57 patients (57%) were males vs 51 patients (51%) in control group (P value = 0.39), the other compared parameters including history and risk factors showed non-significant difference except for ESR and D-dimer which were elevated in COVID group. In COVID group, 5 patients (5%) were having retinal vascular occlusion, 2 patients (2%) were having anterior ischemic optic neuropathy AION, 3 patients (3%) were having uveitis and 2 patients (2%) were having central serous chorioretinopathy CSCR. While in control group, 2 patients (2%) were having retinal vascular occlusion, and none had AION, uveitis or CSCR (P value = 0.006). Conclusion Post-acute COVID-19 syndrome could affect the eyes in the form of coagulation problems, neurological morbidities, and other manifestations. This necessitates meticulous follow-up of recovered patients from COVID-19.
Collapse
Affiliation(s)
- Dalia Tohamy
- Ophthalmology Department, Assiut University, Assiut, Egypt
| | - Mohamed Sharaf
- Ophthalmology Department, Assiut University, Assiut, Egypt
| | | | | | | | - Wael Soliman
- Ophthalmology Department, Assiut University, Assiut, Egypt
| | | | | | - Mohammed A Medhat
- Tropical Medicine and Gastroenterology Department, Assiut University, Assiut, Egypt
| | - Amal M Tohamy
- Neuology Department, Assiut University, Assiut, Egypt
| | - Hany Mahmoud
- Ophthalmology Department, Sohag University, Sohag, Egypt
| |
Collapse
|
208
|
Zhang ML, Jacobsen F, Pepe-Mooney BJ, Mino-Kenudson M, Deshpande V, Shih AR, Mattia AR, Goessling W, Hwabejire JO, Velmahos GC, Misdraji J. Clinicopathologic Findings in COVID-19-Associated Ischemic Enterocolitis. Histopathology 2021; 79:1004-1017. [PMID: 34292620 PMCID: PMC8444633 DOI: 10.1111/his.14457] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/22/2021] [Accepted: 07/20/2021] [Indexed: 12/17/2022]
Abstract
Aims Coronavirus disease 2019 (COVID‐19) has been recognised as a predominantly respiratory tract infection, but some patients manifest severe systemic symptoms/coagulation abnormalities. The aim of this study was to evaluate the impact of severe COVID‐19 infection on the gastrointestinal tract. Methods and results We examined clinicopathological findings in 28 resected ischaemic bowels from 22 patients with severe COVID‐19. Most patients required intubation preoperatively and presented with acute decompensation shortly before surgery. D‐dimer levels were markedly elevated in all measured cases (mean, 5394 ng/ml). Histologically, 25 cases (19 patients) showed evidence of acute ischaemia with necrosis. In this group, the most characteristic finding was the presence of small vessel fibrin thrombi (24 of 25 cases, 96%), which were numerous in 64% of cases. Patients with COVID‐19 were significantly more likely than a control cohort of 35 non‐COVID‐19‐associated acute ischaemic bowels to show isolated small intestine involvement (32% versus 6%, P < 0.001), small vessel fibrin thrombi (100% versus 43%, P < 0.001), submucosal vessels with fibrinous degeneration and perivascular neutrophils (90% versus 54%, P < 0.001), fibrin strands within submucosal vessels (58% versus 20%, P = 0.007), and histological evidence of pneumatosis (74% versus 34%, P = 0.010). Three cases in this cohort had histopathological findings normally seen in the setting of chronic ischaemia, notably prominent fibroblastic proliferation affecting the outer layer of the muscularis propria. Conclusions Herein, we describe the histopathological findings in COVID‐19‐associated ischaemic bowels and postulate a relationship with the hypercoagulable state seen in patients with severe COVID‐19 infection. Additional experience with these cases may further elucidate specific features or mechanisms of COVID‐19‐associated ischaemic enterocolitis.
Collapse
Affiliation(s)
- M Lisa Zhang
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Frank Jacobsen
- Department of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Brian J Pepe-Mooney
- Harvard Medical School, Boston, MA, USA.,Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA.,Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mari Mino-Kenudson
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Vikram Deshpande
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Angela R Shih
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Anthony R Mattia
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Department of Pathology, Newton-Wellesley Hospital, Newton, MA, USA
| | - Wolfram Goessling
- Harvard Medical School, Boston, MA, USA.,Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA.,Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Department of Medicine, Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
| | - John O Hwabejire
- Harvard Medical School, Boston, MA, USA.,Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - George C Velmahos
- Harvard Medical School, Boston, MA, USA.,Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Joseph Misdraji
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| |
Collapse
|
209
|
Paley EL. Towards Understanding COVID-19: Molecular Insights, Co-infections, Associated Disorders, and Aging. J Alzheimers Dis Rep 2021; 5:571-600. [PMID: 34514341 PMCID: PMC8385430 DOI: 10.3233/adr-210010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND COVID-19 can be related to any diseases caused by microbial infection(s) because 1) co-infection with COVID-19-related virus and other microorganism(s) and 2) because metabolites produced by microorganisms such as bacteria, fungi, and protozoan can be involved in necrotizing pneumonia and other necrotizing medical conditions observed in COVID-19. OBJECTIVE By way of illustration, the microbial metabolite of aromatic amino acid tryptophan, a biogenic amine tryptamine inducing neurodegeneration in cell and animal models, also induces necrosis. METHODS This report includes analysis of COVID-19 positivity by zip codes in Florida and relation of the positivity to population density, possible effect of ecological and social factors on spread of COVID-19, autopsy analysis of COVID-19 cases from around the world, serum metabolomics analysis, and evaluation of autoantigenome related to COVID-19. RESULTS In the present estimations, COVID-19 positivity percent per zip code population varied in Florida from 4.65% to 44.3% (February 2021 data). COVID-19 analysis is partially included in my book Microbial Metabolism and Disease (2021). The autoantigenome related to COVID-19 is characterized by alterations in protein biosynthesis proteins including aminoacyl-tRNA synthetases. Protein biosynthesis alteration is a feature of Alzheimer's disease. Serum metabolomics of COVID-19 positive patients show alteration in shikimate pathway metabolism, which is associated with the presence of Alzheimer's disease-associated human gut bacteria. CONCLUSION Such alterations in microbial metabolism and protein biosynthesis can lead to toxicity and neurodegeneration as described earlier in my book Protein Biosynthesis Interference in Disease (2020).
Collapse
Affiliation(s)
- Elena L. Paley
- Expert BioMed, Inc. and Nonprofit Public Charity Stop Alzheimers Corp., Miami-Dade, FL, USA
| |
Collapse
|
210
|
Codd AS, Hanna SJ, Compeer EB, Richter FC, Pring EJ, Gea-Mallorquí E, Borsa M, Moon OR, Scourfield DO, Gallimore AM, Milicic A. Neutrophilia, lymphopenia and myeloid dysfunction: a living review of the quantitative changes to innate and adaptive immune cells which define COVID-19 pathology. OXFORD OPEN IMMUNOLOGY 2021; 2:iqab016. [PMID: 35593707 PMCID: PMC8371938 DOI: 10.1093/oxfimm/iqab016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Destabilization of balanced immune cell numbers and frequencies is a common feature of viral infections. This occurs due to, and further enhances, viral immune evasion and survival. Since the discovery of the Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2), which manifests in coronavirus disease 2019 (COVID-19), a great number of studies have described the association between this virus and pathologically increased or decreased immune cell counts. In this review, we consider the absolute and relative changes to innate and adaptive immune cell numbers, in COVID-19. In severe disease particularly, neutrophils are increased, which can lead to inflammation and tissue damage. Dysregulation of other granulocytes, basophils and eosinophils represents an unusual COVID-19 phenomenon. Contrastingly, the impact on the different types of monocytes leans more strongly to an altered phenotype, e.g. HLA-DR expression, rather than numerical changes. However, it is the adaptive immune response that bears the most profound impact of SARS-CoV-2 infection. T cell lymphopenia correlates with increased risk of intensive care unit admission and death; therefore, this parameter is particularly important for clinical decision-making. Mild and severe diseases differ in the rate of immune cell counts returning to normal levels post disease. Tracking the recovery trajectories of various immune cell counts may also have implications for long-term COVID-19 monitoring. This review represents a snapshot of our current knowledge, showing that much has been achieved in a short period of time. Alterations in counts of distinct immune cells represent an accessible metric to inform patient care decisions or predict disease outcomes.
Collapse
Affiliation(s)
- Amy S Codd
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Stephanie J Hanna
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Ewoud B Compeer
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Felix C Richter
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Eleanor J Pring
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Ester Gea-Mallorquí
- Viral Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Mariana Borsa
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Owen R Moon
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - D Oliver Scourfield
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Awen M Gallimore
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Anita Milicic
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, UK
| |
Collapse
|
211
|
Kritselis M, Yambayev I, Prilutskiy A, Shevtsov A, Vadlamudi C, Zheng H, Elsadwai M, Ma L, Aniskovich E, Kataria Y, Higgins S, Sarita-Reyes C, Zuo T, Zhao Q, Quillen K, Burks EJ. Distinctive pseudopalisaded histiocytic hyperplasia characterizes the transition of exudative to proliferative phase of diffuse alveolar damage in patients dying of COVID-19. Hum Pathol 2021; 116:49-62. [PMID: 34273395 PMCID: PMC8278870 DOI: 10.1016/j.humpath.2021.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/21/2021] [Accepted: 06/24/2021] [Indexed: 12/15/2022]
Abstract
Severe COVID-19 results in a glucocorticoid responsive form of acute respiratory distress (ARDS)/diffuse alveolar damage (DAD). Herein we compare the immunopathology of lung tissue procured at autopsy in patients dying of SARS-CoV-2 with those dying of DAD prior to the COVID-19 pandemic. Autopsy gross and microscopic features stratified by duration of illness in twelve patients who tested positive for SARS-CoV-2 viral RNA, as well as seven patients dying of DAD prior to the COVID-19 pandemic were evaluated with multiplex (5-plex: CD4, CD8, CD68, CD20, AE1/AE3) and SARS-CoV immunohistochemistry to characterize the immunopathologic stages of DAD. We observed a distinctive pseudopalisaded histiocytic hyperplasia interposed between the exudative and proliferative phase of COVID-19 associated DAD, which was most pronounced at the fourth week from symptom onset. Pulmonary macrothrombi were seen predominantly in cases with pseudopalisaded histiocytic hyperplasia and/or proliferative phase DAD. Neither pseudopalisaded histiocytic hyperplasia nor pulmonary macrothrombi was seen in non-COVID-19 DAD cases, whereas microthrombi were common in DAD regardless of etiology. The inflammatory pattern of pseudopalisaded histiocytic hyperplasia may represent the distinctive immunopathology associated with the dexamethasone responsive form of DAD seen in severe COVID-19.
Collapse
Affiliation(s)
- Michael Kritselis
- Department of Pathology & Laboratory Medicine, Boston University School of Medicine, Boston Medical Center, Boston, MA, 02118, USA
| | - Ilyas Yambayev
- Department of Pathology & Laboratory Medicine, Boston University School of Medicine, Boston Medical Center, Boston, MA, 02118, USA
| | - Andrey Prilutskiy
- Department of Pathology & Laboratory Medicine, Boston University School of Medicine, Boston Medical Center, Boston, MA, 02118, USA
| | - Artem Shevtsov
- Department of Pathology & Laboratory Medicine, Boston University School of Medicine, Boston Medical Center, Boston, MA, 02118, USA
| | - Charitha Vadlamudi
- Department of Pathology & Laboratory Medicine, Boston University School of Medicine, Boston Medical Center, Boston, MA, 02118, USA
| | - Hanqiao Zheng
- Department of Pathology & Laboratory Medicine, Boston University School of Medicine, Boston Medical Center, Boston, MA, 02118, USA
| | - Murad Elsadwai
- Department of Pathology & Laboratory Medicine, Boston University School of Medicine, Boston Medical Center, Boston, MA, 02118, USA
| | - Lina Ma
- Department of Pathology & Laboratory Medicine, Boston University School of Medicine, Boston Medical Center, Boston, MA, 02118, USA
| | - Emily Aniskovich
- Department of Pathology & Laboratory Medicine, Boston University School of Medicine, Boston Medical Center, Boston, MA, 02118, USA
| | - Yachana Kataria
- Department of Pathology & Laboratory Medicine, Boston University School of Medicine, Boston Medical Center, Boston, MA, 02118, USA
| | - Sara Higgins
- Department of Pathology & Laboratory Medicine, Boston University School of Medicine, Boston Medical Center, Boston, MA, 02118, USA
| | - Carmen Sarita-Reyes
- Department of Pathology & Laboratory Medicine, Boston University School of Medicine, Boston Medical Center, Boston, MA, 02118, USA
| | - Tao Zuo
- Department of Pathology & Laboratory Medicine, Boston University School of Medicine, Boston Medical Center, Boston, MA, 02118, USA
| | - Qing Zhao
- Department of Pathology & Laboratory Medicine, Boston University School of Medicine, Boston Medical Center, Boston, MA, 02118, USA
| | - Karen Quillen
- Department of Hematology and Oncology, Boston University School of Medicine, Boston Medical Center, Boston, MA, 02118, USA
| | - Eric J Burks
- Department of Pathology & Laboratory Medicine, Boston University School of Medicine, Boston Medical Center, Boston, MA, 02118, USA.
| |
Collapse
|
212
|
Corredor G, Toro P, Bera K, Rasmussen D, Viswanathan VS, Buzzy C, Fu P, Barton LM, Stroberg E, Duval E, Gilmore H, Mukhopadhyay S, Madabhushi A. Computational pathology reveals unique spatial patterns of immune response in H&E images from COVID-19 autopsies: preliminary findings. J Med Imaging (Bellingham) 2021; 8:017501. [PMID: 34268443 DOI: 10.1117/1.jmi.8.s1.017501] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 06/28/2021] [Indexed: 12/22/2022] Open
Abstract
Purpose: We used computerized image analysis and machine learning approaches to characterize spatial arrangement features of the immune response from digitized autopsied H&E tissue images of the lung in coronavirus disease 2019 (COVID-19) patients. Additionally, we applied our approach to tease out potential morphometric differences from autopsies of patients who succumbed to COVID-19 versus H1N1. Approach: H&E lung whole slide images from autopsy specimens of nine COVID-19 and two H1N1 patients were computationally interrogated. 606 image patches ( ∼ 55 per patient) of 1024 × 882 pixels were extracted from the 11 autopsied patient studies. A watershed-based segmentation approach in conjunction with a machine learning classifier was employed to identify two types of nuclei families: lymphocytes and non-lymphocytes (i.e., other nucleated cells such as pneumocytes, macrophages, and neutrophils). Based off the proximity of the individual nuclei, clusters for each nuclei family were constructed. For each of the resulting clusters, a series of quantitative measurements relating to architecture and density of nuclei clusters were calculated. A receiver operating characteristics-based feature selection method, violin plots, and the t-distributed stochastic neighbor embedding algorithm were employed to study differences in immune patterns. Results: In COVID-19, the immune response consistently showed multiple small-size lymphocyte clusters, suggesting that lymphocyte response is rather modest, possibly due to lymphocytopenia. In H1N1, we found larger lymphocyte clusters that were proximal to large clusters of non-lymphocytes, a possible reflection of increased prevalence of macrophages and other immune cells. Conclusion: Our study shows the potential of computational pathology to uncover immune response features that may not be obvious by routine histopathology visual inspection.
Collapse
Affiliation(s)
- Germán Corredor
- Case Western Reserve University, Center for Computational Imaging and Personalized Diagnostics, Cleveland, Ohio, United States.,Louis Stokes Cleveland VA Medical Center, Cleveland, Ohio, United States
| | - Paula Toro
- Case Western Reserve University, Center for Computational Imaging and Personalized Diagnostics, Cleveland, Ohio, United States
| | - Kaustav Bera
- Case Western Reserve University, Center for Computational Imaging and Personalized Diagnostics, Cleveland, Ohio, United States
| | - Dylan Rasmussen
- Case Western Reserve University, Center for Computational Imaging and Personalized Diagnostics, Cleveland, Ohio, United States
| | - Vidya Sankar Viswanathan
- Case Western Reserve University, Center for Computational Imaging and Personalized Diagnostics, Cleveland, Ohio, United States
| | - Christina Buzzy
- Case Western Reserve University, Center for Computational Imaging and Personalized Diagnostics, Cleveland, Ohio, United States
| | - Pingfu Fu
- Case Western Reserve University, Department of Population and Quantitative Health Sciences, Cleveland, Ohio, United States
| | - Lisa M Barton
- Oklahoma Office of the Chief Medical Examiner, Oklahoma City, Oklahoma, United States
| | - Edana Stroberg
- Oklahoma Office of the Chief Medical Examiner, Oklahoma City, Oklahoma, United States
| | - Eric Duval
- Oklahoma Office of the Chief Medical Examiner, Oklahoma City, Oklahoma, United States
| | - Hannah Gilmore
- University Hospitals, Department of Pathology, Cleveland, Ohio, United States
| | | | - Anant Madabhushi
- Case Western Reserve University, Center for Computational Imaging and Personalized Diagnostics, Cleveland, Ohio, United States.,Louis Stokes Cleveland VA Medical Center, Cleveland, Ohio, United States
| |
Collapse
|
213
|
Assimakopoulos SF, Markantes GK, Papageorgiou D, Mamali I, Markou KB, Marangos M, Michalaki MA. Low serum TSH in the acute phase of COVID-19 pneumonia: thyrotoxicosis or a face of "non-thyroidal illness syndrome"? Clin Chem Lab Med 2021; 59:e420-e423. [PMID: 34246200 DOI: 10.1515/cclm-2021-0511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 05/31/2021] [Indexed: 01/08/2023]
Affiliation(s)
- Stelios F Assimakopoulos
- Department of Internal Medicine, University of Patras School of Health Sciences, Patras, Greece.,Division of Infectious Diseases, University of Patras School of Health Sciences, Patras, Greece
| | - Georgios K Markantes
- Division of Endocrinology - Department of Internal Medicine, University of Patras School of Health Sciences, Patras, Greece
| | - Dimitris Papageorgiou
- Department of Internal Medicine, University of Patras School of Health Sciences, Patras, Greece
| | - Irene Mamali
- Division of Endocrinology - Department of Internal Medicine, University of Patras School of Health Sciences, Patras, Greece
| | - Kostas B Markou
- Division of Endocrinology - Department of Internal Medicine, University of Patras School of Health Sciences, Patras, Greece
| | - Markos Marangos
- Department of Internal Medicine, University of Patras School of Health Sciences, Patras, Greece.,Division of Infectious Diseases, University of Patras School of Health Sciences, Patras, Greece
| | - Marina A Michalaki
- Division of Endocrinology - Department of Internal Medicine, University of Patras School of Health Sciences, Patras, Greece
| |
Collapse
|
214
|
Salem AM, Al Khathlan N, Alharbi AF, Alghamdi T, AlDuilej S, Alghamdi M, Alfudhaili M, Alsunni A, Yar T, Latif R, Rafique N, Al Asoom L, Sabit H. The Long-Term Impact of COVID-19 Pneumonia on the Pulmonary Function of Survivors. Int J Gen Med 2021; 14:3271-3280. [PMID: 34267545 PMCID: PMC8276825 DOI: 10.2147/ijgm.s319436] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 06/24/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The sequelae of COVID-19 pneumonia on pulmonary function and airways inflammation are still an area of active research. OBJECTIVE This research aimed to explore the long-term impact of COVID-19 pneumonia on the lung function after three months from recovery. METHODS Fifty subjects (age 18-60 years) were recruited and classified into two groups: the control group (30 subjects) and the post-COVID-19 pneumonia group (20 patients). Pulmonary function tests, spirometry, body plethysmography [lung volumes and airway resistance (Raw)], diffusion capacity for carbon monoxide (DLCO), and fractional exhaled nitric oxide (FeNO), were measured after at least 3 months post-recovery. RESULTS Significant reduction in total lung capacity (TLC), forced vital capacity (FVC), forced expiratory volume (FEV1), FEV1/FEV, and diffusing capacity for carbon monoxide (DLCO) was observed in post-COVID-19 subjects compared to controls. Restrictive lung impairment was observed in 50% of post-COVID-19 cases (n = 10) compared to 20% in the control group (n = 6, P = 0.026). In addition, mild diffusion defect was detected in 35% (n = 7) of the post-COVID-19 group compared to 23.3% (n = 7) in the controls (P = 0.012). CONCLUSION COVID-19 pneumonia has an impact on the lung functions in terms of restrictive lung impairment and mild diffusion defect after three months from recovery. Therefore, a long-term follow-up of the lung function in post-COVID-19 survivors is recommended.
Collapse
Affiliation(s)
- Ayad Mohammed Salem
- Department of Physiology, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Noor Al Khathlan
- Department of respiratory care, College of Applied Medical Sciences, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | | | - Turki Alghamdi
- College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Saleh AlDuilej
- College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Mohammed Alghamdi
- College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Majed Alfudhaili
- College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Ahmed Alsunni
- Department of Physiology, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Talay Yar
- Department of Physiology, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Rabia Latif
- Department of Physiology, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Nazish Rafique
- Department of Physiology, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Lubna Al Asoom
- Department of Physiology, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Hussein Sabit
- Department of Genetics, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| |
Collapse
|
215
|
Sanguedolce F, Zanelli M, Froio E, Bisagni A, Zizzo M, Ascani S, Stallone G, Netti S, Ranieri E, Falagario U, Carrieri G, Cormio L. Pathological diagnosis of Coronavirus-related nephropathy: insight from postmortem studies. Crit Rev Clin Lab Sci 2021; 58:563-575. [PMID: 34236278 DOI: 10.1080/10408363.2021.1944047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
A novel coronavirus pneumonia first occurred in Wuhan, China in early December 2019; the causative agent was identified and named as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) by the World Health Organization (WHO), and the resulting disease termed coronavirus disease 2019 (COVID-19), according to the WHO coronavirus disease situation reports. This condition has spread rapidly all over the world and caused more than 125 million cases globally, with more than 2 million related deaths. Two previous outbreaks due to zoonotic coronaviruses have occurred in the last 20 years, namely the severe acute respiratory syndrome coronavirus (SARS-CoV) and the Middle East respiratory syndrome coronavirus (MERS-CoV), causing high morbidity and mortality in human populations upon crossing the species barriers. SARS-CoV-2, SARS-CoV, and MERS-CoV show several similarities in pathogenicity and clinical presentations, the latter ranging from asymptomatic infection to severe acute respiratory distress syndrome (ARDS) and multiorgan impairment. Acute kidney injury (AKI) has been commonly reported in patients with CoV infections; therefore, pathological analysis of renal parenchyma in these patients has been carried out in order to improve knowledge about underlying mechanisms. Viral infection has been demonstrated in the renal tubular epithelial cells by electron microscopy (EM), immunohistochemistry (IHC), and in situ hybridization (ISH), although with conflicting results. Light microscopy (LM) changes have been described in the renal parenchyma primarily in the form of acute renal tubular damage, possibly due to direct viral cytopathic effect and immune-mediated mechanisms such as cytokine storm syndrome. In this review, we describe and discuss the spectrum of histological, ultrastructural, and molecular findings in SARS-CoV, MERS-CoV, and SARS-CoV-2-related renal pathology obtained from postmortem studies, as well as intrinsic limitations and pitfalls of current diagnostic techniques.
Collapse
Affiliation(s)
| | - Magda Zanelli
- Pathology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Emilia, Italy
| | - Elisabetta Froio
- Pathology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Emilia, Italy
| | - Alessandra Bisagni
- Pathology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Emilia, Italy
| | - Maurizio Zizzo
- Surgical Oncology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Emilia, Italy.,Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Stefano Ascani
- Pathology Unit, Azienda Ospedaliera S. Maria di Terni, University of Perugia, Terni, Italy
| | - Giovanni Stallone
- Nephrology Dialysis and Transplantation Unit, University of Foggia, Foggia, Italy
| | - Stefano Netti
- Clinical Pathology Unit, University of Foggia, Foggia, Italy
| | - Elena Ranieri
- Clinical Pathology Unit, University of Foggia, Foggia, Italy
| | - Ugo Falagario
- Urology and Renal Transplantation Unit, University of Foggia, Foggia, Italy
| | - Giuseppe Carrieri
- Urology and Renal Transplantation Unit, University of Foggia, Foggia, Italy
| | - Luigi Cormio
- Urology and Renal Transplantation Unit, University of Foggia, Foggia, Italy.,Department of Urology, Bonomo Teaching Hospital, Andria, Italy
| |
Collapse
|
216
|
Barth RF, Buja LM, Barth AL, Carpenter DE, Parwani AV. A Comparison of the Clinical, Viral, Pathologic, and Immunologic Features of Severe Acute Respiratory Syndrome (SARS), Middle East Respiratory Syndrome (MERS), and Coronavirus 2019 (COVID-19) Diseases. Arch Pathol Lab Med 2021; 145:1194-1211. [PMID: 34232978 DOI: 10.5858/arpa.2020-0820-sa] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/25/2021] [Indexed: 11/06/2022]
Abstract
CONTEXT -The purpose of this review is to compare three coronavirus diseases: severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS), and coronavirus disease 2019 (COVID-19) caused by SARS-CoV, MERS-CoV, and SARS-CoV-2 viruses, respectively. OBJECTIVE -To cover the following topics: clinical considerations, viral characteristics, pathology, immune response, pathogenesis, and the prognosis associated with each coronavirus human disease in humans. DATA SOURCES -Clinically, flu-like symptoms are usual at the time of presentation for all 3 diseases, but these vary from asymptomatic to severe multi-system involvement. The pathology associated with symptomatic SARS and COVID-19 has been well described, the most prominent of which is diffuse alveolar damage (DAD). The immune response to each of these viruses is highly complex and includes both humoral and cellular components that can have a significant impact on prognosis. In severe cases of COVID-19, a dysregulated innate host immune system can initiate a hyperinflammatory syndrome dominated by endothelial dysfunction that can lead to a hypercoagulable state with microthrombi, resulting in a systemic micro- and macro-vascular disease. CONCLUSIONS -The SARS and MERS epidemics have been limited, involving 7,500 and 2,500 individuals, respectively. In contrast, COVID-19 has resulted in a worldwide pandemic with over 177 million cases and 3.9 million deaths as of June 15, 2021, and fatality rates ranging from <0.1% to ~10% depending upon the country. Ending on a positive note, the development of a number of vaccines, at least six of which now are in clinical use, should mitigate and eventually control the devastating COVID-19 pandemic.
Collapse
Affiliation(s)
- Rolf F Barth
- Department of Pathology (RF Barth, Parwani), S.P. Technical Editor (Retired) Departments of Neurosurgery and Radiation Oncology (Carpenter), The Ohio State University Columbus, Ohio
| | - L Maximillian Buja
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas (Buja)
| | - Alison L Barth
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA (AL Barth)
| | - David E Carpenter
- Department of Pathology (RF Barth, Parwani), S.P. Technical Editor (Retired) Departments of Neurosurgery and Radiation Oncology (Carpenter), The Ohio State University Columbus, Ohio
| | - Anil V Parwani
- Department of Pathology (RF Barth, Parwani), S.P. Technical Editor (Retired) Departments of Neurosurgery and Radiation Oncology (Carpenter), The Ohio State University Columbus, Ohio
| |
Collapse
|
217
|
Sharathkumar AA, Faustino EVS, Takemoto CM. How we approach thrombosis risk in children with COVID-19 infection and MIS-C. Pediatr Blood Cancer 2021; 68:e29049. [PMID: 33955167 PMCID: PMC8206673 DOI: 10.1002/pbc.29049] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/04/2021] [Accepted: 03/22/2021] [Indexed: 01/08/2023]
Abstract
Thrombosis within the microvasculature and medium to large vessels is a serious and common complication among critically ill individuals with coronavirus disease 2019 (COVID-19). While children are markedly less likely to develop severe disease than adults, they remain at risk for thrombosis during acute infection and with the post-acute inflammatory illness termed multisystem inflammatory syndrome in children. Significant knowledge deficits in understanding COVID-19-associated coagulopathy and thrombotic risk pose clinical challenges for pediatric providers who must incorporate expert opinion and personal experience to manage individual patients. We discuss clinical scenarios to provide framework for characterizing thrombosis risk and thromboprophylaxis in children with COVID-19.
Collapse
Affiliation(s)
- Anjali A. Sharathkumar
- Stead Family Department of Pediatrics, Carver College of MedicineUniversity of IowaIowa CityIowaUSA
| | - E. Vincent S. Faustino
- Section of Pediatric Critical Care Medicine, Department of PediatricsYale School of MedicineNew HavenConnecticutUSA
| | - Clifford M. Takemoto
- Division of Clinical HematologySt. Jude Children's Research HospitalMemphisTennesseeUSA
| |
Collapse
|
218
|
Gaussen A, Hornby L, Rockl G, O'Brien S, Delage G, Sapir-Pichhadze R, Drews SJ, Weiss MJ, Lewin A. Evidence of SARS-CoV-2 Infection in Cells, Tissues, and Organs and the Risk of Transmission Through Transplantation. Transplantation 2021; 105:1405-1422. [PMID: 33724248 DOI: 10.1097/tp.0000000000003744] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The emergence of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus responsible for the coronavirus disease 2019 (COVID-19) pandemic has raised concerns for programs overseeing donation and transplantation of cells, tissues, and organs (CTO) that this virus might be transmissible by transfusion or transplantation. Transplant recipients are considered particularly vulnerable to pathogens because of immunosuppression, and SARS-CoV-2 is likely to generate complications if contracted. Several signs and symptoms observed in COVID-19 positive patients reflect damage to multiple organs and tissues, raising the possibility of extrapulmonary SARS-CoV-2 infections and risk of transmission. At the beginning of the pandemic, a consensus has emerged not to consider COVID-19 positive patients as potential living or deceased donors, resulting in a global decrease in transplantation procedures. Medical decision-making at the time of organ allocation must consider safely alongside the survival advantages offered by transplantation. To address the risk of transmission by transplantation, this review summarizes the published cases of transplantation of cells or organs from donors infected with SARS-CoV-2 until January 2021 and assesses the current state of knowledge for the detection of this virus in different biologic specimens, cells, tissues, and organs. Evidence collected to date raises the possibility of SARS-CoV-2 infection and replication in some CTO, which makes it impossible to exclude transmission through transplantation. However, most studies focused on evaluating transmission under laboratory conditions with inconsistent findings, rendering the comparison of results difficult. Improved standardization of donors and CTO screening practices, along with a systematic follow-up of transplant recipients could facilitate the assessment of SARS-CoV-2 transmission risk by transplantation.
Collapse
Affiliation(s)
- Amaury Gaussen
- Medical Affairs and Innovation, Héma-Québec, Québec, QC, Canada
| | - Laura Hornby
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Canadian Blood Services, Ottawa, ON, Canada
| | - Gary Rockl
- Medical Affairs and Innovation, Héma-Québec, Québec, QC, Canada
| | | | - Gilles Delage
- Medical Affairs and Innovation, Héma-Québec, Saint-Laurent, QC, Canada
| | - Ruth Sapir-Pichhadze
- Centre for Outcomes Research and Evaluation (CORE), Research Institute of McGill University Health Centre, Montréal, QC, Canada
- Division of Nephrology and the Multi Organ Transplant Program, Royal Victoria Hospital, McGill University Health Centre, Montréal, QC, Canada
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montréal, QC, Canada
| | - Steven J Drews
- Canadian Blood Services, Edmonton, AB, Canada
- Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Matthew J Weiss
- Population Health and Optimal Health Practices Research Unit, Trauma-Emergency-Critical Care Medicine, CHU de Québec, Université Laval Research Center, Québec, QC, Canada
- Pediatrics Department, Intensive Care Division, Faculté de Médecine, Université Laval, Québec, QC, Canada
- Transplant Québec, Montréal, QC, Canada
| | - Antoine Lewin
- Medical Affairs and Innovation, Héma-Québec, Saint-Laurent, QC, Canada
- Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
219
|
Moshrefi M, Ghasemi-Esmailabad S, Ali J, Findikli N, Mangoli E, Khalili MA. The probable destructive mechanisms behind COVID-19 on male reproduction system and fertility. J Assist Reprod Genet 2021; 38:1691-1708. [PMID: 33977466 PMCID: PMC8112744 DOI: 10.1007/s10815-021-02097-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 01/28/2021] [Indexed: 01/08/2023] Open
Abstract
PURPOSE The present study aims to summarize the current understanding of probable mechanisms and claims of adverse effects of SARS-CoV-2 on male fertility potential. METHODS Our search was including original articles, reviews, guidelines, letters to the editor, comments on guidelines, and editorials, regarding the male reproductive system. We used the words SARS-CoV-2, coronavirus, severe acute respiratory syndrome coronavirus 2, "2019 ncov," testis, sperm, male factor infertility, fertility treatment, semen, assisted reproductive technology (ART), sexual transmission, and ACE2. RESULTS Data showed coronavirus affects men more than women because of more expression of 2019 nCoV receptors (ACE2 and TMPRSS2) in testicular cells. Also, "Bioinformatics Analysis" suggests that sperm production may be damaged, since "Pseudo Time Analysis" has shown disruption in spermatogenesis. "Gene Ontology" (GO) showed an increase in viral reproduction and a decrease in sperm production-related terms. Recently, SARS-COV-2 mRNA and protein were detected in the semen of patients that had recovered from SARS-CoV-2 infection. Therefore, the probable disruption of blood-testis barrier (BTB) in febrile diseases is suspected in the acute phase of the disease enabling viral entry into the testes. Not only is spermatogenesis disturbed, but also disturbs gonadotropin, androgens, and testosterone secretion during SARS-CoV-2 infection. No sexual transmission has been reported yet; however, detection of the virus in semen still makes the sexual transmission an open question. CONCLUSION There is a concern that male fertility may be disturbed after the SARS-CoV-2 infection. Therefore, follow-up of the reproductive functions and male fertility may be necessary in recovered cases, especially in aged men.
Collapse
Affiliation(s)
- Mojgan Moshrefi
- Research and Clinical Center for Infertility, Yazd Reproductive Science Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Reproductive Biology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Medical Nanotechnology & Tissue Engineering Research Center, Yazd Reproductive Science Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | - Jaffar Ali
- Synbios Media, www.synbiosmedia.com, P.O. Box 02042, GPO, Shah Alam, Selangor Malaysia
| | - Necati Findikli
- Bahceci Fulya Assisted Reproduction Center, Istanbul, Turkey
- Department of Bioengineering, Beykent University, Istanbul, Turkey
| | - Esmat Mangoli
- Research and Clinical Center for Infertility, Yazd Reproductive Science Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Reproductive Biology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohammad Ali Khalili
- Research and Clinical Center for Infertility, Yazd Reproductive Science Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Reproductive Biology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
220
|
Saha BK, Chong WH, Austin A, Kathuria R, Datar P, Shkolnik B, Beegle S, Chopra A. Pleural abnormalities in COVID-19: a narrative review. J Thorac Dis 2021; 13:4484-4499. [PMID: 34422375 PMCID: PMC8339774 DOI: 10.21037/jtd-21-542] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 06/25/2021] [Indexed: 12/12/2022]
Abstract
OBJECTIVE This narrative review aims to provide a detailed overview of pleural abnormalities in patients with coronavirus disease 19 or COVID-19. BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-Cov-2) is a novel beta coronavirus responsible for COVID-19. Although pulmonary parenchymal and vascular changes associated with COVID-19 are well established, pleural space abnormalities have not been the primary focus of investigations. METHODS Narrative overview of the medical literature regarding pleural space abnormalities in COVID-19. The appropriate manuscripts were identified by searching electronic medical databases and by hand searching the bibliography of the identified papers. Pleural abnormalities on transverse and ultrasound imaging are discussed. The incidence, clinical features, pathophysiology, and fluid characteristics of pleural effusion are reviewed. Studies reporting pneumothorax and pneumomediastinum are examined to evaluate for pathogenesis and prognosis. A brief comparative analysis of pleural abnormalities among patients with COVID-19, severe acute respiratory syndrome (SARS), and Middle Eastern respiratory syndrome (MERS) has been provided. CONCLUSIONS Radiologic pleural abnormalities are common in COVID-19, but the incidence of pleural effusion appears to be low. Pneumothorax is rare and does not independently predispose the patient to worse outcomes. SARS-CoV-2 infects the pleural space; however, whether the pleural fluid can propagate the infection is unclear.
Collapse
Affiliation(s)
- Biplab K. Saha
- Department of Pulmonary and Critical Care Medicine, Ozarks Medical Center, West Plains, MO, USA
| | - Woon H. Chong
- Department of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY, USA
| | - Adam Austin
- Department of Pulmonary and Critical Care Medicine, University of Florida, Gainesville, FL, USA
| | - Ritu Kathuria
- Department of Infectious Disease, Ozarks Medical Center, West Plains, MO, USA
| | - Praveen Datar
- Department of Pulmonary and Critical Care Medicine, Ozarks Medical Center, West Plains, MO, USA
| | - Boris Shkolnik
- Department of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY, USA
| | - Scott Beegle
- Department of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY, USA
| | - Amit Chopra
- Department of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY, USA
| |
Collapse
|
221
|
Liu K, Chen X, Ren X, Wu Y, Ren S, Qin C. SARS-CoV-2 effects in the genitourinary system and prospects of sex hormone therapy. Asian J Urol 2021; 8:303-314. [PMID: 33282690 PMCID: PMC7703223 DOI: 10.1016/j.ajur.2020.11.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 08/20/2020] [Indexed: 12/20/2022] Open
Abstract
OBJECT Corona virus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which leads to acute respiratory infection symptoms. SARS-CoV-2 infection is not always limited to the respiratory tract, and renal infection and dysfunction have been shown to be specific risk factors for death. In addition, COVID-19 has a higher incidence, severity and mortality in men than women. This disparity is due to biological rather than comorbid or behavioral sex differences. Because the male reproductive system is unique, the function of sex hormones in COVID-19 infection may explain the differences between males and females. Understanding these factors will provide appropriate prevention measures and adequate triage strategies and guide the drug discovery process. METHODS An electronic search was completed in PubMed, ARXIV, MEDRXIV and BIORXIV. The most relevant articles were systematically reviewed. In addition, single cell RNA sequencing analysis of tissue samples from human cell landscape was conducted. RESULTS The influence of SARS-CoV-2 on the urogenital system, the possibility of urinary tract transmission and the functions of sex hormones were discussed in this review. CONCLUSION Corona viruses can invade the genitourinary system, causing urological symptoms. Identifying the potential genitourinary organ impairments and protecting them from damage are necessary. Since sex hormones have potential as specific drugs, the gonadal hormones substitution therapy should be considered in both sexes in the COVID-19 pandemic.
Collapse
Affiliation(s)
| | - Xinglin Chen
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaohan Ren
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuqing Wu
- Medical College, Southeast University, Nanjing, China
| | - Shancheng Ren
- Department of Urology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Chao Qin
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
222
|
Khatam‐Lashgari A, Henningsen MJ, Olsen KB, Jacobsen C, Hasselby JP, Colville‐Ebeling B, Banner J. Autopsies in pandemics - a perspective on barriers and benefits. Is it time for a revival? APMIS 2021; 129:324-339. [PMID: 33645838 PMCID: PMC8013917 DOI: 10.1111/apm.13111] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/21/2020] [Indexed: 01/13/2023]
Abstract
Influenza virus and coronavirus pandemics regularly sweep the globe, at great cost of health and economy. Our aim was to conduct a PubMed search for autopsy studies on influenza and coronavirus to investigate the contribution of autopsies during pandemics, focussing on autopsy methods and procedures and the role of autopsy findings in pandemics. The retrieved autopsy studies generally relied on microscopy, polymerase chain reaction (PCR), immunostaining and electron microscopy. Most were small and reported on lung effects, including diffuse alveolar damage (DAD), pneumonia and tracheobronchitis. Antibiotic therapy has diminished a role for bacterial pneumonia, whereas obesity is an emerging risk factor. Autopsy studies have provided new insights into coronavirus disease 2019 (COVID-19) treatments like anti-coagulative therapy. Unfortunately, autopsies during pandemics are hampered by lack of guidelines, facilities and expertise for handling potentially infectious corpses and by widely varying recommendations for personal protective equipment and procedures. The Department of Forensic Pathology, at the Forensic Institute, at the University of Copenhagen in Denmark has, in collaboration with the Department of Pathology, Rigshospitalet, Copenhagen, initiated a prospective observational study on COVID-19-related deaths encompassing postmortem imaging, standardized autopsy procedures/reporting and extensive tissue sampling for histological, chemical, microbiological and genetic analysis. The study involves a diverse array of research groups at the University of Copenhagen, and the clinical field.
Collapse
Affiliation(s)
- Apameh Khatam‐Lashgari
- Department of Forensic MedicineSection of Forensic PathologyUniversity of CopenhagenCopenhagenDenmark
| | - Mikkel Jon Henningsen
- Department of Forensic MedicineSection of Forensic PathologyUniversity of CopenhagenCopenhagenDenmark
| | - Kristine Boisen Olsen
- Department of Forensic MedicineSection of Forensic PathologyUniversity of CopenhagenCopenhagenDenmark
| | - Christina Jacobsen
- Department of Forensic MedicineSection of Forensic PathologyUniversity of CopenhagenCopenhagenDenmark
| | - Jane Preuss Hasselby
- Department of PathologyUniversity Hospital of Copenhagen (Rigshospitalet)CopenhagenDenmark
| | | | - Jytte Banner
- Department of Forensic MedicineSection of Forensic PathologyUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
223
|
Laveneziana P, Straus C, Meiners S. How and to What Extent Immunological Responses to SARS-CoV-2 Shape Pulmonary Function in COVID-19 Patients. Front Physiol 2021; 12:628288. [PMID: 34267671 PMCID: PMC8276038 DOI: 10.3389/fphys.2021.628288] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 05/31/2021] [Indexed: 12/25/2022] Open
Abstract
COVID-19 is a disease caused by a new coronavirus SARS-CoV-2, primarily impacting the respiratory system. COVID-19 can result in mild illness or serious disease leading to critical illness and requires admission to ICU due to respiratory failure. There is intense discussion around potential factors predisposing to and protecting from COVID-19. The immune response and the abnormal respiratory function with a focus on respiratory function testing in COVID-19 patients will be at the center of this Perspective article of the Frontiers in Physiology Series on “The Tribute of Physiology for the Understanding of COVID-19 Disease.” We will discuss current advances and provide future directions and present also our perspective in this field.
Collapse
Affiliation(s)
- Pierantonio Laveneziana
- Assistance Publique - Hôpitaux de Paris (AP-HP), Groupe Hospitalier Universitaire APHP-Sorbonne Université, Sites Pitié-Salpêtrière, Saint-Antoine et Tenon, Service des Explorations Fonctionnelles de la Respiration, de l'Exercice et de la Dyspnée (Département R3S), Paris, France.,Sorbonne Université, INSERM, UMRS 1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Paris, France
| | - Christian Straus
- Assistance Publique - Hôpitaux de Paris (AP-HP), Groupe Hospitalier Universitaire APHP-Sorbonne Université, Sites Pitié-Salpêtrière, Saint-Antoine et Tenon, Service des Explorations Fonctionnelles de la Respiration, de l'Exercice et de la Dyspnée (Département R3S), Paris, France.,Sorbonne Université, INSERM, UMRS 1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Paris, France
| | - Silke Meiners
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| |
Collapse
|
224
|
Kianzad A, Meijboom LJ, Nossent EJ, Roos E, Schurink B, Bonta PI, van den Berk IAH, Britstra R, Stoker J, Vonk Noordegraaf A, van der Valk P, Thunnissen E, Bugiani M, Bogaard HJ, Radonic T. COVID-19: Histopathological correlates of imaging patterns on chest computed tomography. Respirology 2021; 26:869-877. [PMID: 34159661 PMCID: PMC8447040 DOI: 10.1111/resp.14101] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/22/2021] [Accepted: 06/02/2021] [Indexed: 02/01/2023]
Abstract
BACKGROUND AND OBJECTIVE Patients with coronavirus disease 2019 (COVID-19) pneumonia present with typical findings on chest computed tomography (CT), but the underlying histopathological patterns are unknown. Through direct regional correlation of imaging findings to histopathological patterns, this study aimed to explain typical COVID-19 CT patterns at tissue level. METHODS Eight autopsy cases were prospectively selected of patients with PCR-proven COVID-19 pneumonia with varying clinical manifestations and causes of death. All had been subjected to chest CT imaging 24-72 h prior to death. Twenty-seven lung areas with typical COVID-19 patterns and two radiologically unaffected pulmonary areas were correlated to histopathological findings in the same lung regions. RESULTS Two dominant radiological patterns were observed: ground-glass opacity (GGO) (n = 11) and consolidation (n = 16). In seven of 11 sampled areas of GGO, diffuse alveolar damage (DAD) was observed. In four areas of GGO, the histological pattern was vascular damage and thrombosis, with (n = 2) or without DAD (n = 2). DAD was also observed in five of 16 samples derived from areas of radiological consolidation. Seven areas of consolidation were based on a combination of DAD, vascular damage and thrombosis. In four areas of consolidation, bronchopneumonia was found. Unexpectedly, in samples from radiologically unaffected lung parenchyma, evidence was found of vascular damage and thrombosis. CONCLUSION In COVID-19, radiological findings of GGO and consolidation are mostly explained by DAD or a combination of DAD and vascular damage plus thrombosis. However, the different typical CT patterns in COVID-19 are not related to specific histopathological patterns. Microvascular damage and thrombosis are even encountered in the radiologically normal lung.
Collapse
Affiliation(s)
- Azar Kianzad
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Lilian J Meijboom
- Department of Radiology and Nuclear Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Esther J Nossent
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Eva Roos
- Department of Pathology, Cancer Centre Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Bernadette Schurink
- Department of Pathology, Cancer Centre Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Peter I Bonta
- Department of Pulmonary Medicine, Amsterdam UMC, AMC, Amsterdam, The Netherlands
| | - Inge A H van den Berk
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, AMC, Amsterdam, The Netherlands
| | - Rieneke Britstra
- Department of Pathology, Cancer Centre Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Jaap Stoker
- Department of Radiology and Nuclear Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Anton Vonk Noordegraaf
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Paul van der Valk
- Department of Pathology, Cancer Centre Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Erik Thunnissen
- Department of Pathology, Cancer Centre Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Marianna Bugiani
- Department of Pathology, Cancer Centre Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Harm Jan Bogaard
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Teodora Radonic
- Department of Pathology, Cancer Centre Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
225
|
Patel JC, Singh A, Tulswani R, Sharma YK, Khurana P, Ragumani S. Identification of VEGFA-centric temporal hypoxia-responsive dynamic cardiopulmonary network biomarkers. Life Sci 2021; 281:119718. [PMID: 34147483 DOI: 10.1016/j.lfs.2021.119718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/31/2021] [Accepted: 06/08/2021] [Indexed: 10/21/2022]
Abstract
AIMS Hypoxia, a pathophysiological condition, is profound in several cardiopulmonary diseases (CPD). Every individual's lethality to a hypoxia state differs in terms of hypoxia exposure time, dosage units and dependent on the individual's genetic makeup. Most of the proposed markers for CPD were generally aim to distinguish disease samples from normal samples. Although, as per the 2018 GOLD guidelines, clinically useful biomarkers for several cardio pulmonary disease patients in stable condition have yet to be identified. We attempt to address these key issues through the identification of Dynamic Network Biomarkers (DNB) to detect hypoxia induced early warning signals of CPD before the catastrophic deterioration. MATERIALS AND METHODS The human microvascular endothelial tissues microarray datasets (GSE11341) of lung and cardiac expose to hypoxia (1% O2) for 3, 24 and 48 h were retrieved from the public repository. The time dependent differentially expressed genes were subjected to tissue specificity and promoter analysis to filtrate the noise levels in the networks and to dissect the tissue specific hypoxia induced genes. These filtered out genes were used to construct the dynamic segmentation networks. The hypoxia induced dynamic differentially expressed genes were validated in the lung and heart tissues of male rats. These rats were exposed to hypobaric hypoxia (simulated altitude of 25,000 or PO2 - 282 mm of Hg) progressively for 3, 24 and 48 h. KEY FINDINGS To identify the temporal key genes regulated in hypoxia, we ranked the dominant genes based on their consolidated topological features from tissue specific networks, time dependent networks and dynamic networks. Overall topological ranking described VEGFA as a single node dynamic hub and strongly communicated with tissue specific genes to carry forward their tissue specific information. We named this type of VEGFAcentric dynamic networks as "V-DNBs". As a proof of principle, our methodology helped us to identify the V-DNBs specific for lung and cardiac tissues namely V-DNBL and V-DNBC respectively. SIGNIFICANCE Our experimental studies identified VEGFA, SLC2A3, ADM and ENO2 as the minimum and sufficient candidates of V-DNBL. The dynamic expression patterns could be readily exploited to capture the pre disease state of hypoxia induced pulmonary vascular remodelling. Whereas in V-DNBC the minimum and sufficient candidates are VEGFA, SCL2A3, ADM, NDRG1, ENO2 and BHLHE40. The time dependent single node expansion indicates V-DNBC could also be the pre disease state pathological hallmark for hypoxia-associated cardiovascular remodelling. The network cross-talk and expression pattern between V-DNBL and V-DNBC are completely distinct. On the other hand, the great clinical advantage of V-DNBs for pre disease predictions, a set of samples during the healthy condition should suffice. Future clinical studies might further shed light on the predictive power of V-DNBs as prognostic and diagnostic biomarkers for CPD.
Collapse
Affiliation(s)
- Jai Chand Patel
- Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, India
| | - Ajeet Singh
- Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, India
| | - Rajkumar Tulswani
- Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, India
| | - Yogendra Kumar Sharma
- Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, India
| | - Pankaj Khurana
- Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, India
| | - Sugadev Ragumani
- Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, India.
| |
Collapse
|
226
|
Guan CS, Lv ZB, Li JJ, Du YN, Chen H, Cui T, Guo N, Chen BD, Xie RM. CT appearances, patterns of progression, and follow-up of COVID-19: evaluation on thin-section CT. Insights Imaging 2021; 12:73. [PMID: 34110540 PMCID: PMC8190726 DOI: 10.1186/s13244-021-01019-0] [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: 02/18/2021] [Accepted: 05/20/2021] [Indexed: 12/17/2022] Open
Abstract
Background To retrospectively analyze CT appearances and progression pattern of COVID-19 during hospitalization, and analyze imaging findings of follow-up on thin-section CT. Methods CT findings of 69 patients with COVID-19 were evaluated on initial CT, peak CT, and pre-discharge CT. CT pattern were divided into four types on CT progression. Lesion percentage of pulmonary lobe (lobe score) was graded. Correlation analysis was made between scores and intervals. 53 patients were followed up by CT. Results Among 69 patients, 33.3% exhibited improvement pattern, 65.2% peak pattern, 1.5% deterioration pattern, and 0% fluctuation pattern. The lobe scores were positively correlated with most of intervals. It was more common to observe consolidation, pleural thickening and pleural effusion on the peak CT, and irregular line and reticulation on pre-discharge CT. The peak-initial interval were shortened when the initial CT with consolidation and pleural thickening. The intervals were extended when the irregular lines appeared on peak CT and reticulation on pre-discharge CT. Among 53 follow-up patients, 37.7% showed normal chest CT, and 62.3% showed viral pneumonia remained that mainly included GGO (100.0%) and irregular lines (33.3%). Conclusions COVID-19 displayed different appearances on CT as progressing. The peak pattern was the most common progression pattern. The CT appearances showed closely related to the intervals. The COVID-19 pneumonia can be remained or completely absorbed on CT with follow-up.
Collapse
Affiliation(s)
- Chun-Shuang Guan
- Department of Radiology, Beijing Ditan Hospital, Capital Medical University, No. 8 Jingshun East Street, Chaoyang District, Beijing, China
| | - Zhi-Bin Lv
- Department of Radiology, Beijing Ditan Hospital, Capital Medical University, No. 8 Jingshun East Street, Chaoyang District, Beijing, China
| | - Jing-Jing Li
- Department of Radiology, Beijing Ditan Hospital, Capital Medical University, No. 8 Jingshun East Street, Chaoyang District, Beijing, China
| | - Yan-Ni Du
- Department of Radiology, Beijing Ditan Hospital, Capital Medical University, No. 8 Jingshun East Street, Chaoyang District, Beijing, China
| | - Hui Chen
- Department of Radiology, Beijing Ditan Hospital, Capital Medical University, No. 8 Jingshun East Street, Chaoyang District, Beijing, China
| | - Tao Cui
- Department of Radiology, Beijing Ditan Hospital, Capital Medical University, No. 8 Jingshun East Street, Chaoyang District, Beijing, China
| | - Ning Guo
- Department of Clinical Research, Shukun (Beijing) Technology Co., Ltd., Jinhui Bd, Qiyang Rd, Chaoyang District, Beijing, China
| | - Bu-Dong Chen
- Department of Radiology, Beijing Ditan Hospital, Capital Medical University, No. 8 Jingshun East Street, Chaoyang District, Beijing, China.
| | - Ru-Ming Xie
- Department of Radiology, Beijing Ditan Hospital, Capital Medical University, No. 8 Jingshun East Street, Chaoyang District, Beijing, China.
| |
Collapse
|
227
|
Increased procoagulant platelet levels are predictive of death in COVID-19. GeroScience 2021; 43:2055-2065. [PMID: 34109507 PMCID: PMC8189550 DOI: 10.1007/s11357-021-00385-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 05/13/2021] [Indexed: 02/06/2023] Open
Abstract
Prior research has identified abnormal platelet procoagulant responses in COVID-19. Coated-platelets, a form of procoagulant platelets, support thrombin formation and are elevated in ischemic stroke patients with increased risk for recurrent infarction. Our goal was to examine changes in coated-platelet levels over the course of COVID-19 infection and determine their association with disease severity, thrombosis, and death. Coated-platelet levels were assayed after admission and repeated weekly in COVID-19 patients, and in COVID-19 negative controls. Receiver operator characteristic (ROC) analysis was used to calculate area under the curve (AUC) values for a model including baseline coated-platelets to predict death. Kaplan–Meier and Cox proportional hazards analysis was used to predict risk for death at 90 days. We enrolled 33 patients (22 with moderate and 11 with severe infection) and 20 controls. Baseline coated-platelet levels were lower among moderate (mean ± SD; 21.3 ± 9.8%) and severe COVID-19 patients (28.5 ± 11.9%) compared to controls (38.1 ± 10.4%, p < 0.0001). Coated-platelet levels increased during follow-up in COVID-19 patients by 7% (relative) per day from symptom onset (95% CI 2–12%, p = 0.007). A cut-off of 33.9% for coated-platelet levels yielded 80% sensitivity and 96% specificity for death at 90 days, with resulting AUC of 0.880 (95% CI 0.680–1.0, p = 0.0002). The adjusted hazard ratio for death in patients with coated-platelet levels > 33.9% was 40.99 when compared to those with levels ≤ 33.9% (p < 0.0001). Platelet procoagulant potential is transiently decreased in most patients during COVID-19; however, increased baseline platelet procoagulant levels predict death. Defining the mechanisms involved and potential links with aging may yield novel treatment targets.
Collapse
|
228
|
Shakaib B, Zohra T, Ikram A, Shakaib MB, Ali A, Bashir A, Salman M, Khan MA, Ansari J. A comprehensive review on clinical and mechanistic pathophysiological aspects of COVID-19 Malady: How far have we come? Virol J 2021; 18:120. [PMID: 34098986 PMCID: PMC8182739 DOI: 10.1186/s12985-021-01578-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 05/17/2021] [Indexed: 01/08/2023] Open
Abstract
Since its outbreak in 2019, the coronavirus disease (COVID-19) has become a pandemic, affecting more than 52 million people and causing more than 1 million mortalities globally till date. Current research reveals a wide array of disease manifestations and behaviors encompassing multiple organ systems in body and immense systemic inflammation, which have been summarized in this review. Data from a number of scientific reviews, research articles, case series, observational studies, and case reports were retrieved by utilizing online search engines such as Cochrane, PubMed, and Scopus from December 2019 to November 2020. The data for prevalence of signs and symptoms, underlying disease mechanisms and comorbidities were analyzed using SPSS version 25. This review will discuss a wide range of COVID-19 clinical presentations recorded till date, and the current understanding of both the underlying general as well as system specific pathophysiologic, and pathogenetic pathways. These include direct viral penetration into host cells through ACE2 receptors, induction of inflammosomes and immune response through viral proteins, and the initiation of system-wide inflammation and cytokine production. Moreover, peripheral organ damage and underlying comorbid diseases which can lead to short term and long term, reversible and irreversible damage to the body have also been studied. We concluded that underlying comorbidities and their pathological effects on the body contributed immensely and determine the resultant disease severity and mortality of the patients. Presently there is no drug approved for treatment of COVID-19, however multiple vaccines are now in use and research for more is underway.
Collapse
Affiliation(s)
- Baila Shakaib
- Rawal Institute of Health Sciences, Islamabad, Pakistan
| | | | - Aamer Ikram
- National Institute of Health, Islamabad, Pakistan
| | | | - Amna Ali
- National Institute of Health, Islamabad, Pakistan
| | - Adnan Bashir
- National Institute of Health, Islamabad, Pakistan
| | | | | | - Jamil Ansari
- National Institute of Health, Islamabad, Pakistan
| |
Collapse
|
229
|
Intestinal Abnormalities in Patients With SARS-CoV-2 Infection: Histopathologic Changes Reflect Mechanisms of Disease. Am J Surg Pathol 2021; 46:89-96. [PMID: 34081038 DOI: 10.1097/pas.0000000000001755] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Approximately 20% of patients with symptomatic syndrome-associated coronavirus-2 (SARS-CoV-2) infection have gastrointestinal bleeding and/or diarrhea. Most are managed without endoscopic evaluation because the risk of practitioner infection outweighs the value of biopsy analysis unless symptoms are life-threatening. As a result, much of what is known about the gastrointestinal manifestations of coronavirus disease-2019 (COVID-19) has been gleaned from surgical and autopsy cases that suffer from extensive ischemic injury and/or poor preservation. There are no detailed reports describing any other gastrointestinal effects of SARS-CoV-2 even though >3,000,000 people have died from COVID-19 worldwide. The purpose of this study is to report the intestinal findings related to SARS-CoV-2 infection by way of a small case series including one with evidence of direct viral cytopathic effect and 2 with secondary injury attributed to viral infection. Infection can be confirmed by immunohistochemical stains directed against SARS-CoV-2 spike protein, in situ hybridization for spike protein-encoding RNA, and ultrastructural visualization of viruses within the epithelium. It induces cytoplasmic blebs and tufted epithelial cells without inflammation and may not cause symptoms. In contrast, SARS-CoV-2 infection can cause gastrointestinal symptoms after the virus is no longer detected, reflecting systemic activation of cytokine and complement cascades rather than direct viral injury. Reversible mucosal ischemia features microvascular injury with hemorrhage, small vessel thrombosis, and platelet-rich thrombi. Systemic cytokine elaboration and dysbiosis likely explain epithelial cell injury that accompanies diarrheal symptoms. These observations are consistent with clinical and in vitro data and contribute to our understanding of the protean manifestations of COVID-19.
Collapse
|
230
|
Abstract
The body of a deceased with Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection is considered infectious. In this study, we present the results of infectivity testing of the body and testing of mortuary staff for SARS-CoV-2. We performed real-time quantitative polymerase chain reaction (RT-qPCR) for SARS-CoV-2 on 33 decedents with ante mortem confirmed SARS-CoV-2 infection. Swabs of the body surface from five different body regions and from the body bag or coffin were examined. A subset of the swabs was brought into cell culture. In addition, screening of 25 Institute of Legal Medicine (ILM) personnel for ongoing or past SARS-CoV-2 infection was performed at two different time points during the pandemic. Swabs from all locations of the body surface and the body environment were negative in cases of negative post mortem nasopharyngeal testing (n=9). When the post mortem nasopharyngeal swab tested positive (n=24), between 0 and 5 of the body surface swabs were also positive, primarily the perioral region. In six of the cases, the body bag also yielded a positive result. The longest postmortem interval with positive SARS-CoV-2 RT-qPCR at the body surface was nine days. In no case viable SARS-CoV-2 was found on the skin of the bodies or the body bags. One employee (autopsy technician) had possible occupational infection with SARS-CoV-2; all other employees were tested negative for SARS-CoV-2 RNA or antibody twice. Our data indicate that with adequate management of general safety precautions, transmission of SARS-CoV-2 through autopsies and handling of bodies is unlikely.
Collapse
|
231
|
Maiese A, Manetti AC, La Russa R, Di Paolo M, Turillazzi E, Frati P, Fineschi V. Autopsy findings in COVID-19-related deaths: a literature review. Forensic Sci Med Pathol 2021; 17:279-296. [PMID: 33026628 PMCID: PMC7538370 DOI: 10.1007/s12024-020-00310-8] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2020] [Indexed: 02/07/2023]
Abstract
Although many clinical reports have been published, little is known about the pathological post-mortem findings from people who have died of the novel coronavirus disease. The need for postmortem information is urgent to improve patient management of mild and severe illness, and treatment strategies. The present systematic review was carried out according to the Preferred Reporting Items for Systematic Review (PRISMA) standards. A systematic literature search and a critical review of the collected studies were conducted. An electronic search of PubMed, Science Direct Scopus, Google Scholar, and Excerpta Medica Database (EMBASE) from database inception to June 2020 was performed. We found 28 scientific papers; the total amount of cases is 341. The major histological feature in the lung is diffuse alveolar damage with hyaline membrane formation, alongside microthrombi in small pulmonary vessels. It appears that there is a high incidence of deep vein thrombosis and pulmonary embolism among COVID-19 decedents, suggesting endothelial involvement, but more studies are needed. A uniform COVID-19 post-mortem diagnostic protocol has not yet been developed. In a time in which international collaboration is essential, standardized diagnostic criteria are fundamental requirements.
Collapse
Affiliation(s)
- Aniello Maiese
- Department of Surgical Pathology, Medical, Molecular and Critical Area, Institute of Legal Medicine, University of Pisa, 56126, Pisa, PI, Italy
| | - Alice Chiara Manetti
- Department of Surgical Pathology, Medical, Molecular and Critical Area, Institute of Legal Medicine, University of Pisa, 56126, Pisa, PI, Italy
| | - Raffaele La Russa
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00161, Rome, RM, Italy
| | - Marco Di Paolo
- Department of Surgical Pathology, Medical, Molecular and Critical Area, Institute of Legal Medicine, University of Pisa, 56126, Pisa, PI, Italy
| | - Emanuela Turillazzi
- Department of Surgical Pathology, Medical, Molecular and Critical Area, Institute of Legal Medicine, University of Pisa, 56126, Pisa, PI, Italy
| | - Paola Frati
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00161, Rome, RM, Italy
| | - Vittorio Fineschi
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00161, Rome, RM, Italy.
| |
Collapse
|
232
|
Rosenberg HF, Foster PS. Eosinophils and COVID-19: diagnosis, prognosis, and vaccination strategies. Semin Immunopathol 2021; 43:383-392. [PMID: 33728484 PMCID: PMC7962927 DOI: 10.1007/s00281-021-00850-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/02/2021] [Indexed: 02/07/2023]
Abstract
The unprecedented impact of the coronavirus disease 2019 (COVID-19) pandemic has resulted in global challenges to our health-care systems and our economic security. As such, there has been significant research into all aspects of the disease, including diagnostic biomarkers, associated risk factors, and strategies that might be used for its treatment and prevention. Toward this end, eosinopenia has been identified as one of many factors that might facilitate the diagnosis and prognosis of severe COVID-19. However, this finding is neither definitive nor pathognomonic for COVID-19. While eosinophil-associated conditions have been misdiagnosed as COVID-19 and others are among its reported complications, patients with pre-existing eosinophil-associated disorders (e.g., asthma, eosinophilic gastrointestinal disorders) do not appear to be at increased risk for severe disease; interestingly, several recent studies suggest that a diagnosis of asthma may be associated with some degree of protection. Finally, although vaccine-associated aberrant inflammatory responses, including eosinophil accumulation in the respiratory tract, were observed in preclinical immunization studies targeting the related SARS-CoV and MERS-CoV pathogens, no similar complications have been reported clinically in response to the widespread dissemination of either of the two encapsulated mRNA-based vaccines for COVID-19.
Collapse
Affiliation(s)
- Helene F Rosenberg
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Paul S Foster
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle and Hunter Medical Research Institute (HMRI), New Lambton Heights, New South Wales, 2300, Australia
| |
Collapse
|
233
|
Gholami M, Safari S, Ulloa L, Motaghinejad M. Neuropathies and neurological dysfunction induced by coronaviruses. J Neurovirol 2021; 27:380-396. [PMID: 33983506 PMCID: PMC8117458 DOI: 10.1007/s13365-021-00977-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 01/23/2021] [Accepted: 04/05/2021] [Indexed: 02/03/2023]
Abstract
During the recent years, viral epidemic due to coronaviruses, such as SARS (Severe Acute Respiratory Syndrome), Middle East Respiratory Coronavirus Syndrome (MERS), and COVID-19 (coronavirus disese-19), has become a global problem. In addition to causing cardiovascular and respiratory lethal dysfunction, these viruses can cause neurodegeneration leading to neurological disorders. Review of the current scientific literature reveals the multiple neuropathies and neuronal dysfunction associated with these viruses. Here, we review the major findings of these studies and discuss the main neurological sequels and outcomes of coronavirus infections with SARS, MERS, and COVID-19. This article analyzes and discusses the main mechanisms of coronavirus-induced neurodegeneration according to the current experimental and clinical studies. Coronaviruses can damage the nerves directly through endovascular dysfunctions thereby affecting nerve structures and synaptic connections. Coronaviruses can also induce neural cell degeneration indirectly via mitochondrial dysfunction inducing oxidative stress, inflammation, and apoptosis. Thus, coronaviruses can cause neurological disorders by inducing neurovascular dysfunction affecting nerve structures and synaptic connections, and by inducing inflammation, oxidative stress, and apoptosis. While some of these mechanisms are similar to other RNA viruses, the neurotoxic mechanisms of COVID-19, MERS, and SARS-CoV viruses are unknown and need detailed clinical and experimental studies.
Collapse
Affiliation(s)
- Mina Gholami
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Safari
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Luis Ulloa
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University, NC, 27710, Durham, USA.
| | - Majid Motaghinejad
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
234
|
Nolte KB, Muller TB, Denmark AM, Burstein R, Villalobos YA. Design and Construction of a Biosafety Level 3 Autopsy Laboratory. Arch Pathol Lab Med 2021; 145:407-414. [PMID: 33307551 DOI: 10.5858/arpa.2020-0644-sa] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2020] [Indexed: 11/06/2022]
Abstract
CONTEXT.— Autopsy pathologists, including medical examiners, provide valuable public health support for infectious disease deaths through surveillance for deaths of public health concern including emerging infections, identifying causative organisms for unexplained deaths, and providing insights into the pathology and pathogenesis of novel or unusual infections. However, autopsy poses biosafety risks to workers within and outside the laboratory. The highest rates of laboratory-acquired infections occur in autopsy workers. OBJECTIVE.— To design and construct an appropriately biosafe autopsy laboratory. DESIGN.— We conducted a biosafety risk assessment for autopsy workers using the process developed by the US Centers for Disease Control and Prevention and National Institutes of Health and applied these findings as the basis of laboratory design and construction. RESULTS.— Autopsy workers are unpredictably exposed to a variety of infectious organisms, including hepatitis C virus, HIV, and Mycobacterium tuberculosis. Hazardous autopsy procedures include using and encountering sharp objects and the generation of aerosols from dissection, fluid aspiration, rinsing tissues, and dividing bone with an oscillating saw. CONCLUSIONS.— Exposure to blood-borne and airborne pathogens from procedures that can cause cutaneous inoculation and inhalation of aerosols indicates that human autopsies should be performed at biosafety level 3. We designed a large, entirely biosafety level 3 medical examiner autopsy laboratory using design principles and characteristics that can be scaled to accommodate smaller academic or other hospital-based autopsy spaces. Containment was achieved through a concentric ring design, with access control at interface zones. As new autopsy laboratories are planned, we strongly recommend that they be designed to function uniformly at biosafety level 3.
Collapse
Affiliation(s)
- Kurt B Nolte
- From the Office of the Medical Investigator and Departments of Pathology and Radiology (Nolte [https://orcid.org/0000-0003-0257-6284]), University of New Mexico Health Science Center, Albuquerque
| | - Timothy B Muller
- The Office of Research (Muller), University of New Mexico Health Science Center, Albuquerque
| | - Adam M Denmark
- The Department of Science and Technology, SmithGroup, Phoenix, Arizona (Denmark)
| | - Ron Burstein
- Studio Southwest Architects, Inc, Albuquerque, New Mexico (Burstein)
| | - Yvonne A Villalobos
- The Office of the Medical Investigator (Villalobos), University of New Mexico Health Science Center, Albuquerque
| |
Collapse
|
235
|
Archambault AS, Zaid Y, Rakotoarivelo V, Turcotte C, Doré É, Dubuc I, Martin C, Flamand O, Amar Y, Cheikh A, Fares H, El Hassani A, Tijani Y, Côté A, Laviolette M, Boilard É, Flamand L, Flamand N. High levels of eicosanoids and docosanoids in the lungs of intubated COVID-19 patients. FASEB J 2021; 35:e21666. [PMID: 34033145 PMCID: PMC8206770 DOI: 10.1096/fj.202100540r] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/29/2021] [Accepted: 04/30/2021] [Indexed: 12/13/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 is responsible for coronavirus disease 2019 (COVID-19). While COVID-19 is often benign, a subset of patients develops severe multilobar pneumonia that can progress to an acute respiratory distress syndrome. There is no cure for severe COVID-19 and few treatments significantly improved clinical outcome. Dexamethasone and possibly aspirin, which directly/indirectly target the biosynthesis/effects of numerous lipid mediators are among those options. Our objective was to define if severe COVID-19 patients were characterized by increased bioactive lipids modulating lung inflammation. A targeted lipidomic analysis of bronchoalveolar lavages (BALs) by tandem mass spectrometry was done on 25 healthy controls and 33 COVID-19 patients requiring mechanical ventilation. BALs from severe COVID-19 patients were characterized by increased fatty acids and inflammatory lipid mediators. There was a predominance of thromboxane and prostaglandins. Leukotrienes were also increased, notably LTB4 , LTE4 , and eoxin E4 . Monohydroxylated 15-lipoxygenase metabolites derived from linoleate, arachidonate, eicosapentaenoate, and docosahexaenoate were also increased. Finally yet importantly, specialized pro-resolving mediators, notably lipoxin A4 and the D-series resolvins, were also increased, underscoring that the lipid mediator storm occurring in severe COVID-19 involves pro- and anti-inflammatory lipids. Our data unmask the lipid mediator storm occurring in the lungs of patients afflicted with severe COVID-19. We discuss which clinically available drugs could be helpful at modulating the lipidome we observed in the hope of minimizing the deleterious effects of pro-inflammatory lipids and enhancing the effects of anti-inflammatory and/or pro-resolving lipid mediators.
Collapse
Affiliation(s)
- Anne-Sophie Archambault
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Faculté de médecine, Département de médecine, Université Laval, Québec, QC, Canada.,Canada Excellence Research Chair in the Microbiome-Endocannabinoidome Axis in Metabolic Health, Université Laval, Québec, QC, Canada
| | - Younes Zaid
- Biology Department, Faculty of Sciences, Mohammed V University, Rabat, Morocco.,Cheikh Zaïd Hospital, Abulcasis University of Health Sciences, Rabat, Morocco
| | - Volatiana Rakotoarivelo
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Faculté de médecine, Département de médecine, Université Laval, Québec, QC, Canada.,Canada Excellence Research Chair in the Microbiome-Endocannabinoidome Axis in Metabolic Health, Université Laval, Québec, QC, Canada
| | - Caroline Turcotte
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Faculté de médecine, Département de médecine, Université Laval, Québec, QC, Canada.,Canada Excellence Research Chair in the Microbiome-Endocannabinoidome Axis in Metabolic Health, Université Laval, Québec, QC, Canada
| | - Étienne Doré
- Centre de Recherche du Centre Hospitalier, Universitaire de Québec-Université Laval, Québec, QC, Canada.,Centre de Recherche Arthrite, Université Laval, Québec, QC, Canada
| | - Isabelle Dubuc
- Centre de Recherche du Centre Hospitalier, Universitaire de Québec-Université Laval, Québec, QC, Canada
| | - Cyril Martin
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Faculté de médecine, Département de médecine, Université Laval, Québec, QC, Canada.,Canada Excellence Research Chair in the Microbiome-Endocannabinoidome Axis in Metabolic Health, Université Laval, Québec, QC, Canada
| | - Olivier Flamand
- Centre de Recherche du Centre Hospitalier, Universitaire de Québec-Université Laval, Québec, QC, Canada
| | - Youssef Amar
- Moroccan Foundation for Advanced Science, Innovation & Research (MAScIR), Rabat, Morocco
| | - Amine Cheikh
- Cheikh Zaïd Hospital, Abulcasis University of Health Sciences, Rabat, Morocco
| | - Hakima Fares
- Cheikh Zaïd Hospital, Abulcasis University of Health Sciences, Rabat, Morocco
| | - Amine El Hassani
- Cheikh Zaïd Hospital, Abulcasis University of Health Sciences, Rabat, Morocco
| | - Youssef Tijani
- Faculty of Medicine, Mohammed VI University of Health Sciences, Casablanca, Morocco
| | - Andréanne Côté
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Faculté de médecine, Département de médecine, Université Laval, Québec, QC, Canada
| | - Michel Laviolette
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Faculté de médecine, Département de médecine, Université Laval, Québec, QC, Canada
| | - Éric Boilard
- Centre de Recherche du Centre Hospitalier, Universitaire de Québec-Université Laval, Québec, QC, Canada.,Centre de Recherche Arthrite, Université Laval, Québec, QC, Canada.,Département de Microbiologie-Infectiologie et d'immunologie, Université Laval, Québec, QC, Canada
| | - Louis Flamand
- Centre de Recherche du Centre Hospitalier, Universitaire de Québec-Université Laval, Québec, QC, Canada.,Département de Microbiologie-Infectiologie et d'immunologie, Université Laval, Québec, QC, Canada
| | - Nicolas Flamand
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Faculté de médecine, Département de médecine, Université Laval, Québec, QC, Canada.,Canada Excellence Research Chair in the Microbiome-Endocannabinoidome Axis in Metabolic Health, Université Laval, Québec, QC, Canada
| |
Collapse
|
236
|
Wang M, Fan Y, Chai Y, Cheng W, Wang K, Cao J, Hu X. Association of Clinical and Immunological Characteristics With Disease Severity and Outcomes in 211 Patients With COVID-19 in Wuhan, China. Front Cell Infect Microbiol 2021; 11:667487. [PMID: 34123873 PMCID: PMC8195246 DOI: 10.3389/fcimb.2021.667487] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 05/10/2021] [Indexed: 01/08/2023] Open
Abstract
Background Coronavirus disease 2019 (COVID-19) has posed a great threat to global public health. There remains an urgent need to address the clinical significance of laboratory finding changes in predicting disease progression in COVID-19 patients. We aimed to analyze the clinical and immunological features of severe and critically severe patients with COVID-19 in comparison with non-severe patients and identify risk factors for disease severity and clinical outcome in COVID-19 patients. Methods The consecutive records of 211 patients with COVID-19 who were admitted to Zhongnan Hospital of Wuhan University from December 2019 to February 2020 were retrospectively reviewed. Results Of the 211 patients with COVID-19 recruited, 111 patients were classified as non-severe, 59 as severe, and 41 as critically severe cases. The median age was obviously higher in severe and critically severe cases than in non-severe cases. Severe and critically severe patients showed more underlying comorbidities than non-severe patients. Fever was the predominant presenting symptom in COVID-19 patients, and the duration of fever was longer in critically severe patients. Moreover, patients with increased levels of serum aminotransferases and creatinine (CREA) were at a higher risk for severe and critical COVID-19 presentations. The serum levels of IL-6 in severe and critically severe patients were remarkably higher than in non-severe patients. Lymphopenia was more pronounced in severe and critically severe patients compared with non-severe patients. Lymphocyte subset analysis indicated that severe and critically severe patients had significantly decreased count of lymphocyte subpopulations, such as CD4+ T cells, CD8+ T cells and B cells. A multivariate logistic analysis indicated that older age, male sex, the length of hospital stay, body temperature before admission, comorbidities, higher white blood cell (WBC) counts, lower lymphocyte counts, and increased levels of IL-6 were significantly associated with predicting the progression to severe stage of COVID-19. Conclusion Older age, male sex, underlying illness, sustained fever status, abnormal liver and renal functions, excessive expression of IL-6, lymphopenia, and selective loss of peripheral lymphocyte subsets were related to disease deterioration and clinical outcome in COVID-19 patients. This study would provide clinicians with valuable information for risk evaluation and effective interventions for COVID-19.
Collapse
Affiliation(s)
- Man Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Yongzhen Fan
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, China
| | - Yuqiong Chai
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, China
| | - Wenlin Cheng
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, China
| | - Kun Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Jianlei Cao
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, China
| | - Xiaorong Hu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, China
| |
Collapse
|
237
|
De-Giorgio F, Cittadini F, Cina A, Cavarretta E, Biondi-Zoccai G, Vetrugno G, Natale L, Colosimo C, Pascali VL. Use of post-mortem chest computed tomography in Covid-19 pneumonia. Forensic Sci Int 2021; 325:110851. [PMID: 34090259 PMCID: PMC8154189 DOI: 10.1016/j.forsciint.2021.110851] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/12/2021] [Accepted: 05/19/2021] [Indexed: 02/05/2023]
Abstract
Background and aim COVID-19 is an extremely challenging disease, both from a clinical and forensic point of view, and performing autopsies of COVID-19 deceased requires adequately equipped sectorial rooms and exposes health professionals to the risk of contagion. Among one of the categories that are most affected by SARS-Cov-2 infection are the elderly residents. Despite the need for prompt diagnoses, which are essential to implement all isolation measures necessary to contain the infection spread, deceased subjects in long-term care facilities are still are often diagnosed post-mortem. In this context, our study focuses on the use of post-mortem computed tomography for the diagnosis of COVID-19 infection, in conjunction with post-mortem swabs. The aim of this study was to assess the usefulness of post-mortem whole CT-scanning in identifying COVID-19 pneumonia as a cause of death, by comparing chest CT-findings of confirmed COVID-19 fatalities to control cases. Materials and methods The study included 24 deceased subjects: 13 subjects coming from long-term care facility and 11 subjects died at home. Whole body CT scans were performed within 48 h from death in all subjects to evaluate the presence and distribution of pulmonary abnormalities typical of COVID-19-pneumonia, including: ground-glass opacities (GGO), consolidation, and pleural effusion to confirm the post-mortem diagnosis. Results Whole-body CT scans was feasible and allowed a complete diagnosis in all subjects. In 9 (69%) of the 13 cases from long-term care facility the cause of death was severe COVID 19 pneumonia, while GGO were present in 100% of the study population. Conclusion In the context of rapidly escalating COVID-19 outbreaks, given that laboratory tests for the novel coronavirus is time-consuming and can be falsely negative, the post-mortem CT can be considered as a reliable and safe modality to confirm COVID-19 pneumonia. This is especially true for specific postmortem chest CT-findings that are rather characteristic of COVID-19 fatalities.
Collapse
Affiliation(s)
- Fabio De-Giorgio
- Department of Health Care Surveillance and Bioethics, Section of Legal Medicine, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| | - Francesca Cittadini
- Department of Health Care Surveillance and Bioethics, Section of Legal Medicine, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Alessandro Cina
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy; Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Diagnostic Imaging Area, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Elena Cavarretta
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy; Mediterranea Cardiocentro, Napoli, Italy
| | - Giuseppe Biondi-Zoccai
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy; Mediterranea Cardiocentro, Napoli, Italy
| | - Giuseppe Vetrugno
- Department of Health Care Surveillance and Bioethics, Section of Legal Medicine, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Luigi Natale
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy; Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Diagnostic Imaging Area, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Cesare Colosimo
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy; Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Diagnostic Imaging Area, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Vincenzo L Pascali
- Department of Health Care Surveillance and Bioethics, Section of Legal Medicine, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
238
|
He Y, Wang J, Ren J, Zhao Y, Chen J, Chen X. Effect of COVID-19 on Male Reproductive System - A Systematic Review. Front Endocrinol (Lausanne) 2021; 12:677701. [PMID: 34122351 PMCID: PMC8190708 DOI: 10.3389/fendo.2021.677701] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 04/27/2021] [Indexed: 12/30/2022] Open
Abstract
Background Angiotensin-converting enzyme II (ACE2), a receptor for Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) to enter host cells, is widely expressed in testes and prostate tissues. The testis and prostate produce semen. At present, there are contradictory reports about whether SARS-CoV-2 can exist in the semen of infected men. Objective To provide a comprehensive overview of the topic of whether COVID-19 can impact on male reproductive system. Methods We reviewed the relevant publications on the possible impact of Coronavirus Disease 2019 (COVID-19) on male reproductive system and summarized the latest and most important research results so far. Literature published in English from December 2019 to January 31, 2021 regarding the existence of SARS-CoV-2 in semen, testis, and prostatic fluid and the effects of COVID-19 on male reproductive were included. Results We identified 28 related studies, only one of which reported the presence of SARS-CoV-2 in semen. The study found that the semen quality of patients with moderate infection was lower than that of patients with mild infection and healthy controls. The impaired semen quality may be related to fever and inflammation. Pathological analysis of the testis/epididymis showed that SARS-CoV-2 viral particles were positive in 10 testicular samples, and the spermatogenic function of the testis was impaired. All 94 expressed prostatic secretion (EPS) samples were negative for SARS-CoV-2 RNA. Conclusion The likelihood of SARS-CoV-2 in the semen of COVID-19 patients is very small, and semen should rarely be regarded as a carrier of SARS-CoV-2 genetic material. However, COVID-19 may cause testicular spermatogenic dysfunction via immune or inflammatory reactions. Long-term follow-up is needed for COVID-19 male patients and fetuses conceived during the father's infection period.
Collapse
Affiliation(s)
- Yanfei He
- Health Management Center, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jie Wang
- Health Management Center, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Junlin Ren
- Department of Infection Control, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yubo Zhao
- Department of Urology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jing Chen
- Cadre Clinic of the Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xuejiao Chen
- Scientific Research and Training Office, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
239
|
Sridhar S, Nicholls J. Pathophysiology of infection with SARS-CoV-2-What is known and what remains a mystery. Respirology 2021; 26:652-665. [PMID: 34041821 PMCID: PMC8242464 DOI: 10.1111/resp.14091] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/03/2021] [Indexed: 12/12/2022]
Abstract
Coronavirus disease 2019 (COVID‐19), caused by coronavirus severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2), has caused extensive disruption and mortality since its recent emergence. Concomitantly, there has been a race to understand the virus and its pathophysiology. The clinical manifestations of COVID‐19 are manifold and not restricted to the respiratory tract. Extrapulmonary manifestations involving the gastrointestinal tract, hepatobiliary system, cardiovascular and renal systems have been widely reported. However, the pathophysiology of many of these manifestations is controversial with questionable support for direct viral invasion and an abundance of alternative explanations such as pre‐existing medical conditions and critical illness. Prior research on SARS‐Co‐V and NL63 was rapidly leveraged to identify angiotensin‐converting enzyme 2 (ACE2) receptor as the key cell surface receptor for SARS‐CoV‐2. The distribution of ACE2 has been used as a starting point for estimating vulnerability of various tissue types to SARS‐CoV‐2 infection. Sophisticated organoid and animal models have been used to demonstrate such infectivity of extrapulmonary tissues in vitro, but the clinical relevance of these findings remains uncertain. Clinical autopsy studies are typically small and inevitably biased towards patients with severe COVID‐19 and prolonged hospitalization. Technical issues such as delay between time of death and autopsy, use of inappropriate antibodies for paraffin‐embedded tissue sections and misinterpretation of cellular structures as virus particles on electron micrograph images are additional problems encountered in the extant literature. Given that SARS‐CoV‐2 is likely to circulate permanently in human populations, there is no doubt that further work is required to clarify the pathobiology of COVID‐19.
Collapse
Affiliation(s)
- Siddharth Sridhar
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - John Nicholls
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| |
Collapse
|
240
|
Savchenko SV, Gritsinger VA, Tikhonov VV, Lamanov AN, Novoselov VP, Koshlyak DA. [Clinical and anatomical analysis of mortality from COVID-19 in sudden death and in those who died in health care facilities]. Sud Med Ekspert 2021; 64:5-10. [PMID: 34013687 DOI: 10.17116/sudmed2021640315] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Assessment of pathological and morphological changes in those who died from COVID-19 including persons received therapy in medical and preventive institutions (LPI) and who died suddenly from this pathology at home. The analysis data of the pathological and anatomical changes in 57 deaths from COVID-19 in hospitals and 74 forensic medical examinations where infectious pathology was established as the main cause of death are presented. For microscopy the sections were stained with hemotoxylin and eosin, OCG, immunohistochemical study with markers for CD3, CD 4, CD 20, SK-7. The mixed viral and bacterial lesions of the lungs were detected more often than pure viral infection in those who died suddenly from COVID-19 compared with people whose death occurred in medical facilities. This allows speaking about the lack of adequate antibiotic therapy out-patiently. Features of mononuclear lung infiltration in COVID-19 with a predominance of a moderately pronounced reaction of T-lymphocytes and a mild B-lymphocytic reaction indicate a decrease in immunological reactivity. Conducting clinical and anatomical analysis allows determining the features of pathogenesis and morphogenesis in each specific fatal case and informing the clinicians of health facilities (clinics and hospitals) allows the autopsy doctor (pathologist, forensic physician) to provide significant assistance in improving the quality of diagnosis and treatment of patients with this highly contagious severe viral disease.
Collapse
Affiliation(s)
- S V Savchenko
- Novosibirsk State Medical University, Novosibirsk, Russia
| | - V A Gritsinger
- Novosibirsk Regional Clinical Bureau of Forensic Medical Expertise, Novosibirsk, Russia
| | - V V Tikhonov
- Novosibirsk Regional Clinical Bureau of Forensic Medical Expertise, Novosibirsk, Russia
| | - A N Lamanov
- Novosibirsk Regional Clinical Bureau of Forensic Medical Expertise, Novosibirsk, Russia
| | - V P Novoselov
- Novosibirsk State Medical University, Novosibirsk, Russia
| | - D A Koshlyak
- Novosibirsk Regional Clinical Bureau of Forensic Medical Expertise, Novosibirsk, Russia
| |
Collapse
|
241
|
Emerging spectrum of COVID-19-related cardiopulmonary pathology in adults. ACTA ACUST UNITED AC 2021; 27:317-324. [PMID: 34031637 PMCID: PMC8133386 DOI: 10.1016/j.mpdhp.2021.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
COVID-19 is currently a major cause of morbidity and mortality in adults throughout the world. Given the high infection rate, it is increasingly likely that histopathologists will encounter this disease during their practice. Although COVID-19 is increasingly recognized as a multi-system disease, the lungs and, to a lesser degree, the heart remain the major sites of pathology. This article aims to acquaint the general histopathologist with the main pathological findings in the lungs and heart of adults with COVID-19. It highlights the need for clinicopathological correlation with a discussion of the cardiopulmonary clinical features in COVID-19 and relates those to the pathological findings. In the lungs, diffuse alveolar damage is emphasized with its variety of morphological appearances over time. It concludes with a discussion of the main techniques available to identify the virus in fixed tissues and their potential limitations related specifically to the heart and lungs.
Collapse
|
242
|
Lowery SA, Sariol A, Perlman S. Innate immune and inflammatory responses to SARS-CoV-2: Implications for COVID-19. Cell Host Microbe 2021; 29:1052-1062. [PMID: 34022154 PMCID: PMC8126603 DOI: 10.1016/j.chom.2021.05.004] [Citation(s) in RCA: 190] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
COVID-19 can result in severe disease characterized by significant immunopathology that is spurred by an exuberant, yet dysregulated, innate immune response with a poor adaptive response. A limited and delayed interferon I (IFN-I) and IFN-III response results in exacerbated proinflammatory cytokine production and in extensive cellular infiltrates in the respiratory tract, resulting in lung pathology. The development of effective therapeutics for patients with severe COVID-19 depends on our understanding of the pathological elements of this unbalanced innate immune response. Here, we review the mechanisms by which SARS-CoV-2 both activates and antagonizes the IFN and inflammatory response following infection, how a dysregulated cytokine and cellular response contributes to immune-mediated pathology in COVID-19, and therapeutic strategies that target elements of the innate response.
Collapse
Affiliation(s)
- Shea A Lowery
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - Alan Sariol
- Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - Stanley Perlman
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA; Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
243
|
Pannone G, Caponio VCA, De Stefano IS, Ramunno MA, Meccariello M, Agostinone A, Pedicillo MC, Troiano G, Zhurakivska K, Cassano T, Bizzoca ME, Papagerakis S, Buonaguro FM, Advani S, Muzio LL. Lung histopathological findings in COVID-19 disease - a systematic review. Infect Agent Cancer 2021; 16:34. [PMID: 34001199 PMCID: PMC8127295 DOI: 10.1186/s13027-021-00369-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/23/2021] [Indexed: 02/08/2023] Open
Abstract
Since December 2019, the global burden of the COVID-19 pandemic has increased rapidly and has impacted nearly every country in the world, affecting those who are elderly or with underlying comorbidities or immunocompromised states. Aim of this systematic review is to summarize lung histopathological characteristics of COVID-19, not only for diagnostic purpose but also to evaluate changes that can reflect pathophysiological pathways that can inform clinicians of useful treatment strategies. We identified following histopathological changes among our patients:: hyaline membranes; endothelial cells/ interstitial cells involvement; alveolar cells, type I pneumocytes/ type II pneumocytes involvement; interstitial and/ or alveolar edema; evidence of hemorrhage, of inflammatory cells, evidence of microthrombi; evidence of fibrin deposition and of viral infection in the tissue samples.The scenario with proliferative cell desquamation is typical of Acute Respiratory Distress Syndrome (ARDS) that can be classified as diffuse alveolar damage (DAD) and not DAD-ARDS. The proposed pathological mechanism concerns the role of both innate and adaptive components of the immune system. COVID-19 lethal cases present themselves as a heterogeneous disease, characterized by the different simultaneous presence of different histological findings, which reflect histological phases with corresponding different pathological pathways (epithelial, vascular and fibrotic changes), in the same patient.
Collapse
Affiliation(s)
- Giuseppe Pannone
- Anatomic Pathology Unit, Department of Clinic and Experimental Medicine, University of Foggia, 71122, Foggia, Italy
| | | | - Ilenia Sara De Stefano
- Anatomic Pathology Unit, Department of Clinic and Experimental Medicine, University of Foggia, 71122, Foggia, Italy
| | - Maria Antonietta Ramunno
- Anatomic Pathology Unit, Department of Clinic and Experimental Medicine, University of Foggia, 71122, Foggia, Italy
| | - Mario Meccariello
- Anatomic Pathology Unit, Department of Clinic and Experimental Medicine, University of Foggia, 71122, Foggia, Italy
| | - Alessio Agostinone
- Anatomic Pathology Unit, Department of Clinic and Experimental Medicine, University of Foggia, 71122, Foggia, Italy
| | - Maria Carmela Pedicillo
- Anatomic Pathology Unit, Department of Clinic and Experimental Medicine, University of Foggia, 71122, Foggia, Italy
| | - Giuseppe Troiano
- Department of Clinic and Experimental Medicine, University of Foggia, 71122, Foggia, Italy
| | - Khrystyna Zhurakivska
- Department of Clinic and Experimental Medicine, University of Foggia, 71122, Foggia, Italy
| | - Tommaso Cassano
- Department of Clinic and Experimental Medicine, University of Foggia, 71122, Foggia, Italy
| | - Maria Eleonora Bizzoca
- Department of Clinic and Experimental Medicine, University of Foggia, 71122, Foggia, Italy
| | - Silvana Papagerakis
- Department of Surgery, College of Medicine, Health Sciences Center, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| | - Franco Maria Buonaguro
- Molecular Biology and Viral Oncology Unit Istituto Nazionale, Tumori IRCCS "Fondazione Pascale", 80131, Naples, Italy
| | - Shailesh Advani
- Georgetown University School of Medicine, Georgetown University, Washington, DC, USA
| | - Lorenzo Lo Muzio
- Department of Clinic and Experimental Medicine, University of Foggia, 71122, Foggia, Italy
| |
Collapse
|
244
|
Keshinro SO, Awolola NA, Adebayo LA, Mutiu WB, Saka BA, Abdus-Salam IA. Full autopsy in a confirmed COVID-19 patient in Lagos, Nigeria - A case report. HUMAN PATHOLOGY: CASE REPORTS 2021; 24:200524. [PMID: 34026549 PMCID: PMC8125910 DOI: 10.1016/j.ehpc.2021.200524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 05/09/2021] [Accepted: 05/11/2021] [Indexed: 01/08/2023] Open
Abstract
Objectives To report the postmortem findings of a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) positive individual who died in Lagos (Nigeria) in June 2020 and to investigate the cause, pathogenesis as well as pathological changes noticed during the examination. Methods Complete postmortem examination was performed according to standard procedures in a regular autopsy suite using personal protective equipment including N95 masks, goggles and disposable gowns. The diagnosis of coronavirus disease 2019 (COVID-19) was confirmed by real-time reverse transcription polymerase chain reaction (RT-PCR) testing on postmortem nasopharyngeal swabs. Results A 47-year-old man with a medical history of well controlled hypertension and dyslipidaemia died after long hours of transportation for medical care in a hospital in Lagos. He tested positive for SARS-CoV-2 on ante- and postmortem nasopharyngeal swabs. Autopsy revealed pneumonia with diffuse alveolar damage, disseminated intravascular coagulopathy and hypovolaemic shock. Conclusions Autopsy can be performed on decedents who died from or with SARS-CoV-2 infection in a low resource environment such as ours. A standard autopsy room was used while deploying recommended infection prevention control and regular decontamination. The clinical details, autopsy findings such as diffuse alveolar damage and airway inflammation were consistent with a COVID-19 related pathology. While the decedent had ‘controlled’ co-morbidity, he succumbed to multi-organ failure occasioned by shock and disseminated intravascular coagulopathy.
Collapse
Affiliation(s)
- Samuel Olalekan Keshinro
- Nigeria Police Force, Nigeria Police Medical Services, Police Hospital, Falomo, Ikoyi, Lagos, Nigeria
| | | | | | - Wasiu Bamidele Mutiu
- Lagos State Biobank, Directorate of Epidemiology, Biosecurity & Global Health, Ministry of Health, Lagos State, Nigeria
| | - Babatunde Akeem Saka
- Lagos State Biobank, Directorate of Epidemiology, Biosecurity & Global Health, Ministry of Health, Lagos State, Nigeria
| | | |
Collapse
|
245
|
Bayyoud T, Iftner A, Iftner T, Bartz-Schmidt KU, Ziemssen F, Bösmüller H, Fend F, Rohrbach JM, Ueffing M, Schindler M, Thaler S. Severe acute respiratory Syndrome-Coronavirus-2: Can it be detected in the retina? PLoS One 2021; 16:e0251682. [PMID: 33984050 PMCID: PMC8118466 DOI: 10.1371/journal.pone.0251682] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 05/01/2021] [Indexed: 12/23/2022] Open
Abstract
Background/Objectives The systemic organ involvement of SARS-CoV-2 needs to be thoroughly investigated including the possibility of an ocular reservoir in humans. To examine retinal tissues and vitreous for histopathology and SARS-CoV-2 presence with regard to possible effects on the human retina and/ or vitreous. We performed histopathological analyses and quantitative (q)RT-PCR-testing for SARS-CoV-2 RNA on retinal tissues and vitreous of COVID-19 postmortem donors. Subjects/Methods Included in this study were 10 eyes of 5 deceased COVID-19 patients. The diagnosis of SARS-CoV-2 infection was confirmed via pharyngeal swabs and broncho-alveolar fluids. The highest level of personal protective equipment (PPE) and measures was employed during fluid-tissue procurement and preparation. Histopathological examinations and qRT-PCR-testing were carried out for all retinal tissues and vitreous fluids. Results The histopathological examinations revealed no signs of morphologically identifiable retinal inflammation or vessel occlusions based on hematoxylin and eosin stains. By qRT-PCRs, we detected no significant level of viral RNA in human retina and vitreous. Conclusions In this study, no significant level of SARS-CoV-2-RNA was detected in the human retinal and vitreous fluid samples of deceased COVID-19 patients. Histopathological examinations confirmed no morphological sign of damage to retinal vasculature or tissues. Further studies are needed to confirm or refute the results.
Collapse
Affiliation(s)
- Tarek Bayyoud
- Department of Ophthalmology, University Hospital Tübingen, Tübingen, Germany
- * E-mail:
| | - Angelika Iftner
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital Tübingen, Tübingen, Germany
| | - Thomas Iftner
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital Tübingen, Tübingen, Germany
| | | | - Focke Ziemssen
- Department of Ophthalmology, University Hospital Tübingen, Tübingen, Germany
| | - Hans Bösmüller
- Institute of Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Falko Fend
- Institute of Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | | | - Marius Ueffing
- Department of Ophthalmology, University Hospital Tübingen, Tübingen, Germany
- Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | - Michael Schindler
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital Tübingen, Tübingen, Germany
| | - Sebastian Thaler
- Department of Ophthalmology, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
246
|
Paterson RW, Benjamin LA, Mehta PR, Brown RL, Athauda D, Ashton NJ, Leckey CA, Ziff OJ, Heaney J, Heslegrave AJ, Benedet AL, Blennow K, Checkley AM, Houlihan CF, Mummery CJ, Lunn MP, Manji H, Zandi MS, Keddie S, Chou M, Vinayan Changaradil D, Solomon T, Keshavan A, Barker S, Jäger HR, Carletti F, Simister R, Werring DJ, Spyer MJ, Nastouli E, Gauthier S, Rosa-Neto P. Serum and cerebrospinal fluid biomarker profiles in acute SARS-CoV-2-associated neurological syndromes. Brain Commun 2021; 3:fcab099. [PMID: 34396099 PMCID: PMC8194666 DOI: 10.1093/braincomms/fcab099] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/12/2021] [Accepted: 05/07/2021] [Indexed: 11/24/2022] Open
Abstract
Preliminary pathological and biomarker data suggest that SARS-CoV-2 infection can damage the nervous system. To understand what, where and how damage occurs, we collected serum and CSF from patients with COVID-19 and characterized neurological syndromes involving the PNS and CNS (n = 34). We measured biomarkers of neuronal damage and neuroinflammation, and compared these with non-neurological control groups, which included patients with (n = 94) and without (n = 24) COVID-19. We detected increased concentrations of neurofilament light, a dynamic biomarker of neuronal damage, in the CSF of those with CNS inflammation (encephalitis and acute disseminated encephalomyelitis) [14 800 pg/ml (400, 32 400)], compared to those with encephalopathy [1410 pg/ml (756, 1446)], peripheral syndromes (Guillain-Barré syndrome) [740 pg/ml (507, 881)] and controls [872 pg/ml (654, 1200)]. Serum neurofilament light levels were elevated across patients hospitalized with COVID-19, irrespective of neurological manifestations. There was not the usual close correlation between CSF and serum neurofilament light, suggesting serum neurofilament light elevation in the non-neurological patients may reflect peripheral nerve damage in response to severe illness. We did not find significantly elevated levels of serum neurofilament light in community cases of COVID-19 arguing against significant neurological damage. Glial fibrillary acidic protein, a marker of astrocytic activation, was not elevated in the CSF or serum of any group, suggesting astrocytic activation is not a major mediator of neuronal damage in COVID-19.
Collapse
Affiliation(s)
- Ross W Paterson
- University College London, Queen Square Institute of Neurology, London WC1N 3BG, UK
- National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London WC1N 3BG, UK
- Darent Valley Hospital, Dartford, Kent DA2 8DA, UK
| | - Laura A Benjamin
- National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London WC1N 3BG, UK
- UCL Institute of Neurology, Stroke Research Centre, Russell Square House, London WC1B 5EH, UK
- University of Liverpool, Brain Infections Group, Liverpool, Merseyside L69 3GA, UK
- Laboratory of Molecular and Cell Biology, UCL, London WC1E 6BT, UK
| | - Puja R Mehta
- National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London WC1N 3BG, UK
| | - Rachel L Brown
- National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London WC1N 3BG, UK
- University College London Institute of Immunity and Transplantation, London NW3 2QG, UK
| | - Dilan Athauda
- National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London WC1N 3BG, UK
- Francis Crick Institute, London NW1 1AT, UK
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal 431 41, Sweden
- King’s College London, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London SE5 9RT, UK
| | - Claire A Leckey
- University College London, Queen Square Institute of Neurology, London WC1N 3BG, UK
- National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London WC1N 3BG, UK
| | | | - Judith Heaney
- National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London WC1N 3BG, UK
- Advanced Pathogens Diagnostic Unit, University College London Hospitals NHS Foundation Trust, London WC1H 8NJ, UK
| | - Amanda J Heslegrave
- University College London, Queen Square Institute of Neurology, London WC1N 3BG, UK
- National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London WC1N 3BG, UK
- UK Dementia Research Institute, London WC1E 6BT, UK
| | - Andrea L Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal 431 41, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal 431 41, Sweden
| | - Anna M Checkley
- Department of Infection and Immunity, University College London, London WC1E 6BT, UK
- Hospital for Tropical Diseases, University College Hospitals London, London WC1E 6BT, UK
| | - Catherine F Houlihan
- Department of Infection and Immunity, University College London, London WC1E 6BT, UK
- Department of Clinical Virology, University College London Hospitals NHS Foundation Trust, London WC1H 8NJ, UK
| | - Catherine J Mummery
- University College London, Queen Square Institute of Neurology, London WC1N 3BG, UK
- National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London WC1N 3BG, UK
| | - Michael P Lunn
- University College London, Queen Square Institute of Neurology, London WC1N 3BG, UK
- National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London WC1N 3BG, UK
| | - Hadi Manji
- National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London WC1N 3BG, UK
| | - Michael S Zandi
- University College London, Queen Square Institute of Neurology, London WC1N 3BG, UK
- National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London WC1N 3BG, UK
| | - Stephen Keddie
- University College London, Queen Square Institute of Neurology, London WC1N 3BG, UK
- National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London WC1N 3BG, UK
| | - Michael Chou
- University College London, Queen Square Institute of Neurology, London WC1N 3BG, UK
- National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London WC1N 3BG, UK
| | | | - Tom Solomon
- National Institute for Health Research Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK
- Walton Centre NHS Foundation Trust, Liverpool L9 7LJ, UK
| | - Ashvini Keshavan
- University College London, Queen Square Institute of Neurology, London WC1N 3BG, UK
- National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London WC1N 3BG, UK
| | - Suzanne Barker
- University College London, Queen Square Institute of Neurology, London WC1N 3BG, UK
- National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London WC1N 3BG, UK
| | - Hans Rolf Jäger
- University College London, Queen Square Institute of Neurology, London WC1N 3BG, UK
- National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London WC1N 3BG, UK
| | - Francesco Carletti
- University College London, Queen Square Institute of Neurology, London WC1N 3BG, UK
- National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London WC1N 3BG, UK
| | - Robert Simister
- University College London, Queen Square Institute of Neurology, London WC1N 3BG, UK
- National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London WC1N 3BG, UK
| | - David J Werring
- National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London WC1N 3BG, UK
- UCL Institute of Neurology, Stroke Research Centre, Russell Square House, London WC1B 5EH, UK
| | - Moira J Spyer
- Advanced Pathogens Diagnostic Unit, University College London Hospitals NHS Foundation Trust, London WC1H 8NJ, UK
| | - Eleni Nastouli
- Advanced Pathogens Diagnostic Unit, University College London Hospitals NHS Foundation Trust, London WC1H 8NJ, UK
- Department of Clinical Virology, University College London Hospitals NHS Foundation Trust, London WC1H 8NJ, UK
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Montreal H4H 1R3, Canada
- Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montreal H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal H4H 1R3, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Montreal H4H 1R3, Canada
- Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montreal H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal H4H 1R3, Canada
| |
Collapse
|
247
|
Mahmoud H, Hamody A, M Hefny H, Tohamy D, Awny I. Evaluation of Anti-SARS-CoV-2 IgA in the Conjunctival Secretions of COVID-19 Patients. Clin Ophthalmol 2021; 15:1933-1937. [PMID: 34007145 PMCID: PMC8121674 DOI: 10.2147/opth.s312942] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 04/14/2021] [Indexed: 12/25/2022] Open
Abstract
Purpose To assess the presence of anti-SARS-CoV-2 IgA in the conjunctival secretions of confirmed COVID-19 patients by nasopharyngeal swabs and correlate its presence with the severity of the disease, patient's age, sex and ocular symptoms. Methods This study included 44 positive COVID-19 patients confirmed with nasopharyngeal swabs during the period 17-28 February 2021 at Sohag Tropical Medicine Hospital. Tears and conjunctival secretions were examined for the presence of anti-SARS-CoV-2 IgA. Results While non-reactive results are strongly correlated to low titre and vice versa, severity showed significant correlation with neither IgA reactivity nor titre. Meanwhile, IgA reactivity did not show significant correlation with either age or sex. The reactivity and IgA titre are correlated with ocular symptoms. Conclusion The anti-SARS-CoV-2 IgA could be found in ocular secretions in SARS-CoV-2 patients. There is no correlation with age or sex or severity of the disease; however, they are correlated with ocular symptoms.
Collapse
Affiliation(s)
- Hany Mahmoud
- Department of Ophthalmology, Faculty of Medicine, Sohag University, Sohag, Egypt
| | - Ahmed Hamody
- Anesthesia Department, Sohag University, Sohag, Egypt
| | - Hesham M Hefny
- Clinical Pathology Department, Sohag University, Sohag, Egypt
| | - Dalia Tohamy
- Ophthalmology Department, Assiut University, Assiut, Egypt
| | - Islam Awny
- Department of Ophthalmology, Sohag University, Sohag, Egypt
| |
Collapse
|
248
|
Hooper JE, Padera RF, Dolhnikoff M, da Silva LFF, Duarte-Neto AN, Kapp ME, Lacy JM, Mauad T, Saldiva PHN, Rapkiewicz AV, Wolf DA, Felix JC, Benson P, Shanes E, Gawelek KL, Marshall DA, McDonald MM, Muller W, Priemer DS, Solomon IH, Zak T, Bhattacharjee MB, Fu L, Gilbert AR, Harper HL, Litovsky S, Lomasney J, Mount SL, Reilly S, Sekulic M, Steffensen TS, Threlkeld KJ, Zhao B, Williamson AK. A Postmortem Portrait of the Coronavirus Disease 2019 (COVID-19) Pandemic: A Large Multi-institutional Autopsy Survey Study. Arch Pathol Lab Med 2021; 145:529-535. [PMID: 33449998 DOI: 10.5858/arpa.2020-0786-sa] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2021] [Indexed: 01/04/2023]
Abstract
CONTEXT.— This study represents the largest compilation to date of clinical and postmortem data from decedents with coronavirus disease 2019 (COVID-19). It will augment previously published small series of autopsy case reports, refine clinicopathologic considerations, and improve the accuracy of future vital statistical reporting. OBJECTIVE.— To accurately reflect the preexisting diseases and pathologic conditions of decedents with SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) infection through autopsy. DESIGN.— Comprehensive data from 135 autopsy evaluations of COVID-19-positive decedents is presented, including histologic assessment. Postmortem examinations were performed by 36 pathologists at 19 medical centers or forensic institutions in the United States and Brazil. Data from each autopsy were collected through the online submission of multiple-choice and open-ended survey responses. RESULTS.— Patients dying of or with COVID-19 had an average of 8.89 pathologic conditions documented at autopsy, spanning a combination of prior chronic disease and acute conditions acquired during hospitalization. Virtually all decedents were cited as having more than 1 preexisting condition, encompassing an average of 2.88 such diseases each. Clinical conditions during terminal hospitalization were cited 395 times for the 135 autopsied decedents and predominantly encompassed acute failure of multiple organ systems and/or impaired coagulation. Myocarditis was rarely cited. CONCLUSIONS.— Cause-of-death statements in both autopsy reports and death certificates may not encompass the severity or spectrum of comorbid conditions in those dying of or with COVID-19. If supported by additional research, this finding may have implications for public health decisions and reporting moving forward through the pandemic.
Collapse
Affiliation(s)
- Jody E Hooper
- The Department of Pathology, Johns Hopkins University, Baltimore, Maryland (Hooper)
| | - Robert F Padera
- The Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (Padera Jr, Gawelek, Solomon)
| | - Marisa Dolhnikoff
- Departamento de Patologia, Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil (Dolhnikoff, Ferraz da Silva, Nunes Duarte-Neto, Mauad, Nascimento Saldiva, de Almeida Monteiro)
| | - Luiz Fernando Ferraz da Silva
- Departamento de Patologia, Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil (Dolhnikoff, Ferraz da Silva, Nunes Duarte-Neto, Mauad, Nascimento Saldiva, de Almeida Monteiro)
| | - Amaro Nunes Duarte-Neto
- Departamento de Patologia, Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil (Dolhnikoff, Ferraz da Silva, Nunes Duarte-Neto, Mauad, Nascimento Saldiva, de Almeida Monteiro)
| | - Meghan E Kapp
- The Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (Kapp)
| | - J Matthew Lacy
- Snohomish County Medical Examiner's Office, Everett, Washington (Lacy)
| | - Thais Mauad
- Departamento de Patologia, Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil (Dolhnikoff, Ferraz da Silva, Nunes Duarte-Neto, Mauad, Nascimento Saldiva, de Almeida Monteiro)
| | - Paulo Hilario Nascimento Saldiva
- Departamento de Patologia, Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil (Dolhnikoff, Ferraz da Silva, Nunes Duarte-Neto, Mauad, Nascimento Saldiva, de Almeida Monteiro)
| | - Amy V Rapkiewicz
- The Department of Pathology, NYU Long Island School of Medicine, Mineola, New York (Rapkiewicz)
| | - Dwayne A Wolf
- Harris County Institute of Forensic Sciences, Houston, Texas (Wolf)
| | - Juan C Felix
- The Department of Pathology, Medical College of Wisconsin, Milwaukee (Felix)
| | - Paul Benson
- The Department of Pathology, University of Alabama at Birmingham, Birmingham (Benson, Litovsky, Reilly)
| | - Elisheva Shanes
- The Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Shanes, Muller, Zak, Fu, Lomasney)
| | - Kara L Gawelek
- The Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (Padera Jr, Gawelek, Solomon)
| | - Desiree A Marshall
- The Department of Laboratory Medicine and Pathology, University of Washington, Seattle (Marshall)
| | - Michelle M McDonald
- The Department of Pathology & Laboratory Medicine, University of Texas Health Science Center Houston, McGovern Medical School, Houston (McDonald, Bhattacharjee, Zhao)
| | - William Muller
- The Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Shanes, Muller, Zak, Fu, Lomasney)
| | - David S Priemer
- The Department of Pathology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland (Priemer)
| | - Isaac H Solomon
- The Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (Padera Jr, Gawelek, Solomon)
| | - Taylor Zak
- The Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Shanes, Muller, Zak, Fu, Lomasney)
| | - Meenakshi B Bhattacharjee
- The Department of Pathology & Laboratory Medicine, University of Texas Health Science Center Houston, McGovern Medical School, Houston (McDonald, Bhattacharjee, Zhao)
| | - Lucy Fu
- The Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Shanes, Muller, Zak, Fu, Lomasney)
| | - Andrea R Gilbert
- The Department of Pathology and Laboratory Medicine, University of Texas Health San Antonio, Long School of Medicine, San Antonio (Gilbert)
| | - Holly L Harper
- The Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio (Harper)
| | - Silvio Litovsky
- The Department of Pathology, University of Alabama at Birmingham, Birmingham (Benson, Litovsky, Reilly)
| | - Jon Lomasney
- The Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Shanes, Muller, Zak, Fu, Lomasney)
| | - Sharon L Mount
- The Department of Pathology and Laboratory Medicine, University of Vermont Medical Center, Burlington (Mount, Threlkeld)
| | - Stephanie Reilly
- The Department of Pathology, University of Alabama at Birmingham, Birmingham (Benson, Litovsky, Reilly)
| | - Miroslav Sekulic
- The Department of Pathology and Cell Biology, Columbia University, New York, New York (Sekulic)
| | - Thora S Steffensen
- The Department of Pathology, Tampa General Hospital, Tampa, Florida (Steffensen)
| | - Kirsten J Threlkeld
- The Department of Pathology and Laboratory Medicine, University of Vermont Medical Center, Burlington (Mount, Threlkeld)
| | - Bihong Zhao
- The Department of Pathology & Laboratory Medicine, University of Texas Health Science Center Houston, McGovern Medical School, Houston (McDonald, Bhattacharjee, Zhao)
| | - Alex K Williamson
- The Department of Pathology and Laboratory Medicine, Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York (Williamson)
| |
Collapse
|
249
|
Chinnici CM, Russelli G, Bulati M, Miceli V, Gallo A, Busà R, Tinnirello R, Conaldi PG, Iannolo G. Mesenchymal stromal cell secretome in liver failure: Perspectives on COVID-19 infection treatment. World J Gastroenterol 2021; 27:1905-1919. [PMID: 34007129 PMCID: PMC8108038 DOI: 10.3748/wjg.v27.i17.1905] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/05/2021] [Accepted: 04/05/2021] [Indexed: 02/06/2023] Open
Abstract
Due to their immunomodulatory potential and release of trophic factors that promote healing, mesenchymal stromal cells (MSCs) are considered important players in tissue homeostasis and regeneration. MSCs have been widely used in clinical trials to treat multiple conditions associated with inflammation and tissue damage. Recent evidence suggests that most of the MSC therapeutic effects are derived from their secretome, including the extracellular vesicles, representing a promising approach in regenerative medicine application to treat organ failure as a result of inflammation/fibrosis. The recent outbreak of respiratory syndrome coronavirus, caused by the newly identified agent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has forced scientists worldwide to use all available instruments to fight the infection, including the inflammatory cascade caused by this pandemic disease. The use of MSCs is a valid approach to combat organ inflammation in different compartments. In addition to the lungs, which are considered the main inflammatory target for this virus, other organs are compromised by the infection. In particular, the liver is involved in the inflammatory response to SARS-CoV-2 infection, which causes organ failure, leading to death in coronavirus disease 2019 (COVID-19) patients. We herein summarize the current implications derived from the use of MSCs and their soluble derivatives in COVID-19 treatment, and emphasize the potential of MSC-based therapy in this clinical setting.
Collapse
Affiliation(s)
- Cinzia Maria Chinnici
- Department of Research, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS-ISMETT), Palermo 90127, Italy
- Department of Regenerative Medicine, Fondazione Ri.MED, Palermo 90127, Italy
| | - Giovanna Russelli
- Department of Research, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS-ISMETT), Palermo 90127, Italy
| | - Matteo Bulati
- Department of Research, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS-ISMETT), Palermo 90127, Italy
| | - Vitale Miceli
- Department of Research, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS-ISMETT), Palermo 90127, Italy
| | - Alessia Gallo
- Department of Research, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS-ISMETT), Palermo 90127, Italy
| | - Rosalia Busà
- Department of Research, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS-ISMETT), Palermo 90127, Italy
| | - Rosaria Tinnirello
- Neuroscience Unit, CNR Institute of Biomedicine and Molecular Immunology, Palermo 90146, Italy
| | - Pier Giulio Conaldi
- Department of Research, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS-ISMETT), Palermo 90127, Italy
| | - Gioacchin Iannolo
- Department of Research, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS-ISMETT), Palermo 90127, Italy
| |
Collapse
|
250
|
McMullen P, Pytel P, Snyder A, Smith H, Vickery J, Brainer J, Guzy R, Wu D, Schoettler N, Adegunsoye A, Sperling A, Hart J, Alpert L, Chang A, Gurbuxani S, Krausz T, Husain AN, Mueller J. A series of COVID-19 autopsies with clinical and pathologic comparisons to both seasonal and pandemic influenza. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2021; 7:459-470. [PMID: 33960723 PMCID: PMC8239851 DOI: 10.1002/cjp2.220] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/25/2021] [Accepted: 04/06/2021] [Indexed: 12/24/2022]
Abstract
Autopsies of patients who have died from COVID‐19 have been crucial in delineating patterns of injury associated with SARS‐CoV‐2 infection. Despite their utility, comprehensive autopsy studies are somewhat lacking relative to the global burden of disease, and very few comprehensive studies contextualize the findings to other fatal viral infections. We developed a novel autopsy protocol in order to perform postmortem examinations on victims of COVID‐19 and herein describe detailed clinical information, gross findings, and histologic features observed in the first 16 complete COVID‐19 autopsies. We also critically evaluated the role of ancillary studies used to establish a diagnosis of COVID‐19 at autopsy, including immunohistochemistry (IHC), in situ hybridization (ISH), and electron microscopy (EM). IHC and ISH targeting SARS‐CoV‐2 were comparable in terms of the location and number of infected cells in lung tissue; however, nonspecific staining of bacteria was seen occasionally with IHC. EM was unrevealing in blindly sampled tissues. We then compared the clinical and histologic features present in this series to six archival cases of fatal seasonal influenza and six archival cases of pandemic influenza from the fourth wave of the ‘Spanish Flu’ in the winter of 1920. In addition to routine histology, the inflammatory infiltrates in the lungs of COVID‐19 and seasonal influenza victims were compared using quantitative IHC. Our results demonstrate that the clinical and histologic features of COVID‐19 are similar to those seen in fatal cases of influenza, and the two diseases tend to overlap histologically. There was no significant difference in the composition of the inflammatory infiltrate in COVID‐19 and influenza at sites of acute lung injury at the time of autopsy. Our study underscores the relatively nonspecific clinical features and pathologic changes shared between severe cases of COVID‐19 and influenza, while also providing important caveats to ancillary methods of viral detection.
Collapse
Affiliation(s)
- Phillip McMullen
- Department of Pathology, University of Chicago Medical Center, Chicago, IL, USA
| | - Peter Pytel
- Department of Pathology, University of Chicago Medical Center, Chicago, IL, USA
| | - Alexis Snyder
- Department of Pathology, University of Chicago Medical Center, Chicago, IL, USA
| | - Heather Smith
- Department of Pathology, University of Chicago Medical Center, Chicago, IL, USA
| | - Jasmine Vickery
- Department of Pathology, University of Chicago Medical Center, Chicago, IL, USA
| | - James Brainer
- Department of Pathology, University of Chicago Medical Center, Chicago, IL, USA
| | - Robert Guzy
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, University of Chicago Medical Center, Chicago, IL, USA
| | - David Wu
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, University of Chicago Medical Center, Chicago, IL, USA
| | - Nathan Schoettler
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, University of Chicago Medical Center, Chicago, IL, USA
| | - Ayodeji Adegunsoye
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, University of Chicago Medical Center, Chicago, IL, USA
| | - Anne Sperling
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, University of Chicago Medical Center, Chicago, IL, USA
| | - John Hart
- Department of Pathology, University of Chicago Medical Center, Chicago, IL, USA
| | - Lindsay Alpert
- Department of Pathology, University of Chicago Medical Center, Chicago, IL, USA
| | - Anthony Chang
- Department of Pathology, University of Chicago Medical Center, Chicago, IL, USA
| | - Sandeep Gurbuxani
- Department of Pathology, University of Chicago Medical Center, Chicago, IL, USA
| | - Thomas Krausz
- Department of Pathology, University of Chicago Medical Center, Chicago, IL, USA
| | - Aliya N Husain
- Department of Pathology, University of Chicago Medical Center, Chicago, IL, USA
| | - Jeffrey Mueller
- Department of Pathology, University of Chicago Medical Center, Chicago, IL, USA
| |
Collapse
|